Elsevier

Mechanisms of Ageing and Development

Volume 124, Issues 8–9, August–September 2003, Pages 941-949
Mechanisms of Ageing and Development

Analysis of the effect of aging on the response to hypoxia by cDNA microarray

https://doi.org/10.1016/S0047-6374(03)00166-0Get rights and content

Abstract

Little is known about the detail of hypoxia-responsive gene expression patterns in advanced age, even though aging is thought to be partially associated with a decreased response to hypoxia. In the present study, we identified several hypoxia-inducible genes and investigated the effect of aging on hypoxic gene expression profiles using cDNA microarray analysis of young/old human diploid fibroblasts. Of 7458 genes in the microarray, we found that genes involved in angiogenesis, defense against oxidative stress, and transcription regulation are severely impaired in senescent cell, which is consistent with the fact that aged cells have attenuated responses to various stimuli.

Introduction

Aging is characterized by a deleterious and progressive decline in physiological efficiency. Since multiple factors are involved in the aging progress, it is still difficult to collectively understand the global network involved. Nonetheless, it is known that the aging process primarily occurs due to an altered gene expression by the loss or the altered activities of gene regulatory molecules (Roy et al., 2002). In particular, an aged organism is susceptible to various age-related diseases including vascular diseases and shows impaired angiogenesis under hypoxic conditions, which result from the reduced induction of vascular endothelial growth factor (VEGF) (Rivard et al., 1999), a potent hypoxia-targeted angiogenesis stimulator (Shweiki et al., 1992).

Hypoxia, a limited cellular oxygen tension, is important for the regulation of the expressions of essential genes under specific developmental or physiological conditions: energy metabolism, proliferation, survival/apoptosis, erythropoiesis and angiogenesis (Semenza, 2001). Hypoxia-inducible factor-1 (HIF-1) is a basic–helix–loop–helix protein and forms heterodimer composed of a constitutively expressed HIF-1β subunit and an O2-regulated HIF-1α subunit (Wang et al., 1995). In the normoxic cells, HIF-1α protein, which is hydroxylated at proline residues in its oxygen-dependent degradation (ODD) domain by O2-regulated prolyl hydroxylases, is subjected to ubiquitination and subsequent proteosomal degradation by von Hippel–Lindau (VHL) tumor suppressor protein. However, under hypoxic conditions, HIF-1α escapes from the degradation pathway, and forms a heterodimeric active complex that binds to hypoxia-responsive recognition element (HRE), which leads to the induction of hypoxia-responsive genes such as erythropoietin (EPO), VEGF, and the genes required for metabolic adaptation to an oxygen-limited environment (Semenza, 2001, Wenger, 2002).

Recent reports have suggested that the DNA binding activity of HIF-1α declines in old age, and that this is responsible for reduced VEGF expression and impaired angiogenesis (Frenkel-Denkberg et al., 1999). In addition, it is well known that the metabolic rate in advanced age is gradually reduced. Given that the unique characteristics of impaired angiogenesis and a slow rate of metabolic uptake in an aged organism appear to be deeply involved in the inefficiency of gene regulation under hypoxic conditions, it becomes apparent that aging exerts an influence on global gene expression patterns of hypoxia-targeted genes.

cDNA microarrays becomes a growing technology, which can be used to better understand the global networking of individual genes. Thus, in this study, we used a cDNA microarray to identify hypoxia-related genes expressed in an age-dependent fashion, and by analyzing gene expression profile, we attempt to explain the background of the impaired biological function of old organisms in hypoxia.

Here, we demonstrate that the inductions of genes, responsible for adaptation to hypoxia, are markedly declined in old human diploid fibroblasts (HDFs). Especially, those genes related to angiogenesis such as VEGF, PGAR, thromboplastin and PAI-1 and to those related to oxidative stress like heme oxygenase 1 (HO-1). These genes were found to be differentially expressed in ‘old’ and ‘young’ HDFs, and could be responsible for the impaired angiogenesis and susceptibility to ischemic injury observed in old age. In addition, we report, for the first time, that insulin induced gene 1 (INSIG-1), and bagpipe homeobox homologue 1 (BAPX1) are hypoxia-inducible genes.

Section snippets

Cell culture and hypoxic exposure

HDFs were obtained from newborn foreskins, as described by Boyce and Ham (1983), and maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum and 50 μU of penicillin/streptomycin. Cells with mean population doublings (MPDs) of less than 25 were used as young cells and the cells with MPDs of 65–70 MPD as old cells. Senescent cells were characterized by morphological changes, enhanced β-galactosidase activity, and a reduced rate of proliferation (Yeo et

General presentation of the results

To examine the aging effect on hypoxia-responsive gene expression, we performed cDNA microarray analysis on young and old HDFs (Fig. 1A). EST clones including 215 unassigned ESTs and the 20 ESTs of putative genes, we selected genes showing significant fold-differences between young and old HDFs. The expression fold of genes under hypoxia was plotted (Fig. 1B). The region A includes genes induced by hypoxia in both young and old HDFs and region B includes genes induced preferentially in young

Discussion

Under hypoxic conditions, organisms respond to maintain oxygen homeostasis by increasing oxygen delivery or by providing a metabolic adaptation, and by regulating cellular growth. Moreover, these responses are facilitated at the transcription level and are followed by posttranslational modification. In this study, we investigated the aging effect on the response to hypoxia by comparing the differential gene expressions of young and old HDFs upon exposure to hypoxia.

Of 7458 human ESTs examined,

Acknowledgements

This work was supported by Korean Research Foundation (KRF-2001-015-FP0042).

References (36)

  • S.F. Yan et al.

    Induction of interleukin 6 (IL-6) by hypoxia in vascular cells. Central role of the binding site for nuclear factor-IL-6

    J. Biol. Chem.

    (1995)
  • S.F. Yan et al.

    Hypoxia-associated induction of early growth response-1 gene expression

    J. Biol. Chem.

    (1999)
  • E.J. Yeo et al.

    Reduction of UV-induced cell death in the human senescent fibroblasts

    Mol. Cells

    (2000)
  • W.G. An et al.

    Stabilization of wild-type p53 by hypoxia-inducible factor 1α

    Nature

    (1998)
  • D. Bandyopadhyay et al.

    Down-regulation of p300/CBP histone acetyltransferase activates a senescence checkpoint in human melanocytes

    Cancer Res.

    (2002)
  • S.T. Boyce et al.

    Calcium-regulated differentiation of normal human epidermal keratinocytes in chemically defined clonal culture and serum-free serial culture

    J. Invest. Dermatol.

    (1983)
  • T.A. Chan et al.

    14-3-3σ is required to prevent mitotic catastrophe after DNA damage

    Nature

    (1999)
  • G. Deng et al.

    Is plasminogen activator inhibitor-1 the molecular switch that governs urokinase receptor-mediated cell adhesion and release

    J. Cell Biol.

    (1996)
  • Cited by (26)

    • Hypoxia and brain aging: Neurodegeneration or neuroprotection?

      2021, Ageing Research Reviews
      Citation Excerpt :

      HIF-1 has been identified as a potent modulator of aging in C. elegans (Leiser and Kaeberlein, 2010), an effect mediated by acyl coenzyme A binding proteins (Shamalnasab et al., 2017), and dependent on temperature (Leiser et al., 2011) and oxidative stress (Hwang and Lee, 2011). Moreover, Kim et al. (Kim et al., 2003) demonstrated impaired HIF-1 binding to HREs in aged human diploid fibroblasts. Diminished HIF-1 function also was demonstrated in aging mice (Frenkel-Denkberg et al., 1999).

    • Beaver and Naked Mole Rat Genomes Reveal Common Paths to Longevity

      2020, Cell Reports
      Citation Excerpt :

      One of the candidate genes we identified was PTGIS, which harbored a disease-causing substitution for hypertension (R26Q) that was unique to the beaver (Figure S3). This gene encodes prostacyclin, which plays roles in cardiac function, response to oxidative stress, and hypoxia (Kim et al., 2003; Nakayama et al., 2002). It may have evolved as a diving adaptation, because beavers, like other diving mammals, are known to optimize respiration and cardiac function for staying longer under water (Dolka et al., 2015; Irving and Orr, 1935).

    • Modeling colon adenocarcinomas in vitro: A 3D co-culture system induces cancer-relevant pathways upon tumor cell and stromal fibroblast interaction

      2011, American Journal of Pathology
      Citation Excerpt :

      We used two gene set collections from the Molecular Signature Database (Broad Institute48): the c2 curated gene sets and the c5 GO gene sets. Among the data sets most significantly enriched in the co-cultures were gene sets involved in hypoxia, ras signaling, inflammation, ECM deposition, angiogenesis, tissue development, wound healing, and epithelial mesenchymal transition (EMT) (Table 149–61; see also Supplemental Table S2 at http://ajp.amjpathol.org). The core genes of statistically significant gene sets were used to calculate PCA plots.

    • Fibroblast dysfunction is a key factor in the non-healing of chronic venous leg ulcers

      2008, Journal of Investigative Dermatology
      Citation Excerpt :

      As the present data implies that there is a lack of major enzymic antioxidant induction in CWF, this suggests that the innate mechanisms of protecting against oxidative stress are defective. Previous studies have reported either a delayed antioxidant induction in fibroblasts until premature senescence is fully reached (Chen et al., 2004) or the severely impaired induction of antioxidant defense genes in senescent fibroblast populations (Kim et al., 2003), following oxidative stress exposure. Consequently, these findings would explain the increased susceptibility of CWF to oxidative stress and telomere-independent senescence within the chronic wound environment.

    • Hypoxia stimulates degradation of 3-hydroxy-3-methylglutaryl-coenzyme A reductase through accumulation of lanosterol and hypoxia-inducible factor-mediated induction of insigs

      2007, Journal of Biological Chemistry
      Citation Excerpt :

      In several DNA microarray analyses, transcripts for Insig-1 and/or Insig-2 have been identified among those induced by hypoxia or DMOG treatment (54-57). Importantly, induction of Insig-1 in hypoxic cells was verified in one study by Northern blot analysis (54). The quantitative real-time PCR experiment of Fig. 7A shows a 2-3-fold induction of Insig-1 and Insig-2 mRNAs in C4.5 cells following 12 h of hypoxia or DMOG treatment.

    View all citing articles on Scopus
    View full text