Hypothalamic-Pituitary Axis Regulates Hydrogen Sulfide Production

Christopher Hine, Hyo-Jeong Kim, Yan Zhu, Eylul Harputlugil, Alban Longchamp, Marina Souza Matos, Preeti Ramadoss, Kevin Bauerle, Lear Brace, John M Asara, C Keith Ozaki, Sheue-Yann Cheng, Subhankar Singha, Kyo Han Ahn, Alec Kimmelman, Ffolliott M Fisher, Pavlos Pissios, Dominic J Withers, Colin Selman, Rui Wang, Kelvin Yen, Valter D Longo, Pinchas Cohen, Andrzej Bartke, John J Kopchick, Richard Miller, Anthony N Hollenberg, James R Mitchell, Christopher Hine, Hyo-Jeong Kim, Yan Zhu, Eylul Harputlugil, Alban Longchamp, Marina Souza Matos, Preeti Ramadoss, Kevin Bauerle, Lear Brace, John M Asara, C Keith Ozaki, Sheue-Yann Cheng, Subhankar Singha, Kyo Han Ahn, Alec Kimmelman, Ffolliott M Fisher, Pavlos Pissios, Dominic J Withers, Colin Selman, Rui Wang, Kelvin Yen, Valter D Longo, Pinchas Cohen, Andrzej Bartke, John J Kopchick, Richard Miller, Anthony N Hollenberg, James R Mitchell

Abstract

Decreased growth hormone (GH) and thyroid hormone (TH) signaling are associated with longevity and metabolic fitness. The mechanisms underlying these benefits are poorly understood, but may overlap with those of dietary restriction (DR), which imparts similar benefits. Recently we discovered that hydrogen sulfide (H2S) is increased upon DR and plays an essential role in mediating DR benefits across evolutionary boundaries. Here we found increased hepatic H2S production in long-lived mouse strains of reduced GH and/or TH action, and in a cell-autonomous manner upon serum withdrawal in vitro. Negative regulation of hepatic H2S production by GH and TH was additive and occurred via distinct mechanisms, namely direct transcriptional repression of the H2S-producing enzyme cystathionine γ-lyase (CGL) by TH, and substrate-level control of H2S production by GH. Mice lacking CGL failed to downregulate systemic T4 metabolism and circulating IGF-1, revealing an essential role for H2S in the regulation of key longevity-associated hormones.

Keywords: FGF21; IGF-1; IRS-1; autophagy; cystathionine γ-lyase; growth hormone; hydrogen sulfide; hypopituitary dwarfism; longevity; thyroid hormone.

Copyright © 2017 Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure 1
Figure 1
Increased Hepatic H2S Production in Long-Lived Hypopituitary Dwarf Mice In Vivo (A–D) Hepatic CGL and CBS mRNA expression (n = 3/group) (A), protein expression (n = 3/group) (B), H2S production capacity via the lead sulfide method (n = 3/group) (C), and endogenous H2S production via two-photon florescence microscopy (n = 3/group) (D) in male and female WT or Snell dwarf mice as indicated. Scale bar, 25 μm. Asterisk indicates the significance of the difference between genotypes within sex; ∗p < 0.05. (E) Liver H2S production capacity in 18-month-old female Ames dwarf or WT mice treated +/− growth hormone during postnatal development at weeks 2-8 (n = 9–14/group). The asterisk indicates the significance of the difference between the WT+Saline control group and experimental group, and the # sign indicates the significance of the difference between +Saline and +GH; ∗/#p < 0.05. (F) Liver H2S production capacity in mice treated with saline control or lanreotide (n = 4/group). The asterisks indicate the significance of the difference between treatment groups; ∗p < 0.05. Error bars are ± SEM. See also Figure S1.
Figure 2
Figure 2
Growth Hormone Signaling Inhibits Hepatic H2S Production In vivo (A–C) Hepatic CGL and CBS mRNA expression (n = 3/group) (A), protein expression (n = 3/group) (B), and H2S production capacity (n = 3/group) (C) in male and female growth hormone receptor knockout (GHRKO) mice. The asterisk indicates the significance of the difference between genotypes within sex; ∗p < 0.05. (D) Liver H2S production capacity (n = 8/group) in WT mice treated for 2 weeks with recombinant IGF-1, GH, or saline vehicle as indicated. The asterisk indicates the significance of the difference between GH and saline treatment; ∗p < 0.05. (E and F) Liver H2S production capacity in male and female IRS-1 WT or KO mice (n = 4–11/group) (E) and in male FGF21 WT or overexpressing (OE) mice (n = 6/group) (F). The asterisk indicates the significance of the difference between genotypes within sex; ∗p < 0.05. Error bars are ± SEM. See also Figure S2.
Figure 3
Figure 3
Growth Hormone Receptor Signaling Inhibits Hepatic H2S Production In Vitro (A–C) Endogenous H2S production in primary mouse hepatocytes as measured via two-photon fluorescent microscopy under different medium conditions: (A) +/−growth serum and +/−GH, with the asterisk indicating the significance of the difference between Complete and −Serum, and the # sign indicating the significance of the difference between −Serum and −Serum+GH, ∗/#p < 0.05; (B) +/−growth serum, +/−GH, +/−AZD1480, with the asterisk indicating the significance of the difference between Complete and −Serum in each group, and the # sign indicating the significance of the difference between −Serum (−) (no addition) and −Serum+ZD1480; and the $ sign indicating the significance of the difference between +GH and +GH+ZD1480 for both Complete and −Serum conditions, ∗/#/$p < 0.05; (C) +/−FGF21 in Complete medium containing serum, with the asterisk indicating the significance of the difference between Complete and Complete+FGF21; ∗p < 0.05. (D) mRNA expression of Igf-I, Cgl, and Cbs in mouse primary hepatocytes. The asterisk indicates the significance of the difference between +GH and +GH+ZD1480; ∗p < 0.05. (E and F) Endogenous H2S production in mouse primary hepatocytes by florescent microscopy after overnight treatment under the indicated medium conditions followed by addition of the P3 probe for 1 hr. The asterisk indicates the significance of the difference between Complete and −Serum, and the # sign indicates the significance of the difference between −Serum and −Serum+BAF (E), or −Serum−SAA and −Serum−SAA+CQ (F); ∗#p < 0.05. (G and H) Endogenous H2S production in Hepa1-6 cells with or without knockdown of autophagy components ATG5 and ATG7 by shRNA (G) or in MEFs by genetic knockout of ATG5 and ATG7 (H) detected by UV spectrophotometry after overnight treatment with or without serum followed by addition of the P3 probe for 1 hr. The asterisk indicates the significance of the difference between Complete and −Serum in the Control group, and the # sign indicates the significance of the difference between −Serum Control and −Serum ATG5 deficient or −Serum ATG7 deficient; ∗/#p < 0.05. Each experiment was repeated at least three times. Scale bars, 100 μm (A–C, E, and F). Error bars are ± SEM. See also Figure S3.
Figure 4
Figure 4
Hypothyroidism Increases and Thyroid Hormone Represses Hepatic H2S Production In Vivo (A–D) mRNA expression (n = 4) (A), protein expression (n = 3) (B), H2S production capacity (n = 4) (C), and transmethylation/transsulfuration metabolite levels by liquid chromatography-tandem mass spectrometry (n = 4) (D) in livers of mice under hypo- (PTU+Saline), hyper- (PTU+high dose T3), and eu-(PTU+low dose T3) thyroid states. The asterisks indicate the significance of the difference from the hypothyroid state (PTU+Saline); ∗p < 0.05. (E–G) Analysis of TH-responsive and sulfur amino acid metabolism-associated mRNA levels (n = 4–6) (E), protein expression (n = 5) (F), and H2S production capacity (n = 5) (G) in livers of mice treated with T3 (hyperthyroid) versus vehicle (saline) control (euthyroid). The asterisk indicates the significance of the difference between vehicle and +T3 groups; ∗p < 0.05. Error bars are ± SEM. See also Figure S4.
Figure 5
Figure 5
Thyroid Hormone Signaling through TRβ Represses Hepatic H2S Production In Vivo (A–C) Analysis of TH-responsive and sulfur amino acid metabolism-associated mRNA levels (n = 4–6) (A), protein expression (n = 6) (B), and H2S production capacity (n = 5) (C) in livers of mice treated with GC-1 versus vehicle (saline) control. The asterisk indicates the significance of the difference between vehicle (saline) control (euthyroid) and +GC-1 groups (hyperthyroid); ∗p < 0.05. (D and E) Liver CBS and CGL protein expression (D) and H2S production capacity (E) in mice with indicated TRβ status (WT, homozygous WT; TRβPV/+, Het; TRβPV/PV, homozygous mutant; n = 4–5/group). The asterisk indicates the significance of the difference between WT and TRβPV/PV, and the # sign indicates the significance of the difference between TRβPV/+ and TRβPV/PV; ∗/#p < 0.05. (F) Fold enrichment of TRβ binding to genetic regulator elements in sulfur amino acid metabolism and H2S producing genes in the livers of mice infected with Ad-GFP (control) or Ad-TRβ while on PTU diets +/−T3 injection as indicated (n = 5/group). The asterisk indicates the significance of the difference between the Ad-GFP PTU and Ad-TRβ PTU or Ad-TRβ T3 groups, and the # sign indicates the significance of the difference between the Ad-TRβ PTU and Ad-TRβ T3 groups; ∗/#p < 0.05. (G and H) Liver ATF4 protein expression in mice due to PTU/T3 administration (G) (n = 4/group) or TRβ mutations (H) (n = 4–5/group). The asterisks indicate the significance of the difference between Hyper-T and Eu-T or Hypo-T (G), or WT and TRβPV/+ or TRβPV/PV (H), and the # sign indicates the significance of the difference between TRβPV/+ and TRβPV/PV (H), ∗/#p < 0.05. Error bars are ± SEM. See also Figure S5.
Figure 6
Figure 6
Additive Suppression of Hepatic H2S Production by TH and GH In Vitro (A–C) mRNA levels (n = 3) (A), protein expression (n = 3) (B), and endogenous H2S production (n = 3) (C) in TH-responsive Hepa1-6 cells treated with T3 at the indicated concentration. The asterisk indicates the significance of the difference between vehicle control and T3 treatment and the hash indicates the significance of the difference between different T3 dosage groups; ∗/#p < 0.05. (D) Endogenous H2S production in TH-responsive Hepa1-6 cells grown in Complete medium or −Serum medium +/−GH +/−T3. The asterisk indicates the significance of the difference between Complete and −Serum within the GH/T3 treatment group, and the # sign indicates the significance of the difference between “w/out Additions” and +GH, +T3, or +GH+T3 groups within the −Serum grouping; ∗/#p < 0.05. Scale bars, 100 μm (C and D). Error bars are ± SEM. See also Figure S6.
Figure 7
Figure 7
CGL/H2S Is Required for Negative Regulation of TH and GH/IGF-I Signaling (A and B) Western blot analysis of H2S-producing enzymes CGL, CBS, and 3MST (n = 3) (A) and H2S production capacity in livers of CGL WT and KO mice on Normal and PTU diets (n = 4–5) (B). The asterisk indicates the significance of the difference relative to CGL WT mice on the Normal diet; ∗p < 0.05. (C) Serum T4 levels over a 3-week time course of Normal versus PTU diets in CGL WT and KO mice as indicated (n = 4–5). (D) Pituitary mRNA expression of TSHα and TSHβ in CGL WT and KO mice after 3 weeks of Normal or PTU diet (n = 4–5). The asterisk indicates the significance of difference between diets within genotype, and the # sign indicates the significance of difference between CGL WT and CGL KO mice on the PTU diet; ∗/#p < 0.05. (E) Serum TSH levels after 3 weeks of Normal or PTU diet in CGL WT and KO mice (n = 4–5). The asterisk indicates the significance of difference between diets within genotype, and the # sign indicates the significance of difference between CGL WT and CGL KO mice on the PTU diet; ∗/#p < 0.05. (F) Percent change in serum T4 levels after 3 days of fasting in CGL WT and CGL KO mice (n = 3–4). The asterisk indicates the significance of the difference between day 0 and day 3 T4 levels; ∗p < 0.05. (G–I) Pituitary GH mRNA expression (n = 4–5) (G), liver GHR and IGF-I mRNA expression (n = 4–5) (H), and serum IGF-1 (n = 4–5) (I) in CGL WT and KO mice after 3 weeks on a Normal or PTU diet as indicated. The asterisk indicates the significance of the difference between diets within genotype, and the # sign indicates the significance of the difference between genotypes on the PTU diet; ∗/#p < 0.05. (J) Fold change in serum IGF-I between day 0 and day 3 of a 3-day fast in CGL WT and KO mice (n = 3–4/group). The asterisk indicates the significance of the difference between day 3 and day 0; ∗p < 0.05. (K) Serum IGF-1 levels in mice 7 days after adenoviral infection with Ad-Null control or Ad-CGL overexpression adenovirus expressed relative to Ad-Null control (n = 4/group). The asterisk indicates the significance of the difference between Ad-Null and Ad-CGL; ∗p < 0.05. (L) Fold change in serum IGF-1 after a 2-day fast in female CGL WT and KO mice with +/−NaHS supplementation in the CGL KOs (n = 4–5/group). The asterisks indicate the significance of the difference between serum IGF-1 levels on day 2 compared with day 0; ∗p < 0.05. (M) Relationship between diet, GH/TH, and H2S production. Error bars are ± SEM. See also Figure S7.

References

    1. Abe K., Kimura H. The possible role of hydrogen sulfide as an endogenous neuromodulator. J. Neurosci. 1996;16:1066–1071.
    1. Ahluwalia A., Clodfelter K.H., Waxman D.J. Sexual dimorphism of rat liver gene expression: regulatory role of growth hormone revealed by deoxyribonucleic acid microarray analysis. Mol. Endocrinol. 2004;18:747–760.
    1. Arum O., Bonkowski M.S., Rocha J.S., Bartke A. The growth hormone receptor gene-disrupted mouse fails to respond to an intermittent fasting diet. Aging Cell. 2009;8:756–760.
    1. Atzmon G., Barzilai N., Surks M.I., Gabriely I. Genetic predisposition to elevated serum thyrotropin is associated with exceptional longevity. J. Clin. Endocrinol. Metab. 2009;94:4768–4775.
    1. Bartke A., Brown-Borg H. Life extension in the dwarf mouse. Curr. Top. Dev. Biol. 2004;63:189–225.
    1. Bartke A., Wright J.C., Mattison J.A., Ingram D.K., Miller R.A., Roth G.S. Extending the lifespan of long-lived mice. Nature. 2001;414:412.
    1. Bian J.S., Yong Q.C., Pan T.T., Feng Z.N., Ali M.Y., Zhou S., Moore P.K. Role of hydrogen sulfide in the cardioprotection caused by ischemic preconditioning in the rat heart and cardiac myocytes. J. Pharmacol. Exp. Ther. 2006;316:670–678.
    1. Biddinger S.B., Hernandez-Ono A., Rask-Madsen C., Haas J.T., Alemán J.O., Suzuki R., Scapa E.F., Agarwal C., Carey M.C., Stephanopoulos G. Hepatic insulin resistance is sufficient to produce dyslipidemia and susceptibility to atherosclerosis. Cell Metab. 2008;7:125–134.
    1. Blackstone E., Roth M.B. Suspended animation-like state protects mice from lethal hypoxia. Shock. 2007;27:370–372.
    1. Bonkowski M.S., Rocha J.S., Masternak M.M., Al Regaiey K.A., Bartke A. Targeted disruption of growth hormone receptor interferes with the beneficial actions of calorie restriction. Proc. Natl. Acad. Sci. USA. 2006;103:7901–7905.
    1. Brown-Borg H.M. The somatotropic axis and longevity in mice. Am. J. Physiol. Endocrinol. Metab. 2015;309:E503–E510.
    1. Brown-Borg H.M., Rakoczy S.G., Wonderlich J.A., Rojanathammanee L., Kopchick J.J., Armstrong V., Raasakka D. Growth hormone signaling is necessary for lifespan extension by dietary methionine. Aging Cell. 2014;13:1019–1027.
    1. Cai W.J., Wang M.J., Moore P.K., Jin H.M., Yao T., Zhu Y.C. The novel proangiogenic effect of hydrogen sulfide is dependent on Akt phosphorylation. Cardiovasc. Res. 2007;76:29–40.
    1. Chiellini G., Apriletti J.W., Yoshihara H.A., Baxter J.D., Ribeiro R.C., Scanlan T.S. A high-affinity subtype-selective agonist ligand for the thyroid hormone receptor. Chem. Biol. 1998;5:299–306.
    1. Coschigano K.T., Holland A.N., Riders M.E., List E.O., Flyvbjerg A., Kopchick J.J. Deletion, but not antagonism, of the mouse growth hormone receptor results in severely decreased body weights, insulin, and insulin-like growth factor I levels and increased life span. Endocrinology. 2003;144:3799–3810.
    1. Dacks P.A., Moreno C.L., Kim E.S., Marcellino B.K., Mobbs C.V. Role of the hypothalamus in mediating protective effects of dietary restriction during aging. Front. Neuroendocrinol. 2013;34:95–106.
    1. Dauncey M.J., Burton K.A., White P., Harrison A.P., Gilmour R.S., Duchamp C., Cattaneo D. Nutritional regulation of growth hormone receptor gene expression. FASEB J. 1994;8:81–88.
    1. Dieguez C., Jordan V., Harris P., Foord S., Rodriguez-Arnao M.D., Gomez-Pan A., Hall R., Scanlon M.F. Growth hormone responses to growth hormone-releasing factor (1-29) in euthyroid, hypothyroid and hyperthyroid rats. J. Endocrinol. 1986;109:53–56.
    1. Do A., Menon V., Zhi X., Gesing A., Wiesenborn D.S., Spong A., Sun L., Bartke A., Masternak M.M. Thyroxine modifies the effects of growth hormone in Ames dwarf mice. Aging (Albany NY) 2015;7:241–255.
    1. Dozmorov I., Galecki A., Chang Y., Krzesicki R., Vergara M., Miller R.A. Gene expression profile of long-lived Snell dwarf mice. J. Gerontol. A Biol. Sci. Med. Sci. 2002;57:B99–B108.
    1. Elrod J.W., Calvert J.W., Morrison J., Doeller J.E., Kraus D.W., Tao L., Jiao X., Scalia R., Kiss L., Szabo C. Hydrogen sulfide attenuates myocardial ischemia-reperfusion injury by preservation of mitochondrial function. Proc. Natl. Acad. Sci. USA. 2007;104:15560–15565.
    1. Fan N.Y., Powers C.A., Stier C.T. Lack of antidiuretic activity of lanreotide in the diabetes insipidus rat. J. Pharmacol. Exp. Ther. 1996;276:875–881.
    1. Fontana L., Klein S., Holloszy J.O., Premachandra B.N. Effect of long-term calorie restriction with adequate protein and micronutrients on thyroid hormones. J. Clin. Endocrinol. Metab. 2006;91:3232–3235.
    1. Fontana L., Weiss E.P., Villareal D.T., Klein S., Holloszy J.O. Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell. 2008;7:681–687.
    1. Gesing A., Bartke A., Masternak M.M., Lewiński A., Karbownik-Lewińska M. Decreased thyroid follicle size in dwarf mice may suggest the role of growth hormone signaling in thyroid growth regulation. Thyroid Res. 2012;5:7.
    1. Gesing A., Al-Regaiey K.A., Bartke A., Masternak M.M. Growth hormone abolishes beneficial effects of calorie restriction in long-lived Ames dwarf mice. Exp. Gerontol. 2014;58:219–229.
    1. Gu L., Liao Z., Hoang D.T., Dagvadorj A., Gupta S., Blackmon S., Ellsworth E., Talati P., Leiby B., Zinda M. Pharmacologic inhibition of Jak2-Stat5 signaling By Jak2 inhibitor AZD1480 potently suppresses growth of both primary and castrate-resistant prostate cancer. Clin. Cancer Res. 2013;19:5658–5674.
    1. Guevara-Aguirre J., Balasubramanian P., Guevara-Aguirre M., Wei M., Madia F., Cheng C.W., Hwang D., Martin-Montalvo A., Saavedra J., Ingles S. Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci. Transl. Med. 2011;3:70ra13.
    1. Harputlugil E., Hine C., Vargas D., Robertson L., Manning B.D., Mitchell J.R. The TSC complex is required for the benefits of dietary protein restriction on stress resistance in vivo. Cell Rep. 2014;8:1160–1170.
    1. Hauck S.J., Hunter W.S., Danilovich N., Kopchick J.J., Bartke A. Reduced levels of thyroid hormones, insulin, and glucose, and lower body core temperature in the growth hormone receptor/binding protein knockout mouse. Exp. Biol. Med. (Maywood) 2001;226:552–558.
    1. Hine C., Harputlugil E., Zhang Y., Ruckenstuhl C., Lee B.C., Brace L., Longchamp A., Treviño-Villarreal J.H., Mejia P., Ozaki C.K. Endogenous hydrogen sulfide production is essential for dietary restriction benefits. Cell. 2015;160:132–144.
    1. Inagaki T., Lin V.Y., Goetz R., Mohammadi M., Mangelsdorf D.J., Kliewer S.A. Inhibition of growth hormone signaling by the fasting-induced hormone FGF21. Cell Metab. 2008;8:77–83.
    1. Kabil O., Vitvitsky V., Xie P., Banerjee R. The quantitative significance of the transsulfuration enzymes for H2S production in murine tissues. Antioxid. Redox Signal. 2011;15:363–372.
    1. Kato Y., Ying H., Willingham M.C., Cheng S.Y. A tumor suppressor role for thyroid hormone beta receptor in a mouse model of thyroid carcinogenesis. Endocrinology. 2004;145:4430–4438.
    1. Kharitonenkov A., Shiyanova T.L., Koester A., Ford A.M., Micanovic R., Galbreath E.J., Sandusky G.E., Hammond L.J., Moyers J.S., Owens R.A. FGF-21 as a novel metabolic regulator. J. Clin. Invest. 2005;115:1627–1635.
    1. Kimura Y., Kimura H. Hydrogen sulfide protects neurons from oxidative stress. FASEB J. 2004;18:1165–1167.
    1. Koenig R.J., Brent G.A., Warne R.L., Larsen P.R., Moore D.D. Thyroid hormone receptor binds to a site in the rat growth hormone promoter required for induction by thyroid hormone. Proc. Natl. Acad. Sci. USA. 1987;84:5670–5674.
    1. Kuhn J.M., Legrand A., Ruiz J.M., Obach R., De Ronzan J., Thomas F. Pharmacokinetic and pharmacodynamic properties of a long-acting formulation of the new somatostatin analogue, lanreotide, in normal healthy volunteers. Br. J. Clin. Pharmacol. 1994;38:213–219.
    1. Lee J.I., Dominy J.E., Sikalidis A.K., Hirschberger L.L., Wang W., Stipanuk M.H. HepG2/C3A cells respond to cysteine deprivation by induction of the amino acid deprivation/integrated stress response pathway. Physiol. Genomics. 2008;33:218–229.
    1. Lee Z.W., Zhou J., Chen C.S., Zhao Y., Tan C.H., Li L., Moore P.K., Deng L.W. The slow-releasing hydrogen sulfide donor, GYY4137, exhibits novel anti-cancer effects in vitro and in vivo. PLoS One. 2011;6:e21077.
    1. Lee C., Wan J., Miyazaki B., Fang Y., Guevara-Aguirre J., Yen K., Longo V., Bartke A., Cohen P. IGF-I regulates the age-dependent signaling peptide humanin. Aging Cell. 2014;13:958–961.
    1. Li W., Li X., Miller R.A. ATF4 activity: a common feature shared by many kinds of slow-aging mice. Aging Cell. 2014;13:1012–1018.
    1. Liang L., Zhou T., Jiang J., Pierce J.H., Gustafson T.A., Frank S.J. Insulin receptor substrate-1 enhances growth hormone-induced proliferation. Endocrinology. 1999;140:1972–1983.
    1. Liu H., Bravata D.M., Olkin I., Nayak S., Roberts B., Garber A.M., Hoffman A.R. Systematic review: the safety and efficacy of growth hormone in the healthy elderly. Ann. Intern. Med. 2007;146:104–115.
    1. Liu Y., Yang R., Liu X., Zhou Y., Qu C., Kikuiri T., Wang S., Zandi E., Du J., Ambudkar I.S. Hydrogen sulfide maintains mesenchymal stem cell function and bone homeostasis via regulation of Ca(2+) channel sulfhydration. Cell Stem Cell. 2014;15:66–78.
    1. Majtan T., Pey A.L., Fernández R., Fernández J.A., Martínez-Cruz L.A., Kraus J.P. Domain organization, catalysis and regulation of eukaryotic cystathionine beta-synthases. PLoS One. 2014;9:e105290.
    1. Miller D.L., Roth M.B. Hydrogen sulfide increases thermotolerance and lifespan in Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA. 2007;104:20618–20622.
    1. Miller R.A., Buehner G., Chang Y., Harper J.M., Sigler R., Smith-Wheelock M. Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell. 2005;4:119–125.
    1. Mistry R.K., Murray T.V., Prysyazhna O., Martin D., Burgoyne J.R., Santos C., Eaton P., Shah A.M., Brewer A.C. Transcriptional regulation of cystathionine-γ-lyase in endothelial cells by NADPH oxidase 4-dependent signaling. J. Biol. Chem. 2016;291:1774–1788.
    1. Mustafina A.N., Yakovlev A.V., Gaifullina ., Weiger T.M., Hermann A., Sitdikova G.F. Hydrogen sulfide induces hyperpolarization and decreases the exocytosis of secretory granules of rat GH3 pituitary tumor cells. Biochem. Biophys. Res. Commun. 2015;465:825–831.
    1. Nakano S., Ishii I., Shinmura K., Tamaki K., Hishiki T., Akahoshi N., Ida T., Nakanishi T., Kamata S., Kumagai Y. Hyperhomocysteinemia abrogates fasting-induced cardioprotection against ischemia/reperfusion by limiting bioavailability of hydrogen sulfide anions. J. Mol. Med. (Berl.) 2015;93:879–889.
    1. Olson K.R., Dombkowski R.A., Russell M.J., Doellman M.M., Head S.K., Whitfield N.L., Madden J.A. Hydrogen sulfide as an oxygen sensor/transducer in vertebrate hypoxic vasoconstriction and hypoxic vasodilation. J. Exp. Biol. 2006;209:4011–4023.
    1. Panici J.A., Harper J.M., Miller R.A., Bartke A., Spong A., Masternak M.M. Early life growth hormone treatment shortens longevity and decreases cellular stress resistance in long-lived mutant mice. FASEB J. 2010;24:5073–5079.
    1. Papapetropoulos A., Pyriochou A., Altaany Z., Yang G., Marazioti A., Zhou Z., Jeschke M.G., Branski L.K., Herndon D.N., Wang R. Hydrogen sulfide is an endogenous stimulator of angiogenesis. Proc. Natl. Acad. Sci. USA. 2009;106:21972–21977.
    1. Paul B.D., Sbodio J.I., Xu R., Vandiver M.S., Cha J.Y., Snowman A.M., Snyder S.H. Cystathionine γ-lyase deficiency mediates neurodegeneration in Huntington's disease. Nature. 2014;509:96–100.
    1. Ramadoss P., Abraham B.J., Tsai L., Zhou Y., Costa-e-Sousa R.H., Ye F., Bilban M., Zhao K., Hollenberg A.N. Novel mechanism of positive versus negative regulation by thyroid hormone receptor β1 (TRβ1) identified by genome-wide profiling of binding sites in mouse liver. J. Biol. Chem. 2014;289:1313–1328.
    1. Rozing M.P., Houwing-Duistermaat J.J., Slagboom P.E., Beekman M., Frölich M., de Craen A.J., Westendorp R.G., van Heemst D. Familial longevity is associated with decreased thyroid function. J. Clin. Endocrinol. Metab. 2010;95:4979–4984.
    1. Selman C., Lingard S., Choudhury A.I., Batterham R.L., Claret M., Clements M., Ramadani F., Okkenhaug K., Schuster E., Blanc E. Evidence for lifespan extension and delayed age-related biomarkers in insulin receptor substrate 1 null mice. FASEB J. 2008;22:807–818.
    1. Selman C., Partridge L., Withers D.J. Replication of extended lifespan phenotype in mice with deletion of insulin receptor substrate 1. PLoS One. 2011;6:e16144.
    1. Sikalidis A.K., Stipanuk M.H. Growing rats respond to a sulfur amino acid-deficient diet by phosphorylation of the alpha subunit of eukaryotic initiation factor 2 heterotrimeric complex and induction of adaptive components of the integrated stress response. J. Nutr. 2010;140:1080–1085.
    1. Sikalidis A.K., Lee J.I., Stipanuk M.H. Gene expression and integrated stress response in HepG2/C3A cells cultured in amino acid deficient medium. Amino Acids. 2011;41:159–171.
    1. Singha S., Kim D., Moon H., Wang T., Kim K.H., Shin Y.H., Jung J., Seo E., Lee S.J., Ahn K.H. Toward a selective, sensitive, fast-responsive, and biocompatible two-photon probe for hydrogen sulfide in live cells. Anal. Chem. 2015;87:1188–1195.
    1. Szabó C., Papapetropoulos A. Hydrogen sulphide and angiogenesis: mechanisms and applications. Br. J. Pharmacol. 2011;164:853–865.
    1. Tahiliani P., Kar A. The combined effects of Trigonella and Allium extracts in the regulation of hyperthyroidism in rats. Phytomedicine. 2003;10:665–668.
    1. Tsukada A., Ohkubo T., Sakaguchi K., Tanaka M., Nakashima K., Hayashida Y., Wakita M., Hoshino S. Thyroid hormones are involved in insulin-like growth factor-I (IGF-I) production by stimulating hepatic growth hormone receptor (GHR) gene expression in the chicken. Growth Horm. IGF Res. 1998;8:235–242.
    1. Vella K.R., Ramadoss P., Lam F.S., Harris J.C., Ye F.D., Same P.D., O'Neill N.F., Maratos-Flier E., Hollenberg A.N. NPY and MC4R signaling regulate thyroid hormone levels during fasting through both central and peripheral pathways. Cell Metab. 2011;14:780–790.
    1. Wang R. Physiological implications of hydrogen sulfide: a whiff exploration that blossomed. Physiol. Rev. 2012;92:791–896.
    1. Wang M., Miller R.A. Fibroblasts from long-lived mutant mice exhibit increased autophagy and lower TOR activity after nutrient deprivation or oxidative stress. Aging Cell. 2012;11:668–674.
    1. Yakar S., Liu J.L., Stannard B., Butler A., Accili D., Sauer B., LeRoith D. Normal growth and development in the absence of hepatic insulin-like growth factor I. Proc. Natl. Acad. Sci. USA. 1999;96:7324–7329.
    1. Yang W., Yang G., Jia X., Wu L., Wang R. Activation of KATP channels by H2S in rat insulin-secreting cells and the underlying mechanisms. J. Physiol. 2005;569:519–531.
    1. Yang G., Wu L., Jiang B., Yang W., Qi J., Cao K., Meng Q., Mustafa A.K., Mu W., Zhang S. H2S as a physiologic vasorelaxant: hypertension in mice with deletion of cystathionine gamma-lyase. Science. 2008;322:587–590.
    1. Yang L., Li P., Fu S., Calay E.S., Hotamisligil G.S. Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Cell Metab. 2010;11:467–478.
    1. Yuan M., Breitkopf S.B., Yang X., Asara J.M. A positive/negative ion-switching, targeted mass spectrometry-based metabolomics platform for bodily fluids, cells, and fresh and fixed tissue. Nat. Protoc. 2012;7:872–881.
    1. Zambrano A., García-Carpizo V., Gallardo M.E., Villamuera R., Gómez-Ferrería M.A., Pascual A., Buisine N., Sachs L.M., Garesse R., Aranda A. The thyroid hormone receptor β induces DNA damage and premature senescence. J. Cell Biol. 2014;204:129–146.
    1. Zanardo R.C., Brancaleone V., Distrutti E., Fiorucci S., Cirino G., Wallace J.L. Hydrogen sulfide is an endogenous modulator of leukocyte-mediated inflammation. FASEB J. 2006;20:2118–2120.
    1. Zhang Y., Xie Y., Berglund E.D., Coate K.C., He T.T., Katafuchi T., Xiao G., Potthoff M.J., Wei W., Wan Y. The starvation hormone, fibroblast growth factor-21, extends lifespan in mice. Elife. 2012;1:e00065.
    1. Zhao W., Zhang J., Lu Y., Wang R. The vasorelaxant effect of H(2)S as a novel endogenous gaseous K(ATP) channel opener. EMBO J. 2001;20:6008–6016.
    1. Zhao L., Zhu X., Won Park J., Fozzatti L., Willingham M., Cheng S.Y. Role of TSH in the spontaneous development of asymmetrical thyroid carcinoma in mice with a targeted mutation in a single allele of the thyroid hormone-β receptor. Endocrinology. 2012;153:5090–5100.
    1. Zhao K., Li H., Li S., Yang G. Regulation of cystathionine gamma-lyase/H2S system and its pathological implication. Front. Biosci. (Landmark Ed.) 2014;19:1355–1369.
    1. Zhou L., Ding S., Li Y., Wang L., Chen W., Bo T., Wu K., Li C., Liu X., Zhao J. Endoplasmic reticulum stress may play a pivotal role in lipid metabolic disorders in a novel mouse model of subclinical hypothyroidism. Sci. Rep. 2016;6:31381.

Source: PubMed

3
Předplatit