Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

In Vitro Cell.Dev.Biol.

—Animal (2008) 44:45–50


DOI 10.1007/s11626-007-9079-4

An improved protocol for primary culture of cardiomyocyte


from neonatal mice
P. Sreejit & Suresh Kumar & Rama S. Verma

Received: 13 September 2007 / Accepted: 11 December 2007 / Published online: 23 February 2008 / Editor: J. Denry Sato.
# The Society for In Vitro Biology 2008

Abstract The primary culture of neonatal mice cardiomyo- The neonatal murine cardiomyocyte culture was first
cyte model enables researchers to study and understand the described by Harary and Farley (1963); since then, several
morphological, biochemical, and electrophysiological char- modifications have been developed and reported in the past
acteristics of the heart, besides being a valuable tool for 40 yr. Various groups working with adult murine cardio-
pharmacological and toxicological studies. Because cardio- myocyte cultures have elaborated on issues like finding and
myocytes do not proliferate after birth, primary myocardial formulating conditions for proper tissue dissociation such
culture is recalcitrant. The present study describes an as enzymes and duration of enzyme treatment, in selecting
improved method for rapid isolation of cardiomyocytes from various growth supplements and appropriate serum con-
neonatal mice, as well as the maintenance and propagation of centrations for cell attachment, and also appropriate
such cultures for the long term. Immunocytochemical and attachment surfaces especially for long-term cell cultures
gene expression data also confirmed the presence of several (Kruppenbacher et al. 1993; Chlopclkova et al. 2001).
cardiac markers in the beating cells during the long-term Neonatal murine cardiomyocyte cultures have been used as
culture condition used in this protocol. The whole culture a model for studying contraction, ischemia, and hypoxia at
process can be effectively shortened by reducing the enzyme the cellular level (Yamashita et al. 1994; Bahi et al. 2006).
digestion period and the cardiomyocyte enrichment step. Murine cardiomyocyte cultures have also been used to
study the toxicology of drugs and their transport (Limaye
Keywords Primary cell culture . Neonatal mice . and Shaikh 1999; Wang and Kang 1999).
Murine cardiomyocyte enrichment . Immunostaining . In this report, we describe a simple and rapid method for
Gene expression the isolation of cardiomyocytes from neonatal mice heart
and their maintenance in primary cultures that consistently
yielded long-term cardiomyocyte cultures in our laboratory.
This protocol does not involves the addition of growth
Electronic supplementary material The online version of this article supplements like carnitine, creatinine, taurine, and non-
(doi:10.1007/s11626-007-9079-4) contains supplementary material, muscle cells proliferation inhibitors such as 9-β-D-arabino-
which is available to authorized users. furanosyl cytosine and 5-bromo-2′-deoxyuridine or by
P. Sreejit : S. Kumar : R. S. Verma increasing the Ca2+ concentration in the washing and
Stem Cell and Molecular Biology Laboratory, incubation mediums (Kruppenbacher et al. 1993; Nuss
Department of Biotechnology, and Marban 1994; Remião et al. 2001; Fu et al. 2005). This
Indian Institute of Technology Madras,
Chennai 600036 TN, India
protocol reduces the processing time considerably. For
culturing the neonatal cardiomyocytes in vitro, cardiomyo-
R. S. Verma (*) cyte enrichment and the enzyme digestion steps are the two
201, Bhupat and Jyoti Mehta School of Biosciences, critical steps. The cardiomyocyte enrichment step, which
Department of Biotechnology,
Indian Institute of Technology Madras,
removes nonmuscle cells, is crucial for obtaining a homog-
Chennai 600 036 TN, India enous yield of myocytes for culture. Several techniques,
e-mail: vermars@iitm.ac.in including the differential attachment technique (Polinger
46 SREEJIT ET AL.

Table 1. Comparison of various protocols used for cultivation of neonatal mouse cardiomyocytes

Research Ingredient variations Protocol variations


group
(year of Media and supplements Enzyme used Supplements/ Enzyme treatment and Cardiomyocyte
publication) used inhibitors number of repeats enrichment

Our study DMEM/F12 (1:1) with 0.5% trypsin EDTA (~1 ml of 2 mM L-glutamine, 37° C/4 min in a water bath Differential
(2007) in fetal calf serum (20%) trypsin for every 100 mg of 0.1 mM nonessential with stirring and intermittent attachment
BALB/c and horse serum (5%) tissue) amino acids, 3 mM pipeting; solution allowed to technique at
mice with antibiotics sodium pyruvate stand for 1 min; supernatant 37° C for 2–
and bovine insulin collected into 15-ml falcon 3h
(1 μg/ml) tube on ice, to which 2 ml
media supplemented with
20% fetal calf serum was
added; digested suspensions
were pooled and centrifuged
at 3,500 rpm/10 min/4° C.
Repeated three to five times
till tissue is digested
completely
Wang and MEM with 20% FBS 0.25% trypsin in HBSS – 37° C /15 min; centrifuged at Differential
Kang with antibiotics (without Ca++ & Mg++) at 200×g/8 min repeated till the attachment
(1999) in pH 7.4 tissue is completely dissolved technique at
FVB 37° C for 2 h
mice
Song et al. M199 with 10% fetal calf 0.625 mg/ml collagenase type 5 mM D-glucose 37° C /40 min; filtered through Differential
(2000) in serum II a polypropylene macroporous attachment
C57BL6 filter (mesh opening technique for
and iNOS 105 μm); centrifuged at 40 min
(2/2) 1,200 rpm/5 min
mutant
mice
Nuss and DMEM (nutrient mixture 0.25 mg/ml collagenase in – 37° C with slow rotation of the –
Marban F-12 HAM) with 10% Joklik MEM with following tube containing the digestion
(1994) in fetal bovine serum additions (mM unless solution; supernatant during
CD-1 otherwise given): HEPES, 10; repeated collagenase
mice sodium pyruvate, 10; L- digestions pooled to digestion
glutamine, 5; nicotinamide, solution (without
1; L-ascorbate, 0.4; collagenase) on ice; fractions
adenosine, 1; D-ribose, 1; centrifuged at low speed;
MgCl2, 1; taurine, 1; DL- resuspended in digestion
carnitine, 2; potassium solution with 0.2% fetal
bicarbonate, 26; CaCl2, 0.5; bovine serum; filtered with
gentamycin, 10 μg/ml cell strainer, repeated
Rosenblatt 3:1 mix of DMEM and 0.45 mg/ml collagenase and – – Differential
et al. M 199 with 10% horse 1 mg/ml pancreatin in buffer attachment
(2005) in serum and 5% fetal technique for
C57BL/6 bovine serum with 45 min
mice antibiotics
Nickson et 4:1 mix of DMEM:M199 0.03% Collagenase type II and 10 mM glutamine Enzyme treatment for 20 min Two rounds of
al. (2007) with 10% heat- 0.06% pancreatin in a and 0.2 mM at 37° C; supernatant differential
in wild- inactivated horse serum balanced salt solution bromodeoxyuridine collected and cells isolated attachment
type and and 5% heat-inactivated containing MgSO4 7H20 by centrifugation at technique for
Puma fetal bovine serum with (0.2 g/l), NaCl (6.8 g/l), KCl 1,400 rpm/4 min; 2h
knockout antibiotics (0.4 g/l), NaH2PO4H2O resuspension in 2 ml of
mice (1.5 g/l), glucose (1 g/l), and newborn calf serum at
HEPES (4.76 g/l) at pH 7.5 37° C; repeated four times
IMPROVED PROTOCOL FOR CULTURE OF NEONATAL MICE CARDIOMYOCYTE 47

1970; Blondel et al. 1971), deprivation of serum (Shields McKoy et al. 2007). Because both mice and rats belong to
et al. 1988), density gradient centrifugation (Flanders et al. the murine family, protocols for neonatal mice cardiomyo-
1995), radiation (Desmond and Harary 1972), glutamine cyte culture used by many groups are much similar to those
deprivation (Clark 1976), and use of chemical reagents to used with neonatal rats (Wang and Kang 1999; Song et al.
inhibit nonmyocyte proliferation (Simpson and Savion 2000; Pellieux et al. 2001; Matsuura et al. 2004; Rosenblatt
1982), have been used to culture of neonatal murine et al. 2005; Nickson et al. 2007). A comparison of the
cardiomyocytes. The enzyme digestion step for dissociating protocols is shown along with the report (Table 1). The
cells from heart tissue too is critical during culturing of present report describes a convenient, reliable, and time-
neonatal cardiomyocytes. It has been reported that short saving protocol, which has been successfully demonstrated
repetitive trypsinization of heart tissue helps in obtaining a for consistent cardiomyocyte cultures. It involves decreas-
higher yield of undamaged myocytes, rather than a single ing the duration of enzymatic digestion to a maximum of
digestion for longer period (Mark and Strasser 1966). The 3 h during which nonmyocytes will be removed too.
incubation period with proteolytic enzyme depends on the BALB/c neonatal and adult mice were procured from
amount of intact tissue left during dissociation into single Tamil Nadu Veterinary and Animal Sciences University,
cells. Although repetitive digestion of heart tissue with Chennai. All procedures, approved by the Institutional
proteolytic enzymes gives good yield, the cell viability Animal Ethics Committee (IIT Madras, India) and the
decreases if the tissue is overexposed to proteolytic Committee for the Purpose of Control and Supervision of
enzymes. Cardiomyocytes lose their ability to proliferate Experiments on Animals, Government of India, were
shortly after birth, and growth of heart tissue is governed by performed under the Rule 5(a) of the Breeding and
cell growth rather than by proliferation (Ahuja et al. 2007; Experiments on Animals (Control and Supervision) Rules

Figure 1. Phase-contrast photo-


micrograph of cultures showing
different morphologies of beat-
ing cardiomyocytes in culture
(a, b, c, d; magnification: ×400).
Supplementary data—
videographs 1a, 1b, 1c, 1d.
48 SREEJIT ET AL.

from the heart with a sterile forceps, to prevent clot


formation inside the lumen of heart. The excised hearts were
again washed with chilled PBS and followed by sterile ice-
cold balanced salt solution (20 mM hydroxyethylpiperazi-
neethanesulfonic acid–NaOH [pH 7.6], 130 mM NaCl,
1 mM NaH2PO4, 4 mM glucose, 3 mM KCl), in which
the tissue was kept for 10 min. The ventricles were excised
in a sterile 60-mm Petri dish, and the auricles were carefully
removed and discarded. The tissues were then minced with
a sterile scalpel blade into small pieces less than or equal to
1 mm3 in 0.05% trypsin ethylenediamine tetraacetic acid
(EDTA; Invitrogen, Carlsbad, CA; 0.1 ml per heart) and
transferred into sterile 15-ml falcon tubes. The myocardial
cells were dispersed by incubating with 0.5% Trypsin
EDTA (~1 ml of Trypsin for every 100 mg of tissue), which
was then mixed by intermittent pipeting along with stirring
at 37° C in a water bath for 4 min. The cell suspension was
allowed to stand for 1 min. The supernatant containing
single cells was collected into a 15-ml falcon tube kept on
ice, to which 2 ml Dulbecco’s modified Eagle’s medium
(DMEM)/F12 (1:1) medium (Gibco, Carlsbad, CA) supple-
mented with 20% fetal calf serum (Gibco) was added, and
the digestion step was repeated three times. The cell
suspensions from each digestion was pooled and centri-
fuged at 3,500 rpm for 10 min at 4° C.
The cell pellet was resuspended in a maintenance
medium containing DMEM/F12 (1:1) medium supple-
mented with fetal calf serum (20%), horse serum (Gibco)
(5%), penicillin (100 U/ml), and streptomycin (100 mg/ml;
Cambrex, Verviers, Belgium), 2 mM L-glutamine (Cam-
Figure 2. Immunocytochemical analysis of GATA-4 and Nkx-2.5 in
brex), 0.1 mM nonessential amino acids (Gibco), 3 mM
cardiomyocytes (double staining). (a) Phase-contrast photomicrograph
of culture. (b) Positive staining with anti-GATA-4 antibody. (c) Positive sodium pyruvate (Gibco), and bovine insulin (1 μg/ml;
staining with anti-Nkx-2.5 antibody (original magnification, ×200). USV, India). Viability of cardiomyocytes was assessed by
Trypan blue exclusion test.
(1998). Neonatal mice 1–3-d-old were killed by cervical The cells were plated on plates precoated with 1%
dislocation. Whole hearts were excised and immediately gelatin and incubated in 95% air and 5% CO2 at 37° C for
transferred into ice-cold phosphate-buffered saline (PBS; ~2–3 h, to allow the differential attachment of nonmyo-
Ca2+ and Mg2+ free). The blood was gently squeezed out cardial cells. The nonadhesive cells (cardiomyocytes) were

Figure 3. Reverse transcrip-


tion-polymerase chain reaction
(RT-PCR) analysis for cardiac
transcription factors (Nkx-2.5,
GATA-4, and MEF-2C), as well
as cardiac structural and func-
tional proteins (α-Cardiac Actin,
Cardiac Troponin T, Connexin
43) in cardiomyocytes on dif-
ferent days of primary culture.
Adult mouse heart was used as a
positive control, and β-actin
was used as a loading internal
control.
IMPROVED PROTOCOL FOR CULTURE OF NEONATAL MICE CARDIOMYOCYTE 49

transferred into a sterile tube. The viability of cells (85– amino acids and glutamine reduced the overgrowth of
90%) was assessed by the Trypan blue exclusion test. After nonmyocyte cell-like fibroblasts (Fioramonti et al. 1955),
counting, the myocyte-enriched suspension was plated on to when compared to the maintenance medium containing
culture dishes at a density of 2×104 cells per cm2. The cells M199, which was used by earlier workers (Simpson and
were incubated in a humid 5% CO2 incubator at 37° C. The Savion 1982; Song et al. 2000; Nickson et al. 2007). It has
Medium was replenished after 72 h and there after every already been known that α2-macroglobulins from horse
48 h. Beating myocardial cells were clearly observed on serum in combination with seromucoids from fetal calf
third day in culture (Fig. 1) through an inverted microscope serum promote rapid growth of embryonic mouse cells
(Nikon Eclipse TS100, Melville, NY), and video graphs of (Healy and Parker 1966). Although the fetal calf serum-
beating cells were recorded with a digital camera (Nikon containing medium was used primarily for inactivation of
Coolpix5400; supplementary data—videographs 1a, 1b, 1c, trypsin, it has an added advantage that it also contains
1d). Cells were maintained up to 4 wk for further study. several growth factors besides differentiation factors needed
To demonstrate that cultured cells are cardiomyocytes, we for the growth and survival of cells (Bryja et al. 2006).
characterized beating cells by immunocytochemistry using Cells can be plated on dishes pretreated with collagen,
primary antibodies: mouse monoclonal anti-GATA-4 (1:100; fibronectin, laminin, and gelatin or by using Primaria
Santa Cruz Biotechnology, Santa Cruz, CA) and rabbit (Falcon Plastics) or Pronectin (Promega, Madison, WI)-
polyclonal anti-Nkx-2.5 (1:100; Santa Cruz Biotechnology) coated culture plates (Chlopclkova et al. 2001). It has been
and appropriate secondary antibodies: R-Phycoerythrin- reported that laminin pretreatment was a less suitable
labelled goat anti-mouse IgG (1:300; Sigma, St. Louis, matrix for the neonatal murine cardiomyocyte cultures,
MO) and fluorescein isothiocyanate-labeled goat anti-rabbit while cardiogel remains the most suitable matrix (Bick
IgG (1:300; Sigma). The immunofluorescent staining of cells et al. 1998). In our study and previous reports also (Song et
showed the presence of cardiac-specific proteins; the cells al. 2000; Matsuura et al. 2004), 1% gelatin has been used to
were stained positive for GATA-4 (Fig. 2b) and Nkx-2.5 coat culture dishes, as fibroblasts easily adheres to a
(Fig. 2c) in the nucleus. collagenous extracellular matrix like gelatin (Haas et al.
Furthermore, we have also analyzed the time course 1984). Thus, by using gelatin and in the absence of growth
expression of cardiomyocyte-specific transcription factors supplements as well as nonmyocyte growth inhibitors, we
and structural and functional proteins by reverse transcrip- have developed a protocol for culturing neonatal mice
tase–polymerase chain reaction (RT-PCR). Total ribonu- cardiomyocytes for a longer period of time, which is
cleic acid (RNA) from cardiomyocytes on different days of efficient as well as cost efficient. This modified protocol
primary culture and adult mouse heart were isolated by for culture of neonatal cardiomyocyte may help to study the
using the acid guanidinium thiocyanate–phenol–chloroform various stress conditions such as hypoxia.
extraction method (Chomczynski and Sacchi 1987). RNA
was subjected to RT-PCR by using M-MLV Reverse Acknowledgments This work is supported by grants to R.S.V. by
the Ministry of Human Resource Development (MHRD—BIO/2005–
Transcriptase (New England Biologicals, Beverly, MA). 2006/007/MHRD/RAMS/859) and Department of Biotechnology,
Primers are as follows: Nkx-2.5, GATA-4, MEF-2C, α- Ministry of Science and Technology (DBT-BT/PR5392/MED/14/
Cardiac Actin, Cardiac Troponin T, Connexin 43 (Supple- 693/2004).
mentary Table 1). RT-PCR results revealed that cells
expressed cardiac transcription factors (Nkx-2.5, GATA-4,
and MEF-2C), as well as cardiac structural and functional
proteins (α-Cardiac Actin, Cardiac Troponin T, Connexin References
43; Fig. 3). Thus, our immunocytochemical and gene
expression data confirmed the presence of several cardiac Ahuja, P.; Sdek, P.; MacLellan, W.R. Cardiac myocyte cell cycle
control in development, disease, and regeneration. Physiol. Rev.
markers in the beating cells. 87(2):521–544; 2007.
Insulin was added to protect the cardiomyocytes from Bahi, N.; Zhang, J.; Llovera, M.; Ballester, M.; Comella, J.X.;
stress by enhancing myocardial metabolic recovery. How- Sanchis, D. Switch from caspase-dependent to caspase-indepen-
ever, the stimulation of glycolysis during ischemia may be dent death during heart development: essential role of endonu-
clease G in ischemia-induced DNA processing of differentiated
detrimental because of an accumulation of metabolic end cardiomyocytes. J. Biol. Chem. 281(32):22943–22952; 2006.
products. Because insulin stimulates pyruvate dehydroge- Bick, R.J.; Snuggs, M.B.; Poindexter, B.J.; Buja, L.M.; Van Winkle,
nase activity, the addition of sodium pyruvate in the W.B. Physical, contractile and calcium handling properties of
medium may help in directing glycolysis and preventing neonatal cardiac myocytes cultured on different matrices. Cell
Adhes. Commun. 6(4)301–310; 1998.
accumulation of metabolic end products like lactate, thus Blondel, B.; Roijen, I.; Cheneval, J.P. Heart cells in culture: a simple
resulting in improved aerobic metabolism (Rao et al. 1998; method for increasing the proportion of myoblasts. Experientia
Duarte et al. 2006). Horse serum along with nonessential 27:356–358; 1971.
50 SREEJIT ET AL.

Bryja, V.; Bonilla, S.; Cajánek, L.; Parish, C.L.; Schwartz, C.M.; Luo, Matsuura, K.; Wada, H.; Nagai, T.; Iijima, Y.; Minamino, T.; Sano, M.;
Y.; et al. An efficient method for the derivation of mouse et al. Cardiomyocytes fuse with surrounding noncardiomyocytes
embryonic stem cells. Stem. Cells 24(4)844–849; 2006. and reenter the cell cycle. J. Cell Biol. 167(2):351–363; 2004.
Chlopclkova, Š.; Psotova, J.; Miketova, P. Neonatal rat cardiomyo- McKoy, G.; Bicknell, K.A.; Patel, K.; Brooks, G. Developmental
cytes—a model for the study of morphological, biochemical and expression of myostatin in cardiomyocytes and its effect on foetal
electrophysiological characteristic of the heart. Biomed. Papers and neonatal rat cardiomyocyte proliferation. Cardiovasc. Res.
145:49–55; 2001. 74(2):304–312; 2007.
Chomczynski, P.; Sacchi, N. Single-step method of RNA isolation by Nickson, P.; Toth, A.; Erhardt, P. PUMA is critical for neonatal
acid guanidinium thiocyanate–phenol–chloroform extraction. cardiomyocyte apoptosis induced by endoplasmic reticulum
Anal. Biochem. 162(1):156–159; 1987. stress. Cardiovasc. Res. 73(1):48–56; 2007.
Clark, W.J. Selective control of fibroblast proliferation and its effect Nuss, H.B.; Marban, E. Electrophysiological properties of neonatal
on cardiac muscle differentiation in vitro. Dev. Biol. 52:263–282; mouse cardiac myocytes in primary culture. J. Physiol. 479
1976. (2):265–279; 1994.
Desmond, W.J.; Harary I. In vitro studies of beating heart cells in Pellieux, C.; Foletti, A.; Peduto, G.; Aubert, J. F.; Nussberger, J.;
culture. XV. Myosin turnover and the effect of serum. Arch. Beermann, F.; et al. Dilated cardiomyopathy and impaired
Biochem. Biophys. 151:285–294; 1972. cardiac hypertrophic response to angiotensin II in mice lacking
Duarte, A.I.; Proença, T.; Oliveira, C.R.; Santos, M.S.; Rego C. FGF-2. J. Clin. Invest. 108:1843–1851; 2001.
Insulin restores metabolic function in cultured cortical neurons Polinger, I.S. Separation of cell types in embryonic heart cell cultures.
subjected to oxidative stress. Diabetes 55:2863–2870; 2006. Exp. Cell Res. 63:78–82; 1970.
Fioramonti, M.C.; Bryant, J.C.; Mcquilkin, W.T.; Evans, V.J.; Sanford, Rao, V.; Merante, F.; Weisel, R.D.; Shirai, T.; Ikonomidis, J.S.; Cohen,
K.K.; Earle, W.R. The effect of horse serum residue and G.; et al. Insulin stimulates pyruvate dehydrogenase and protects
chemically defined supplements on proliferation of Strain L human ventricular cardiomyocytes from simulated ischemia. J.
Clone 929 Cells from the Mouse. Cancer Res. 15(11):763–766; Thorac. Cardiovasc. Surg. 116(3):485–494; 1998.
1955. Remião, F.; Carmo, H.; Carvalho, F.; Bastos, M.L. Cardiotoxicity
Flanders, K.C.; Holder, M.G.; Winokur, T.S. Autoinduction of mRNA studies using freshly isolated calcium-tolerant cardiomyocytes
and protein expression for transforming growth factor-βs in from adult rat. In Vitro Cell Dev. Biol.—Animal 37:1–4; 2001.
cultured cardiac cells. J. Mol. Cell Cardiol. 27(2):805–812; 1995. Rosenblatt, V.N.; Lepore, M.G.; Cartoni, C.; Beermann, F.; Pedrazzini,
Fu, J.; Gao, J.; Pi, R.; Liu, P. An optimized protocol for culture of T. FGF-2 controls the differentiation of resident cardiac precursors
cardiomyocyte from neonatal rat. Cytotechnology 49:109–116; into functional cardiomyocytes. J. Clin. Invest. 115(7):1724–
2005. 1733; 2005.
Haas, R.; Banerji, S.S.; Culp, L.A. Adhesion site composition of Shields, P.P.; Dixon, J.E.; Glembotski, C.C. The secretion of atrial
murine fibroblasts cultured on gelatin-coated substrata. J. Cell natriuretic factor-(99–126) by cultured cardiac myocytes is
Physiol. 120(2):117–125; 1984. regulated by glucocorticoids. J. Biol. Chem. 26:3126–3128; 1988.
Harary, I.; Farley, B. In vitro studies on single beating rat heart cells II Simpson, P.; Savion, S. Differentiation of myocytes in single cell
intercellular communication. Exp. Cell Res. 29:466–474; 1963. cultures with and without proliferating nonmyocardial cells. Circ.
Healy, G.M.; Parker, R.C. Cultivation of mammalian cells in defined Res. 50:101–116; 1982. Circ. Res. 50:101–116; 1982.
media with protein and nonprotein supplements. J. Cell Biol. 30 Song, W.; Lu, X.; Feng, Q. Tumor necrosis factor-alpha induces
(3):539–553; 1966. apoptosis via inducible nitric oxide synthase in neonatal mouse
Kruppenbacher, J.P.; May, T.; Eggers, H.J.; Piper, H.M. Cardiomyo- cardiomyocytes. Cardiovasc. Res. 45(3):595–602; 2000.
cytes of adult mice in long-term culture. Naturwissenschaften Wang, G.W.; Kang, Y.J. Inhibition of doxorubicin toxicity in cultured
80:132–134; 1993. neonatal mouse cardiomyocytes with elevated metallothionein
Limaye, D.A.; Shaikh, Z.A. Cytotoxicity of cadmium and characteristics levels. J. Pharmacol. Exp. Ther. 288(3):938–944; 1999.
of its transport in cardiomyocytes. Toxicol. Appl. Pharmacol. 154 Yamashita, N.; Nishida, M.; Hoshida, S.; Kuzuya, T.; Hori, M.;
(1):59–66; 1999. Taniguchi N.; et al. Induction of manganese superoxide dis-
Mark, G.E.; Strasser, F.F. Pacemaker activity and mitosis in cultures of mutase in rat cardiac myocytes increases tolerance to hypoxia
newborn rat heart ventricle cells. Exp. Cell Res. 44:217–233; 1966. 24 h after preconditioning. J. Clin. Invest. 94:2193–2199; 1994.

You might also like