Skip to main content
Erschienen in: Molecular Cancer 1/2010

Open Access 01.12.2010 | Research

Carnosine retards tumor growth in vivo in an NIH3T3-HER2/neu mouse model

verfasst von: Christof Renner, Nadine Zemitzsch, Beate Fuchs, Kathrin D Geiger, Matthias Hermes, Jan Hengstler, Rolf Gebhardt, Jürgen Meixensberger, Frank Gaunitz

Erschienen in: Molecular Cancer | Ausgabe 1/2010

Abstract

Background

It was previously demonstrated that the dipeptide carnosine inhibits growth of cultured cells isolated from patients with malignant glioma. In the present work we investigated whether carnosine also affects tumor growth in vivo and may therefore be considered for human cancer therapy.

Results

A mouse model was used to investigate whether tumor growth in vivo can be inhibited by carnosine. Therefore, NIH3T3 fibroblasts, conditionally expressing the human epidermal growth factor receptor 2 (HER2/neu), were implanted into the dorsal skin of nude mice, and tumor growth in treated animals was compared to control mice. In two independent experiments nude mice that received tumor cells received a daily intra peritoneal injection of 500 μl of 1 M carnosine solution. Measurable tumors were detected 12 days after injection. Aggressive tumor growth in control animals, that received a daily intra peritoneal injection of NaCl solution started at day 16 whereas aggressive growth in mice treated with carnosine was delayed, starting around day 19. A significant effect of carnosine on tumor growth was observed up to day 24. Although carnosine was not able to completely prevent tumor growth, a microscopic examination of tumors revealed that those from carnosine treated animals had a significant lower number of mitosis (p < 0.0003) than untreated animals, confirming that carnosine affects proliferation in vivo.

Conclusion

As a naturally occurring substance with a high potential to inhibit growth of malignant cells in vivo, carnosine should be considered as a potential anti-cancer drug. Further experiments should be performed in order to understand how carnosine acts at the molecular level.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-9-2) contains supplementary material, which is available to authorized users.
Christof Renner, Nadine Zemitzsch contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CR and NZ carried out the animal experiments and the cell based assays. BF carried out the MALDI experiments. KG carried out the microscopic study and analysed mitotic activity. MH and JH advised the animal experiments and supplied know how as well as the NIH3T3-Her2/neu cells. RG and JM helped to draft the manuscript. FG designed and coordinated the study, performed animal experiments and cell based assays, performed statistical analysis and drafted the pictures and the manuscript. All authors read and approved the manuscript.

Background

Carnosine, a dipeptide discovered more than 100 years ago [1] is a naturally occurring substance synthesized by endogenous carnosine synthetase. It is present in mammalian brain at a concentration between 0.7 and 2.0 mM [2] and reaches concentrations of up to 20 mM in skeletal muscle [3]. However, not much is known about its physiological function but several possible roles have been considered since its first discovery (for detailed review see [4, 5]). Among these functions ph-buffering [6], metal chelation [7] or neurotransmitter function [8] have been discussed and the currently most intensively debated aspects are its potential protective effect against oxidative stress [9], its likely role as a therapeutic agent for the treatment of Alzheimer's disease [10] and its use as a potential anti-senescence drug [11]. Just recently, it was demonstrated that carnosine is also able to inhibit growth of cultured cells isolated from human brain tumors in cell culture. This effect is not accompanied by apoptotis or necrosis, but is rather caused by reduced proliferation [12]. In fact, more recently published data indicates that reduced proliferation is mediated by an influence on glycolytic energy metabolism [13]. These interesting results indicate that carnosine may have an anti tumorigenic effect. In order to ask, whether carnosine might also be able to inhibit growth of tumors in vivo, we were looking for an experimental model in order to test a potential therapeutic applicability of carnosine. We decided to employ an approved animal model. This model uses murine NIH3T3 fibroblasts conditionally expressing human HER2/neu under the control of a tetracycline-responsive promoter, that allows turning off of expression under the influence of tetracycline or its derivatives [14]. HER2/neu, also termed neu or erbB-2, belongs to the epidermal growth factor receptors, involved in cell cycle control and cell differentiation. Its activation induces several signal transduction pathways including the ras/MAP kinase pathway [15, 16] and the phosphatidyl inositol 3,4,5-triphosphate kinase/AKT pathway [17]. In the experiments performed, fibroblasts expressing the HER2/neu receptor were subcutaneously injected into the dorsal skin of nude mice, resulting in the growth of solid tumors. Two series of experiments were performed with animals, that were treated daily with and without carnosine after the subcutaneous injection of HER2/neu expressing fibroblasts. For several weeks, tumor size of animals, receiving carnosine or NaCl as control, was determined daily.

Methods

Chemicals and reagents

If not stated otherwise all chemicals were purchased from Sigma (Taufkirchen, Germany).

Cell culture

The tetracycline-controlled cell line NIH3T3-HER2/neu [14] was cultivated in 75-cm2 flasks (TPP, Trasading, Germany) in DMEM-Medium (high glucose, Invitrogen, Karlsruhe, Germany) supplemented, with 10% fetal bovine serum (FCS Gold; PAA, Cölbe, Germany) and antibiotics (50 μg/ml streptomycin and 30 μg/ml penicillin) at 37°C and 5% CO2 in humidified air. Harvest of cells was performed using accutase (PAA) after cells reached 90% confluency.

Animals

Female nude mice Crl:CD®-1-Foxn1nu were purchased from Charles River Laboratories (Sulzfeld, Germany). The animals had access to standard chow (Altrumin pellets; Altrumin GmbH, Lage, Germany) and received water ad libitum.

Cell based assays

For the determination of ATP concentration and dehydrogenase activity, cells were cultivated in 96 well plates (black, clear bottom; μClear, Greiner Bio One, Frickenhausen, Germany) in 200 μl of medium. After 4 hours, medium was exchanged and fresh medium containing carnosine was added. After 70 hours, intracellular adenosine triphosphate (ATP) concentration was measured by the CellTiter-Glo Assay (Promega, Mannheim, Germany) as described [18]. Briefly, after incubation in the absence or presence of carnosine, medium was removed and exchanged for 50 μl of fresh medium without carnosine. At this point it should be noted, that removal of medium is not necessary and does not change the results. We included this step in order to make the experiments absolutely comparable to previously published experiments [12]. Then, 50 μl of CellTiter-Glo reagent were added. Ten minutes after addition of reagent and incubation at room temperature, luminescence was determined using a SpectraMax M5 Multilabel Reader (Molecular Devices, Ismaning, Germany) in luminescence mode with an integration time of 500 ms.
For the determination of dehydrogenase activities the CellTiter-Blue assay (Promega) was employed. Briefly, before the start of the assay, cells received 100 μl of fresh medium without carnosine and 20 μl of CellTiter-Blue reagent. Then, cells were incubated for 90 min at 37°C, 5% CO2 and in humidified air in an incubator. Finally, fluorescence was determined using an M5 Multilabel Reader with an excitation wave length of 560 nm and an emission wave length of 590 nm.

Analysis of mitotic activity

Tumors were excised and fixed in formalin for at least 24 hrs. Then the tissues were dehydrated and embedded in paraffin wax. Five μm thick serial sections of whole tissue were cut with a rotation microtome (Leica, Germany), deparaffinized and stained with Hematoxylin-Eosin (Merck, Germany). The number of mitoses was counted at 40 × magnification on a Zeiss Axioplan 2- microscope (Carl Zeiss, Oberkochen, Germany) with a graded ocular lens (10 × 10 squares, corresponding to 250 × 250 μm at 40 × magnification). All areas of the tissues were counted (maximum 10 High Power fields (HPF)) and the rate of mitosis was calculated as mean per one High-Power field. Statistical analysis used the students t-test.

Determination of carnosine concentrations in serum

Serum samples were applied onto a MALDI target as 1 μl droplets. The matrix 2,5-dihydroxybenzoic acid (DHB) was added as 1 μl droplet (0.5 mol/l stock solution in methanol) containing pentaglycine (Gly-Gly-Gly-Gly-Gly) as 5 mmol/l reference standard onto the sample. Positive ion MALDI-TOF mass spectra were acquired on a Bruker Daltonics Autoflex workstation (Bruker, Germany). The system utilizes a pulsed nitrogen laser emitting at 337 nm and spectra were recorded in the reflector mode under "delayed extraction" conditions. The extraction voltage was 20 kV and 128 single laser shots were averaged for each mass spectrum. The sum of the intensities of the H+, Na+ and K+ adducts of carnosine (m/z = 227, 249 and 265) and of pentaglycine (m/z = 304, 326 and 342) were determined for quantitative analysis using the software "Flex Analysis" version 2.2 (Bruker Daltonics, Germany).

Statistics

All data are expressed as Mean ± standard deviation. Analysis of significance was performed using the two sample t-test implemented in the Origin 7.5 Software (OriginLab Corporation, Northampton, USA).

Results

Carnosine inhibits growth of NIH3T3-HER2/neu cells in culture

Before the experiments with animals were initiated, it was asked, whether NIH3T3-HER2/neu cells are influenced by carnosine in culture, as demonstrated for cells from human glioblastoma [12]. Therefore, NIH3T3-HER2/neu cells were cultivated at an initial density of 500 cells per well in 96-well plates. Four hours after the start of the culture, carnosine was added at different concentrations, and cells were cultivated for 70 hours. Carnosine was added from a 1 M stock solution in 0.7% NaCl with pH adjusted to that of cell culture medium (ph = 7.4). Besides, we showed that addition of carnosine to culture medium does not influence pH under the culture conditions (37°C and 5% CO2 [12]). Control cells received a medium containing the corresponding amount of 0.7% NaCl solution. After the incubation time, the concentration of ATP was measured and dehydrogenase activity was determined. As can be seen in Fig. 1, carnosine reduces dehydrogenase activity by ~20% at 20 mM, by ~60% at 40 mM and by ~80% at 50 mM carnosine. The concentration of ATP is reduced by ~30% at 20 mM, by ~70% at 40 mM and by ~80% at 50 mM carnosine. This result is in good agreement with the previously published data from tumor cells derived from patients with malignant glioma and it encouraged to ask whether the effect of carnosine on tumor cell growth may also be observable in vivo.

Intraperitoneal delivered carnosine is detectable in the blood

In order to answer the question whether intraperitoneal delivered carnosine will enter the circulation, 3 animals received an intraperitoneal injection of 500 μl carnosine (1 M) dissolved in NaCl-Solution (0.7%), ph 7.4. One μl samples of blood from the tails were analysed for the presence of carnosine in serum by MALDI TOF mass spectrometry. The concentrations determined after injection were 20 ± 5 mM after 15 min, 32 ± 4.2 mM after 30 min, 10 ± 0 mM after 60 min and 15 ± 0 mM after two hours. In mice, not treated with carnosine, the mean concentration determined was 0.097 ± 0,031 mM and therefore already below the limit of quantification (0.2 mM) but above the limit of detection (0.05 mM).

Tumor growth in the NIH3T3-HER2/neu animal model under the influence of carnosine

In two independent experiments (each with 11 animals) 7 × 106 NIH3T3-HER2/neu cells in 100 μl sterile phosphate-buffered saline (PBS: 2.7 mM KCl, 1.5 mM KH2PO4, 140 mM NaCl, 6.5 mM Na2HPO4, pH 7.4) were injected subcutaneous into the dorsal skin of female nude mice. The eleven animals in each of the two series were divided into two groups. One group (6 animals) received a daily intraperitoneal injection of 500 μl carnosine (1 M), dissolved in NaCl-Solution (0.7%), ph 7.4. The second group (5 animals) was used as control and received a daily injection of 500 μl of 0.9% NaCl, pH 7.4. All injections were performed for six days with a one day break, starting again for six days. Measurable tumors were detected 12 days after injection, and tumor size was determined by measuring the maximum and minimum diameters with a caliber rule. The product of these two diameters was defined as the tumor size with the dimension mm2. The result of the two experiments is presented in Fig. 2. As can be seen, progressive tumor development starts at day 16 in control mice and at day 19 in carnosine mice. Hypothesis testing using students t-test indicated significances between treated and control animals from day 17 to day 24 in the first series and from day 17 to day 23, as indicated by the numbers in Fig. 2. Statistical analysis was not performed after 24 days, since at this time some animals already reached a tumor size in the control population, that was not bearable with animal care. When the tumors reached the critical size (>220 mm2), the animals were treated with anhydrotetracycline (animals from first series; Fig. 2a) or were sacrificed for microscopic analysis of tumors (animals from second series; Fig. 2b).

Development of tumors after switching off HER2/neu expression

In the first experimental series (Fig. 2a) the animals were not sacrificed but were further treated. The treatment protocol and the detailed tumor progression is presented as additional material. Briefly, the animals received subcutaneous injections of 100 μl anhydrotetracycline (12 mg/ml) every second day, after tumors reached a critical size (>220 mm2). These injections turn off HER2/neu expression and result in the disappearance of tumors. In previous experiments, a reoccurrence of tumors was observed after 20 to 45 days after anhydrotetracycline administration [14]. Since these new tumors were reported do be independent from HER2/neu expression, we wondered whether carnosine may also have an effect on the reoccurrence of tumors. In our experiments, all tumors treated with anhydrotetracycline disappeared but no recurrence of tumors was detected. In addition, one mouse had a spontaneously diminishing tumor. This animal never received anhydrotetracycline, but was continuously treated with carnosine, until a second tumor developed, that reached a critical size around day 60. At this time anhydrotetracycline was injected followed by a significant decrease of tumor size, indicating that this tumour was still resulting from the injected NIH3T3-HER2/neu cells.

Mitotic activity is reduced in tumors from animals treated with carnosine

In the second series of experiments (Fig. 2b) the animals were not treated with anhydrotetracycline. Instead, the animals were sacrificed and the tumours removed in order to analyse mitotic activity. Paraffin imbedded sections were analysed as described in the Materials and Methods section. Microscopy revealed that all tumors showed a highly malignant mostly sarcoma-like morphology. Compared to the tumours from non-treated animals, the nuclei of carnosine treated tumours were less pleomorph (Fig. 3). Most interestingly, the tumours in the carnosine treated group exhibited a significantly lower number of mitoses per high power field (14.0 ± 1.58 mitoses/HPF) compared to the tumors from control mice (21.6 ± 2.19 mitoses/HPF; P < 0.0003).

Discussion

The search for potent anticancer drugs is still a huge challenge. The two most challenging aspects are on one hand to find drugs with minor side effects and on the other hand to find substances that are able to prevent growth of tumors that can not be successfully treated yet. Carnosine is a highly interesting candidate with regard to both aspects. Firstly, carnosine is a naturally occurring substance that is present in muscle and other organs. Secondly, experiments with cultured cells from non-curable malignant glioma demonstrated that carnosine is able to diminish proliferation of these cells. Just recently, it was demonstrated that carnosine inhibits glycolysis and ATP synthesis in tumor cells in culture [13]. This observation is in contrast to the well-known fact that in normal cells carnosine improves both processes [19]. The basic difference with regard to energy metabolism between tumor cells and normal cells have been described almost a century ago and are known as the Warburg effect [20]. The molecular mechanisms of this effect are still not completely understood. In fact, the study of the mechanisms, on how carnosine affects normal and tumor cell energy metabolism differentially, may help to exploit the basic differences, and may suggest a way for the creation of a knowledge-based strategy to fight cancer without affecting normal cells [21]. However, and first of all, it had to be asked whether carnosine can be administered as a drug. In the present study, we therefore investigated whether carnosine may in fact prevent growth of tumor cells in an established mouse model based on NIH3T3 cells expressing the oncogene HER2/neu [14, 22]. HER2/neu belongs to the human epidermal growth factor receptor family (HER) of tyrosine kinases. It was first described to be overexpressed in 25-30% of breast cancers, but its overexpression is also seen in subsets of gastric, esophagal and endometric cancers and in some cancers of the oropharynx, the lung and the bladder (for review see [23]). Just rececently, it was demonstrated that HER2 is also expressed in glioblastoma patient samples [24]. In addition, Herceptin, a recombinant humanised anti-HER2/neu antibody, can induce cell death in cell lines derived from glioblastomas [25]. In the mouse model employed in the present study, expression of HER2/neu is strongly required for tumor growth. Switching off expression by the administration of anhydrotetracycline results in rapid tumor regression. Therefore, it is tempting to speculate that carnosine also affects HER2/neu expression, but recent data more strongly indicates that carnosine exhibits its effect at the level of glycolysis, depleting ATP production, finally resulting in reduced proliferation [13]. In the present work the anti proliferative effect of carnosine in vivo is demonstrated by the reduced growth of tumors in treated animals and by the smaller number of mitotic cells in treated tumors. One interesting challenge is now, how to enhance the local concentration of carnosine in hope to get a stronger effect on tumor growth. In fact, the carnosine concentration in the serum of treated animals after ~26 days of treatment and 24 hours after the last injection is not significantly different from the carnosine concentration in the serum of animals not treated with carnosine (data not shown). We also do not know whether the continuous administration of carnosine may induce carnosinase, the degrading enzyme, that in humans is known to be secreted from the liver into the serum [26]. Of course, an intratumoral injection of carnosine might have been a possibility but this would have made the determination of tumor size in the experiments presented difficult. Considering a clinical application of carnosine, an intratumoral injection of carnosine or the implantation of wafers (e.g. into the resection cavity of a surgically excised tumor in the case of malignant gliomas), that continuously release carnosine, may be discussed. However, discussing a potential clinical application, we should not withhold, that two minor tumors developed in two carnosine treated mice of the first series and one minor tumor in two carnosine treated mice of the second series, close to the time, when the primary tumor already had reached its maximum size (data not shown). The additional tumors were very small and regressed after administration of anhydrotetracycline. Although they may have been initiated in the vicinity of the primary tumor by a small number of cells subcutaneously introduced by the injection, it cannot be ruled out, that they originated from cells that have migrated from the primary tumor. Whether carnosine may induce a migratory potential, is pure speculative and needs further experimental data.
One puzzling observation in the course of the experiments was the complete disappearance of one tumor in a mouse, that was continuously treated with carnosine. A careful examination of the mouse did not reveal any sign of an accidental injection that is usually detectable even after several days. In addition, regression needs several repeated injections and cannot just be caused by one accidental injection. Therefore, it must be concluded that the tumor regressed under the continuous treatment with carnosine. Although this tumor reappeared after 50 days of treatment, this observation is a highly interesting one with regard to a possible use of carnosine in human tumor therapy.

Conclusions

The data presented prove that carnosine has an antiproliferative effect on malignant cells in vivo as demonstrated before in culture. Therefore, carnosine should be considered as a potential anticancer agent, especially since it is a naturally occurring substance. It is now very important to analyse how carnosine inhibits proliferation and whether it may be a useful drug for anticancer therapy in humans.

Acknowledgements

We like to thank Mrs. Anne-Kathrin Krause from the Rudolf-Boehm-Institut of Pharmakology und Toxikology for expert animal care.
This work was supported by a grant from the German Research Council to Beate Fuchs (DFG Fu 771/1-1).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

CR and NZ carried out the animal experiments and the cell based assays. BF carried out the MALDI experiments. KG carried out the microscopic study and analysed mitotic activity. MH and JH advised the animal experiments and supplied know how as well as the NIH3T3-Her2/neu cells. RG and JM helped to draft the manuscript. FG designed and coordinated the study, performed animal experiments and cell based assays, performed statistical analysis and drafted the pictures and the manuscript. All authors read and approved the manuscript.
Literatur
1.
Zurück zum Zitat Gulewitsch W, Amiradzibi S: Ueber das Carnosin, eine neue organische Base des Fleischextraktes. Ber Deut Chem Ges. 1900, 33: 1902-1903. 10.1002/cber.19000330275.CrossRef Gulewitsch W, Amiradzibi S: Ueber das Carnosin, eine neue organische Base des Fleischextraktes. Ber Deut Chem Ges. 1900, 33: 1902-1903. 10.1002/cber.19000330275.CrossRef
2.
Zurück zum Zitat Margolis FL: Carnosine: an olfactory neuropeptide. The Role of Peptides in Neuronal Function. Edited by: Barker JL, Smith TG Jr. 1980, 545-572. New York: Marcel Decker Margolis FL: Carnosine: an olfactory neuropeptide. The Role of Peptides in Neuronal Function. Edited by: Barker JL, Smith TG Jr. 1980, 545-572. New York: Marcel Decker
3.
Zurück zum Zitat Mannion AF, Jakeman PM, Dunnett M, Harris RC, Willan PL: Carnosine and anserine concentrations in the quadriceps femoris muscle of healthy humans. Eur J Appl Physiol Occup Physiol. 1992, 64: 47-50. 10.1007/BF00376439CrossRefPubMed Mannion AF, Jakeman PM, Dunnett M, Harris RC, Willan PL: Carnosine and anserine concentrations in the quadriceps femoris muscle of healthy humans. Eur J Appl Physiol Occup Physiol. 1992, 64: 47-50. 10.1007/BF00376439CrossRefPubMed
4.
Zurück zum Zitat Boldyrev AA: Problems and perspectives in studying the biological role of carnosine. Biochemistry (Mosc). 2000, 65: 751-756. Boldyrev AA: Problems and perspectives in studying the biological role of carnosine. Biochemistry (Mosc). 2000, 65: 751-756.
5.
Zurück zum Zitat Guiotto A, Calderan A, Ruzza P, Borin G: Carnosine and carnosine-related antioxidants: a review. Curr Med Chem. 2005, 12: 2293-2315. 10.2174/0929867054864796CrossRefPubMed Guiotto A, Calderan A, Ruzza P, Borin G: Carnosine and carnosine-related antioxidants: a review. Curr Med Chem. 2005, 12: 2293-2315. 10.2174/0929867054864796CrossRefPubMed
7.
Zurück zum Zitat Baran EJ: Metal complexes of carnosine. Biochemistry (Mosc). 2000, 65: 789-797. Baran EJ: Metal complexes of carnosine. Biochemistry (Mosc). 2000, 65: 789-797.
8.
Zurück zum Zitat Nadi NS, Hirsch JD, Margolis FL: Laminar distribution of putative neurotransmitter amino acids and ligand binding sites in the dog olfactory bulb. J Neurochem. 1980, 34: 138-146. 10.1111/j.1471-4159.1980.tb04632.xCrossRefPubMed Nadi NS, Hirsch JD, Margolis FL: Laminar distribution of putative neurotransmitter amino acids and ligand binding sites in the dog olfactory bulb. J Neurochem. 1980, 34: 138-146. 10.1111/j.1471-4159.1980.tb04632.xCrossRefPubMed
9.
Zurück zum Zitat Fontana M, Pinnen F, Lucente G, Pecci L: Prevention of peroxynitrite-dependent damage by carnosine and related sulphonamido pseudodipeptides. Cell Mol Life Sci. 2002, 59: 546-551. 10.1007/s00018-002-8446-2CrossRefPubMed Fontana M, Pinnen F, Lucente G, Pecci L: Prevention of peroxynitrite-dependent damage by carnosine and related sulphonamido pseudodipeptides. Cell Mol Life Sci. 2002, 59: 546-551. 10.1007/s00018-002-8446-2CrossRefPubMed
10.
Zurück zum Zitat Hipkiss AR: Could carnosine or related structures suppress Alzheimer's disease?. J Alzheimers Dis. 2007, 11: 229-240.PubMed Hipkiss AR: Could carnosine or related structures suppress Alzheimer's disease?. J Alzheimers Dis. 2007, 11: 229-240.PubMed
11.
Zurück zum Zitat Gallant S, Semyonova M, Yuneva M: Carnosine as a potential anti-senescence drug. Biochemistry (Mosc). 2000, 65: 866-868. Gallant S, Semyonova M, Yuneva M: Carnosine as a potential anti-senescence drug. Biochemistry (Mosc). 2000, 65: 866-868.
12.
Zurück zum Zitat Renner C, Seyffarth A, de Arriba S, Meixensberger J, Gebhardt R, Gaunitz F: Carnosine Inhibits Growth of Cells Isolated from Human Glioblastoma Multiforme. Int J Pept Res Ther. 2008, 14: 127-135. 10.1007/s10989-007-9121-0.CrossRef Renner C, Seyffarth A, de Arriba S, Meixensberger J, Gebhardt R, Gaunitz F: Carnosine Inhibits Growth of Cells Isolated from Human Glioblastoma Multiforme. Int J Pept Res Ther. 2008, 14: 127-135. 10.1007/s10989-007-9121-0.CrossRef
13.
Zurück zum Zitat Renner C, Asperger A, Seyffarth A, Meixensberger J, Gebhardt R, Gaunitz F: Carnosine inhibits ATP production in cells from malignant glioma. Neurol Res. 2009 Renner C, Asperger A, Seyffarth A, Meixensberger J, Gebhardt R, Gaunitz F: Carnosine inhibits ATP production in cells from malignant glioma. Neurol Res. 2009
14.
Zurück zum Zitat Schiffer IB, Gebhard S, Heimerdinger CK, Heling A, Hast J, Wollscheid U, Seliger B, Tanner B, Gilbert S, Beckers T: Switching off HER-2/neu in a tetracycline-controlled mouse tumor model leads to apoptosis and tumor-size-dependent remission. Cancer Res. 2003, 63: 7221-7231.PubMed Schiffer IB, Gebhard S, Heimerdinger CK, Heling A, Hast J, Wollscheid U, Seliger B, Tanner B, Gilbert S, Beckers T: Switching off HER-2/neu in a tetracycline-controlled mouse tumor model leads to apoptosis and tumor-size-dependent remission. Cancer Res. 2003, 63: 7221-7231.PubMed
15.
Zurück zum Zitat Busse D, Doughty RS, Arteaga CL: HER-2/neu (erbB-2) and the cell cycle. Semin Oncol. 2000, 27: 3-8.PubMed Busse D, Doughty RS, Arteaga CL: HER-2/neu (erbB-2) and the cell cycle. Semin Oncol. 2000, 27: 3-8.PubMed
16.
Zurück zum Zitat Harari D, Yarden Y: Molecular mechanisms underlying ErbB2/HER2 action in breast cancer. Oncogene. 2000, 19: 6102-6114. 10.1038/sj.onc.1203973CrossRefPubMed Harari D, Yarden Y: Molecular mechanisms underlying ErbB2/HER2 action in breast cancer. Oncogene. 2000, 19: 6102-6114. 10.1038/sj.onc.1203973CrossRefPubMed
17.
Zurück zum Zitat Zhou BP, Hu MC, Miller SA, Yu Z, Xia W, Lin SY, Hung MC: HER-2/neu blocks tumor necrosis factor-induced apoptosis via the Akt/NF-kappaB pathway. J Biol Chem. 2000, 275: 8027-8031. 10.1074/jbc.275.11.8027CrossRefPubMed Zhou BP, Hu MC, Miller SA, Yu Z, Xia W, Lin SY, Hung MC: HER-2/neu blocks tumor necrosis factor-induced apoptosis via the Akt/NF-kappaB pathway. J Biol Chem. 2000, 275: 8027-8031. 10.1074/jbc.275.11.8027CrossRefPubMed
18.
Zurück zum Zitat Gaunitz F, Heise K: HTS compatible assay for antioxidative agents using primary cultured hepatocytes. Assay Drug Dev Technol. 2003, 1: 469-477. 10.1089/154065803322163786CrossRefPubMed Gaunitz F, Heise K: HTS compatible assay for antioxidative agents using primary cultured hepatocytes. Assay Drug Dev Technol. 2003, 1: 469-477. 10.1089/154065803322163786CrossRefPubMed
19.
Zurück zum Zitat Qureshi Y, Wood T: The effect of carnosine on glycolysis. Biochim Biophys Acta. 1962, 60: 190-192. 10.1016/0006-3002(62)90390-6CrossRefPubMed Qureshi Y, Wood T: The effect of carnosine on glycolysis. Biochim Biophys Acta. 1962, 60: 190-192. 10.1016/0006-3002(62)90390-6CrossRefPubMed
20.
Zurück zum Zitat Warburg O, Posener K, Negelein E: Ueber den Stoffwechsel der Carcinomzelle. Biochemische Zeitschrift. 1924, 152: 309-344. Warburg O, Posener K, Negelein E: Ueber den Stoffwechsel der Carcinomzelle. Biochemische Zeitschrift. 1924, 152: 309-344.
21.
Zurück zum Zitat Pelicano H, Martin DS, Xu RH, Huang P: Glycolysis inhibition for anticancer treatment. Oncogene. 2006, 25: 4633-4646. 10.1038/sj.onc.1209597CrossRefPubMed Pelicano H, Martin DS, Xu RH, Huang P: Glycolysis inhibition for anticancer treatment. Oncogene. 2006, 25: 4633-4646. 10.1038/sj.onc.1209597CrossRefPubMed
22.
Zurück zum Zitat Hermes M, Schormann W, Brulport M, Uhlemann K, Lupatsch F, Horn LC, Schumann A, Allgaier C, Weishaupt M, Engeland K: Trastuzumab therapy vs tetracycline controlled ERBB2 downregulation: influence on tumour development in an ERBB2-dependent mouse tumour model. British Journal of Cancer. 2008, 98: 1525-1532. 10.1038/sj.bjc.6604318PubMedCentralCrossRefPubMed Hermes M, Schormann W, Brulport M, Uhlemann K, Lupatsch F, Horn LC, Schumann A, Allgaier C, Weishaupt M, Engeland K: Trastuzumab therapy vs tetracycline controlled ERBB2 downregulation: influence on tumour development in an ERBB2-dependent mouse tumour model. British Journal of Cancer. 2008, 98: 1525-1532. 10.1038/sj.bjc.6604318PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Moasser MM: The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene. 2007, 26: 6469-6487. 10.1038/sj.onc.1210477PubMedCentralCrossRefPubMed Moasser MM: The oncogene HER2: its signaling and transforming functions and its role in human cancer pathogenesis. Oncogene. 2007, 26: 6469-6487. 10.1038/sj.onc.1210477PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Mineo JF, Bordron A, Baroncini M, Maurage CA, Ramirez C, Siminski RM, Berthou C, Hieu PD: Low HER2-expressing glioblastomas are more often secondary to anaplastic transformation of low-grade glioma. Journal of Neuro-Oncology. 2007, 85: 281-287. 10.1007/s11060-007-9424-1CrossRefPubMed Mineo JF, Bordron A, Baroncini M, Maurage CA, Ramirez C, Siminski RM, Berthou C, Hieu PD: Low HER2-expressing glioblastomas are more often secondary to anaplastic transformation of low-grade glioma. Journal of Neuro-Oncology. 2007, 85: 281-287. 10.1007/s11060-007-9424-1CrossRefPubMed
25.
Zurück zum Zitat Mineo JF, Bordron A, Quintin-Roue I, Loisel S, Ster KL, Buhe V, Lagarde N, Berthou C: Recombinant humanised anti-HER2/neu antibody (Herceptin (R)) induces cellular death of glioblastomas. British Journal of Cancer. 2004, 91: 1195-1199.PubMedCentralPubMed Mineo JF, Bordron A, Quintin-Roue I, Loisel S, Ster KL, Buhe V, Lagarde N, Berthou C: Recombinant humanised anti-HER2/neu antibody (Herceptin (R)) induces cellular death of glioblastomas. British Journal of Cancer. 2004, 91: 1195-1199.PubMedCentralPubMed
26.
Zurück zum Zitat Teufel M, Saudek V, Ledig JP, Bernhardt A, Boularand S, Carreau A, Cairns NJ, Carter C, Cowley DJ, Duverger D: Sequence identification and characterization of human carnosinase and a closely related non-specific dipeptidase. J Biol Chem. 2003, 278: 6521-6531. 10.1074/jbc.M209764200CrossRefPubMed Teufel M, Saudek V, Ledig JP, Bernhardt A, Boularand S, Carreau A, Cairns NJ, Carter C, Cowley DJ, Duverger D: Sequence identification and characterization of human carnosinase and a closely related non-specific dipeptidase. J Biol Chem. 2003, 278: 6521-6531. 10.1074/jbc.M209764200CrossRefPubMed
Metadaten
Titel
Carnosine retards tumor growth in vivo in an NIH3T3-HER2/neu mouse model
verfasst von
Christof Renner
Nadine Zemitzsch
Beate Fuchs
Kathrin D Geiger
Matthias Hermes
Jan Hengstler
Rolf Gebhardt
Jürgen Meixensberger
Frank Gaunitz
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2010
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-9-2

Weitere Artikel der Ausgabe 1/2010

Molecular Cancer 1/2010 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.