ArticlePDF Available

Abstract and Figures

Despite well-documented cardiotoxic effects, doxorubicin remains a major anticancer agent. To study the role of myocardial apoptosis follow- ing doxorubicin administration, male Wistar rats were exposed to 1.25, 2.5, and 5 mg/kg of i.p. doxorubicin and terminated on days 1-7 in groups of five. Doxorubicin caused a significant (P < 0.001) and dose-dependent induction of cardiomyocyte apoptosis at 24 - 48 h after the injection. Repeated injections of 2.5 mg/kg given every other day resulted in peaks of apoptosis at 24 h after each injection. However, no additive effect of repeated dosing was noted. In histological samples, alterations in the cytoskeletal apparatus with focal loss of contractile elements were seen after a single injection. Myocyte necrosis was absent. Thus, acute doxo- rubicin-induced cardiotoxicity involves cardiomyocyte apoptosis, a poten- tially preventable form of myocardial tissue loss.
Content may be subject to copyright.
2000;60:1789-1792. Cancer Res
Olli J. Arola, Antti Saraste, Kari Pulkki, et al.
Apoptosis
Acute Doxorubicin Cardiotoxicity Involves Cardiomyocyte
Updated version
http://cancerres.aacrjournals.org/content/60/7/1789
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/60/7/1789.full.html#ref-list-1
This article cites by 20 articles, 3 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/60/7/1789.full.html#related-urls
This article has been cited by 49 HighWire-hosted articles. Access the articles at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
on June 5, 2013. © 2000 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
[CANCER RESEARCH 60, 1789–1792, April 1, 2000]
Advances in Brief
Acute Doxorubicin Cardiotoxicity Involves Cardiomyocyte Apoptosis
1
Olli J. Arola, Antti Saraste, Kari Pulkki, Markku Kallajoki, Martti Parvinen, and Liisa-Maria Voipio-Pulkki
2
Departments of Medicine [O. J. A., L-M. V-P.], Anatomy [A. S., M. P.], Clinical Chemistry [K. P.], and Pathology [M. K.], University of Turku, FIN-20521 Turku, Finland
Abstract
Despite well-documented cardiotoxic effects, doxorubicin remains a
major anticancer agent. To study the role of myocardial apoptosis follow-
ing doxorubicin administration, male Wistar rats were exposed to 1.25,
2.5, and 5 mg/kg of i.p. doxorubicin and terminated on days 1–7 in groups
of five. Doxorubicin caused a significant (P<0.001) and dose-dependent
induction of cardiomyocyte apoptosis at 24–48 h after the injection.
Repeated injections of 2.5 mg/kg given every other day resulted in peaks
of apoptosis at 24 h after each injection. However, no additive effect of
repeated dosing was noted. In histological samples, alterations in the
cytoskeletal apparatus with focal loss of contractile elements were seen
after a single injection. Myocyte necrosis was absent. Thus, acute doxo-
rubicin-induced cardiotoxicity involves cardiomyocyte apoptosis, a poten-
tially preventable form of myocardial tissue loss.
Introduction
The anthracycline antibiotic DOX
3
is an important antineoplastic
agent because of its high antitumor efficacy in hematological as well
as in solid malignancies. Its use is limited by the not infrequent
induction of dose-dependent chronic cardiomyopathy. The mecha-
nisms of DOX cardiotoxicity include (a) the formation of free reactive
oxygen radicals, (b) direct DNA damage and/or interference with
DNA repair (1, 2), and (c) induction of immune reactions involving
antigen-presenting cells in the heart (3). In addition to nucleic acids
and cellular membranes, the cytotoxic action by anthracyclines in-
volves the cytoskeleton of both tumor cells and cardiomyocytes (4).
Cytoskeletal changes following DOX administration include reduc-
tion in the density of myofibrillar bundles (5), alterations on the Z-disc
structure, and disarray and depolymerization of actin filaments (6, 7).
Histologically, cytoplasmic vacuolization due to dilation of the sar-
cotubules and loss of myofibrils characterize DOX cardiomyopathy
(8). Recently, myocardial apoptosis has been suggested as a common
mechanism of acute and chronic myocyte loss (9–11). DOX induces
apoptosis in several cell lines (12) and, in a rat model, in the kidney
and the intestine (13). Whether DOX-induced cardiotoxicity involves
CA remains unclear. In a rat model, no CA was observed in samples
obtained after a 12-week cumulative DOX treatment (13). In human
heart samples, however, contraction and ring formation of the nuclei
suggestive of apoptosis have been documented within 24 h after DOX
injection (14). Furthermore, apoptosis-type cell morphology with
shrunken focal cardiomyocytes has been observed in connection with
myofibrillar lysis in the classic study by Billingham et al. (7). We
therefore designed an experimental study to investigate whether acute
DOX administration induces CA in vivo, the possible dose-response
of such an acute effect, and the relationship of CA to ensuing histo-
logical myocardial damage typical of anthracycline cardiomyopathy.
Materials and Methods
Experimental Protocol. Adult male Wistar rats weighing 300–470 g (total
n110) were purchased from the Central Animal Laboratory, Turku Uni-
versity, Finland. The rats had free access to standard rodent chow and water.
All procedures for animal care and housing were in compliance with the
contemporary guidelines. The Laboratory Animal Committee and the Social
and Health Department of the Provincial State Office of Western Finland
approved the experimental protocol. DOX was obtained from Pharmacia SPA
(Milan, Italy). DOX cardiomyopathy was induced as described by Iliskovic
and Singal (15). In brief, after rats received s.c. buprenorphin (0.05 mg/kg;
Reckitt & Colman, Hull, England) to provide analgesia, the first 2.5 mg/kg
dose of DOX was injected i.p., and the animals were allowed to recover for
48 h before the procedure was repeated as determined by the subprotocols.
Control animals (n10) were treated with only buprenorphin and i.p. saline.
To first study the occurrence of CA after a single versus multiple DOX
injections, a pilot study with 15 animals was performed. The rats were given
2.5 mg/kg of DOX every other day up to a maximal cumulative dose of 15
mg/kg over 2 weeks. The rats were killed with carbon dioxide on days 1, 3, and
14 after the first injection and on days 1, 7, and 18 after the last DOX injection
(n3–5 in each group).
Being guided by the results from the pilot study, we then studied the early
time course and dose-dependence of apoptosis occurrence. Groups of 15–25
rats received single injections of 1.25, 2.5, or 5 mg/kg of DOX. The rats were
killed in groups of five on days 1, 2, and 3 after the injection. For the 2.5 mg/kg
injections, rats were also killed at 6 h and on day 7. Finally, to study the effects
of consecutive injections, three injections of 2.5 mg/kg each were given every
other day. Ten rats were killed on the day after each injection, and five rats
were killed on days 2 and 3 after the respective injections.
Immediately post mortem, the entire heart was excised. The heart was sliced
transversally, and a midventricular slice was fixed in neutral-buffered 10%
formalin for 24 h, embedded in paraffin, and cut into 5-
m sections for
assessment of apoptosis and histological features.
In Situ Assay for Apoptosis. Apoptotic cardiomyocytes were detected
with the TUNEL assay and previously described methodology (9, 11, 16).
In brief, nuclear DNA strand breaks were end-labeled with digoxigenin-
conjugated dideoxy-UTP by terminal transferase and visualized immuno-
histochemically with digoxigenin antibody conjugated to alkaline phospha-
tase. The assay was carefully standardized using adjacent tissue sections of
each sample, which were pretreated with DNase I (1 unit/ml for 15 min at
37°C; positive control of DNA breaks). The staining of each section was
interrupted when intense positivity in the DNase I-treated section appeared.
This approach provides optimal sensitivity for double-strand DNA breaks
and normalizes the results for differences in tissue permeability of the
reagents. The cardiomyocyte origin of TUNEL-positive nuclei was identi-
fied by the presence of surrounding myofilaments. The validity of this
approach was confirmed by immunofluorescence staining with antimyosin
(cat. no. M-8421; Sigma, St. Louis, MO) of randomly selected TUNEL-
stained sections. To further confirm the specificity of TUNEL staining in
respect to apoptosis, attention was paid to the morphology of positive
nuclei and cells.
Quantification of Apoptotic Cells. The number of apoptotic cardiomyo-
cytes in TUNEL-stained sections was counted by use of light microscopy with
an ocular grid (250 magnification; area of the field, 0.25 mm
2
). An average
of 184 and 210 microscopic fields were analyzed per DOX-treated and control
animal, respectively. The average number of cardiomyocytes per field was
Received 11/1/99; accepted 2/15/00.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance with
18 U.S.C. Section 1734 solely to indicate this fact.
1
This study was supported financially by the Aarno Koskelo Foundation, Turku
University Foundation, and Finnish Cultural Foundation.
2
To whom requests for reprints should be addressed, at Department of Medicine,
Turku University Central Hospital, P.O. Box 52, 20521 Turku, Finland. E-mail:
olli.arola@utu.fi.
3
The abbreviations used are: DOX, doxorubicin; CA, cardiomyocyte apoptosis;
TUNEL, terminal transferase-mediated DNA nick end labeling.
1789
on June 5, 2013. © 2000 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
287 42 (mean SD) in DOX-treated animals and 320 45 in controls. The
results are expressed as the percentage of apoptotic nuclei of the total number
of labeled cardiomyocyte nuclei, which were counted in the corresponding
DNase I-treated section. No apoptotic nuclei in the interstitial space or in the
endothelial lining of blood vessels were found. The correlation between the
results of repeated procedures with this method is 0.88 (P0.01; Ref. 11).
Histological Analysis. Formalin-fixed, paraffin-embedded heart tissue sec-
tions were stained with van Gieson to demonstrate general histological fea-
tures. To visualize the cytoskeletal elements, formalin-fixed, paraffin-embed-
ded sections were stained for the intermediate filament protein desmin.
Sections were first deparaffinized with xylol, and then boiled twice for 5 min
in a microwave oven in 10 mMcitrate buffer (pH 6.0). The primary antibody
mouse antidesmin (Zymed Laboratories Inc., San Francisco, CA) was diluted
1:30, and the bound antibody was detected with a biotin-avidin-peroxidase
complex (Vectastain ABC kit; Vector Laboratories Inc., Burlingame, CA). The
sections were counterstained with hematoxylin.
Statistical Analysis. Quantitative results are expressed as means SD.
Percentages of apoptotic cardiomyocytes between DOX-treated and control
rats were compared using one-way ANOVA followed by Dunnett’s two-sided
ttest. In the case of multiple comparisons (one dose versus multiple doses,
different dosages, or consecutive injections) the least significant difference
post hoc test with Bonferroni correction was used. The software used was
Fig. 1. van Gieson-stained sections of rat myocardium from a control animal (A) and 24 h after a single injection of 2.5 mg/kg DOX (B). Note the vacuolization of cardiomyocytes
(arrows) and edema ()inB.C, dark brown-colored TUNEL-positive nucleus in a sample obtained 24 h after injection of 2.5 mg/kg DOX. Note the condensation of the nucleus and
shrunken appearance of the cell. D, double staining with antimyosin shows the same nucleus located within the cardiomyocyte. E–G, immmunohistochemical staining for desmin. E,
control rat myocardium; F, myocardium after a single injection of 2.5 mg/kg DOX; G, myocardium after the cumulative DOX dose of 15 mg/kg and a follow-up for 7 days. Note the
gradual decrease in the intensity of staining and disorganization of cross-striated staining pattern (). Magnification: A,B,E–G,200; bar 50
m; Cand D,1000; bar 10
m.
1790
DOXORUBICIN AND CARDIOMYOCYTE APOPTOSIS
on June 5, 2013. © 2000 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
SPSS 8.0 for Windows (SPSS Inc, Chicago, IL). Differences were considered
significant at P0.05.
Results
General Effects and Histological Analysis. After the total cumu-
lative DOX dose of l5 mg/kg over 2 weeks (pilot study), attenuation
in weight gain and signs of clinical DOX-induced cardiomyopathy
(ascites, hepatic enlargement, and some degree of general lethargy)
were observed as originally described by Iliskovic and Singal (15).
The clinical cardiomyopathy syndrome became gradually more evi-
dent during the 18-day observation time following the last injection;
in particular, greater ascites formation was noted. The pilot study
revealed the presence of TUNEL positivity in myocardial samples
obtained early during the course of DOX treatment. However, in
samples obtained 1–2 weeks after the cumulative treatment, the num-
ber of TUNEL-positive cells declined and was not significantly dif-
ferent from controls (data not shown).
In the van Gieson-stained light microscopic sections, alterations
consistent with DOX-induced toxicity were seen: cardiomyocyte vac-
uolization (Fig. 1, Aand B) was present after a single injection of 2.5
mg/kg DOX. Notably, necrotic cardiomyocytes were not seen. Neither
polymorphonuclear inflammatory cell infiltration nor lymphocyte in-
filtration was present.
Immunohistochemical staining of desmin, which is an intermediate
filament protein important in the anchorage of the contractile elements
in cardiac myocytes (17), was profoundly altered after a single injec-
tion of DOX. The uniformly intense staining for desmin vanished, and
the normal cross-striation of the cardiomyocytes (Fig. 1E) disappeared
in a focal manner (Fig. 1F). After the cumulative 15 mg/kg of DOX
and follow-up for 7 days, the desmin staining showed even more
striking changes: the staining of cardiomyocytes appeared patchy; and
the sarcomeres and intercalated discs were hardly visible. These
heavily damaged cells were, however, often adjacent to cells that
appeared normal.
Single DOX Injection: Time Course and Dose-dependent In-
duction of TUNEL Positivity. Microscopic examination of the
TUNEL-stained sections showed the presence of sparse single posi-
tive nuclei scattered across the ventricular wall. These were sur-
rounded by cardiomyocyte myofilaments (Fig. 1C) as visualized by
double staining with antimyosin (Fig. 1D). In general, TUNEL-
positive cardiomyocytes showed condensation of nuclei and,
sometimes, shrinking of the cytoplasm consistent with apoptotic mor-
phology (Fig. 1, Cand D). Only very few TUNEL-positive inflam-
matory and other non-myocyte cells were observed equally in all
hearts, including the control samples.
TUNEL-positive CA was a very rare event in control animals
[0.0065 0.0022% (mean SD)]. A significant induction of CA to
0.033 0.012% (P0.001) of total cardiomyocytes was seen at 24 h
after a single 2.5 mg/kg injection of DOX. The percentage of CA
peaked on the first day after the injection and declined thereafter (Fig.
2A). The onset of TUNEL positivity was very rapid: at 6 h, the mean
percentage was increased to 0.034 0.027% (P0.05). The gradual
decline of TUNEL positivity continued at day 7, confirming prelim-
inary observations from the pilot study.
In response to 1.25, 2.5, and 5 mg/kg DOX, a dose-dependent
induction of CA was observed. As shown in Fig. 2B, the highest
relative percentages of apoptotic cardiomyocytes were observed on
day 1 after the 1.25 and 2.5 mg/kg doses and on day 2 after 5.0 mg/kg.
Compared with sham-injected controls (day 0; 0.0065 0.0022%),
the peak amount of TUNEL-positive nuclei was statistically signifi-
cantly increased after the 2.5 mg/kg (0.033 0.012%; P0.001) and
Fig. 2. A, time course of apoptosis occurrence after a single injection of 2.5 mg/kg DOX. B, dose-dependent induction of apoptosis following single injections of 1.25, 2.5, and 5.0
mg/kg DOX.
Fig. 3. Three consecutive injections of 2.5 mg/kg DOX administered 1 day apart. Note
that the increase of apoptosis is cumulative but not additive. Arrows indicate injections.
1791
DOXORUBICIN AND CARDIOMYOCYTE APOPTOSIS
on June 5, 2013. © 2000 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
5.0 mg/kg (0.066 0.020%; P0.001). A gradual decline of
TUNEL positivity was observed on day 3, but the percentages still
remained above control values.
Consecutive Injections. Fig. 3 shows the amount of CA after three
consecutive injections of 2.5 mg/kg DOX administered 1 day apart.
After each injection, a new increase in the mean amount of CA was
observed (0.033, 0.042, and 0.049% on days 1, 3, and 5, respectively;
Fig. 3). Notably, the effect of each new DOX dose was attenuated so
that the relative increase was 7-fold after the first injection, 33% after
the second injection, but only 16% after the third injection.
Discussion
Our data demonstrate a significant induction of CA in the early
phase following DOX administration. The percentage of TUNEL-
positive cells peaked within 2448 h after a single injection, followed
by a gradual decline to baseline levels by day 7 after the moderate 2.5
mg/kg dose. The number of TUNEL-positive cardiomyocytes also
declined rapidly during the follow-up after a cumulative high-dose
DOX treatment. Thus, our findings are not inconsistent with those
obtained by Zhang et al. (13), who could not demonstrate CA after
prolonged (42–84 days) DOX administration and follow-up.
The fact that DOX is toxic to and interferes with DNA raises
concern regarding the specificity of the TUNEL assay in assessing
apoptosis as a mechanism of DOX cardiotoxicity. The validity of our
method has been discussed previously in detail (9, 11, 16). In brief,
the standardization procedure using DNase I-treated control sections
results in optimization of the TUNEL assay for apoptosis-specific
double-strand DNA breaks. During microscopic examination, typical
nuclear and cellular morphological features of apoptosis were found
in association with TUNEL positivity. Recently, induction of signif-
icant CA by DOX in a rat ventricular cell line was reported by Delpy
et al. (18). A protective effect by thermal preconditioning in this in
vitro model was also described by Ito et al. (19). In addition to the
TUNEL assay, these researchers used also agarose DNA gel electro-
phoresis (DNA ladder assay). In our experience, DNA ladders are
only demonstrable in the TUNEL-positive areas when the amount of
positive cells exceeds 0.04% (9).
A theoretical basis for reduced toxicity by fractional DOX dosing is
provided by the observation that the single high dose of 5 mg/kg
increased TUNEL positivity more (0.066 0.020%) than did two 2.5
mg/kg injections (0.051 0.018%). The induction of TUNEL posi-
tivity was cumulative after repeated injections, but the relative in-
crease was blunted. This further supports dose fractionation and
suggests induction of protective mechanisms. Because the number of
apoptotic cardiomyocytes was reduced to nonsignificant levels 3 days
after the cumulative dose was achieved, induction of apoptosis by
DOX seems to be an acute effect in DOX cardiotoxicity. Cardio-
myocyte necrosis is characterized by swelling of the cell, which
eventually leads to leakage of cellular components, causing an inflam-
matory response in adjacent areas. In principle, cells may proceed to
or transform to a necrotic type of cell death. However, immediate or
delayed histological signs of necrosis did not develop in our model
even after a dose considered to be pharmacological or after three
repeated injections.
Myocardial apoptosis is not, however, the only mechanism of
deteriorating contractile force because the remaining myocardium is
also dysfunctional. In addition to the apparent loss of cellular ele-
ments, our data show that the very first injection of DOX induces
changes in the cytoskeleton and in the contractile element of the
cardiomyocyte. The rapid damage observed in the cytoskeleton is
likely to cause myocardial dysfunction and contribute to clinical heart
failure syndrome (20). Whether this effect is due to direct DOX
toxicity or a failure in protein synthesis reflecting DNA damage
remains unclear. These cytoskeletal interferences are likely to favor
cell shrinkage and affect tension development (20). This effect can
modify or even speed up apoptosis.
Attenuation of apoptotic cell death by a caspase inhibitor has been
demonstrated in a myocardial reperfusion injury model. The caspase
inhibitor pretreatment effect was achieved in cells receiving a signal
that usually promotes apoptosis (21). In cultured cardiomyocytes,
exposition to thermal preconditioning before DOX administration
attenuates apoptosis occurrence (19). Thus, myocardial apoptosis is a
potentially modifiable and preventable form of myocardial tissue loss.
The acute cardiotoxicity of DOX is multifactorial. The very first
injection of DOX alters the organization of the cardiomyocyte; in
addition, an early induction of apoptosis is observed. This potentially
novel mechanism is transient, but it may be of key importance to the
ensuing heart failure. Inhibition and modification of this mechanism
warrants further study.
References
1. Gianni, L., and Myers, C. E. The role of free radical formation in the cardiotoxicity
of anthracycline. In: F. M. Muggia, M. D. Green, and J. L. Speyer (eds.), Cancer
Treatment and the Heart, pp. 9–46. Baltimore: The John Hopkins University Press,
1992.
2. Dorr, R. T. Cytoprotective agents for anthracyclines. Semin. Oncol., 23 (Suppl. 8):
23–34, 1996.
3. Zhang, J., Clark, J. R., Herman, E. H., and Ferrans, V. J. Dendritic cells in the hearts
of spontaneously hypertensive rats treated with doxorubicin with or without ICRF-
187. Am. J. Pathol., 142: 1916–1926, 1993.
4. Molinari, A., Calcabrini, A., Crateri, P., and Arancia, G. Interaction of anthracyclinic
antibiotics with cytoskeletal components of cultured carcinoma cells (CG5). Exp.
Mol. Pathol., 53: 11–33, 1990.
5. Jaenke, R. S. An anthracycline antibiotic-induced cardiomyopathy in rabbits. Lab.
Investig., 30: 292–304, 1974.
6. Lewis, W., and Gonzales, B. Anthracycline effects on actin and actin-containing thin
filaments in cultured neonatal rat myocardial cells. Lab. Investig., 54: 416–423,
1986.
7. Billingham, M. E., Mason, J. W., Bristow, M. R., and Daniels, J. R. Anthracycline
cardiomyopathy monitored by morphologic changes. Cancer Treat. Rep., 62: 865–
872, 1978.
8. Lefrak, E. A., Pitha, J., Rosenheim, S., and Gottlieb, J. A. A clinicopathologic
analysis of Adriamycin cardiotoxicity. Cancer (Phila.), 32: 302–314, 1973.
9. Saraste, A., Pulkki, K., Henriksen, K., Kallajoki, M., Parvinen, M., and Voipio-
Pulkki, L-M. Apoptosis in human acute myocardial infarction. Circulation, 95:
320–323, 1997.
10. Olivetti, G., Abbi, R., Quaini, F., Kajstura, J., Cheng, W., Nitahara, J. A., Quaini, E.,
di Loreto, C., Beltrami, C. A., Krajewski, S., Reed, J. C., and Anversa, P. Apoptosis
in the failing human heart. N. Engl. J. Med., 336: 1131–1141, 1997.
11. Saraste, A., Pulkki, K., Kallajoki, M., Heikkila¨, P., Laine, P., Mattila, S., Nieminen,
M. S., Parvinen, M., and Voipio-Pulkki, L-M. Cardiomyocyte apoptosis and progres-
sion of heart failure to transplantation. Eur. J. Clin. Investig., 29: 380–386, 1999.
12. Ling, Y. H., Priebe, W., and Perez-Soler, R. Apoptosis induced by anthracycline
antibiotics in P388 parent and multidrug-resistant cells. Cancer Res., 53: 1845–1852,
1993.
13. Zhang, J., Clark, J. R., Herman, E. H., and Ferrans, V. J. Doxorubicin-induced
apoptosis in spontaneously hypertensive rats: differential effects in heart, kidney and
intestine, and inhibition by ICRF-187. J. Mol. Cell. Cardiol., 28: 1931–1943, 1996.
14. Unverferth, B. J., Magorien, R. D., Balcerzak, S. P., Leier, C. V., and Unverferth,
D. V. Early changes in human myocardial nuclei after doxorubicin. Cancer (Phila.),
52: 215–221, 1983.
15. Iliskovic, N., and Singal, P. K. Lipid lowering: an important factor in preventing
Adriamycin-induced heart failure. Am. J. Pathol., 150: 727–734, 1997.
16. Saraste, A. Morphologic criteria and detection of apoptosis. Herz, 24: 189–195, 1999.
17. Lockard, V. G., and Bloom, S. Trans-cellular desmin-lamin B intermediate filament
network in cardiac myocytes. J. Mol. Cell. Cardiol., 25: 303–309, 1993.
18. Delpy, E., Hatem, S. N., Andrieu, N., Ru¨cker-Martin, C., Jaffre´zou, J-P., Levade, T.,
Laurent, G., and Mercadier, J-J. The anthracycline doxorubicin induces ceramide
accumulation and apoptosis in rat ventricular myocytes. Eur. Heart J., 19: 259, 1998.
19. Ito, H., Shimojo, T., Fujisaki, H., Tamamori, M., Ishiyama, S., Adachi, S., Abe, S.,
Marumo, F., and Hiroe, M. Thermal preconditioning protects rat cardiac muscle cells
from doxorubicin-induced apoptosis. Life Sci., 64: 755–761, 1999.
20. Chen, J., and Chien, K. R. Complexity in simplicity: monogenic disorders and
complex cardiomyopathies. J. Clin. Investig., 103: 1483–1485, 1999.
21. Yaoita, H., Ogawa, K., Machara, K., and Maruyama, Y. Attenuation of ischaemia/
reperfusion injury in rats by a caspase inhibitor. Circulation, 97: 276–281, 1998
1792
DOXORUBICIN AND CARDIOMYOCYTE APOPTOSIS
on June 5, 2013. © 2000 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from
... Bnip3 was found to be a key effector of DOX-induced mitochondrial damage and cardiomyocyte necrosis, and it was highly enriched in DOX-treated cardiomyocytes, and knockdown or reduction of Bnip3 prevented DOX-induced cell necrosis [49]. Cardiomyocyte apoptosis is a key process in the occurrence and development of DOX-induced cardiotoxicity [50,51]. Inhibition of apoptosis may provide a new way to alleviate DOX-related cardiotoxicity [52]. ...
Article
Full-text available
Aims This study aimed to evaluate the global research trend in the prevention and treatment of cardiotoxicity caused by anthracyclines from 2000 to 2023, and to explore international cooperation, research hotspots, and frontier trends. Methods The articles on the prevention and treatment of anthracycline-induced cardiotoxicity published from 2000 to 2023 were searched by Web of Science. The bibliometrics software CiteSpace was used for visual analysis of countries, institutions, journals, authors, cited authors, cited references, and keywords. Results This study analyzed the current status of global research on the prevention and treatment of cardiotoxicity caused by anthracyclines. A total of 3,669 papers were searched and 851 studies were included. The number of publications increased gradually throughout the years. Cardiovascular Toxicology (15) is the journal with the most publications. Circulation (547) ranked first among cited journals. In this field, the country with the most publications is the United States (229), and the institution with the most publications is Charles Univ Prague (18). In the analysis of the authors, Tomas S (10) ranked first. Cardinale D (262) ranked first among cited authors. In the ranking of cited literature frequency, the article ranked first is “Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy” (121). The keywords “heart failure” (215) and “oxidative stress” (212) were the most frequent. “Enalapril”, “inflammation”, “cell death”, “NF-κB” and “Nrf2” were the advanced research contents in 2019–2023. Conclusions This study provided valuable information for cardio-oncology researchers to identify potential collaborators and institutions, discover hot topics, and explore new research directions. The prevention and treatment of anthracycline-induced cardiotoxicity focuses on early detection and timely treatment. The results of the current clinical studies on the treatment of anthracycline-induced cardiotoxicity are contradictory, and more studies are needed to provide more reliable clinical evidence in the future.
... S2A-S2C). Due to the role of reactive oxygen species (ROS) and DNA damage in the doxorubicin-induced cardiotoxicity [24,25], we measured the levels of ROS and γ-H2AX, a DNA double-strand break marker, in the cardiomyocytes. We found that doxorubicin increased ROS levels and γ-H2AX expressions in vivo ( Fig. 3A and C) and in vitro (Fig. 3B and D), which were reversed by semaglutide (Fig. 3A-D). ...
Article
Full-text available
Aims Doxorubicin is a powerful chemotherapeutic agent for cancer, whose use is limited due to its potential cardiotoxicity. Semaglutide (SEMA), a novel analog of glucagon-like peptide-1 (GLP-1), has received widespread attention for the treatment of diabetes. However, increasing evidence has highlighted its potential therapeutic benefits on cardiac function. Therefore, the objective of this study was to examine the efficacy of semaglutide in ameliorating doxorubicin-induced cardiotoxicity. Methods and results Doxorubicin-induced cardiotoxicity is an established model to study cardiac function. Cardiac function was studied by transthoracic echocardiography and invasive hemodynamic monitoring. The results showed that semaglutide significantly ameliorated doxorubicin-induced cardiac dysfunction. RNA sequencing suggested that Bnip3 is the candidate gene that impaired the protective effect of semaglutide in doxorubicin-induced cardiotoxicity. To determine the role of BNIP3 on the effect of semaglutide in doxorubicin-induced cardiotoxicity, BNIP3 with adeno-associated virus serotype 9 (AAV9) expressing cardiac troponin T (cTnT) promoter was injected into tail vein of C57/BL6J mice to overexpress BNIP3, specifically in the heart. Overexpression of BNIP3 prevented the improvement in cardiac function caused by semaglutide. In vitro experiments showed that semaglutide, via PI3K/AKT pathway, reduced BNIP3 expression in the mitochondria, improving mitochondrial function. Conclusion Semaglutide ameliorates doxorubicin-induced mitochondrial and cardiac dysfunction via PI3K/AKT pathway, by reducing BNIP3 expression in mitochondria. The improvement in mitochondrial function reduces doxorubicin-mediated cardiac injury and improves cardiac function. Therefore, semaglutide is a potential therapy to reduce doxorubicin-induced acute cardiotoxicity.
... Apoptosis of cardiomyocytes is a critical aspect of the cardiac damage induced by anthracyclines. Recent studies underline the significant role of this apoptosis in the development of drug-induced cardiomyopathy [2][3][4]. To counteract the oxidative stress provoked by anthracycline medications, the use of antioxidants, whether natural or synthetic, has been proposed. ...
Article
Full-text available
Anthracyclines, including epirubicin (Epi), are effective chemotherapeutics but are known for their cardiotoxic side effects, primarily inducing cardiomyocyte apoptosis. This study investigates the protective role of hesperetin (HSP) against cardiomyopathy triggered by Epi in a murine model. Male CD1 mice were divided into four groups, with the Epi group receiving a cumulative dose of 12 mg/kg intraperitoneally, reflecting a clinically relevant dosage. The co-treatment group received 100 mg/kg of HSP daily for 13 days. After the treatment period, mice were euthanized, and heart tissues were collected for histopathological, immunofluorescence/immunohistochemistry, and transmission electron microscopy (TEM) analyses. Histologically, Epi treatment led to cytoplasmic vacuolization, myofibril loss, and fiber disarray, while co-treatment with HSP preserved cardiac structure. Immunofluorescent analysis of Bcl-2 family proteins revealed Epi-induced upregulation of the pro-apoptotic protein Bax and a decrease in anti-apoptotic Bcl-2, which HSP treatment reversed. TEM observations confirmed the preservation of mitochondrial ultrastructure with HSP treatment. Moreover, in situ detection of DNA fragmentation highlighted a decrease in apoptotic nuclei with HSP treatment. In conclusion, HSP demonstrates a protective effect against Epi-induced cardiac injury and apoptosis, suggesting its potential as an adjunctive therapy in anthracycline-induced cardiomyopathy. Further studies, including chronic cardiotoxicity models and clinical trials, are warranted to optimize its therapeutic application in Epi-related cardiac dysfunction.
... Previous studies suggested that oxidative stress and inflammation are the leading causes of DOX-induced cardiac injury [2]. Although the pathogenesis of DOX-induced acute cardiotoxicity is somewhat complex, there appears to be a direct link between apoptosis and cardiac injury [3]. Apoptosis is a vital physiological process that plays a crucial role in maintaining homeostasis in organisms [4]. ...
Article
Full-text available
Doxorubicin (DOX) possesses strong anti-tumor effects but is limited by its irreversible cardiac toxicity. The relationship between exercise, a known enhancer of cardiovascular health, and DOX-induced cardiotoxicity has been a focus of recent research. Exercise has been suggested to mitigate DOX's cardiac harm by modulating the Yes-associated protein (YAP) and Signal transducer and activator of transcription 3 (STAT3) pathways, which are crucial in regulating cardiac cell functions and responses to damage. This study aimed to assess the protective role of exercise preconditioning against DOX-induced cardiac injury. We used Sprague–Dawley rats, divided into five groups (control, DOX, exercise preconditioning (EP), EP-DOX, and verteporfin + EP + DOX), to investigate the potential mechanisms. Our findings, including echocardiography, histological staining, Western blot, and q-PCR analysis, demonstrated that exercise preconditioning could alleviate DOX-induced cardiac dysfunction and structural damage. Notably, exercise preconditioning enhanced the nuclear localization and co-localization of YAP and STAT3. Our study suggests that exercise preconditioning may counteract DOX-induced cardiotoxicity by activating the YAP/STAT3 pathway, highlighting a potential therapeutic approach for reducing DOX's cardiac side effects.
... DOX-induced cardiomyopathy involves a complex interplay of various cell death mechanisms. Among these, the induction of cardiomyocyte apoptosis is recognized as a signi cant contributor to the pathogenesis of DOX-induced cardiotoxicity [32,52], and the main cause of cardiac tissue loss and cardiac insu ciency during mitochondria-dependent intrinsic apoptosis [53]. Studies have reported that ...
Preprint
Full-text available
Background Liguzinediol (Lig) has emerged as a promising candidate for mitigating Doxorubicin (DOX)-induced cardiotoxicity, a significant limitation in the clinical application of this widely used antineoplastic drug known for its efficacy. This study aimed to explore the effects and potential mechanisms underlying Lig's protective role against DOX-induced cardiotoxicity. Methods C57BL/6 mice were treated with DOX. Cardiac function changes were observed by echocardiography. Cardiac structure changes were observed by HE and Masson staining. Immunofluorescence was applied to visualize the cardiomyocyte apoptosis. Western blotting was used to detect the expression levels of AMPK, SIRT3, Caspase-3 and GSDME. These experiments confirmed that Liguzinediol had a ameliorative effect on DOX-induced cardiotoxicity in mice. Results The results demonstrated that Lig effectively countered myocardial oxidative stress by modulating intracellular levels of reactive oxygen species (ROS), malondialdehyde (MDA), and superoxide dismutase (SOD). Lig reduced levels of creatine kinase (CK), lactate dehydrogenase (LDH), and ameliorated histopathological changes while improving electrocardiogram profiles in vivo. Furthermore, the study revealed that Lig activated the AMP-activated protein kinase (AMPK)/sirtuin 3 (SIRT3) pathway, thereby enhancing mitochondrial function and attenuating myocardial cell apoptosis. In experiments with H9C2 cells treated with DOX, co-administration of the AMPK inhibitor compound C (CC) led to a significant increase in intracellular ROS levels. Lig intervention reversed these effects, along with the downregulation of gasdermin E N-terminal fragment (GSDME-N), interleukin-1β (IL-1β), and interleukin-6 (IL-6), suggesting a potential role of Lig in mitigating Caspase-3/GSDME-mediated pyroptosis. Conclusions The findings of this study suggest that Lig effectively alleviates DOX-induced cardiotoxicity through the activation of the AMPK/SIRT3 pathway, thereby presenting itself as a natural product with therapeutic potential for preventing DOX-associated cardiotoxicity. This novel approach may pave the way for the development of alternative strategies in the clinical management of DOX-induced cardiac complications.
... Recent research has shown that Doxorubicin induces apoptosis in the rat heart [79,80], and these processes have been linked, at least partially, to mitochondria-mediated pathways [81,82]. This underscores the significance of understanding the molecular mechanisms underlying doxorubicin-induced cell death, particularly within mitochondrial function and the delicate balance between pro-and antiapoptotic factors. ...
Article
Full-text available
One of the significant side effects of doxorubicin treatment for cancer patients is cognitive impairment, and the mechanisms underlying this impairment must be investigated to treat or prevent it. The current study investigates the impact of virgin coconut oil and carvedilol administration on neurobehaviour brain apoptotic and inflammatory markers in doxorubicin-treated mice. 32 male and 32 female mice were randomly assigned to four groups of 8 animals each, and the treatment lasted for twenty-eight days. Group 1 animals in both the male and female groups were the controls. In comparison, the Group 2 animals in both groups received doxorubicin dosage (3.75mg/kg body weight) intraperitoneally weekly as a single dose on days 5, 12, 19 and 26 to make up for the desired amount (15 mg/kg body weight). Group 3 animals received doxorubicin and were orally treated with virgin coconut oil (5ml/kg body weight) for 28 days. Group 4 animals received doxorubicin and were treated with carvedilol (5 mg/kg body weight) weekly for three days (days 5-7 for four weeks). The histological analysis of the brain tissue was done by staining the tissues with haematoxylin and eosin. The data was analyzed using GraphPad Prism 9.0. Analysis of Variance (ANOVA) was used to compare between groups. All results were presented as mean±SEM. Both virgin coconut oil and carvedilol demonstrated ameliorative effects on neurobehaviour and apoptotic and inflammatory brain markers.
... Ключевыми молекулярными медиаторами пироптоза являются каспазы и Bnip3 [77]. В то время как роль апоптоза и некроза в доксорубициновой кардиотоксичности достаточно хорошо исследована [75,78], аутофагия кардиомиоцитов в генезе их гибели в условиях лечения доксорубицином остается предметом дискуссии [79][80][81][82]. В последние несколько лет ряд исследований продемонстрировали, что доксорубицин ослабляет аутофагический поток на нескольких этапах, включающих формирование аутофагосом и лизосомальную деградацию посредством Akt-зависимого и Akt-независимого механизма [83][84][85]. ...
Article
Introduction . Long-term use of anthracyclines during cancer chemotherapy has been associated with the development of potentially life-threatening cardiotoxicity. Despite researches ongoing since the middle of the last century, approaches to the choice of therapy remain limited. Text . Doxorubicin currently is the most widely used chemotherapy. The leading side effect mechanism of the drug is the formation of reactive oxygen species in mitochondria with the mediated development of oxidative stress, which contributes to myocardial damage. However, despite the huge number of scientific papers devoted to various aspects of doxorubicin cardiotoxicity, its prevention and treatment, this issue requires detailed investigation in order to develop more advanced methods for early diagnosis and timely cardioprotective therapy. Conclusion . The current review discusses the pathogenetic mechanisms of cardiotoxicity associated with the use of doxorubicin chemotherapy. The pathogenesis of the cardiomyocytes death mechanism will provide an opportunity to develop new diagnostic and therapeutic approaches in the clinical practice.
... However, the clinical usefulness of doxorubicin is limited by a dose-related cardiac toxicity. Acute doxorubicin induced cardiotoxicity alters the organization of the cardiomyocytes and induces apoptosis , which is a potentially modifiable and preventable form of myocardial tissue loss (14 ) .Mitochondria have been identified as one of the targets in ADR-induced subcellular damage in heart tissues ( 15 ) . Doxorubicin is an anthracycline, which is widely used for the treatment of various cancers. ...
Article
Background : Doxorubicin based regimen is the most common treatment of breast cancer which is highly complicated by cardiotoxicity.Aim : To clarify the possible effects of carvedilol on echocardiographic ejection fraction and fraction shortening in doxorubicin based regimen in females with breast cancer.Patients and Methods : A total of 16 females with breast cancer were included in this study. The patients were randomized into 2 groups , 8 patients each. Group I included patients were treated with doxorubicin based regimen for 6 cycles with 21 day apart. Group II included patients were received doxorubicin based regimen with carvedilol 3.125 mg , orally , twice daily for 5 days , for 6 cycles. Echocardiography was done to measure ejection fraction and fraction shortening at zero time and 3 days after 2nd , 4th and 6th cycles.Results: Treatment with CAF regimen caused highly significant decrease in echocardiographic ejection fraction and fraction shortening after 2nd , 4th and 6th cycles in comparison to baseline readings ( P < 0.01 ). Combined CAF + Carvedilol 3.125 mg orally twice daily for 5 days caused highly significant increment in echocardiographic ejection fraction and fraction shortening compared with that of CAF regimen group ( P < 0.01 ).Conclusion: Carvedilol causes significant increase in echocardiographic ejection fraction and fraction shortening in doxorubicin treated patients.
Article
Full-text available
Aims The clinical application of doxorubicin (DOX), a potent anthracycline anticancer drug that effectively treats various malignancies, is limited by its side effects, such as cardiomyopathy. Apurinic/apyrimidinic endonuclease 1/redox factor‐1 (APE1/Ref‐1) is a multifunctional protein that can be secreted and is a promising target for the reduction of DOX‐induced inflammation and oxidative stress. We aimed to investigate the protective role of secretory APE1/Ref‐1 against DOX‐induced cardiac injury. Methods and results Designated adenoviral preprotrypsin‐leading sequence APE1/Ref‐1 (Ad‐PPTLS‐APE1/Ref‐1) was used to overexpress secretory APE1/Ref‐1 and assess its role in preventing DOX‐induced cardiomyopathy in vitro . Our findings revealed that exposure to secretory APE1/Ref‐1 significantly decreased N‐terminal pro‐B‐type natriuretic peptide levels in DOX‐treated H9C2 cells. In addition, secretory APE1/Ref‐1 reduced the severity of cardiomyocyte injury and apoptosis in both in vitro and in vivo DOX‐induced cardiotoxicity models. The observed cardioprotective effects of secretory APE1/Ref‐1 were mediated via inhibition of the p53 signalling pathway and enhancement of cell viability through attenuation of oxidative stress in DOX‐treated cardiomyocytes. Conclusions Our study provides evidence that secretory APE1/Ref‐1 has the potential to inhibit DOX‐induced cardiac toxicity by inhibiting oxidative stress and p53 related apoptosis both in vitro and in vivo . These findings suggest that secretory APE1/Ref‐1 supplementation is a promising strategy to attenuate DOX‐induced cardiomyocyte damage in a preclinical model. Further clinical investigations are essential to validate the therapeutic efficacy and safety of the intervention in human subjects.
Article
Full-text available
The effect of the topoisomerase II inhibitor doxorubicin and its non-cross-resistant analogue annamycin on DNA degradation and programmed cell death was examined in murine leukemia P388 cells. P388 parental cells exposed to various concentrations of doxorubicin and annamycin for 24 h displayed dose-dependent DNA cleavage: at 1 microM, both doxorubicin and annamycin were effective in inducing DNA breakdown, but at 10 microM, the effect was markedly decreased or totally absent. In multidrug-resistant P388/Dox cells, doxorubicin did not cause DNA cleavage, while 10 microM annamycin had a significant effect. By agarose gel analysis, drug-induced DNA fragmentation showed the characteristic pattern of internucleosomal ladder. Morphologically, P388 cells treated with 1 microM doxorubicin or annamycin for 24 h showed a reduction in cell volume and condensation of nuclear structures. Similar changes were observed in P388/Dox cells exposed to 10 microM annamycin for 24 h but not in cells exposed to 10 microM doxorubicin. Time course studies demonstrated that DNA fragmentation was detected 12 h after incubation with 1 microM doxorubicin or annamycin, while loss of membrane integrity appeared at 24 h, thus indicating that DNA degradation was a preceding event. DNA fragmentation caused by doxorubicin and annamycin was inhibited by the RNA synthesis inhibitor actinomycin D, the protein synthesis inhibitor cycloheximide, and the endonuclease inhibitor aurintricarboxylic acid. Drug-induced cell death was partially prevented by cycloheximide and aurintricarboxylic acid, thus suggesting that the apoptotic process caused by these drugs requires gene expression, synthesis of new proteins, and activation of endogenous nucleases. In contrast, DNA cleavage was not affected by incubating cells with 1 mM ethylene glycol-bis(2-aminoethyl ether)-N,N,N',N'-tetraacetic acid, thus indicating that intracellular calcium depletion does not affect anthracycline-induced apoptosis. The results obtained demonstrate that the cell killing effect of anthracyclines is mediated, at least in part, by the induction of apoptosis.
Article
Seventy-six endomyocardial biopsies obtained from 60 patients receiving adriamycin (ADM) and other anthracycline analogs were studied. The biopsies were studied by light and electron microscopy. Two main types of myocyte degeneration were consistently present, the lesions were focal, and inflammatory infiltrate was absent. The severity of pathologic changes was graded on a scale from 0 (normal) to 3 (marked abnormality). Twelve patients receiving previous mediastinal irradiation (600-5700 rads) showed a mean pathology grade (2.0 +/- 0.89) that was significantly higher than in those patients receiving a comparable dose of ADM but who were not irradiated (1.18 +/- 0.23) (P less than 0.01). This study indicated that radiation, even if remote, enhances ADM-induced cardiotoxicity and evokes a "recall" phenomenon of latent acute irradiation changes. Our data suggest that a specific, progressive, subclinical injury to the heart occurs with anthracycline therapy that cannot be detected reliably by conventional tests. Anthracycline-induced cardiotoxicity in rabbits, monkeys, and dogs shows the same basic cellular lesions as in man. The analogs, adria-DNA and rubidazone, also show lesions similar to those produced by ADM in the human heart. The endomyocardial biopsy is a reliable method for monitoring cardiac damage due to anthracyclines in man.
Article
The effects of doxorubicin (adriamycin, ADR) and daunorubicin (daunomycin, DAU), two anthracyclinic antibiotics, on a human breast carcinoma cell line (CG5) were studied by cytochemical and morphological methods. Both ADR and DAU were capable of inducing the multinucleation and spreading phenomena, associated with a decrease of the cell growth rate. DAU appeared to be more effective than ADR at the tested concentrations (10(-5), 5 x 10(-5) mM), in affecting the cell growth as well as in inducing multinucleation. As revealed by scanning electron microscopy, spreading and multinucleation were accompanied by a remarkable redistribution of surface structures. Moreover, a dose- and time-dependent rearrangement of the underlying cytoskeletal components was clearly detected. In addition, both ADR and DAU at 5 x 10(-5) mM seemed to favor the rebuilding of microtubules after treatment with colcemid, while a higher dose (10(-4) mM) exerted the opposite effect. Furthermore, both anthracyclines prevented the action of the antimicrotubular agent. When recovered after treatment with cytochalasin B, in presence of ADR (or DAU) (5 x 10(-5), 10(-4) mM), cells showed a microfilament pattern rearranged differently as compared to that of cells recovered in anthracycline-free medium. The results reported here strongly suggest the involvement of actin and tubulin in CG5 cell response to ADR and DAU treatments. Thus, the cytoskeletal apparatus is confirmed as another target involved in the mechanism of action of anthracyclines.
Article
Adriamycin (ADR, Doxorubicin) effects on actin and other proteins in cultured neonatal rat cardiac myocytes were investigated. Heart cells were exposed to ADR in doses of 10(-8) M to 10(-5) M for 24 hours. Cells were harvested in 2 mM of Tris buffer containing Triton X-100, homogenized and centrifuged in a microfuge. Parallel dishes of cultured cardiac myocytes were washed in buffered saline and were fixed at 4 degrees C in Karnovsky's fixative. The supernatant solutions were dialyzed and then incubated with pancreatic DNAase I to quantify actin by enzyme inhibition. In parallel studies, both cell supernatant solutions and pellets were subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis and the stained polypeptide bands were quantified by densitometry. Results showed that heart cells exposed to 10(-6) M of ADR for 24 hours had unpolymerized actin levels reduced to 7.7 micrograms/10(6) cells (as measured by DNAase I inhibition or by sodium dodecyl sulfate polyacrylamide gel electrophoresis along with densitometry) compared to 11.0 micrograms/10(6) cells in untreated culture heart cells. When ADR concentration was 10(-7) or 10(-8) M, unpolymerized actin levels were similar to the levels of untreated heart cells. Protein content of extract solutions of untreated and ADR-treated myocytes were 1.2 mg/ml and 0.8 mg/ml, respectively. Gel densitometry of electrophoretograms showed actin to account for 12 to 16% of total density of bands on sodium dodecyl sulfate polyacrylamide gel electrophoresis. Comparative densitometry of ADR-treated cells treated with 10(-6) M of ADR show depolymerized actin to account for 77% of total actin. Ultrastructural results show a large clear cytoplasmic zone of disorganized 12 to 14-nm filaments in cultured myocytes exposed to 10(-6) M ADR. Little change in myocyte ultrastructure was seen at 10(-7) M or 10(-8) M ADR exposure. Data support ADR as a cellular disruptor with toxic effects on cardiac cytoplasmic and contractile proteins and filaments. This ADR effect on heart cells in culture is dose-related.
Article
The cardiotoxic effects of adriamycin were studied in 399 patients treated for far-advanced carcimma. Forty-five patients (11%) exhibited transient electrocardiographs changes. Eleven others developed severe congestive heart failure. Eight of these latter patients died within 3 weeks of the onset of the cardiac decompensation. The diffuse nature of this myocardiopathy was suggested by: 1. a conspicuous decrease in the QRS voltage on the electrocardiograms; 2. rapidly occurring cardiac dilatation and ventricular failure, and 3. refractoriness to inoiropic drugs and mechanical ventricular assistance. Postmortem examination of the hearts in two cases showed a striking decrease in the number of cardiac: muscle cells present, degeneration of the remaining myocardial cells, loss o contractile substance, mitochondrial swelling, and intramitochondrial dense inclusion bodies. Congestive heart failure occurred only once in the 366 patients who were treated with less than 550 mg/m2 of adriamycin (0.27%), but there were 10 cases of cardiac failure in the 33 patients who received more than 550 mg/m2 of this drug (30%). Therefore, until more direct means are established to prevent adriamycin-induced congestive heart failure, it is suggested that the total dose of adriamycin should be limited to less than 550 mg/m2 to permit safer use of this efficacious cancer chemotherapeutic agent.
Article
Delayed cardiomyopathy, similar to that observed in cancer patients during chemotherapy with the anthracycline antibiotics, daunorubicin and doxorubicin was produced in the rabbit. In chronically treated rabbits this syndrome was characterized by congestive myocardial failure and focal to disseminated myofiber degeneration and necrosis. Alterations in myocardial cells were characterized by expansion of the intracellular membrane bound compartments, the selective isolation and degradation of degenerating mitochondria accompanied by myofibrillar breakdown, and eventual complete myolysis and fibrosis. The spectrum of myofiber alterations observed suggests two possible mechanisms which might explain the pathogenesis of the lesions observed in myocardial cells of chronically treated subjects. The interference of anthracycline antibiotics with DNA dependent RNA synthesis and the presence of nucleolar alterations suggest that the myocardial lesions may be related to normal or accelerated subcellular necrobiosis and a subsequent failure of the cell to renew mitochondrial and myofibrillar proteins. However, the similarity of these lesions to those produced by a wide variety of myocardial and general cellular toxins also suggests that more direct injury to sarcoplasmic components may be important in the pathogenesis of daunorubicin and doxorubicin cardiomyopathies.
Article
Ten nuclei from the endomyocardial biopsies for each of the following 32 patients were examined by electron microscopy: seven patients before and then four and 24 hours after treatment with first-dose doxorubicin; seven patients before and four and 24 hours after treatment with first-dose doxorubicin plus N-acetyl cysteine; nine patients with doxorubicin induced cardiomyopathy; and nine patients with idiopathic congestive cardiomyopathy. Five criteria were used to semiquantitatively compare nuclei and nucleoli from each group. The most dramatic changes in nuclear and nucleolar morphology were seen four hours after doxorubicin administration. Nucleoli were smaller, contracted or segregated and contained fewer fibrillar centers and a collapsed or fragmented nucleolonema. The addition of N-acetylcysteine to treatment did not alter these results. By 24 hours, nuclei had returned to the pre-treatment status. Long-term doxorubicin therapy produced increased chromatin clumping and slightly contracted nucleoli. The idiopathic congestive cardiomyopathic nuclei differed significantly from these doxorubicin cardiomyopathic nuclei in the decreased amount of chromatin clumping and the increase in fibrillar centers and nucleonema pattern. It is concluded from this study that: (1) doxorubicin markedly alters the morphology of the human myocardial nucleus and nucleolus four hours after treatment, but these changes diminish by 24 hours; (2) N-acetylcysteine treatment fails to prevent these changes; and (3) the nuclei and nucleoli of chronic doxorubicin-induced cardiomyopathy differ significantly from other congestive cardiomyopathies, but do resemble changes seen four hours after the first dose of doxorubicin.
Article
Histological and immunohistochemical studies using specific monoclonal antibodies were made to evaluate the severity of the chronic cardiomyopathy and the quantitative changes in interstitial dendritic cells (antigen-presenting cells), T helper lymphocytes, T cytotoxic/suppressor lymphocytes, and macrophages in the hearts of spontaneously hypertensive rats (SHRs) treated with doxorubicin at 1 mg/kg per week for 3, 6, 9 or 12 weeks. In addition, an assessment was made of the modifications of the responses of these cell populations by pretreatment of the SHR with ICRF-187, which protects against doxorubicin cardiotoxicity. The number of interstitial dendritic cells/mm2 of section of left ventricle was similar in saline-treated control SHRs (76 +/- 6) and in those treated with ICRF-187 alone (75 +/- 2) but increased markedly (319 +/- 33) in animals receiving a total cumulative dose of 12 mg/kg doxorubicin. Treatment with ICRF-187 prior to each administration of doxorubicin attenuated in a dose-dependent manner the increase in numbers of dendritic cells induced by doxorubicin (231 +/- 47, 174 +/- 11, and 100 +/- 16 cells/mm2) after treatment with 6.25, 12.5, and 25 mg of ICRF-187, respectively. Doxorubicin also induced increases in the numbers of T helper lymphocytes and macrophages but not of T cytotoxic/suppressor lymphocytes. These increases were also attenuated by pretreatment with ICRF-187. These data were interpreted as indicating that doxorubicin cardiotoxicity results in the release of substances that initiate immune reactions involving the antigen-presenting cells of the heart and that such reactions are attenuated by pretreatment with ICRF-187.
Article
Excessive stretch of heart muscle is thought to be a determinant of myocardial hypertrophy. Because cell shape and nuclear shape are closely coupled in cardiac myocytes, we hypothesize that excessive stretch causes physical deformation of the nucleus which might be responsible for some molecular events leading to hypertrophy. Cell shape and nuclear shape are most likely to be coupled by cytoskeletal elements. With this in mind, we have used immunogold labeling to examine the topological associations of desmin cytoskeletal and lamin B nucleoskeletal intermediate filaments with various intracellular structures in mammalian cardiac myocytes. We found that desmin filaments form a sarcoplasmic network radiating from the sarcolemma to the nuclear surface. Perpendicular to the long axis of the cell, strands of desmin filaments traverse the interfibrillary space in a co-linear arrangement with Z-discs. The desmin filament strands extend between peripheral regions of adjacent Z-discs. Desmin filaments traversing the interfibrillary space closely associate with the surface of mitochondria. At the cell surface, desmin filaments extend from Z-discs to terminate immediately beneath the sarcolemma. Close to the nucleus, desmin filaments extend from Z-discs towards nuclear pores. At the same time, lamin B filaments, which co-localize with heterochromatin immediately beneath the inner nuclear membrane, encircle the inner aspect of each nuclear pore. We hypothesize that desmin and lamin B are functionally anchored to each other at the nuclear pore, either directly or through anchorage proteins within the pore complex.(ABSTRACT TRUNCATED AT 250 WORDS)