Elsevier

Molecular Genetics and Metabolism

Volume 92, Issues 1–2, September–October 2007, Pages 78-87
Molecular Genetics and Metabolism

Involvement of endoplasmic reticulum stress in a novel Classic Galactosemia model

https://doi.org/10.1016/j.ymgme.2007.06.005Get rights and content

Abstract

Inherited deficiency of galactose-1-phosphate uridyltransferase (GALT) activity in humans leads to a potentially lethal disorder called Classic Galactosemia. It is well known that patients often accumulate high levels of galactose metabolites such as galactose-1-phosphate (gal-1-p) in their tissues. However, specific targets of gal-1-p and other accumulated metabolites remain uncertain. In this study, we developed a new model system to study this toxicity using primary fibroblasts derived from galactosemic patients. GALT activity was reconstituted in these primary cells through lentivirus-mediated gene transfer. Gene expression profiling showed that GALT-deficient cells, but not normal cells, responded to galactose challenge by activating a set of genes characteristic of endoplasmic reticulum (ER) stress. Western blot analysis showed that the master regulator of ER stress, BiP, was up-regulated at least threefold in these cells upon galactose challenge. We also found that treatment of these cells with galactose, but not glucose or hexose-free media reduced Ca2+ mobilization in response to activation of Gq-coupled receptors. To explore whether the muted Ca2+ mobilization is related to reduced inositol turnover, we discovered that gal-1-p competitively inhibited human inositol monophosphatase (hIMPase1). We hypothesize that galactose intoxication under GALT-deficiency resulted from accumulation of toxic galactose metabolite products, which led to the accumulation of unfolded proteins, altered calcium homeostasis, and subsequently ER stress.

Introduction

Classic Galactosemia (G/G) is an autosomal recessive, multi-system disorder caused by deleterious mutations of the human galactose-1-phosphate uridyltransferase (GALT) gene, which results in inability to metabolize galactose [1], [2], [3] (Fig. 1). Although a galactose-restricted diet prevents the neonatal lethality of this disorder, many well-treated patients continue to develop debilitating complications such as premature ovarian failure (POF), mental retardation, and some neurological defects [4], [5]. The causes of these unsatisfactory outcomes remain unclear. Recent studies, however, showed that patients placed on a galactose-restricted diet are never truly free of galactose insult, as a significant amount of galactose is found in non-dairy foodstuffs such as vegetables and fruits [6], [7]. More importantly, galactose moieties can be produced endogenously from UDP-glucose via the UDP-4-galactose epimerase (GALE) reaction (Fig. 1), and natural turnover of glycoproteins/glycolipids. In fact, using isotopic labeling, Berry et al. elegantly demonstrated that a 50 kg adult male could produce up to 2 g of galactose per day [8], [9]. Once the galactose is formed intracellularly, it will be converted to galactose-1-phosphate (gal-1-p) by galactokinase (GALK).

Paradoxically, patients with an inherited deficiency of galactokinase (GALK) do not manifest either the acute toxicity syndrome or chronic complications seen in galactosemic patients [3]. Since GALK-deficient patients do not accumulate gal-1-p in their tissues [3], [10], researchers have long proposed that gal-1-p plays a significant role in the pathogenesis of Classic Galactosemia [11], [12], [13], [14]. Yet the in vivo targets of this presumably toxic intermediate have never been identified. Concurrently, the lack of overt galactose toxicity in the GALT-knockout mouse model has continued to hamper the efforts to delineate the molecular mechanism of this disease [15], [16], [17].

In this study, we developed a novel isogenic human cell model of GALT-deficiency, and showed that galactose challenge of these cells resulted in accumulation of gal-1-p and distinct signs of endoplasmic reticulum (ER) stress. Thus, our findings not only shed light on the pathogenic mechanisms of Classic Galactosemia, but also expanded the contemporary metabolomics model that depicted the fundamental genetic/physical interactions between galactose utilization and other metabolic pathways [18].

Section snippets

Cell lines and culture conditions

Three primary human fibroblasts cell lines derived from galactosemic patients were obtained from Coriell Cell Repositories (Camden, NJ, USA): GM01703, GM01417, and GM00528. All three lines were confirmed by DNA sequencing to be homozygous for the Q188R mutation in the GALT gene, which results in complete loss of enzyme activity [19], [20], [21]. For routine maintenance and propagation, the cells were grown in high glucose Dulbecco’s minimum Eagle’s medium (DMEM) (Gibco 11995-073) supplemented

Reconstitution of GALT activity in GALT-deficient primary fibroblasts prevented galactose toxicity

The lack of explicit galactose toxicity in the GALT-knockout mouse model [15], [16], [17] has prompted us to focus on cell models of human origin. In this study, we examined how GALT-deficient and GALT-positive cells behave differently in the presence of galactose. To rule out variation in genetic background between cells derived from healthy and galactosemic individuals, we reconstituted GALT enzyme activity in the fibroblasts derived from the same galactosemic patients using a lentiviral

Discussion

Despite the advances made in the biochemical characterization of Classic Galactosemia and the molecular/structural biology of the human GALT gene in the past four decades [3], [20], [41], [42], the pathogenesis of this disease remains largely unknown and the long-term outcome continues to be poor [4], [5], [43]. Faced with the lack of overt galactose toxicity in the GALT-knockout mouse model [15], [16], [17], researchers nonetheless proposed that galactose-1-phosphate (gal-1-p) plays a major

Acknowledgments

The authors thank Dr. Louis J. Elsas, Dr. Steve Roper, Simone Sandiford, Alejandro Ocampo, Helene Klapper, and the staff of University of Miami Microarray Facility for their valuable consultation. Grant support to K.L.: NIH Grant 1R01 HD054744, American Heart Association South-East Affiliate Scientist Development Grant No. 0435267B, The Woman’s Cancer Association of The University of Miami. Grant support to V.Z.S.: NIH Grant GM 060019 and American Heart Association South-East Affiliate

References (60)

  • S. Draghici

    Global functional profiling of gene expression

    Genomics

    (2003)
  • J.S. Cox et al.

    A novel mechanism for regulating activity of a transcription factor that controls the unfolded protein response

    Cell

    (1996)
  • M. Schroder et al.

    ER stress and the unfolded protein response

    Mutat. Res.

    (2005)
  • K.K. Eriksson

    EDEM contributes to maintenance of protein folding efficiency and secretory capacity

    J. Biol. Chem.

    (2004)
  • D.A. Applegarth

    Study of a family with Los Angeles, Duarte, and classical galactosemia variants of galactose-1-phosphate uridyl transferase

    Biochem. Med.

    (1976)
  • E. Little

    Induction of glucose-regulated protein (glucose-regulated protein 78/BiP and glucose-regulated protein 94) and heat shock protein 70 transcripts in the immature rat brain following status epilepticus

    Neuroscience

    (1996)
  • J.B. Tillman

    Structure and regulation of the mouse GRP78 (BiP) promoter by glucose and calcium ionophore

    Gene

    (1995)
  • M. Hashii et al.

    Bradykinin-induced cytosolic Ca2+ oscillations and inositol tetrakisphosphate-induced Ca2+ influx in voltage-clamped ras-transformed NIH/3T3 fibroblasts

    J. Biol. Chem.

    (1993)
  • R. Parthasarathy et al.

    Brain inositol monophosphatase identified as a galactose 1-phosphatase

    Brain Res.

    (1997)
  • R. Gitzelmann et al.

    Galactitol and galactose-1-phosphate in the lens of a galactosemic infant

    Exp. Eye Res.

    (1967)
  • R. Gitzelmann

    Deficiency of erythrocyte galactokinase in a patient with galactose diabetes

    Lancet

    (1965)
  • T. Slepak

    Intracellular galactose-1-phosphate accumulation leads to environmental stress response in yeast model

    Mol. Genet. Metab.

    (2005)
  • J. Jaeken et al.

    Serum lysosomal enzyme abnormalities in galactosaemia

    Lancet

    (1992)
  • K.J. Travers

    Functional and genomic analyses reveal an essential coordination between the unfolded protein response and ER-associated degradation

    Cell

    (2000)
  • J.P. Navarro-Avino et al.

    Yeast inositol mono- and trisphosphate levels are modulated by inositol monophosphatase activity and nutrients

    Biochem. Biophys. Res. Commun.

    (2003)
  • M.J. Berridge et al.

    Neural and developmental actions of lithium: a unifying hypothesis

    Cell

    (1989)
  • K.J. Isselbacher

    Congenital galactosemia, a single enzymatic block in galactose metabolism

    Science

    (1956)
  • S. Segal et al.

    Disorders of galactose metabolism

  • D. Waggoner et al.

    Long-term prognosis in Galactosemia: results of a survey of 350 cases

    Journal of Inherited Metabolic Disorders

    (1990)
  • D. Waggoner et al.

    Long-term complications in treated galactosemia—175 U.S. cases

    Int. Pediatr.

    (1993)
  • Cited by (0)

    View full text