Altered methionine metabolism in long living Ames dwarf mice

https://doi.org/10.1016/S0531-5565(03)00008-1Get rights and content

Abstract

Ames dwarf mice (df/df) are deficient in growth hormone, prolactin, and thyroid-stimulating hormone and live significantly longer than their normal siblings. In the current study, we found that the hormone deficiencies affect methionine metabolism. We previously reported that the dwarf mice exhibit enzyme activities and levels that combat oxidative stress more efficiently than those of normal mice. Moreover, methionine or metabolites of methionine are involved in antioxidative processes. Thus, we performed an experiment that compared various parameters of methionine metabolism between 18-month old male dwarf (N=6) and wild type (N=5) mice. The specific activity of liver methionine adenosyltransferase (MAT) was significantly elevated (205%, p<0.0001) in the dwarf mice, as were cystathionine synthase (50%, p<0.01), cystathionase (83%, p<0.001), and glycine N-methyltransferase (GNMT, 91%, p<0.001) activities. Even though the activities of MAT and GNMT were elevated, the concentration of liver S-adenosylmethionine was decreased (24%, p<0.001) and S-adenosylhomocysteine increased (113%, p<0.001) in the dwarf mice. These data indicate that dwarf mice, compared to wild type mice, have a markedly different metabolism of methionine. Altered methionine metabolism may partially explain earlier reports indicating less oxidative damage to proteins in dwarf mice. Taken together, the data suggest that methionine metabolism may play a role in oxidative defense in the dwarf mouse and should be studied as a potential mechanism of extended lifespan.

Introduction

Long-lived Ames dwarf mice have a homozygous recessive mutation at the Prop-1 locus, which leads to a lack of differentiation of somatotrophic, lactotrophic, and thyrotrophic pituitary cells. As a result, these mice lack growth hormone (GH), prolactin, and thyroid stimulating hormone (Hauck et al., 2001). Recent studies by Brown-Borg (Brown-Borg et al., 1999, Brown-Borg and Rakoczy, 2000, Brown-Borg et al., 2001) have focused on the relevance of reduced GH signaling to longevity and lower oxidative stress in the dwarf mice. The significantly longer life span exhibited by these GH-deficient mice is contrasted with reduced life span and increased free radical processes in transgenic mice that overexpress GH (Rollo et al., 1996, Brown-Borg and Rakoczy, 2000). In further support of a role for GH in longevity determination, two additional mouse lines, the GH-receptor/binding protein knockout (Laron dwarf) mouse and a line of transgenic mice that overexpress a GH antagonist also live significantly longer than normal animals from the same strain (Bartke, 1998, Coschigano et al., 2000).

Brown-Borg reported that concentrations of hepatic inorganic peroxides and mitochondrial H2O2 were decreased and liver and kidney catalase were markedly increased in Ames dwarf mice (Brown-Borg et al., 1999, Brown-Borg and Rakoczy, 2000, Brown-Borg et al., 2001). Catalase activity and protein were significantly elevated in livers from dwarf mice at 3, 6, 13–15, and 24 months of age when compared to age-matched wild type mice. Kidneys from old dwarf mice exhibited significantly increased catalase activity (22%), protein (16%) and mRNA expression (59%) compared to wild type mice (Brown-Borg and Rakoczy, 2000). Hypothalamic catalase has also been found to be elevated in these mice (Hauck and Bartke, 2000). The results of these studies suggested that GH status modulates antioxidative mechanisms and that catalase is important in overall defense capacity with respect to lifespan in the Ames dwarf mouse.

Growth hormone can also affect other metabolic pathways including methionine metabolism (Fig. 1). For example, glycine N-methyltransferase (GNMT), which plays a crucial role in the regulation of tissue concentrations of S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH), is regulated by GH (Aida et al., 1997).

Methionine metabolism also changes with aging. With age, the activities of Methionine adenosyltransferase (MAT), betaine homocysteine methyltransferase (BHMT), and methionine synthase decrease with age while cystathionine synthase (CS) and cystathionase (CTH) increase (Finkelstein, 1962, Finkelstein et al., 1971). MAT also decreases in male rats after puberty (Oscarsson et al., 2001). The concentration of SAM in the lens decreases with increasing age (Geller et al., 1988). The concentration of SAH in rat liver and cerebral cortex increases while brain SAM decreases with age (Hoffman et al., 1979, Trolin et al., 1994). Therefore, the expression of metabolites in the methionine metabolic pathway is tissue and age dependent.

As mentioned, the Ames dwarf mouse is lacking in GH, prolactin and thyroid stimulating hormone. These mice also have decreased concentrations of glucose, insulin, and thyroid hormones, and a reduced core body temperature (Borg et al., 1995, Hunter et al., 1999). Hypothyroid rats have decreased total plasma homocysteine and increased hepatic activities of CS and CTH; thyroid hormone administration to thyroidectomized rats normalized the plasma homocysteine and the activities of the enzymes (Jacobs et al., 2000). Pan and Tarver (Pan and Tarver, 1967) showed that thyroidectomized rats have increased activities of liver MAT and that administration of bovine GH to hypophysectomized rats resulted in a decrease in the activity of the enzyme. They also reported that administration of T3 to normal rats resulted in a decreased activity of MAT. Keating et al. (Keating et al., 1988) also reported that the activity of MAT is elevated in hypothyroidism and that this resulted in an increased concentration of SAM.

Thus, the hormonal changes found in the Ames dwarf mouse may affect methionine metabolism. The objective of our studies was to determine whether or not methionine metabolism is indeed altered and if so, whether this may help explain the extended survival of these mice.

Section snippets

Animals

Ames dwarf and age-matched wild type mice were maintained at the University of North Dakota (UND) vivarium facilities under controlled conditions of photoperiod (12 h light:12 h dark) and temperature (22±1 °C) with free access to food (PMI Nutrition Intl., St. Louis, MO; Laboratory rodent diet) and water. The Ames dwarf (df/df) mice used in this study were derived from a closed colony with heterogeneous background (over 20 years). Dwarf mice were generated by mating either homozygous (df/df) or

Results

As expected, body weights of dwarf mice were significantly lower when compared to the wild type mice (15.8±2.3 vs 44.0±4.8 g; p<0.0001).

Liver SAM was significantly decreased (24%) and SAH considerably increased (113%) in the dwarf compared to wild type mice resulting in a marked decrease in SAM/SAH ratio (64%) in the dwarf (Fig. 2). S-adenosylhomocysteine was also increased (18%) in brain tissues of dwarf mice over that of wild type mice (P<0.02; Fig. 3). However, brain SAM and the SAM/SAH ratio

Discussion

The results show that Ames dwarf mice, which live 49–64% longer than their normal counterparts, have a markedly altered methionine metabolism (Fig. 1). We found a significant increase in the specific activity of liver GNMT in the dwarf mice compared to wild type. Aida et al. (Aida et al., 1997) showed that hypophysectomized mice had high expression of GNMT and that treatment with GH decreased this expression; however, no mechanism for regulation of GNMT by GH was proposed. The Ames dwarf mice

Acknowledgements

The authors thank Kim Baurichter, Tom Zimmerman, Rhonda Poellot, and Sharlene Rakoczy for technical assistance and Cindy Davis for the DNA methylation work.

References (45)

  • S.J. Hauck et al.

    Effects of growth hormone on hypothalamic catalase and Cu/Zn superoxide dismutase

    Free Radic. Biol. Med.

    (2000)
  • D.W. Horne et al.

    Effect of dietary methyl group deficiency on folate metabolism in rats

    J. Nutr.

    (1989)
  • W.S. Hunter et al.

    Low body temperature in long-lived Ames dwarf mice at rest and during stress

    Physiol. Behav.

    (1999)
  • J.N. Keating et al.

    Effect of thiouracil in modifying folate function in severe vitamin B12 deficiency

    Arch. Biochem. Biophys.

    (1988)
  • S.H. Mudd et al.

    Transsulfuration in mammals microassays and tissue distribution of three enzymes of the pathway

    J. Biol. Chem.

    (1965)
  • H. Ogawa et al.

    Structure, function and physiological role of glycine N-methyltransferase

    Int. J. Biochem. Cell Biol.

    (1998)
  • F. Pan et al.

    Effects of diet and other factors on methionine adenosyltransferase levels in rat liver

    J. Nutr.

    (1967)
  • M.H. Stipanuk

    Effect of excess dietary methionine on the catabolism of cysteine in rats

    J. Nutr.

    (1979)
  • M. Suda et al.
  • C.G. Trolin et al.

    Brain ATP: l-methionine S-adenosyltransferase (MAT), S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH): regional distribution and age-related changes

    Eur. Neuropsychopharmacol.

    (1994)
  • C. Wagner et al.

    Inhibition of glycine N-methyltransferase activity by folate derivatives: implications for regulation of methyl group metabolism

    Biochem. Biophys. Res. Com.

    (1985)
  • J. Wagner et al.

    A rapid high-performance liquid chromatographic procedure for the simultaneous determination of methionine, ethionine, S-adenosylmethionine, S-adenosylhomocysteine, and the natural polyamines in rat tissues

    Anal. Biochem.

    (1984)
  • Cited by (63)

    • Hydrogen sulfide in longevity and pathologies: Inconsistency is malodorous

      2021, Ageing Research Reviews
      Citation Excerpt :

      The flux of Met to the transsulfuration pathway is also enhanced in the Ames dwarf mice and Snell dwarf mice (Uthus and Brown-Borg, 2006; Vitvitsky et al., 2013). Compared to wild type, the Ames mice had increased expression of Met metabolism genes including CBS and CSE (Uthus and Brown-Borg, 2003, 2006). Compared to control mice the Snell mice exhibited higher CBS activity in the brain (∼1.4-fold) and female kidney (by 78 %) (Vitvitsky et al., 2013).

    • Functional changes of the liver in the absence of growth hormone (GH) action – Proteomic and metabolomic insights from a GH receptor deficient pig model

      2020, Molecular Metabolism
      Citation Excerpt :

      In contrast, the major hepatic SAM utilizing enzyme, glycine N-methyltransferase (GNMT), that converts SAM (along with glycine) to S-adenosyl-l-homocysteine and sarcosine, was significantly more abundant in the GHR-KO samples (l2fc 2.58, p = 0.0134). Notably, GH-deficient Ames dwarf mice revealed significantly increased MAT and GNMT levels [62] that were reduced upon GH treatment [63]. Increased GNMT levels in GHR-KO liver may decrease the availability of SAM and increase downstream products of the methionine cycle, such as homocysteine, cystathionine, and glutathione (reviewed in [64,65]).

    View all citing articles on Scopus

    Presented in part at American Aging Association (AGE) 2002, June 7-10, 2002, San Diego, CA [Uthus, E.O. and Brown-Borg, H.M (2002) Altered methionine metabolism in long living Ames dwarf mice. American Aging Association 31st Annual Meeting, p. 28]. The US Department of Agriculture, Agricultural Research Service, Northern Plains Area is an equal opportunity/affirmative action employer and all agency services are available without discrimination. Mention of a trademark or proprietary product does not constitute a guarantee of warranty of the product by the United States Department of Agriculture and does not imply its approval to the exclusion of other products that may also be suitable.

    View full text