Abstract
Fibrosis is a pathologic condition resulting from aberrant wound healing responses that lead to excessive accumulation of extracellular matrix components, distortion of organ architecture, and loss of organ function. Fibrotic disease can affect every organ system; moreover, fibrosis is an important microenvironmental component of many cancers, including pancreatic, cervical, and hepatocellular cancers. Fibrosis is also an independent risk factor for cancer. Taken together, organ fibrosis contributes to up to 45% of all deaths worldwide. There are no approved therapies that halt or reverse fibrotic disease, highlighting the great need for novel therapeutic targets. At the heart of almost all fibrotic disease is the TGF-β-mediated differentiation of fibroblasts into myofibroblasts, the primary cell type responsible for the production of collagen and other matrix proteins and distortion of tissue architecture. Recent advances, particularly in the field of lung fibrosis have highlighted the role that metabolic reprogramming plays in the pathogenic phenotype of myofibroblasts, particularly the induction of de novo amino acid synthesis pathways that are required to support collagen matrix production by these cells. In this review, we will discuss the metabolic changes associated with myofibroblast differentiation, focusing on the de novo production of glycine and proline, two amino acids which compose over half of the primary structure of collagen protein. We will also discuss the important role that synthesis of these amino acids plays in regulating cellular redox balance and epigenetic state.
Keywords: Fibrosis, metabolism, amino acid, glutamine, proline and glycine
Fibrosis
Fibrosis is defined by the excessive deposition of extracellular matrix that replaces normal healthy tissue with scar tissue. While matrix deposition is required for normal wound healing responses, severe or repetitive injury may lead to the continued accumulation of matrix proteins, which ultimately impair tissue function. Fibrosis can occur in any organ system and is associated with a broad range of chronic diseases, including heart disease, interstitial lung disease, scleroderma, hepatitis, inflammatory bowel disease, and diabetes. Organ fibrosis is estimated to be associated with as many as 45% of all deaths (Rockey et al. 2015; Henderson et al. 2020; Distler et al. 2019; Jun and Lau 2018). Although genetic susceptibility loci have been identified for some fibrotic diseases and exposures such as alcohol, cigarette smoke, asbestos, silica, and viral infection are associated with increased risk, the etiology of most fibrotic diseases remains unknown. Furthermore, there are no effective therapies for halting or reversing fibrotic progression in any tissue. Thus, there is tremendous need to identify novel targets for the treatment of organ fibrosis.
Fibrosis and Cancer
Similar to fibrosis, cancer has also been described as “a wound that does not heal” (Dvorak 1986). Solid tumors are closely associated non-transformed vasculature, immune cells, and fibroblasts that constitute the stromal compartment. The aberrant matrix environment of the tumor stroma enhances cellular proliferation, promotes angiogenesis, inhibits anti-tumor immune responses, and promotes metastasis and resistance to therapeutics (Piersma et al. 2020; Cox and Erler 2014; Valkenburg et al. 2018). The importance of a wound-like environment has long been appreciated to promote cancer growth. For instance, Rous sarcoma virus injection into chickens leads to tumor formation only at the injection site (a wound) despite viremia. Wounding at distal sites resulted in tumor formation at these sites (Dolberg et al. 1985). Similarly, it has been shown that fibrotic lung disease is an independent risk factor for lung cancer and that stromal gene signatures indicate prognosis of pancreatic and colorectal cancer (Karampitsakos et al. 2017; Ballester et al. 2019; Moffitt et al. 2015; Calon et al. 2015). The increased understanding of the role of the stroma in promoting cancer progression has led to therapeutics that target the stromal compartment (Valkenburg et al. 2018).
Myofibroblasts
While fibrosis is a complex disease involving multiple cell types, myofibroblasts represent the cellular site of pathologic matrix production that leads to distortion of tissue architecture and loss of function (Darby et al. 2016). Myofibroblasts are also present in the tumor stroma and are responsible for the aberrant matrix environment of tumors (Piersma et al. 2020; Liu et al. 2019). Myofibroblasts are contractile cells with features of both fibroblasts and smooth muscle cells that promote wound closure during normal wound healing. These cells are eliminated by apoptosis during normal wound closure; however, they persist during hypertrophic scar development and in fibrotic disease (Hinz and Lagares 2020). Myofibroblasts are not present in healthy tissues and their cell(s) of origin remain a topic of debate; however, tissue resident fibroblasts have been shown to give rise to myofibroblasts in all injured organs (Pakshir et al. 2020). This transformation is largely governed by signaling downstream of Transforming Growth Factor-β (TGF-β), which is highly expressed in fibrotic tissues and is sufficient to induce myofibroblastic differentiation and collagen matrix production in vitro, and to induce fibrosis in vivo (Sporn et al. 1983; Ignotz and Massague 1986; Roberts et al. 1986; Sime et al. 1997).
Recent single cell RNA sequencing and lineage tracing experiments have highlighted heterogeneity and plasticity in fibroblast populations in multiple fibrotic tissues as well as in cancer stroma (Shaw and Rognoni 2020; Biffi and Tuveson 2021). This complexity may complicate the design of myofibroblast-specific therapies; thus, targeting of specific pathologic functions such as matrix production may hold promise for developing new therapies for fibrotic disease. Aberrant production of matrix proteins by myofibroblasts results in an altered mechanical environment that is sufficient to promote further myofibroblast activation in a feed forward cycle (Tschumperlin et al. 2018). Thus, the myofibroblast and its production of matrix proteins is an important target of fibrosis therapy.
Structure of Collagen Matrix Proteins
Collagen is the most abundant component of extracellular matrix and comprises one third of all protein in humans (Shoulders and Raines 2009; Ricard-Blum 2011). The structural stability of collagen is derived from its quaternary structure consisting of three parallel polypeptide strands forming triple helices known as tropocollagens. This helical structure of collagen depends on triple-helical domains consisting of a repeating G-X-Y primary structure where G represents glycine at every third amino acid residue and X and Y can represent any amino acid but are most commonly proline at the X position and hydroxyproline at the Y position. Together, glycine, proline, and hydroxyproline (which is produced through posttranslational hydroxylation of collagen proline residues), constitute over half of all amino acids in the primary structure of collagen protein (Bradley et al. 1974) (Figure 1). The repeating glycine residues allow for interstrand hydrogen bonds while post-translational proline hydroxylation at the Y position provides increased thermostability through stereoelectronic effects. All collagens contain at least one triple-helical domain. Fibril-forming collagens such as collagen I, III, and V which are the primary collagens produced by myofibroblasts, contain one major triple-helical domain. Other collagens are characterized by the presence of several smaller triple-helical domains interrupted by non-helical domains which are associated with molecular recognition and give these collagens plasticity and flexibility. Thus, collagens are able to impart both structural and signaling cues that regulate both normal and pathogenic cellular behavior (Karsdal et al. 2017; Xu et al. 2019).
Figure 1. Percentages of amino acids in human collagen 1.
Amino acid composition of (a) collagen proteins and (2) non-collagen proteins in human lung (Bradley et al. 1974). While glycine and proline/hydroxyproline make up 33.37% and 20.51% of total amino acid residues in collagens, their percentage is much lower in other proteins.
Metabolic Reprogramming during Myofibroblast Differentiation
Glycolysis and de novo glycine synthesis
Myofibroblastic differentiation is associated with transcriptomic and morphologic changes as cells develop an extensive stress fiber network, expand their rough endoplasmic reticulum, and produce extracellular matrix proteins. These cells also alter the way in which they use metabolites in order to support this contraction and matrix production. Upon treatment with TGF-β, normal human lung fibroblasts exhibit increased rates of glycolysis and mitochondrial oxygen consumption (Nigdelioglu et al. 2016; Xie et al. 2015; Bernard et al. 2015; Hamanaka et al. 2019). This increase in metabolic activity supports not only the energetic needs of myofibroblasts but also plays a crucial role in producing the molecular building blocks required by myofibroblasts for the production of matrix proteins.
Inhibition of glycolysis with 2-deoxyglucose prevents TGF-β-induced collagen protein production by lung fibroblasts independent of any effect on collagen mRNA expression, which remains induced by TGF-β (Nigdelioglu et al. 2016). While glycolytic metabolism likely serves many roles in myofibroblasts, including production of ATP, one crucial role for glucose in these cells is as a substrate for de novo glycine synthesis. TGF-β induces the expression of all of the enzymes required for the conversion of the glycolytic intermediate 3-phosphoglycerate to the amino acid glycine using an amino group donated from glutamate. These enzymes include phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase 1 (PSAT1), phosphoserine phosphatase (PSPH), and serine hydroxymethyltransferase 2 (SHMT2) (Nigdelioglu et al. 2016) (Figure 2). TGF-β-induced collagen synthesis occurs independent of extracellular glycine and knockdown of these enzymes is sufficient to inhibit collagen production even in the presence of extracellular glycine, suggesting that de novo-synthesized glycine is preferentially used by myofibroblasts for collagen protein production (Nigdelioglu et al. 2016; Hamanaka et al. 2019). Supporting this notion, metabolic tracing experiments using 13C-glucose in the presence of extracellular 12C-glycine, demonstrated that TGF-β stimulates the production of glycine from glucose with glucose-derived glycine comprising up to 80% of cellular glycine pools (Schworer et al. 2020). Furthermore, carbon derived from glucose is incorporated into collagen protein (Nigdelioglu et al. 2016; Schworer et al. 2020; Selvarajah et al. 2019; Tsurufuji and Mori 1965).
Figure 2. Contributions of glycolytic intermediates and glutamine to collagen protein synthesis.
Both glucose and glutamine contribute to the synthesis of two most abundant amino acids (glycine and proline) in collagen protein. Glycine can be synthesized from the glycolytic intermediate, 3-phosphoglycerate (3P glycerate) by the enzymes of serine glycine synthesis pathway including phosphoglycerate dehydrogenase (PHGDH), phosphoserine aminotransferase 1 (PSAT1), phosphoserine phosphatase (PSPH), and serine hydroxymethyltransferase 2 (SHMT2). Glutamine is converted to glutamate by glutaminase (GLS) in glutaminolysis. Glutamate is not only a precursor for proline but also contributes to the generation of glycine by donating nitrogen through the PSAT1 transamination reaction.
Glutamine Metabolism
Glutamine is the most abundant amino acid found in human plasma and a key substrate used by cells to replenish the TCA cycle to support mitochondrial metabolism (Altman et al. 2016). Glutamine is metabolized in a pathway termed glutaminolysis in which glutamine is first converted to glutamate by the enzyme glutaminase (GLS). Glutamate is then metabolized to the TCA cycle intermediate α-ketoglutarate by either glutamate dehydrogenase (GLUD) or by aminotransferase enzymes, including PSAT1 (Figure 2). The glutaminolytic pathway has been studied extensively in the context of cancer and GLS inhibitors are in clinical trials for colorectal and non-small cell lung cancers.
Multiple reports have demonstrated that GLS expression is induced in lung fibroblasts after TGF-β treatment (Bernard et al. 2018; Ge et al. 2018; Hamanaka et al. 2019; Choudhury et al. 2020). Inhibition of glutaminolysis by either culturing cells in the absence of extracellular glutamine or by inhibition of GLS prevents TGF-β-induced myofibroblast differentiation (Bernard et al. 2018; Ge et al. 2018; Hamanaka et al. 2019; Choudhury et al. 2020). Interestingly, while glutaminolysis is an important metabolic pathway that feeds the TCA cycle, it appears that this anaplerotic role of glutamine is dispensable for myofibroblast differentiation. TGF-β increases the entry of carbons from extracellular glutamine into the TCA cycle (Schworer et al. 2020); however, knockdown of α-ketoglutarate dehydrogenase, the TCA cycle enzyme that converts α-ketoglutarate to succinyl-CoA, had no effect on TGF-β-induced expression of collagen or α-SMA protein (Hamanaka et al. 2019). Furthermore, while TGF-β increases mitochondrial oxygen consumption in lung fibroblasts, this increase was shown to be independent of the presence of glutamine in the media or on the activity of GLS (Hamanaka et al. 2019). Thus, mitochondrial glutamine catabolism, although induced by TGF-β, is not required for myofibroblast differentiation.
Glutamine-Dependent Glycine and Proline Biosynthesis
In addition to its role as a mitochondrial substrate, glutamine-derived glutamate can be used in anabolic pathways, supporting glutathione synthesis, cellular import of cystine, and as a precursor for amino acid biosynthesis, including the de novo synthesis of glycine and proline. As mentioned above, glutamate is required for the activity of PSAT1 which transfers the amino group of glutamate to 3-phosphohydroxypyruvate (the product of PHGDH), producing 3-phosphoserine, a precursor for glycine (Figure 2). Metabolic labeling experiments using glutamine labeled on its alpha nitrogen demonstrated that TGF-β increases the incorporation of glutamine-derived nitrogen into glycine (Schworer et al. 2020). Twenty-five percent of cellular glycine was labeled after culture in 15N-glutamine while no label was found in glycine after culture in 13C-glutamine. Thus, glycine synthesis in lung fibroblasts utilizes carbon derived from glucose and nitrogen derived from glutamate. The role of glutamate sources other than extracellular glutamine has not been investigated; however, knockdown of PSAT1 in the presence of extracellular glycine, inhibited TGF-β-induced collagen protein production in lung fibroblasts, consistent with the importance of this pathway for collagen synthesis (Hamanaka et al. 2019).
Glutamine-derived glutamate is also an important precursor for de novo synthesis of proline. TGF-β induces the expression of all enzymes required to convert glutamate to proline, including pyrroline-5-carboxylate synthase (P5CS), which converts glutamate to pyrroline-5-carboxylate (P5C), as well as P5C reductases (PYCR1, PYCR2, PYCRL/PYCR3), which convert P5C to proline (Hamanaka et al. 2019; Schworer et al. 2020) (Figure 3). Total cellular levels of P5C and proline are increased in cells after TGF-β and carbon labeling shows that TGF-β increases the percentage of cellular proline derived from extracellular glutamine (Hamanaka et al. 2019; Schworer et al. 2020). Glutamine-derived proline is incorporated into collagen protein, and knockdown of P5CS prevents TGF-β-induced collagen protein production (Hamanaka et al. 2019; Schworer et al. 2020). Interestingly, flux analysis showed that TGF-β increases the incorporation of glutamine-derived carbon into proline by 7-fold and into α-ketoglutarate by only 2-fold, indicating that proline is a major cellular destination for glutamine metabolism in myofibroblasts (Schworer et al. 2020).
Figure 3. Metabolic pathways for the biosynthesis of proline.
Proline is synthesized from pyrroline-5-carboxylate (P5C) by P5C reductases (PYCR1, PYCR2, PYCRL/PYCR3). 5PC is generated from either glutamine or arginine. Glutamine is converted to glutamate and then 5PC by glutaminase (GLS) and pyrroline-5-carboxylate synthase (P5CS), respectively. Arginine is converted to ornithine and then 5PC by arginase 1 and 2 (ARG1/2) and ornithine aminotransferase (OAT), respectively.
While metabolic labeling experiments in fibroblasts suggest TGF-β treatment leads to 70% of cellular proline being derived from extracellular glutamine, these experiments were done in the absence of extracellular proline (Schworer et al. 2020). It is unknown how TGF-β affects the use of extracellular proline; however, the finding that endogenous proline synthesis supports collagen protein production is consistent with older reports using radioactive tracers to study the effect of amino acids on cellular proline pools and incorporation of extracellular proline into collagen. When rat granulomas were cultured in the presence of 3H-proline and 14C-glutamate, incorporation of both 3H and 14C was noted in collagen hydroxyproline (Aalto et al. 1973). 14C-proline is taken up by cultured chicken embryo tibiae and incorporated into proteins. Addition of glutamine to the medium reduced the radioactivity of proline and hydroxyproline hydrolyzed from these tibiae (Blumenkrantz and Asboe-Hansen 1973). Similarly, when human fibroblasts were cultured in 14C-proline, increasing the medium glutamine concentration from 2 mM to 8 mM reduced the radioactivity of intracellular free proline as well as that of collagens without affecting the total concentration of collagen (Bellon et al. 1985). It has also been noted that the specific activity of intracellular free proline never equilibrates with extracellular labeled proline and that the specific activity of prolyl-tRNAs and collagen hydroxyproline is lower than that of intracellular free proline (Hildebran et al. 1981; Bellon et al. 1987). This lack of correlation with extracellular proline was abolished in CHO-K1 cells which are proline auxotrophs (Hildebran et al. 1981). Glutamine was shown to compete with extracellular proline for incorporation into prolyl-tRNA and collagen independent of free intracellular proline concentration (Bellon et al. 1987). Similarly, tissue slices from cirrhotic rat livers contained 4 times more 14C-hydroxyproline in collagen protein when cultured in 14C-glutamate than when cultured in 14C-proline (Rojkind and Diaz de Leon 1970). Thus, glutamine and glutamate are important precursors for the proline used for collagen production, even in the presence of extracellular proline.
Ornithine-Dependent Proline Biosynthesis
The non-proteinogenic amino acid ornithine is also an important precursor for proline biosynthesis. Ornithine is a product of the breakdown of arginine by the urea cycle enzyme arginase (Figure 3). To continue the urea cycle, ornithine transcarbamylase (OTC) catalyzes the reaction of carbamoyl phosphate with ornithine, producing citrulline. Alternatively, ornithine can be decarboxylated by ornithine decarboxylase (ODC) producing putrescine, the committed step in polyamine synthesis. Lastly, ornithine aminotransferase (OAT) can transfer the side chain amino group of ornithine to α-ketoglutarate, producing glutamate and P5C (Morris 2002) (Figure 3).
While relatively less is known about the contribution of ornithine-derived proline to collagen synthesis than that from glutamate-derived proline, this pathway may also contribute significantly to de novo synthesis of proline and collagen. When rat xiphoid cartilage was cultured in 3H-proline and 14C-ornithine, 20% of protein-incorporated proline was found to be derived from ornithine (Smith and Phang 1978). Furthermore, the rate of 14C-labeled proline incorporation was not inhibited by elevation of extracellular proline concentration. Labeling of lung fibroblasts with 14C-arginine or 14C-glutamate showed that although glutamate-derived proline accounted for 2–5 times as much cellular proline as that from arginine, 14C-arginine led to significant labeling of cellular free proline, protein-incorporated proline, and hydroxyproline (Shen and Strecker 1975). Moreover, treatment of either NIH-3T3 cells or rat vascular smooth muscle cells with TGF-β resulted in the upregulation of arginase expression, the inhibition of which reduced collagen protein synthesis at the posttranscriptional level (Durante et al. 2001; Kitowska et al. 2008).
In Vivo Evidence of a role for De Novo Amino Acid Biosynthesis for Collagen Production
The work outlined above using cultured cells and tissues has suggested that de novo synthesis of glycine and proline plays a major role in the production of the amino acids incorporated into collagen, independent of extracellular levels of glycine and proline, and suggesting a compartmentalization of these endogenously-synthesized amino acids for protein production. Further work has suggested that collagen-producing cells depend on these same pathways in vivo.
Enzymes of the de novo glycine synthesis pathway, including PHGDH, PSAT1, and SHMT2, have been shown to be highly expressed in fibrotic lung tissue from humans and mice (Nigdelioglu et al. 2016; Hamanaka et al. 2018; Hamanaka et al. 2019). Lung tissue from IPF patients was shown to have significantly elevated levels of glycolytic intermediates and glycine than control donor tissues (Kang et al. 2016). Further, inhibition of de novo synthesis of glycine from glucose with the PHGDH inhibitor NCT503 reduced bleomycin-induced lung fibrosis in mice (Hamanaka et al. 2018).
Tissue levels of proline have also been found to be elevated in IPF lung tissue (Kang et al. 2016). Similar findings were found in liver tissue from cirrhosis patients (Kershenobich et al. 1970). This was independent of any effect of cirrhosis on plasma proline levels, suggesting local production of proline increases the liver free proline levels. Infusion of radiolabeled leucine, tyrosine, or proline into rabbits led to accumulation of these free amino acids in the skin. While leucine and tyrosine were readily incorporated into collagen proteins, tissue free proline proved to be a poor precursor for collagen. Calculations suggested that less than 5% of the proline used for collagen production in the skin was at equilibrium with the free proline in the blood (Robins 1979). Thus, de novo proline synthesis contributes to collagen protein production in vivo as it does in culture.
GLS, P5CS, and OAT expression have all been shown to be elevated in the lung tissue of IPF patients (Ge et al. 2018; Hamanaka et al. 2019; Schworer et al. 2020; Kang et al. 2016; Nojima et al. 2020). The expression of cytoplasmic arginase 1 and mitochondrial arginase 2 is induced in the lungs of mice after bleomycin instillation; however, the evidence of arginase expression in IPF patients is less robust (Endo et al. 2003; Kitowska et al. 2008; Nojima et al. 2020). Expression levels of ALDH18A1 (the gene encoding P5CS) mRNA in IPF lung tissue correlates negatively with pulmonary function measurements including forced vital capacity (FVC) and diffusing capacity for carbon monoxide (DLCO) (Schworer et al. 2020). OAT expression also correlates negatively with FVC (Kang et al. 2016). While inhibition of GLS either pharmacologically, or by conditionally deleting its expression in fibroblasts, reduces bleomycin-induced lung fibrosis in mice (Cui et al. 2019; Choudhury et al. 2020), it remains to be determined whether this is due to the effect of GLS inhibition on proline biosynthesis.
Why do Myofibroblasts Rely on De Novo Amino Acid Biosynthesis?
Redox Balance
The question remains as to why collagen-producing cells such as myofibroblasts engage in de novo amino acid biosynthesis for production of collagen protein even in the presence of extracellular glycine and proline. While no one answer is likely to completely explain this phenomenon, metabolite flux through these metabolic pathways may provide benefits to the cells other than production of the amino acids themselves. One major benefit that glycine and proline synthesis is maintenance of cellular redox balance. Myofibroblasts are characterized by elevated levels of cellular reactive oxygen species (ROS) that are produced both by mitochondria and NADPH oxidases (Jain et al. 2013; Hecker et al. 2009). These ROS act as signaling molecules which are required for the transcriptional events downstream of TGF-β; however, cellular ROS production must be carefully balanced by cellular antioxidants and other compensatory mechanisms so that excessive ROS accumulation does not occur (Hamanaka and Chandel 2010). The de novo synthesis of both glycine and proline is intimately linked with cellular redox status and this link may be beneficial for cells, particularly under stress conditions.
De novo glycine synthesis promotes cellular antioxidant function through reduction of NADP+ to NADPH in the downstream one-carbon pathway. NADPH provides reducing equivalents for cellular glutathione and thioredoxin antioxidant systems. Cells lacking SHMT2 are characterized by increased levels of oxidized glutathione, oxidative stress, and increased sensitivity to oxidative stressors (Ye et al. 2014). De novo production of proline is also associated with protection from oxidative stress, a process extensively studied in plants (Szabados and Savoure 2010). While it has been suggested that proline can act as a direct ROS scavenger (Krishnan et al. 2008; Kaul et al. 2008), another mechanism by which proline synthesis can protect against oxidative stress is by acting as a mitochondrial vent to prevent ROS production in the first place. As mentioned above, myofibroblast differentiation is associated with increased mitochondrial oxygen consumption and elevations of TCA cycle intermediates including citrate, α-ketoglutarate, succinate, fumarate, and malate (Xie et al. 2015; Bernard et al. 2015; Schworer et al. 2020; Hamanaka et al. 2019; Selvarajah et al. 2019; O’Leary et al. 2020). Increased oxidation of substrates in mitochondria can lead to an NADH/NAD+ ratio that exceeds ATP demand and the capacity of complex IV to transfer the electrons to molecular oxygen. This can lead to premature leaking of single electrons to oxygen, producing superoxide. De novo proline production removes both carbon and reducing equivalents from mitochondria. Instead of oxidizing glutamine-derived glutamate (via α-ketoglutarate), which transfers electrons to NAD+ in the TCA cycle, conversion of glutamate to P5C prevents glutamine-derived carbon from entering the TCA cycle. Further, conversion of P5C to proline by mitochondria-localized PYCR1 and PYCR2 oxidizes NADH, reducing the number of electrons transferred to complex I of the electron transport chain. Indeed, when P5CS is knocked out in fibroblasts using CRISPR/Cas9, cells exhibit increased levels of TCA cycle intermediates and cellular ROS content after stimulation with TGF-β (Schworer et al. 2020).
Epigenetic Regulation
While the contribution of amino acid biosynthesis to cellular redox homeostasis is likely a major reason for this dependence, other mechanisms, including epigenetic regulation are also likely to be involved. It is increasingly understood that cellular epigenetic state is intimately linked with metabolic state. This is due to the fact that metabolite cofactors including acetyl-CoA and S-adenosylmethionine (SAM) are required for the activity of chromatin-modifying enzymes such as acetylases and methyltransferases, respectively (Su et al. 2016; Ye et al. 2014). Removal of acetyl and methyl marks is also regulated by metabolites as the sirtuin family of deacetylases are sensitive to NAD+ levels and TET DNA demethylases and Jumanji C (JMJC) histone demethylases require α-ketoglutarate, oxygen, and vitamin C as cofactors (Houtkooper et al. 2012; Islam et al. 2018).
Very little is known about epigenetic regulation during myofibroblastic differentiation or during the pathogenesis of fibrotic disease; however, TGF-β-induced myofibroblastic differentiation as well as fibrotic lung tissue from mice and IPF patients are associated with reductions in histone acetylation and changes in DNA methylation patterns (Jones et al. 2019; Li et al. 2017; Rabinovich et al. 2012; Sanders et al. 2012). De novo amino acid production may be an important regulator of these epigenetic changes. One-carbon metabolism downstream of de novo glycine synthesis supports SAM production and is associated with changes in DNA methylation (Maddocks et al. 2016; Kottakis et al. 2016; Reina-Campos et al. 2019). Furthermore, α-ketoglutarate produced by PSAT1 has been shown to be essential for maintenance of DNA methylation patterns and stemness in mouse embryonic stem cells (mESCs) (Hwang et al. 2016)
Proline metabolism and collagen synthesis may also regulate epigenetic state in myofibroblasts. Treatment of mESCs with proline leads to the development of a mesenchymal phenotype associated with genome-wide increases in histone and DNA methylation (Comes et al. 2013; D’Aniello et al. 2017). This transition is reversed by vitamin C and interestingly does not occur in mESCs with the genes for collagen prolyl hydroxylases knocked out (D’Aniello et al. 2019). Collagen prolyl hydroxylases are α-ketoglutarate-dependent dioxygenases, the same family of enzymes that includes TET DNA demethylases and JMJC histone demethylases (Islam et al. 2018). Thus, collagen synthesis and proline hydroxylation appear to be in competition with chromatin-modifying enzymes for the vitamin C and possibly α-ketoglutarate that are required for their activity. It remains to be determined whether similar regulation occurs through de novo proline production in myofibroblasts; however proline abundance is regulated by P5CS induction as knockout of ALDH18A1 reduces cellular proline levels while overexpression increases them (Schworer et al. 2020).
Caveats and Future Directions
The totality of the evidence suggests that de novo synthesis of glycine and proline is crucial for TGF-β-induced collagen protein production and for organ fibrosis. Genetic approaches have identified metabolic pathways that are regulated by TGF-β in fibroblasts. Metabolic tracing experiments demonstrate incorporation of de novo synthesized glycine and proline into collagen protein, and inhibition of the relevant enzymes prevents TGF-β-induced collagen protein production in vitro and attenuate fibrosis in vivo. It is important to note that the in vitro experiments conducted have been performed in defined environments with defined metabolic conditions that do not accurately reflect in vivo conditions in humans during either health or disease. While much can be learned from monocultures, the complexities of cell-cell interactions as well as the microenvironmental nutrient availability are not representative of the in vivo environment. Culture conditions have been shown to play a major role in regulating the way cells use metabolites. Metabolic pathways that are important in cultured cells are not necessarily important in vivo (Muir et al. 2018; Muir and Vander Heiden 2018). Thus, further study of the nutrient microenvironment found in fibrotic tissues in vivo will be crucial to fully understand the metabolism of myofibroblasts.
Mutations in ALDH18A1 (the gene encoding P5CS) and PYCR1 are associated with the connective tissue disorder cutis laxa, consistent with a role of proline synthesis in collagen and elastin production. PHGDH and PSAT1 mutations are also associated with skin defects, among other clinical manifestations. Whether these defects are the result of an aberrant matrix environment is unknown (Rumping et al. 2020; El-Hattab 2016). While pharmacologic inhibition of either PHGDH or GLS prevents lung fibrosis after bleomycin instillation in vivo, fibroblast-specific knockout of PHGDH or P5CS will be needed to conclusively demonstrate the role of these de novo amino acid synthesis in fibroblasts for the fibrotic response in vivo. Studies of in vivo metabolism of extracellular metabolites after infusion of stable isotope-labeled metabolites will also strengthen the evidence for de novo amino acid synthesis in fibrosis. This technique has been used in tumor-bearing mice as well as in cancer patients to determine how tumors use metabolites in situ (Davidson et al. 2016; Maher et al. 2012; Faubert et al. 2017; Hensley et al. 2016).
Diet has been shown to play a role in microenvironmental metabolite concentrations in cancer models and dietary provision of glycine, proline, and hydroxyproline (which can be converted to glycine in the kidney) has been shown to be important for collagen production and growth of livestock (Sullivan et al. 2019; Li and Wu 2018; Wu 2020; Wu et al. 2019). Elimination of serine and glycine from diets of mice lowers plasma serine and glycine levels to an extent that inhibits cancer cell growth; however, serine and glycine deprivation had no effect on the ability of bleomycin instillation to induce fibrosis in mouse lungs (Maddocks et al. 2013; Maddocks et al. 2017; Hamanaka et al. 2018). Thus, the effect of dietary nutrients on plasma amino acid levels, tissue microenvironment, and fibrotic responses in vivo warrants further investigation. It will also be of interest to examine the relative contributions of dietary versus de novo amino acid synthesis for both homeostatic collagen synthesis and fibrotic collagen synthesis.
Finally, while studies have focused on anabolic pathways, which convert glucose and glutamine into amino acids required for collagen synthesis, the role of catabolic pathways should also be investigated. The branched chain amino acids leucine, isoleucine, and valine can be deaminated by branched chain aminotransferases, producing glutamate which may be used to produce glycine or proline as discussed above (Selwan and Edinger 2017). Another source of amino acids which may be important for collagen production may be breakdown products of extracellular and intracellular proteins. Macropinocytosis has been shown to contribute significantly to the amino acid pools of Kras-driven pancreatic cancer cells (Kamphorst et al. 2015; Davidson et al. 2017). It has also been suggested that recycling of proline from collagen breakdown products by the enzyme prolidase may play a role in supporting collagen synthesis during fibrosis (Karna et al. 2020). Whether this effect is myofibroblast-intrinsic remains to be demonstrated.
Conclusions
Current evidence strongly suggests that de novo amino acid biosynthesis is a major regulator of matrix production by myofibroblasts. These pathways represent novel targets for the treatment of fibrotic diseases and, as inhibitors of these pathways are currently in development for cancer therapy, these drugs may be repurposed for fibrosis therapy. It will be of interest to determine how targeting these pathways affects not only cancer cells, but also the interplay between these cells and the stroma. Cancer-associated fibroblasts participate in a metabolic interplay with tumor cells, producing metabolites such as lactate, alanine, and asparagine that are taken up and metabolized by tumor cells (Li and Simon 2020). Catabolism of collagen peptides produced by stromal cells may also be an important source of nutrients for certain cancer cells (Olivares et al. 2017). Thus, inhibition of collagen synthesis by cancer-associated fibroblasts may deprive cancer cells of the mechanical and microenvironmental niche required for tumorigenesis as well as essential fuel required for cell growth.
While the physical building blocks for collagen protein are a major output of these amino acid synthesis pathways, the requirement for their de novo production even in the presence of extracellular amino acids suggests the non-proteinogenic functions of these pathways also plays a major role in myofibroblasts. Prokaryotes possess the ability to produce all 20 amino acids from carbon and nitrogen sources. Mammals have lost the ability to synthesize 9 of these molecules, making these dietary necessities. The retention of the pathways for the synthesis of the remaining 11 nonessential amino acids suggests that the process of producing these amino acids provides a cellular benefit that is of equal importance to the cell as the actual end product. It has become clear that these amino acid biosynthesis pathways are important regulators of cellular redox homeostasis and epigenetic state. These meta-roles of amino acid synthesis pathways may play important roles in gene expression and apoptosis resistance in myofibroblasts. Continued study of these pathways will thus provide a greater understanding of the biology of fibrosis and has potential to lead to novel drugs to target fibrotic disease.
Acknowledgments
Funding:
NIH R01HL151680 (RBH), and NIH P01HL144454, R01ES010524, U01ES026718, and P30ES027792 and Department of Defense W81XWH-16-1-0711(GMM).
Footnotes
Conflicts of interest/Competing interests: The authors declare no conflict of interest.
Declarations
Availability of data and material: Not applicable
Code availability Not applicable
Ethics approval: Not applicable
Consent to participate: Not applicable
Consent for publication: Not applicable
Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.
References
- Aalto M, Lampiaho K, Pikkarainen J, Kulonen E (1973) Amino acid metabolism of experimental granulation tissue in vitro. Biochem J 132 (4):663–671. doi: 10.1042/bj1320663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Altman BJ, Stine ZE, Dang CV (2016) From Krebs to clinic: glutamine metabolism to cancer therapy. Nat Rev Cancer 16 (10):619–634. doi: 10.1038/nrc.2016.71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ballester B, Milara J, Cortijo J (2019) Idiopathic Pulmonary Fibrosis and Lung Cancer: Mechanisms and Molecular Targets. Int J Mol Sci 20 (3). doi: 10.3390/ijms20030593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bellon G, Monboisse JC, Randoux A, Borel JP (1987) Effects of preformed proline and proline amino acid precursors (including glutamine) on collagen synthesis in human fibroblast cultures. Biochim Biophys Acta 930 (1):39–47. doi: 10.1016/0167-4889(87)90153-4 [DOI] [PubMed] [Google Scholar]
- Bellon G, Randoux A, Borel JP (1985) A study of collagen metabolism in cell cultures by fluorometric determination of proline and hydroxyproline. Coll Relat Res 5 (5):423–435. doi: 10.1016/s0174-173x(85)80030-3 [DOI] [PubMed] [Google Scholar]
- Bernard K, Logsdon NJ, Benavides GA, Sanders Y, Zhang J, Darley-Usmar VM, Thannickal VJ (2018) Glutaminolysis is required for transforming growth factor-beta1-induced myofibroblast differentiation and activation. J Biol Chem 293 (4):1218–1228. doi: 10.1074/jbc.RA117.000444 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bernard K, Logsdon NJ, Ravi S, Xie N, Persons BP, Rangarajan S, Zmijewski JW, Mitra K, Liu G, Darley-Usmar VM, Thannickal VJ (2015) Metabolic Reprogramming Is Required for Myofibroblast Contractility and Differentiation. J Biol Chem 290 (42):25427–25438. doi: 10.1074/jbc.M115.646984 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Biffi G, Tuveson DA (2021) Diversity and Biology of Cancer-Associated Fibroblasts. Physiol Rev 101 (1):147–176. doi: 10.1152/physrev.00048.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blumenkrantz N, Asboe-Hansen G (1973) Effect of amino acids on collagen biosynthesis. In Vitro 8 (5):342–346. doi: 10.1007/BF02619058 [DOI] [PubMed] [Google Scholar]
- Bradley KH, McConnell SD, Crystal RG (1974) Lung collagen composition and synthesis. Characterization and changes with age. J Biol Chem 249 (9):2674–2683 [PubMed] [Google Scholar]
- Calon A, Lonardo E, Berenguer-Llergo A, Espinet E, Hernando-Momblona X, Iglesias M, Sevillano M, Palomo-Ponce S, Tauriello DV, Byrom D, Cortina C, Morral C, Barcelo C, Tosi S, Riera A, Attolini CS, Rossell D, Sancho E, Batlle E (2015) Stromal gene expression defines poor-prognosis subtypes in colorectal cancer. Nat Genet 47 (4):320–329. doi: 10.1038/ng.3225 [DOI] [PubMed] [Google Scholar]
- Choudhury M, Yin X, Schaefbauer KJ, Kang JH, Roy B, Kottom TJ, Limper AH, Leof EB (2020) SIRT7-mediated modulation of glutaminase 1 regulates TGF-beta-induced pulmonary fibrosis. FASEB J. doi: 10.1096/fj.202000564R [DOI] [PubMed] [Google Scholar]
- Comes S, Gagliardi M, Laprano N, Fico A, Cimmino A, Palamidessi A, De Cesare D, De Falco S, Angelini C, Scita G, Patriarca EJ, Matarazzo MR, Minchiotti G (2013) L-Proline induces a mesenchymal-like invasive program in embryonic stem cells by remodeling H3K9 and H3K36 methylation. Stem Cell Reports 1 (4):307–321. doi: 10.1016/j.stemcr.2013.09.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cox TR, Erler JT (2014) Molecular pathways: connecting fibrosis and solid tumor metastasis. Clin Cancer Res 20 (14):3637–3643. doi: 10.1158/1078-0432.CCR-13-1059 [DOI] [PubMed] [Google Scholar]
- Cui H, Xie N, Jiang D, Banerjee S, Ge J, Sanders YY, Liu G (2019) Inhibition of Glutaminase 1 Attenuates Experimental Pulmonary Fibrosis. Am J Respir Cell Mol Biol 61 (4):492–500. doi: 10.1165/rcmb.2019-0051OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- D’Aniello C, Cermola F, Palamidessi A, Wanderlingh LG, Gagliardi M, Migliaccio A, Varrone F, Casalino L, Matarazzo MR, De Cesare D, Scita G, Patriarca EJ, Minchiotti G (2019) Collagen Prolyl Hydroxylation-Dependent Metabolic Perturbation Governs Epigenetic Remodeling and Mesenchymal Transition in Pluripotent and Cancer Cells. Cancer Res 79 (13):3235–3250. doi: 10.1158/0008-5472.CAN-18-2070 [DOI] [PubMed] [Google Scholar]
- D’Aniello C, Habibi E, Cermola F, Paris D, Russo F, Fiorenzano A, Di Napoli G, Melck DJ, Cobellis G, Angelini C, Fico A, Blelloch R, Motta A, Stunnenberg HG, De Cesare D, Patriarca EJ, Minchiotti G (2017) Vitamin C and l-Proline Antagonistic Effects Capture Alternative States in the Pluripotency Continuum. Stem Cell Reports 8 (1):1–10. doi: 10.1016/j.stemcr.2016.11.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darby IA, Zakuan N, Billet F, Desmouliere A (2016) The myofibroblast, a key cell in normal and pathological tissue repair. Cell Mol Life Sci 73 (6):1145–1157. doi: 10.1007/s00018-015-2110-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davidson SM, Jonas O, Keibler MA, Hou HW, Luengo A, Mayers JR, Wyckoff J, Del Rosario AM, Whitman M, Chin CR, Condon KJ, Lammers A, Kellersberger KA, Stall BK, Stephanopoulos G, Bar-Sagi D, Han J, Rabinowitz JD, Cima MJ, Langer R, Vander Heiden MG (2017) Direct evidence for cancer-cell-autonomous extracellular protein catabolism in pancreatic tumors. Nat Med 23 (2):235–241. doi: 10.1038/nm.4256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davidson SM, Papagiannakopoulos T, Olenchock BA, Heyman JE, Keibler MA, Luengo A, Bauer MR, Jha AK, O’Brien JP, Pierce KA, Gui DY, Sullivan LB, Wasylenko TM, Subbaraj L, Chin CR, Stephanopolous G, Mott BT, Jacks T, Clish CB, Vander Heiden MG (2016) Environment Impacts the Metabolic Dependencies of Ras-Driven Non-Small Cell Lung Cancer. Cell Metab 23 (3):517–528. doi: 10.1016/j.cmet.2016.01.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Distler JHW, Gyorfi AH, Ramanujam M, Whitfield ML, Konigshoff M, Lafyatis R (2019) Shared and distinct mechanisms of fibrosis. Nat Rev Rheumatol 15 (12):705–730. doi: 10.1038/s41584-019-0322-7 [DOI] [PubMed] [Google Scholar]
- Dolberg DS, Hollingsworth R, Hertle M, Bissell MJ (1985) Wounding and its role in RSV-mediated tumor formation. Science 230 (4726):676–678. doi: 10.1126/science.2996144 [DOI] [PubMed] [Google Scholar]
- Durante W, Liao L, Reyna SV, Peyton KJ, Schafer AI (2001) Transforming growth factor-beta(1) stimulates L-arginine transport and metabolism in vascular smooth muscle cells: role in polyamine and collagen synthesis. Circulation 103 (8):1121–1127. doi: 10.1161/01.cir.103.8.1121 [DOI] [PubMed] [Google Scholar]
- Dvorak HF (1986) Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 315 (26):1650–1659. doi: 10.1056/NEJM198612253152606 [DOI] [PubMed] [Google Scholar]
- El-Hattab AW (2016) Serine biosynthesis and transport defects. Mol Genet Metab 118 (3):153–159. doi: 10.1016/j.ymgme.2016.04.010 [DOI] [PubMed] [Google Scholar]
- Endo M, Oyadomari S, Terasaki Y, Takeya M, Suga M, Mori M, Gotoh T (2003) Induction of arginase I and II in bleomycin-induced fibrosis of mouse lung. Am J Physiol Lung Cell Mol Physiol 285 (2):L313–321. doi: 10.1152/ajplung.00434.2002 [DOI] [PubMed] [Google Scholar]
- Faubert B, Li KY, Cai L, Hensley CT, Kim J, Zacharias LG, Yang C, Do QN, Doucette S, Burguete D, Li H, Huet G, Yuan Q, Wigal T, Butt Y, Ni M, Torrealba J, Oliver D, Lenkinski RE, Malloy CR, Wachsmann JW, Young JD, Kernstine K, DeBerardinis RJ (2017) Lactate Metabolism in Human Lung Tumors. Cell 171 (2):358–371 e359. doi: 10.1016/j.cell.2017.09.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ge J, Cui H, Xie N, Banerjee S, Guo S, Dubey S, Barnes S, Liu G (2018) Glutaminolysis Promotes Collagen Translation and Stability via alpha-Ketoglutarate-mediated mTOR Activation and Proline Hydroxylation. Am J Respir Cell Mol Biol 58 (3):378–390. doi: 10.1165/rcmb.2017-0238OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamanaka RB, Chandel NS (2010) Mitochondrial reactive oxygen species regulate cellular signaling and dictate biological outcomes. Trends Biochem Sci 35 (9):505–513. doi: 10.1016/j.tibs.2010.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamanaka RB, Nigdelioglu R, Meliton AY, Tian Y, Witt LJ, O’Leary E, Sun KA, Woods PS, Wu D, Ansbro B, Ard S, Rohde JM, Dulin NO, Guzy RD, Mutlu GM (2018) Inhibition of Phosphoglycerate Dehydrogenase Attenuates Bleomycin-induced Pulmonary Fibrosis. Am J Respir Cell Mol Biol 58 (5):585–593. doi: 10.1165/rcmb.2017-0186OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamanaka RB, O’Leary EM, Witt LJ, Tian Y, Gokalp GA, Meliton AY, Dulin NO, Mutlu GM (2019) Glutamine Metabolism Is Required for Collagen Protein Synthesis in Lung Fibroblasts. Am J Respir Cell Mol Biol 61 (5):597–606. doi: 10.1165/rcmb.2019-0008OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hecker L, Vittal R, Jones T, Jagirdar R, Luckhardt TR, Horowitz JC, Pennathur S, Martinez FJ, Thannickal VJ (2009) NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury. Nat Med 15 (9):1077–1081. doi: 10.1038/nm.2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Henderson NC, Rieder F, Wynn TA (2020) Fibrosis: from mechanisms to medicines. Nature 587 (7835):555–566. doi: 10.1038/s41586-020-2938-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hensley CT, Faubert B, Yuan Q, Lev-Cohain N, Jin E, Kim J, Jiang L, Ko B, Skelton R, Loudat L, Wodzak M, Klimko C, McMillan E, Butt Y, Ni M, Oliver D, Torrealba J, Malloy CR, Kernstine K, Lenkinski RE, DeBerardinis RJ (2016) Metabolic Heterogeneity in Human Lung Tumors. Cell 164 (4):681–694. doi: 10.1016/j.cell.2015.12.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hildebran JN, Airhart J, Stirewalt WS, Low RB (1981) Prolyl-tRNA-based rates of protein and collagen synthesis in human lung fibroblasts. Biochem J 198 (2):249–258. doi: 10.1042/bj1980249 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hinz B, Lagares D (2020) Evasion of apoptosis by myofibroblasts: a hallmark of fibrotic diseases. Nat Rev Rheumatol 16 (1):11–31. doi: 10.1038/s41584-019-0324-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houtkooper RH, Pirinen E, Auwerx J (2012) Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol 13 (4):225–238. doi: 10.1038/nrm3293 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hwang IY, Kwak S, Lee S, Kim H, Lee SE, Kim JH, Kim YA, Jeon YK, Chung DH, Jin X, Park S, Jang H, Cho EJ, Youn HD (2016) Psat1-Dependent Fluctuations in alpha-Ketoglutarate Affect the Timing of ESC Differentiation. Cell Metab 24 (3):494–501. doi: 10.1016/j.cmet.2016.06.014 [DOI] [PubMed] [Google Scholar]
- Ignotz RA, Massague J (1986) Transforming growth factor-beta stimulates the expression of fibronectin and collagen and their incorporation into the extracellular matrix. J Biol Chem 261 (9):4337–4345 [PubMed] [Google Scholar]
- Islam MS, Leissing TM, Chowdhury R, Hopkinson RJ, Schofield CJ (2018) 2-Oxoglutarate-Dependent Oxygenases. Annu Rev Biochem 87:585–620. doi: 10.1146/annurev-biochem-061516-044724 [DOI] [PubMed] [Google Scholar]
- Jain M, Rivera S, Monclus EA, Synenki L, Zirk A, Eisenbart J, Feghali-Bostwick C, Mutlu GM, Budinger GR, Chandel NS (2013) Mitochondrial reactive oxygen species regulate transforming growth factor-beta signaling. J Biol Chem 288 (2):770–777. doi: 10.1074/jbc.M112.431973 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jones DL, Haak AJ, Caporarello N, Choi KM, Ye Z, Yan H, Varelas X, Ordog T, Ligresti G, Tschumperlin DJ (2019) TGFbeta-induced fibroblast activation requires persistent and targeted HDAC-mediated gene repression. J Cell Sci 132 (20). doi: 10.1242/jcs.233486 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jun JI, Lau LF (2018) Resolution of organ fibrosis. J Clin Invest 128 (1):97–107. doi: 10.1172/JCI93563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kamphorst JJ, Nofal M, Commisso C, Hackett SR, Lu W, Grabocka E, Vander Heiden MG, Miller G, Drebin JA, Bar-Sagi D, Thompson CB, Rabinowitz JD (2015) Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res 75 (3):544–553. doi: 10.1158/0008-5472.CAN-14-2211 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kang YP, Lee SB, Lee JM, Kim HM, Hong JY, Lee WJ, Choi CW, Shin HK, Kim DJ, Koh ES, Park CS, Kwon SW, Park SW (2016) Metabolic Profiling Regarding Pathogenesis of Idiopathic Pulmonary Fibrosis. J Proteome Res 15 (5):1717–1724. doi: 10.1021/acs.jproteome.6b00156 [DOI] [PubMed] [Google Scholar]
- Karampitsakos T, Tzilas V, Tringidou R, Steiropoulos P, Aidinis V, Papiris SA, Bouros D, Tzouvelekis A (2017) Lung cancer in patients with idiopathic pulmonary fibrosis. Pulm Pharmacol Ther 45:1–10. doi: 10.1016/j.pupt.2017.03.016 [DOI] [PubMed] [Google Scholar]
- Karna E, Szoka L, Huynh TYL, Palka JA (2020) Proline-dependent regulation of collagen metabolism. Cell Mol Life Sci 77 (10):1911–1918. doi: 10.1007/s00018-019-03363-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karsdal MA, Nielsen SH, Leeming DJ, Langholm LL, Nielsen MJ, Manon-Jensen T, Siebuhr A, Gudmann NS, Ronnow S, Sand JM, Daniels SJ, Mortensen JH, Schuppan D (2017) The good and the bad collagens of fibrosis - Their role in signaling and organ function. Adv Drug Deliv Rev 121:43–56. doi: 10.1016/j.addr.2017.07.014 [DOI] [PubMed] [Google Scholar]
- Kaul S, Sharma SS, Mehta IK (2008) Free radical scavenging potential of L-proline: evidence from in vitro assays. Amino Acids 34 (2):315–320. doi: 10.1007/s00726-006-0407-x [DOI] [PubMed] [Google Scholar]
- Kershenobich D, Fierro FJ, Rojkind M (1970) The relationship between the free pool of proline and collagen content in human liver cirrhosis. J Clin Invest 49 (12):2246–2249. doi: 10.1172/JCI106443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kitowska K, Zakrzewicz D, Konigshoff M, Chrobak I, Grimminger F, Seeger W, Bulau P, Eickelberg O (2008) Functional role and species-specific contribution of arginases in pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 294 (1):L34–45. doi: 10.1152/ajplung.00007.2007 [DOI] [PubMed] [Google Scholar]
- Kottakis F, Nicolay BN, Roumane A, Karnik R, Gu H, Nagle JM, Boukhali M, Hayward MC, Li YY, Chen T, Liesa M, Hammerman PS, Wong KK, Hayes DN, Shirihai OS, Dyson NJ, Haas W, Meissner A, Bardeesy N (2016) LKB1 loss links serine metabolism to DNA methylation and tumorigenesis. Nature 539 (7629):390–395. doi: 10.1038/nature20132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krishnan N, Dickman MB, Becker DF (2008) Proline modulates the intracellular redox environment and protects mammalian cells against oxidative stress. Free Radic Biol Med 44 (4):671–681. doi: 10.1016/j.freeradbiomed.2007.10.054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li F, Simon MC (2020) Cancer Cells Don’t Live Alone: Metabolic Communication within Tumor Microenvironments. Dev Cell 54 (2):183–195. doi: 10.1016/j.devcel.2020.06.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li M, Zheng Y, Yuan H, Liu Y, Wen X (2017) Effects of dynamic changes in histone acetylation and deacetylase activity on pulmonary fibrosis. Int Immunopharmacol 52:272–280. doi: 10.1016/j.intimp.2017.09.020 [DOI] [PubMed] [Google Scholar]
- Li P, Wu G (2018) Roles of dietary glycine, proline, and hydroxyproline in collagen synthesis and animal growth. Amino Acids 50 (1):29–38. doi: 10.1007/s00726-017-2490-6 [DOI] [PubMed] [Google Scholar]
- Liu T, Zhou L, Li D, Andl T, Zhang Y (2019) Cancer-Associated Fibroblasts Build and Secure the Tumor Microenvironment. Front Cell Dev Biol 7:60. doi: 10.3389/fcell.2019.00060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maddocks OD, Berkers CR, Mason SM, Zheng L, Blyth K, Gottlieb E, Vousden KH (2013) Serine starvation induces stress and p53-dependent metabolic remodelling in cancer cells. Nature 493 (7433):542–546. doi: 10.1038/nature11743 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maddocks OD, Labuschagne CF, Adams PD, Vousden KH (2016) Serine Metabolism Supports the Methionine Cycle and DNA/RNA Methylation through De Novo ATP Synthesis in Cancer Cells. Mol Cell 61 (2):210–221. doi: 10.1016/j.molcel.2015.12.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maddocks ODK, Athineos D, Cheung EC, Lee P, Zhang T, van den Broek NJF, Mackay GM, Labuschagne CF, Gay D, Kruiswijk F, Blagih J, Vincent DF, Campbell KJ, Ceteci F, Sansom OJ, Blyth K, Vousden KH (2017) Modulating the therapeutic response of tumours to dietary serine and glycine starvation. Nature 544 (7650):372–376. doi: 10.1038/nature22056 [DOI] [PubMed] [Google Scholar]
- Maher EA, Marin-Valencia I, Bachoo RM, Mashimo T, Raisanen J, Hatanpaa KJ, Jindal A, Jeffrey FM, Choi C, Madden C, Mathews D, Pascual JM, Mickey BE, Malloy CR, DeBerardinis RJ (2012) Metabolism of [U-13 C]glucose in human brain tumors in vivo. NMR Biomed 25 (11):1234–1244. doi: 10.1002/nbm.2794 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moffitt RA, Marayati R, Flate EL, Volmar KE, Loeza SG, Hoadley KA, Rashid NU, Williams LA, Eaton SC, Chung AH, Smyla JK, Anderson JM, Kim HJ, Bentrem DJ, Talamonti MS, Iacobuzio-Donahue CA, Hollingsworth MA, Yeh JJ (2015) Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet 47 (10):1168–1178. doi: 10.1038/ng.3398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris SM Jr. (2002) Regulation of enzymes of the urea cycle and arginine metabolism. Annu Rev Nutr 22:87–105. doi: 10.1146/annurev.nutr.22.110801.140547 [DOI] [PubMed] [Google Scholar]
- Muir A, Danai LV, Vander Heiden MG (2018) Microenvironmental regulation of cancer cell metabolism: implications for experimental design and translational studies. Dis Model Mech 11 (8). doi: 10.1242/dmm.035758 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muir A, Vander Heiden MG (2018) The nutrient environment affects therapy. Science 360 (6392):962–963. doi: 10.1126/science.aar5986 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nigdelioglu R, Hamanaka RB, Meliton AY, O’Leary E, Witt LJ, Cho T, Sun K, Bonham C, Wu D, Woods PS, Husain AN, Wolfgeher D, Dulin NO, Chandel NS, Mutlu GM (2016) Transforming Growth Factor (TGF)-beta Promotes de Novo Serine Synthesis for Collagen Production. J Biol Chem 291 (53):27239–27251. doi: 10.1074/jbc.M116.756247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nojima Y, Takeda Y, Maeda Y, Bamba T, Fukusaki E, Itoh MN, Mizuguchi K, Kumanogoh A (2020) Metabolomic analysis of fibrotic mice combined with public RNA-Seq human lung data reveal potential diagnostic biomarker candidates for lung fibrosis. FEBS Open Bio. doi: 10.1002/2211-5463.12982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Leary EM, Tian Y, Nigdelioglu R, Witt LJ, Cetin-Atalay R, Meliton AY, Woods PS, Kimmig LM, Sun KA, Gokalp GA, Mutlu GM, Hamanaka RB (2020) TGF-beta Promotes Metabolic Reprogramming in Lung Fibroblasts via mTORC1-dependent ATF4 Activation. Am J Respir Cell Mol Biol 63 (5):601–612. doi: 10.1165/rcmb.2020-0143OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olivares O, Mayers JR, Gouirand V, Torrence ME, Gicquel T, Borge L, Lac S, Roques J, Lavaut MN, Berthezene P, Rubis M, Secq V, Garcia S, Moutardier V, Lombardo D, Iovanna JL, Tomasini R, Guillaumond F, Vander Heiden MG, Vasseur S (2017) Collagen-derived proline promotes pancreatic ductal adenocarcinoma cell survival under nutrient limited conditions. Nat Commun 8:16031. doi: 10.1038/ncomms16031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pakshir P, Noskovicova N, Lodyga M, Son DO, Schuster R, Goodwin A, Karvonen H, Hinz B (2020) The myofibroblast at a glance. J Cell Sci 133 (13). doi: 10.1242/jcs.227900 [DOI] [PubMed] [Google Scholar]
- Piersma B, Hayward MK, Weaver VM (2020) Fibrosis and cancer: A strained relationship. Biochim Biophys Acta Rev Cancer 1873 (2):188356. doi: 10.1016/j.bbcan.2020.188356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rabinovich EI, Kapetanaki MG, Steinfeld I, Gibson KF, Pandit KV, Yu G, Yakhini Z, Kaminski N (2012) Global methylation patterns in idiopathic pulmonary fibrosis. PLoS One 7 (4):e33770. doi: 10.1371/journal.pone.0033770 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reina-Campos M, Linares JF, Duran A, Cordes T, L’Hermitte A, Badur MG, Bhangoo MS, Thorson PK, Richards A, Rooslid T, Garcia-Olmo DC, Nam-Cha SY, Salinas-Sanchez AS, Eng K, Beltran H, Scott DA, Metallo CM, Moscat J, Diaz-Meco MT (2019) Increased Serine and One-Carbon Pathway Metabolism by PKClambda/iota Deficiency Promotes Neuroendocrine Prostate Cancer. Cancer Cell 35 (3):385–400 e389. doi: 10.1016/j.ccell.2019.01.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ricard-Blum S (2011) The collagen family. Cold Spring Harb Perspect Biol 3 (1):a004978. doi: 10.1101/cshperspect.a004978 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roberts AB, Sporn MB, Assoian RK, Smith JM, Roche NS, Wakefield LM, Heine UI, Liotta LA, Falanga V, Kehrl JH, et al. (1986) Transforming growth factor type beta: rapid induction of fibrosis and angiogenesis in vivo and stimulation of collagen formation in vitro. Proc Natl Acad Sci U S A 83 (12):4167–4171. doi: 10.1073/pnas.83.12.4167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robins SP (1979) Metabolism of rabbit skin collagen. Differences in the apparent turnover rates of type-I-and type-III-collagen precursors determined by constant intravenous infusion of labelled amino acids. Biochem J 181 (1):75–82. doi: 10.1042/bj1810075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rockey DC, Bell PD, Hill JA (2015) Fibrosis--a common pathway to organ injury and failure. N Engl J Med 372 (12):1138–1149. doi: 10.1056/NEJMra1300575 [DOI] [PubMed] [Google Scholar]
- Rojkind M, Diaz de Leon L (1970) Collagen biosynthesis in cirrhotic rat liver slices a regulatory mechanism. Biochim Biophys Acta 217 (2):512–522. doi: 10.1016/0005-2787(70)90548-4 [DOI] [PubMed] [Google Scholar]
- Rumping L, Vringer E, Houwen RHJ, van Hasselt PM, Jans JJM, Verhoeven-Duif NM (2020) Inborn errors of enzymes in glutamate metabolism. J Inherit Metab Dis 43 (2):200–215. doi: 10.1002/jimd.12180 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sanders YY, Ambalavanan N, Halloran B, Zhang X, Liu H, Crossman DK, Bray M, Zhang K, Thannickal VJ, Hagood JS (2012) Altered DNA methylation profile in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 186 (6):525–535. doi: 10.1164/rccm.201201-0077OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schworer S, Berisa M, Violante S, Qin W, Zhu J, Hendrickson RC, Cross JR, Thompson CB (2020) Proline biosynthesis is a vent for TGFbeta-induced mitochondrial redox stress. EMBO J 39 (8):e103334. doi: 10.15252/embj.2019103334 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selvarajah B, Azuelos I, Plate M, Guillotin D, Forty EJ, Contento G, Woodcock HV, Redding M, Taylor A, Brunori G, Durrenberger PF, Ronzoni R, Blanchard AD, Mercer PF, Anastasiou D, Chambers RC (2019) mTORC1 amplifies the ATF4-dependent de novo serine-glycine pathway to supply glycine during TGF-beta1-induced collagen biosynthesis. Sci Signal 12 (582). doi: 10.1126/scisignal.aav3048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Selwan EM, Edinger AL (2017) Branched chain amino acid metabolism and cancer: the importance of keeping things in context. Transl Cancer Res 6 (Suppl 3):S578–S584. doi: 10.21037/tcr.2017.05.05 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shaw TJ, Rognoni E (2020) Dissecting Fibroblast Heterogeneity in Health and Fibrotic Disease. Curr Rheumatol Rep 22 (8):33. doi: 10.1007/s11926-020-00903-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen TF, Strecker HJ (1975) Synthesis of proline and hydroxyproline in human lung (WI-38) fibroblasts. Biochem J 150 (3):453–461. doi: 10.1042/bj1500453 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shoulders MD, Raines RT (2009) Collagen structure and stability. Annu Rev Biochem 78:929–958. doi: 10.1146/annurev.biochem.77.032207.120833 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sime PJ, Xing Z, Graham FL, Csaky KG, Gauldie J (1997) Adenovector-mediated gene transfer of active transforming growth factor-beta1 induces prolonged severe fibrosis in rat lung. J Clin Invest 100 (4):768–776. doi: 10.1172/JCI119590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith RJ, Phang JM (1978) Proline metabolism in cartilage: the importance of proline biosynthesis. Metabolism 27 (6):685–694. doi: 10.1016/0026-0495(78)90006-9 [DOI] [PubMed] [Google Scholar]
- Sporn MB, Roberts AB, Shull JH, Smith JM, Ward JM, Sodek J (1983) Polypeptide transforming growth factors isolated from bovine sources and used for wound healing in vivo. Science 219 (4590):1329–1331. doi: 10.1126/science.6572416 [DOI] [PubMed] [Google Scholar]
- Su X, Wellen KE, Rabinowitz JD (2016) Metabolic control of methylation and acetylation. Curr Opin Chem Biol 30:52–60. doi: 10.1016/j.cbpa.2015.10.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sullivan MR, Danai LV, Lewis CA, Chan SH, Gui DY, Kunchok T, Dennstedt EA, Vander Heiden MG, Muir A (2019) Quantification of microenvironmental metabolites in murine cancers reveals determinants of tumor nutrient availability. Elife 8. doi: 10.7554/eLife.44235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Szabados L, Savoure A (2010) Proline: a multifunctional amino acid. Trends Plant Sci 15 (2):89–97. doi: 10.1016/j.tplants.2009.11.009 [DOI] [PubMed] [Google Scholar]
- Tschumperlin DJ, Ligresti G, Hilscher MB, Shah VH (2018) Mechanosensing and fibrosis. J Clin Invest 128 (1):74–84. doi: 10.1172/JCI93561 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsurufuji S, Mori Y (1965) The Conversion of D-(14c-6)Glucose to Glycine and Its Incorporation into Collagen. Biochim Biophys Acta 97:149–151. doi: 10.1016/0304-4165(65)90281-3 [DOI] [PubMed] [Google Scholar]
- Valkenburg KC, de Groot AE, Pienta KJ (2018) Targeting the tumour stroma to improve cancer therapy. Nat Rev Clin Oncol 15 (6):366–381. doi: 10.1038/s41571-018-0007-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu G (2020) Important roles of dietary taurine, creatine, carnosine, anserine and 4-hydroxyproline in human nutrition and health. Amino Acids 52 (3):329–360. doi: 10.1007/s00726-020-02823-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Z, Hou Y, Dai Z, Hu CA, Wu G (2019) Metabolism, Nutrition, and Redox Signaling of Hydroxyproline. Antioxid Redox Signal 30 (4):674–682. doi: 10.1089/ars.2017.7338 [DOI] [PubMed] [Google Scholar]
- Xie N, Tan Z, Banerjee S, Cui H, Ge J, Liu RM, Bernard K, Thannickal VJ, Liu G (2015) Glycolytic Reprogramming in Myofibroblast Differentiation and Lung Fibrosis. Am J Respir Crit Care Med 192 (12):1462–1474. doi: 10.1164/rccm.201504-0780OC [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu S, Xu H, Wang W, Li S, Li H, Li T, Zhang W, Yu X, Liu L (2019) The role of collagen in cancer: from bench to bedside. J Transl Med 17 (1):309. doi: 10.1186/s12967-019-2058-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye J, Fan J, Venneti S, Wan YW, Pawel BR, Zhang J, Finley LW, Lu C, Lindsten T, Cross JR, Qing G, Liu Z, Simon MC, Rabinowitz JD, Thompson CB (2014) Serine catabolism regulates mitochondrial redox control during hypoxia. Cancer Discov 4 (12):1406–1417. doi: 10.1158/2159-8290.CD-14-0250 [DOI] [PMC free article] [PubMed] [Google Scholar]



