ArticlePDF Available

Down-Regulation of the Expression of the FIH1 and ARD1 Genes at the Transcriptional Level by Nickel and Cobalt in the Human Lung Adenocarcinoma A549 Cell Line

MDPI
International Journal of Environmental Research and Public Health (IJERPH)
Authors:

Abstract and Figures

Although nickel and cobalt compounds have been known to cause induction of the transcription factor hypoxia-inducible factor 1 (HIF-1) and activation of a battery of hypoxia-inducible genes in the cell, the molecular mechanisms of this induction remain unclear. The post-translational modification of HIF-1a, the oxygen-sensitive subunit of HIF-1, regulates stabilization, nuclear translocation, DNA binding activity, and transcriptional activity of the protein. Among the enzymes regulating the post-translational modification of HIF-1a, the factor inhibiting HIF-1 (FIH-1) hydroxylates the protein at asparagine 803, suppressing the interaction of HIF-1a with transcription coactivators p300/CBP and reducing the transcriptional activity of the protein. ARD-1, the acetyltransferase, acetylates HIF-1a at lysine 532, which enhances the interaction of HIF-1a with pVHL. Therefore, FIH-1 and ARD-1 negatively regulate the transcriptional activity and the stability of HIF-1a. We examined the mRNA levels of FIH-1 and ARD-1 genes after exposure nickel (II) or cobalt (II) to the cell and found that both genes were down-regulated by the chemical treatment, which may lead to reduced levels of both proteins and result in increased level of HIF-1a and its transcriptional activity.
Content may be subject to copyright.
Int. J. Environ. Res. Public Health 2005, 2(1), 10–13
International Journal of
Environmental Research and Public Health
ISSN 1660-4601
www.ijerph.org
© 2005 by MDPI
© 2005 MDPI. All rights reserved.
Down-Regulation of the Expression of the FIH-1 and ARD-1 Genes at
the Transcriptional Level by Nickel and Cobalt in the Human Lung
Adenocarcinoma A549 Cell Line
Qingdong Ke
1
, Thomas Kluz
1
, and Max Costa
1*
1
Nelson Institute of Environmental Medicine, New York University, School of Medicine, 57 Old Forge Road, Tuxedo,
New York 10987, USA
*Correspondence to Dr. Max Costa. E-mail: Costam01@nyu.edu
Received: 15 November 2004 / Accepted: 06 February 2005 / Published: 30 April 2005
Abstract: Although nickel and cobalt compounds have been known to cause induction of the transcription factor
hypoxia-inducible factor 1 (HIF-1) and activation of a battery of hypoxia-inducible genes in the cell, the
molecular mechanisms of this induction remain unclear. The post-translational modification of HIF-1a, the
oxygen-sensitive subunit of HIF-1, regulates stabilization, nuclear translocation, DNA binding activity, and
transcriptional activity of the protein. Among the enzymes regulating the post-translational modification of HIF-
1a, the factor inhibiting HIF-1 (FIH-1) hydroxylates the protein at asparagine 803, suppressing the interaction of
HIF-1a with transcription coactivators p300/CBP and reducing the transcriptional activity of the protein. ARD-1,
the acetyltransferase, acetylates HIF-1a at lysine 532, which enhances the interaction of HIF-1a with pVHL.
Therefore, FIH-1 and ARD-1 negatively regulate the transcriptional activity and the stability of HIF-1a. We
examined the mRNA levels of FIH-1 and ARD-1 genes after exposure nickel (II) or cobalt (II) to the cell and
found that both genes were down-regulated by the chemical treatment, which may lead to reduced levels of both
proteins and result in increased level of HIF-1a and its transcriptional activity.
Keywords: nickel, cobalt, hypoxia, HIF-1a, FIH-1, ARD-1
Introduction
According to the International Agency for Research
on Cancer (IARC 1990), both soluble and insoluble
nickel compounds have long been established as human
and animal carcinogens [1]. And cobalt compounds are
carcinogenic in animals [2]. Epidemiological studies
have shown that environmental or occupational exposure
to nickel or cobalt compounds could cause lung and
nasal cancers, asthma, fibrosis, pneumonitis and some
other lung injuries [1-5]. Despite the various differences
among the compounds of these two metals regarding the
biochemical and molecular mechanisms of their toxicity
and carcinogenicity, they mimic hypoxia to induce the
HIF-1a transcription factor and hypoxia-inducible genes
[5, 6], which are believed to play important roles in
carcinogenesis. However, the complete mechanisms by
which nickel and cobalt compounds induce HIF-1a are
still unknown, although several sites of their impact on
HIF-1a have been described [7-9].
The transcription factor hypoxia-inducible factor 1
(HIF-1) plays an essential role in cellular oxygen
homeostasis [10, 11]. HIF-1 is a heterodimeric complex
composed of alpha and beta two subunits [12]. The beta
subunit is also the heterodimerization partner for the aryl
hydrocarbon receptor (AhR) and thereby called aryl
hydrocarbon receptor nuclear translocator (ARNT) that
is constitutively expressed; whereas the alpha subunit
(HIF-1a) is highly oxygen-sensitive and is rarely
detectable under normal oxygen tension but is
dramatically induced with hypoxia [12]. Under reduced
oxygen tension, HIF-1a is stabilized and translocates to
the nucleus, where it dimerizes with ARNT. Then the
active HIF-1 stimulates the transcription of genes
involved in angiogenesis, cell survival, glucose transport
and metabolism [13, 14].
HIF-1a is regulated by a reduced oxygen level largely
at its post-translational modifications, resulting in
stabilization, nuclear translocation, DNA binding
activity, and transcriptional activity of the protein. The
Int. J. Environ. Res. Public Health 2005, 2(1)
11
post-translational modifications of HIF-1a include prolyl
hydroxylation at proline 402 and 564 within the oxygen-
dependent degradation (ODD) domain by HIF-prolyl
hydroxylases (HPHs) [9, 15-17], asparaginyl hydroxylation
at asparagine 803 in the C-terminal activation domain
(C-TAD) by factor inhibiting HIF-1 (FIH-1) [8, 18, 19],
acetylation of lysine 532 in the ODD domain by an
acetyltransferase ARD-1[20], phosphorylation induced
by p42/p44 mitogen-activated protein kinase (MAPK)
activity [21], as well as ubiquitination by the von Hippel-
lindau (pVHL) complex [22-25].
In normoxia, proline 402 and 564 of HIF-1a are
hydroxylated, which is required for the binding of pVHL
complex and leads to the ubiquitination of the protein,
resulting in targeting of HIF-1a for proteasomal
degradation [15-17, 24]. The interaction of HIF-1a with
pVHL is enhanced by ARD-1-mediated acetylation at
lysine 532 [20]. The acetylation of this lysine residue by
ARD-1 is critical to the proteasomal degradation of HIF-
1a since a mutant with arginine substituting lysine 532
shown no acetylation by ARD-1 was stabilized and had a
decreased interaction with pVHL [20]. At the same time,
hydroxylation of asparagine 803 during normoxia
suppresses interaction of HIF-1a CAD with transcription
coactivators p300/CBP and reduces the transcriptional
activity of the protein [18, 26]. In addition to interacting
with HIF-1a, the asparaginyl hydroxylase FIH-1 also
interacts with pVHL, allowing the formation of
complexes containing HIF-1a, FIH-1, and pVHL [27].
In hypoxia, decreased level of prolyl hydroxylation
due to the limiting oxygen prevents pVHL binding to
HIF-1a, resulting in rapid accumulation of HIF-1a
protein [15, 16, 24]. The acetylation level of HIF-1
gradually decreases as the length of hypoxic exposure
time increases, which is due to the reduced expression of
ARD-1[20]. Meanwhile, stabilized HIF-1a protein is
able to bind to p300/CBP to execute its transcriptional
activity due to decreased level of hydroxylation at
asparagine 803[18, 19]. In addition, during hypoxia,
p42/p44 MAPK activity induces phosphorylation of
HIF-1a and promotes its transcriptional activity [21]. As
a consequence, HIF-1a accumulates and promotes
hypoxic tolerance by activating gene transcription.
Among the enzymes that regulate HIF-1a, prolyl
hydroxylases (HPHs) and asparaginyl hydroxylase (FIH-1)
belong to the family of 2-oxoglutarate-dependent
dioxygenases and require Fe2+, 2-oxoglutarate, O2, and
ascorbate for their reactions [8, 15, 16]. ARD-1
acetyltransferase acetylates HIF-1a by transferring an acetyl
group from acetyl-CoA [20].
Nickel (II) and cobalt (II) exposure in the presence of
oxygen causes accumulation of HIF-1a protein and
induction of its transcriptional activity [28-30], in part
resulting from the inability of HIF-1a binding to pVHL
due to the inhibition of HIF-1a hydroxylation by the
metals [24, 31]. Furthermore, Cobalt (II) has been shown
to inhibit activities of recombinant asparaginyl and
prolyl hydroxylase in vitro [8, 9].
In this study, we have examined the effects of nickel
(II) and cobalt (II) on the gene expression of FIH-1 and
ARD-1 using reverse transcriptase PCR (RT-PCR). Both
genes were down-regulated in the metal-exposed cells,
which might lead to reduced level of both proteins and
result in increased level of HIF-1a and its transcriptional
activity.
Materials and Methods
Cell Culture
Human lung adenocarcinoma A549 cells (CCL185)
were purchased from American Type Culture Collection
(Manassas, VA). Cells were maintained in F-12K
medium (Life
Technologies, Inc., Gaithersburg, MD)
supplemented with 10% fetal
bovine serum and 1%
penicillin/streptomycin (equivalent to 100 units/ml
and
100 µg/ml, respectively) at 37°C as monolayers
in a
humidified atmosphere containing 5% CO
2
.
Chemicals and Cell Exposure
Nickel chloride hexahydrate and cobalt chloride
hexahydrate were purchased from Sigma (St. Louis,
MO). Cells were seeded into 100-mm dishes and
allowed to attach overnight. When cells reached 70–80%
confluence, nickel chloride hexahydrate (0.5 mM,
1.0mM), or cobalt chloride hexahydrate (0.2mM, 0.4
mM) was added to the medium for 24h.
RNA Extraction and RT-PCR
At the end of the treatment, total RNA was isolated
from the cell using the Trizol reagent (Invitrogen,
Carlsbad, CA). The mRNA was isolated using Oligotex
kit (Qiagen, Germany). Reverse transcription was carried
out with SuperScript first-Strand Synthesis System for
RT–PCR (Invitrogen) and 1 µg of mRNA was used for
first-strand cDNA synthesis according to the
manufacturer’s protocol. The PCR was carried out in a
total volume of 50µl and 1µl of first-strand cDNA was
used for amplifying genes. The primers used for the
PCRs were: FIH-1 forward primer,
5'-
GCCAGCACCCACAAGTTCTT-3'; FIH-1 reverse primer,
5'-CCTGTTGGACCTCGGCTTAA-3' ARD-1 forward
primer, 5’-TGGGGTGAGGAGGGGATGG-3’; ARD-1
reverse primer, 5’-GGGAAGATTGTGGGGTATG-3’.
Primers for amplifying GAPDH gene were purchased from
BD Biosciences Clontech. Amplification conditions for
FIH-1 and ARD-1 genes were 95°C for 2 min, 22 cycles
at 95°C for 45 s, 57°C for 45 s, 72°C for 1 min, and
72°C for 5 min; the same for GAPDH gene except that
16 cycles were performed. PCR products were then
resolved on 1.5% agarose gels containing Ethidium
bromide.
Results
Nickel (II) and Cobalt (II) Down-Regulate the Gene
Expression of FIH-1and ARD-1 Genes
The investigation on the mechanisms of the induction
of HIF-1a by nickel (II) and cobalt (II) has been focused
on the effects of the metals on the enzyme activities of
HPHs and FIH-1. Besides the possible effects of the
metals on the enzyme activities, nickel (II) and cobalt
(II) might affect the gene expression of the enzymes, so
Int. J. Environ. Res. Public Health 2005, 2(1)
12
that the levels of the enzymes in the cell could be
affected. Therefore, we examined mRNA levels of FIH-
1 and ARD-1 by exposure A549 cells to nickel (II) and
cobalt (II). As shown in figure 1, nickel (II) and cobalt
(II) decreased FIH-1 and ARD-1 mRNA levels in a
dose-dependent manner. GAPDH gene served as an
internal control. The house keep gene 60S acidic
ribosomal protein has also been used for measuring the
loading control, which gave the same pattern as that of
GAPDH gene (data not shown).
Figure 1: Nickel (II) and Cobalt (II) down-regulate
mRNA levels of FIH-1 and ARD-1genes in A549 cells
A549 cells were treated with NiCl
2
·6H
2
O (0.5mM and 1
mM) or CoCl
2
·6H
2
O (0.2 mM and 0.4mM) for 24 h. The
cells were harvested at the end of the treatment and the
mRNA was isolated. The FIH-1, ARD-1, and GAPDH
genes were amplified by RT-PCR.
Discussion
Nickel (II) and cobalt (II) have long been known to
induce hypoxia-like stress by activating HIF-1a [28-30].
However, the mechanisms of this induction are still
unclear, although several studies have shown effects at
sites of HIF-1a regulation, particularly focusing on the
effects of the metals on the hydroxylases activities[7-9].
Since the HIF-1a hydroxylases require iron (II) as a co-
factor for their activities and iron, cobalt, and nickel are
adjacent in the transition metal group, it has been
suggested that nickel (II) and cobalt (II) may substitute
for the iron (II) in the hydroxylases, thereby, causing the
loss of the enzymatic activity[7]. Unfortunately, there is
no direct evidence to support this hypothesis. Recently,
the cellular ascorbate, another co-factor required for the
hydroxylases activities, has been shown to be depleted
by both metals. Since the role of ascorbate is maintaining
iron in its reduced state (iron II), this depletion may
favor enzyme-bound iron oxidation, which may lead to
the inactivation of the hydroxylases[31].
Besides their effects, direct or indirect, on the
hydroxylases activities, nickel (II) and cobalt (II) are
very likely to induce HIF-1a at additional sites. We
demonstrated here that the mRNA levels of FIH-1 and
ARD-1 genes were down-regulated by both metals,
which could result in reduced levels of the protein
products of these genes. Since both FIH-1 and ARD-1
proteins negatively regulate HIF-1a, decreasing of them
would lead to accumulation of HIF-1a and increase of its
transcriptional activity.
Furthermore, we suspect that nickel (II) or cobalt (II)
may affect the acetylation of HIF-1a not only by down-
regulating the acetyltransferase ARD-1, but also by
decreasing the level of the cellular acetyl-CoA, the
supply of the acetyl group, by the way of shutting down
the Kreb cycle as well as increasing the ratio of
NAD/NADH thereby causing the deacetylation of
histones. Since both nickel and cobalt have previously
been shown to inhibit histone acetylation and increase
the methylation of histone H3 at lysine 9, this effect may
play a role in the down-regulation of these two important
genes which impact negatively upon HIF-1a activation
(Costa et al. Mutation Res. in press).
Our study revealed a possible new mechanism of the
nickel- and cobalt- induction of hypoxia-like stress in the
cell, contributing insight to better understanding of their
carcinogenesis.
References
1. IARC, IARC Monographs on the Evaluation of
Carcinogenic Risks to Humans, Chromium, Nickel
and Welding, 1990, 49, p. 677-691.
2. Natl. Toxicol. Program. Tech. Rep. Ser, 1998, 471,
1-268.
3. Costa, M.: Mechanism of nickel genotoxicity and
carcinogenicity. 1996, 245-251.
4. Kelleher, P.; Pacheco, K.; Newman, L. S.: Inorganic
dust pneumonias: the metal-related parenchymal
disorders. Environ Health Perspect, 2000,108 Suppl
4, 685-96.
5. Huang, L. E.; Ho, V.; Arany, Z.; Krainc, D.; Galson,
D.; Tendler, D.; Livingston, D. M.; Bunn, H. F.:
Erythropoietin gene regulation depends on heme-
dependent oxygen sensing and assembly of
interacting transcription factors. Kidney Int, 1997,
51(2), 548-52.
6. Salnikow, K.; Davidson, T.; Costa, M.: The role of
hypoxia-inducible signaling pathway in nickel
carcinogenesis. Environ Health Perspect, 2002, 770
Suppl 5, 831-4.
7. Goldberg, M. A.; Dunning, S. P.; Bunn, H. F.:
Regulation of the erythropoietin gene: evidence that
the oxygen sensor is a heme protein. Science, 1988,
242(4884), 1412-5.
8. Hewitson, K. S.; McNeill, L. A.; Riordan, M. V.;
Tian, Y. M.; Bullock, A. N.; Welford, R. W.; Elkins,
J. M.; Oldham, N. J.; Bhattacharya, S.; Gleadle, J.
M.; Ratcliffe, P. J.; Pugh, C. W.; Schofield, C. J.:
Hypoxia-inducible factor (HEF) asparagines
hydroxylase is identical to factor inhibiting HIF
(FIH) and is related to the cupin structural family. J.
Biol Chem, 2002, 277(29), 26351-5.
9. Epstein, A. C; Gleadle, J. M.; McNeill, L. A.;
Hewitson, K. S.; Oliourke, J.; Mole, D. R.;
Mukherji, M.; Metzen, E.; Wilson, M. I.; Dhanda, A.;
Tian, Y. M.; Masson, N.; Hamilton, D. L.; Jaakkola,
P.; Barstead, R.; Hodgkin, J.; Maxwell, P. H.; Pugh, C.
W.; Schofield, C. J.; Ratcliffe, P. J.: C. elegans EGL-9
and mammalian homologs define a family of
Int. J. Environ. Res. Public Health 2005, 2(1)
13
dioxygenases that regulate HIF by prolyl
hydroxylation. Cell, 2001, 107(1), 43-54.
10. Semenza, G.L., HIF-1: mediator of physiological
and pathophysiological responses to hypoxia. J Appl
Physiol, 2000, 88(4), 1474-80.
11. Iyer, N. V.; Kotch, L. E.; Agani, F.; Leung, S. W.;
Laughner, E.; Wenger, R. H.; Gassmann, M.;
Gearhart, J. D.;Lawler, A. M.; Yu, A. Y.; Semenza,
G. L.: Cellular and developmental control of O
2
homeostasis by hypoxia-inducible factor 1 alpha.
Genes Dev, 1998, 12(2), 149-62.
12. Wang, G. L.; Jiang, B. H.; Rue, E. A.; Semenza, G.
L.: Hypoxia-inducible factor 1 is a basic-helix-loop-
helix-PAS heterodimer regulated by cellular O2
tension. Proc Natl Acad Sci USA, 1995, 92(12),
5510-4.
13. Semenza, G. L.: Targeting HIF-1 for cancer therapy.
Nat Rev Cancer, 2003, 3(10), 721-32.
14. Acker, T.; Plate, K. H.: A role for hypoxia and
hypoxia-inducible transcription factors in tumor
physiology. J Mol Med,, 2000, 80(9), 562-75.
15. Ivan, M.; Kondo, K.; Yang, H.; Kim, W.; Valiando,
J.; Ohh, M.; Salic, A.; Asara, J. M.; Lane, W. S.;
Kaelin Jr., W. G.: HJFalpha targeted for VHL-
mediated destruction by proline hydroxylation:
implications for O
2
sensing. Science, 2001, 292(5516),
464-8.
16. Jaakkola, P.; Mole, D. R.; Tian, Y. M.; Wilson, M.
I.; Gielbert, J.; Gaskell, S. J.; Kriegsheim, A.;
Hebestreit, H. F.; Mukherji, M.; Schofield, C. J.;
Maxwell, P. H.; Pugh, C. W.; Ratcliffe, P. J.:
Targeting of HIF-alpha to the von Hippel-Lindau
ubiquitylation complex by O2-regulated prolyl
hydroxylation. Science, 2001, 292(5516), 468-72.
17. Bruick, R. K.; McKnight, S. L.: A conserved family
of prolyM-hydroxylases that modify HIF. Science,
2001, 294(5545), 1337-10.
18. Lando, D.; Peet, D. J.; Whelan, D. A.; Gorman, J. J.;
Whitelaw, M. L.: Asparagine hydroxylation of the
HIF transactivation domain a hypoxic switch.
Science, 2002, 295(5556), 858-61.
19. Lando, D.; Peet, D. J.; Gorman, J. J.; Whelan, D. A.;
Whitelaw, M. L.; Bruick, R. K.: FIH-1 is an
asparaginyl hydroxylase enzyme that regulates the
transcriptional activity of hypoxia-inducible factor.
Genes Dev, 2002, 16(12), 1466-71.
20. Jeong, J. W.; Bae, M. K.; Ann, M. Y.; Kim, S. H.;
Sohn, T. K.; Bae, M. H.; Yoo, M. A.; Song, E. J.;
Lee, K. J.; Kim, K. W.: Regulation and
destabilization of HLF-lalpha by ARD1-mediated
acetylation. Cell, 2002, 111(5), 709-20.
21. Richard, D. E.; Berra, E.; Gothie, E.; Roux, D.;
Pouyssegur, J.: p42/p44 mitogen-activated protein
kinases phosphorylate hypoxia-inducible factor 1
alpha (HIF-1 alpha) and enhance the transcriptional
activity of HIF-1. J. Biol Chem, 1999, 274(46), 32631-7.
22. Salceda, S.; Caro, J.: Hypoxia-inducible factor
lalpha (HIF-lalpha) protein is rapidly degraded by
the ubiquitin-proteasome system under normoxic
conditions. Its stabilization by hypoxia depends on
redox-induced changes. J Biol Chem, 1997, 272(36),
22642-7.
23. Huang, L. E.; Gu, J.; Schau, M.; Bunn, H. F.:
Regulation of hypoxia-inducible factor lalpha is
mediated by an O
2
-dependent degradation domain
via the ubiquitin-proteasome pathway. Proc Natl
Acad Sci. USA, 1998, 95(14), 7987-92.
24. Maxwell, P. H.; Wiesener, M. S.; Chang, G. W.;
Clifford, S. C; Vaux, E. C; Cockman, M. E.;
Wykoff, C. C; Pugh, C. W.; Maher, E. R.; Ratcliffe, P.
J.: The tumour suppressor protein VHL targets
hypoxia-inducible factors for oxygen-dependent
proteolysis. Nature, 1999, 399(6733), 271-5.
25. Berra, E.; Roux, D.; Richard, D. E.; Pouyssegur, J.:
Hypoxia-inducible factor-alpha (HIF-1 alpha)
escapes O
2
-driven proteasomal degradation
irrespective of its subcellular localization: nucleus
or cytoplasm. EMBO Rep, 2001, 2(7), 615-20.
26. Arany, Z.; Huang, L. E.; Eckner, R.; Bhattacharya,
S.; Jiang, C; Goldberg, M. A.; Bunn, H. F.;
Livingston, D. M.: An essential role or p300/CBP in
the cellular response to hypoxia. Proc Natl Acad Sci
USA, 1996, 93(23), 12969-73.
27. Mahon, P. C; Hirota, K.; Semenza, G. L.: FIH-1: a
novel protein that interacts with HIF-lalpha and
VHL to mediate repression of HIF-1 transcriptional
activity. Genes Dev, 2001, 15(20), 2675-86.
28. Salnikow, K.; Blagosklonny, M. V.; Ryan, H.;
Johnson, R.; Costa, M.: Carcinogenic nickel induces
genes involved with hypoxic stress. Cancer Res,
2000, 60(1), 38-41.
29. Salnikow, K.; An, W. G.; Melillo, G.;
Blagosklonny, M. V.; Costa, M.: Nickel-induced
transformation shifts the balance between HIF-1 and
p53 transcription factors. Carcinogenesis, 1999,
20(9), 1819-23.
30. Wang, G. L.; Semenza, G. L.: Purification and
characterization of hypoxia-inducible factor 1. J. Biol.
Chem., 1995, 270(3), 1230-7.
31. Salnikow, K.; Donald, S. P.; Bruick, R. K.;
Zhitkovich, A.; Phang, J. M.; Kasprzak, K. S.:
Depletion of intracellular ascorbate by the
carcinogenic metals nickel and cobalt results in the
induction of hypoxic stress. J Biol Chem, 2004.
... CoCl 2 downregulates the transcripts of FIH and ARD-1 (Ke, Kluz, & Costa, 2005). ARD-1-mediated acetylation at lysine 532 of HIF-1α enhances the binding of HIF-1α with pVHL , and it has been suggested that this acetylation is critical to proteasomal degradation. ...
... CoCl 2 inhibits histone acetylation and increases the methylation of histone H3 at lysine 9. This effect may play a role in the downregulation of these two important genes with a negative effect on HIF-1α activation(Ke et al., 2005).All these hypotheses show different mechanisms of CoCl 2 on HIF stabilization that could explain why this metal is a specific inductor of HIF-1α/2α.As explained before, CoCl 2 is used to mimic hypoxia by the stabilization of HIF-1α/2α, inducing a transcriptional profile mediated by these transcriptional factors. However, users of the CoCl 2 -induced hypoxia model have always wondered to what extent this chemically induced model is similar to hypoxic conditions. ...
Article
The use of hypoxia models in cell culture has allowed the characterization of the hypoxia response at the cellular, biochemical and molecular levels. Although a decrease in oxygen concentration is the optimal hypoxia model, the problem faced by many researchers is access to a hypoxia chamber or a CO2 incubator with regulated oxygen levels, which is not possible in many laboratories. Several alternative models have been used to mimic hypoxia. One of the most commonly used models is cobalt chloride‐induced chemical hypoxia because it stabilizes hypoxia inducible factors 1α and 2α under normoxic conditions. This model has several advantages, and currently, there is a substantial amount of scattered information about how this model works. This review describes the characteristics of the model, as well as the biochemical and molecular bases that support it. The regulation of hypoxia inducible factors by oxygen and the role of CoCl2 are explained to understand the most accepted bases of the CoCl2‐induced hypoxia model. The different current hypotheses that explain the establishment of hypoxic conditions using CoCl2 are also described. Finally, based on the different observations reported in the literature, we provide a critical review about the scope and limitations of this widely used chemical hypoxia model to be informative to all researchers interested in the field.
Article
Full-text available
Background: Danlou tablet (DLT), the traditional Chinese medicine has been commonly used for dyslipidemia, atherosclerosis, and coronary heart disease. Whether it was effective against vascular injury caused by CIH has remained unknown. The aim of the current study was to observe the effects of DLT on chronic intermittent hypoxia (CIH)-induced vascular injury via regulation of blood lipids and to explore potential mechanisms. Methods: Sixteen 12-week-old male ApoE-/- mice were randomly divided into four groups. The sham group was exposed to normal room air, whereas the other three groups were exposed to CIH. Mice in the CIH + normal saline (NS) group were gavaged with NS. Mice in the CIH + Angptl4-ab group were intraperitoneally injected with Angptl4-antibody. Mice in the CIH + DLT group were gavaged with DLT. After four weeks of intervention, serum lipid concentrations, and serum lipoprotein lipase (LPL) activity were detected. The changes in atherosclerosis in vascular tissue were detected by hematoxylin and eosin (H&E) staining. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analysis were applied to detect the expression levels of hypoxia-induciblefactor-1 (HIF-1), factor-inhibiting HIF-1 (FIH-1), angiopoietin-like 4 (Angptl4), and LPL in different tissues. Results: CIH exposure increases serum lipid levels, decreases serum LPL activity, and exacerbates atherosclerosis. Both Angptl4-ab and DLT treatment reversed the changes in lipid concentration, LPL activity, and atherosclerosis caused by CIH. In the epididymal fat pad, CIH exposure decreased the expression of FIH-1 and increased the expression of HIF-1, whereas DLT treatment increased the expression of FIH-1 and LPL and inhibited the expression of HIF-1 and Angptl4. In heart tissue, the expression levels of LPL and Angptl4 were not affected by modeling or treatment. Conclusions: DLT improved vascular damage by improving the increase in blood lipids induced by CIH, potentially by upregulating FIH-1 and downregulating HIF-1 and Angptl4 in adipose tissue. Therefore, DLT may be a promising agent for the prevention and treatment of CIH-induced vascular injury.
Article
Epidemiological studies have demonstrated a strong association of ambient fine particulate matter (PM2.5) exposure with the increasing mortality by ischemic heart disease (IHD), but the involved mechanisms remain poorly understood. Herein, we found that the chronic exposure of real ambient PM2.5 led to the upregulation of hypoxia-inducible factor-1 alpha (HIF-1α) protein in the myocardium of mice, accompanied by obvious myocardial injury and hypertrophy. Further data from the hypoxia-ischemia cellular model indicated that PM2.5-induced HIF-1α accumulation was responsible for the promotion of myocardial hypoxia injury. Moreover, the declined ATP level due to the HIF-1α-mediated energy metabolism remodeling from β-oxidation to glycolysis had a critical role in the PM2.5-increased myocardial hypoxia injury. The in-depth analysis delineated that PM2.5 exposure decreased the binding of prolyl hydroxylase domain enzyme (PHD2) and HIF-1α and subsequent ubiquitin protease levels, thereby leading to the accumulation of HIF-1α. Meanwhile, factor-inhibiting HIF1 (FIH1) expression was down-regulated by PM2.5, resulting in the enhanced translocation of HIF-1α to the nucleus. Overall, our study provides valuable insight into the regulatory role of oxygen sensor-mediated HIF-1α stabilization and translocation in PM-exacerbated myocardial hypoxia injury, we suggest this adds significantly to understanding the mechanisms of haze particle-caused burden of cardiovascular disease.
Chapter
This chapter will summarize some of the molecular responses exhibited by cells that come into contact with toxic metals. We will consider the transport of toxic metals into cells and how this interferes with the transport of essential metals. Toxic metals also interfere with the intracellular action of essential metals and may cause toxicity and cancer by this mechanism. Some metals have very specialized effects on enzymes, and there are proteins that can bind toxic metals such as metallothionein, which will also be discussed. A number of metals are also slightly mutagenic or genotoxic and these effects will be reviewed. Since most metals that are carcinogenic, such as arsenic (As), beryllium (Be), cadmium (Cd), and nickel (Ni), with the exception of chromium, do not interact with DNA and are not mutagenic, we discuss other mechanisms such as epigenetic effects to account for their carcinogenic activity, as well as how they affect the expression of genes. Metals also interfere with cell signaling and we will discuss the hypoxic signaling pathway, in particular, as well as those involving PI3K, Akt, reactive oxygen species mitogen-activated protein kinase, NF-κB, NF-AT, and AP-1. Finally, the basis of the interaction of toxic metals with all cellular constituents is their coordination with biological ligands, which will be addressed throughout the chapter.
Article
Full-text available
The repair and treatment of articular cartilage injury is a huge challenge of orthopedics. Currently, most of the clinical methods applied in treating cartilage injuries are mainly to relieve pains rather than to cure them, while the strategy of tissue engineering is highly expected to achieve the successful repair of osteochondral defects. Clear understandings of the physiological structures and mechanical properties of cartilage, bone and osteochondral tissues have been established, but the understanding of their physiological heterogeneity still needs further investigation. Apart from the gradients in the micromorphology and composition of cartilage-to-bone extracellular matrixes, an oxygen gradient also exists in natural osteochondral tissue. The response of hypoxia-inducible factor (HIF)-mediated cells to oxygen would affect the differentiation of stem cells and the maturation of osteochondral tissue. This article reviews the roles of oxygen level and HIF signaling pathway in the development of articular cartilage tissue, and their prospective applications in bone and cartilage tissue engineering. The strategies for regulating HIF signaling pathway and how these strategies finding their potential applications in the regeneration of integrated osteochondral tissue are also discussed.
Article
Full-text available
Cobalt exists widely in nature and is one of the essential functional elements in human body, performing in organic or inorganic forms. The lackness of adequate bone integration is a main issue to limit the biomedical substitute materials using widely in clinic. However, introducing cobalt element onto the surface of the materials can significantly change the biological behavior of the implants, which is a good way to solve the above problem. In this paper, the effects of doped cobalt ions on the biological properties of different materials were reviewed, and the development trend of cobalt ion doped biomedical device was prospected.
Article
The mineral constituent of the bone contains non-stoichiometric hydroxyapatite with various substitutional ions. Iron (Fe) and cobalt (Co) are fundamental cofactors, which influence cell respiration and mitosis, respectively. In this study, undoped, Fe³⁺-, Co²⁺-, and dually-doped HA nanoparticles (NPs) were synthesized by the hydrothermal method. The Fe+CoCa+Fe+Co molar ratio was varied between 0.02 to 0.2. Samples were analyzed using X-ray diffraction (XRD), field-emission scanning electron microscopy (FESEM), vibrating sample magnetometry (VSM), Fourier-transformed infrared spectroscopy (FT-IR), inductively coupled plasma optical emission spectroscopy (ICP-OES), and biological assessment. XRD results confirmed the formation of the hexagonal HA crystal structure. The crystallite size was reduced from 34.6 nm to 4.4 nm. Also, the c-axis dimension and the degree of crystallinity were reduced in all the doped samples. The FESEM results showed a change of morphology from rod-like in the undoped nHA into a sphere-like morphology in the doped-samples. With the addition of ferromagnetic dopant, the magnetic behavior of samples altered from diamagnetic to paramagnetic-like and ferromagnetic behavior. Bioactivity assessment indicated the formation of the HA particle on the surface of all synthesized materials. The antibacterial test demonstrated antibacterial activity against gram-negative (-) E. coli. Moreover, the antibiotic-impregnated samples had a synergistic effect on both gram (-), E. coli, as well as gram (+), S. aureus, bacteria. The MTT evaluation revealed the enhanced cell viability of osteoblast-like cells in the dually-doped NPs. Based on this survey, we can strongly emphasize the potential for bone bioactivity and antibacterial feature of these newly developed samples.
Article
Impact statement: Copper promotes angiogenesis, but the mechanistic insights have not been fully elucidated until recently. In addition, the significance of copper promotion of angiogenesis in myocardial regeneration was increasingly revealed. Copper critically participates in the regulation of hypoxia-inducible factor 1 (HIF-1) of angiogenic gene expression. Interestingly, myocardial ischemia causes copper efflux from the heart, leading to suppression of angiogenesis, although HIF-1α, the critical subunit of HIF-1, remains accumulated in the ischemic myocardium. Strategies targeting copper specific delivery to the ischemic myocardium lead to selective activation of HIF-1-regulated angiogenic gene expression. Vascularization of the ischemic myocardium re-establishes the tissue injury microenvironment, and rebuilds the conduit for communication between the tissue injury signals and the remote regenerative responses including stem cells. This process promotes myocardial regeneration. Thus, a simple and effective copper supplementation to the ischemic myocardium would become a novel therapeutic approach to the treatment of patients with ischemic heart diseases.
Article
Cobalt (Co) is vital for cells in trace amounts, but excessive exposure to Co is possible due to surgical devices such as artificial metal-on-metal joints. Cobalt(II) chloride (CoCl2) has also been shown to imitate hypoxic conditions in cells by stabilizing the transcription factor hypoxia-inducible factor-1α (HIF-1α). The purpose of the present study was to investigate the possible immunomodulatory action of CoCl2 by investigating its effects on the expression of inflammatory genes in macrophages. The following factors were assessed: inducible nitric oxidase synthase (iNOS), NADPH oxidase 2 (NOX2), interleukin 6 (IL-6), arginase-1 and HIF-1α. In the absence of exogenous cytokines, Co enhanced alternative (M2) macrophage activation as evidenced by increased arginase-1 expression, but had no direct effect on inflammatory factors associated with classical (M1) activation. Interestingly, in LPS-stimulated macrophages, Co modified the M1 type activation profile by increasing iNOS expression and nitric oxide production and decreasing NOX2 and IL-6. Also, Co increased HIF-1α levels in unstimulated and LPS-stimulated cells as expected. In conclusion, we showed that Co enhanced alternative (M2) activation in resting macrophages. In addition, Co was found to remodel the classical M1 phenotype of macrophage activation by changing the balance of iNOS, NOX2 and IL-6. This article is protected by copyright. All rights reserved.
Article
Full-text available
p300 and CBP are homologous transcription adapters targeted by the E1A oncoprotein. They participate in numerous biological processes, including cell cycle arrest, differentiation, and transcription activation. p300 and/or CBP (p300/CBP) also coactivate CREB. How they participate in these processes is not yet known. In a search for specific p300 binding proteins, we have cloned the intact cDNA for HIF-1 alpha. This transcription factor mediates hypoxic induction of genes encoding certain glycolytic enzymes, erythropoietin (Epo), and vascular endothelial growth factor. Hypoxic conditions lead to the formation of a DNA binding complex containing both HIF-1 alpha and p300/CBP. Hypoxia-induced transcription from the Epo promoter was specifically enhanced by ectopic p300 and inhibited by E1A binding to p300/CBP. Hypoxia-induced VEGF and Epo mRNA synthesis were similarly inhibited by E1A. Hence, p300/CBP-HIF complexes participate in the induction of hypoxia-responsive genes, including one (vascular endothelial growth factor) that plays a major role in tumor angiogenesis. Paradoxically, these data, to our knowledge for the first time, suggest that p300/ CBP are active in both transformation suppression and tumor development.
Article
All organisms can sense O(2) concentration and respond to hypoxia with adaptive changes in gene expression. The large body size of mammals necessitates the development of multiple complex physiological systems to ensure adequate O(2) delivery to all cells under normal conditions. The transcriptional regulator hypoxia-inducible factor 1 (HIF-1) is an essential mediator of O(2) homeostasis. HIF-1 is required for the establishment of key physiological systems during development and their subsequent utilization in fetal and postnatal life. HIF-1 also appears to play a key role in the pathophysiology of cancer, cardiovascular disease, and chronic lung disease, which represent the major causes of mortality among industrialized societies. Genetic or pharmacological modulation of HIF-1 activity in vivo may represent a novel therapeutic approach to these disorders.
Article
Eukaryotic cells sense oxygen and adapt to hypoxia by regulating a number of genes. Hypoxia-inducible factor 1 (HIF-1) is the 'master' in this pleiotypic response. HIF-1 comprises two members of the basic helix–loop–helix transcription factor family, HIF-1α and HIF-1β. The HIF-1α protein is subject to drastic O2-dependent proteasomal control. However, the signalling components regulating the 'switch' for 'escaping' proteasomal degradation under hypoxia are still largely unknown. The rapid nuclear translocation of HIF-1α could represent an efficient way to escape from this degradation. We therefore asked, where in the cell is HIF-1α degraded? To address this question, we trapped HIF-1α either in the cytoplasm, by fusing HIF-1α to the cytoplasmic domain of the Na+-H+ exchanger (NHE-1), or in the nucleus, by treatment with leptomycin B. Surprisingly, we found that HIF-1α is stabilized by hypoxia and undergoes O2-dependent proteasomal degradation with an identical half-life (5–8 min) in both cellular compartments. Therefore, HIF-1α entry into the nucleus is not, as proposed, a key event that controls its stability. This result markedly contrasts with the mechanism that controls p53 degradation via MDM2.
Article
Hypoxia is an essential developmental and physiological stimulus that plays a key role in the pathophysiology of cancer, heart attack, stroke, and other major causes of mortality. Hypoxia inducible factor 1 (HIF-1) is the only known mammalian transcription factor expressed uniquely in response to physiologically relevant levels of hypoxia. We now report that in Hif1 alpha(-/-) embryonic stem cells that did not express the O-2-regulated HIF-1 alpha subunit, levels of mRNAs encoding glucose transporters and glycolytic enzymes were reduced, and cellular proliferation was impaired. Vascular endothelial growth factor mRNA expression was also markedly decreased in hypoxic Hif1 alpha(-/-) embryonic stem cells and cystic embryoid bodies. Complete deficiency of HIF-1 alpha resulted in developmental arrest and lethality by E11 of Hif1 alpha(-/-) embryos that manifested neural tube defects, cardiovascular malformations, and marked cell death within the cephalic mesenchyme. In Hif1 alpha(+/+) embryos, HIF-1 alpha expression increased between E8.5 and E9.5, coincident with the onset of developmental defects and cell death in Hif1 alpha(-/-)embryos. These results demonstrate that HIF-1 alpha is a master regulator of cellular and developmental O-2 homeostasis.
Article
Erythropoietin (Epo), the hormone that stimulates red blood cell production, is synthesized in the kidney and liver in response to hypoxia. The human hepatoma cell line Hep3B regulates its production of Epo in a physiologic manner. Either hypoxia or cobalt chloride markedly increases expression of Epo mRNA as well as production of biologically active and immunologically distinct Epo protein. New protein synthesis is required before the induction of increased levels of hypoxia- or cobalt-induced Epo mRNA. Hypoxia, cobalt chloride, and nickel chloride appear to stimulate Epo production through a common pathway. The inhibition of Epo production at low partial pressures of oxygen by carbon monoxide provides evidence that a heme protein is integrally involved in the oxygen-sensing mechanism. This hypothesis is further supported by the finding that when heme synthesis is blocked, hypoxia-, cobalt-, and nickel-induced Epo production are all markedly inhibited. A model is proposed in which a ligand-dependent conformational change in a heme protein accounts for the mechanism by which hypoxia as well as cobalt and nickel stimulate the production of Epo.
Article
Hypoxia-inducible factor 1 (HIF-1) is found in mammalian cells cultured under reduced O2 tension and is necessary for transcriptional activation mediated by the erythropoietin gene enhancer in hypoxic cells. We show that both HIF-1 subunits are basic-helix-loop-helix proteins containing a PAS domain, defined by its presence in the Drosophila Per and Sim proteins and in the mammalian ARNT and AHR proteins. HIF-1 alpha is most closely related to Sim. HIF-1 beta is a series of ARNT gene products, which can thus heterodimerize with either HIF-1 alpha or AHR. HIF-1 alpha and HIF-1 beta (ARNT) RNA and protein levels were induced in cells exposed to 1% O2 and decayed rapidly upon return of the cells to 20% O2, consistent with the role of HIF-1 as a mediator of transcriptional responses to hypoxia.
Article
Hypoxia-inducible factor 1 (HIF-1) is a DNA-binding protein that activates erythropoietin (Epo) gene transcription in Hep3B cells subjected to hypoxia or cobalt chloride treatment. HIF-1 DNA binding activity is also induced by hypoxia or cobalt in non-Epo-producing cells, suggesting a general role for HIF-1 in hypoxia signal transduction and transcriptional regulation. Here we report the biochemical purification of HIF-1 from Epo-producing Hep3B cells and non-Epo-producing HeLa S3 cells. HIF-1 protein was purified 11,250-fold by DEAE ion-exchange and DNA affinity chromatography. Analysis of HIF-1 isolated from a preparative gel shift assay revealed four polypeptides. Peptide mapping of these HIF-1 components demonstrated that 91-, 93-, and 94-kDa polypeptides had similar tryptic maps, whereas the 120-kDa polypeptide had a distinct profile. Glycerol gradient sedimentation analysis suggested that HIF-1 exists predominantly in a heterodimeric form and to a lesser extent as a heterotetramer. Partially purified HIF-1 bound specifically to the wild-type HIF-1 binding site from the EPO enhancer but not to a mutant sequence that lacks hypoxia-inducible enhancer activity. UV cross-linking analysis with purified HIF-1 indicated that both subunits of HIF-1 contact DNA directly. We conclude that in both cobalt chloride-treated HeLa cells and hypoxic Hep3B cells HIF-1 is composed of two different subunits: 120-kDa HIF-1α and 91-94-kDa HIF-1β.
Article
Studies on erythropoietin (Epo) gene expression have been useful in investigating the mechanism by which cells and tissues sense hypoxia. Both in vivo and in Hep3B cells. Epo production is induced not only by hypoxia but also by certain transition metal (cobalt and nickel) and by iron chelation. When Hep3B cells were incubated in an iron deficient medium, Epo mRNA expression was enhanced fourfold compared to Hep3B cells in iron enriched medium. Epo induction by cobalt was inversely related to iron concentration in the medium, indicating competition between the two metals. Under hyperbaric oxygen, cobalt induction of erythropoietin mRNA was modestly suppressed while nickel induction was markedly enhanced. These recent observations support the proposal that the oxygen sensor is a heme protein in which cobalt and nickel can substitute for iron in the porphyrin ring. The up-regulation of Epo gene transcription by hypoxia depends on at least two known DNA binding transcription factors, HIF-1 and HNF-4, which bind to cognate response elements in a critical approximately 50 bp 3' enhancer. Hypoxia induces HIF-1 binding. HNF-4, an orphan nuclear receptor constitutively expressed in kidney and liver, binds downstream of HIF-1 and cooperates with HIF-1, contributing importantly to high level and perhaps tissue specific expression. The C-terminal activation domain of HNF-4 binds to the beta subunit of HIF-1. The C-terminal portion of the alpha subunit of HIF-1 binds specifically to p300, a general transcriptional activator. Hypoxic induction of the endogenous Epo gene in Hep3B cells as well as an Epo-reporter gene was fully inhibited by E1A, an adenovirus protein that binds to and inactivates p300, but only slightly by a mutant E1A that fails to bind to p300. Moreover, overexpression of p300 enhanced hypoxic induction. Thus, it is likely that in hypoxic cells, p300 or a related family member plays a critical role in forming a macromolecular assembly with HIF-1 and HNF-4, enabling transduction from the Epo 3' enhancer to the apparatus on the promoter responsible for the initiation of transcription.