Skip to main content

Immunohistochemical expression of the glucose transporters Glut-1 and Glut-3 in human malignant melanomas and benign melanocytic lesions

Abstract

Background

Reported data indicate that cancer cells have increased rates of glucose metabolism, as determined by 18FDG-PET imaging in patients with malignancies. The results of many studies have demonstrated that the expression of glucose transporters, especially Glut-1, is increased in a variety of malignancies. This study was undertaken to assess the differential expression of Glut-1 and Glut-3 by benign and malignant melanocytic lesions.

Methods

Immunohistochemical staining for Glut-1 and Glut-3 was performed on paraffin-embedded tissue sections prepared from melanocytic nevi (12 cases), Spitz nevi (12 cases) and primary cutaneous malignant melanomas (20 cases).

Results

We observed immunoreactivity for Glut-1 in all melanocytic nevi, 9 of the 12 Spitz nevi and in 9 of the 20 malignant melanomas, whereas Glut-3 was expressed in all the melanocytic lesions, both benign and malignant.

Conclusion

These findings indicate that the glucose transporters Glut-1 and Glut-3 play a role in the glucose metabolism of melanocytic cells. Glut-1 was present in the majority of benign nevi, whereas its expression was downregulated in 55% of malignant melanomas. Our results suggest that glucose transporter Glut-1 expression can significantly discriminate between human malignant melanoma and benign melanocytic nevi, and support the idea that additional mechanisms other than Glut-1 may contribute to glucose uptake in melanomas.

Background

Glucose transporters (Glut-1-14) belong to a family of structurally-related proteins that mediate energy-independent glucose transport across the plasma membrane. These transporters differ in their tissue distribution and affinity for glucose [1, 2]. Glut-1 was the first member of the facilitated glucose transporter family identified [1]. It is mostly expressed in erythrocytes, endothelial cells of the blood-brain barrier and placental cells [3]. The human Glut-1 gene, which has been localized to the short arm of chromosome 1 (1p34.2), is 35 kb in length and contains 10 exons that encode a protein of 492 aminoacids. This protein is highly conserved among different species that include human, rat, mouse and pig [4].

There is evidence that human malignancies, in which glucose metabolism is increased, express higher levels of Glut-1 than do normal cells. In particular, human Glut-1 is overexpressed in malignant cells and in a variety of tumours, that include the breast, pancreas, cervix, endometrium, lung, mesothelium, colon, bladder, thyroid, bone, soft tissues, and oral cavity [3, 5–14]. Specifically, Glut-1 expression has been associated with increased malignant potential, invasiveness, and a poor prognosis [6, 9, 14, 15]. Glut-3 is also expressed in human malignant tissue, but there are discrepancies among the reported results [9, 16, 17]. To the best of our knowledge, immunoreactivity of Glut-1 has not previously been shown for melanocytic lesions, and immunoreactivity for Glut-3 has not been investigated on these lesions [18]. We used immunohistochemistry to evaluate the expression of Glut-1 and Glut-3 in human benign nevi and malignant melanomas.

Methods

Tissue samples

We selected, retrospectively, 44 specimens from patients who had undergone surgery for either benign or malignant melanocytic lesions. The project was approved by the Committee of Ethics at the Catholic University "Sacro Cuore", Faculty of Medicine, Rome, Italy. Cases were retrieved from the archives of the Pathology Laboratory, Columbus Clinic, Catholic University "Sacro Cuore" (Rome, Italy). The histological diagnoses were as follows: melanocytic nevus, n = 12; Spitz nevus, n = 12; and primary cutaneous malignant melanoma, n = 20. The patients with melanomas were subdivided into two groups: the first contained ten patients whose tumour thickness was less than or equal to 1 mm, whereas the second contained ten patients whose tumour thickness was more than 1 mm. All diagnoses had been made on the basis of histopathological features that were evident in sections routinely stained with haematoxylin and eosin. The criterion for lesion selection was the almost absence or paucity of melanin to allow a good immunohistochemical evaluation. Table 1 summarizes the clinical and pathological characteristics of the patients who were affected by malignant melanomas. A sentinel lymph node biopsy was performed on all patients with melanoma.

Table 1 Clinical and pathological findings of the patients affected by malignant melanoma

Immunohistochemistry

Immunohistochemical analysis was performed on 4 μm paraffin sections of the tumours using an automatic immunostainer (BenchMark; Ventana Medical Systems, Tucson, AZ, USA). The sections were incubated for 24 min at 37°C with a rabbit polyclonal antibody against Glut-1 (1:200; Diapath, Bergamo, Italy) and a rabbit monoclonal antibody against Glut-3 (1:1000; Diapath). The immunostaining was visualized using Xt Ultra View Red (Diapath) as a chromogen, according to the manufacturer's instructions. Cell nuclei were lightly counterstained with standard Mayer's haematoxylin. Erythrocytes and keratinocytes served as internal positive controls for Glut-1. Samples of placenta and from gray matter of the cerebral hemispheres were used as positive controls for Glut-3. For negative controls, the primary antibodies were omitted.

Sections that had been immunostained for Glut-1 or Glut-3 were independently assessed by two observers (PP and GM). Where the results were ambiguous, an agreement was reached after discussion. The results of the immunohistochemical analysis were evaluated according to the intensity of the staining as follows: 0, absent or barely-detectable staining; 1, weak staining; 2, moderate staining; and 3, strong staining. The Glut-1 staining intensity was considered strong when it was equal to that in red blood cells in the same sections.

Statistical analysis

Two-tailed Fisher's exact test was used to analyze the contingency tables and P < 0.05 was considered to be statistically significant. Statistical analyses were performed by JMP software (SAS Institute Inc., CARY, NC, USA).

Results

The normal keratinizing epithelium in samples from all the patients showed immunoreactivity for Glut-1 and Glut-3 in the basal cell and malpighian layers: the signal for Glut-1 was strong, whereas that for Glut-3 appeared weak to moderate. The immunoreactivity of Glut-1 was always strongly intense in erythrocytes, eccrine sweat glands and the perineurium of small nerve trunks. In contrast, the antibody against Glut-3 stained the vascular endothelium together with a number of dendritic cells throughout the squamous cell layer. The immunohistochemical results for all the melanocytic lesions are summarized in Table 2. In all the cases studied, staining of Glut-1 and Glut-3 occurred in both the plasma membrane and the cytoplasm of the tumour cells. Within the cytoplasm, the staining pattern was granular.

Table 2 Immunohistochemical expression of Glut-1 and Glut-3 in benign melanocytic lesions and malignant melanomas

Benign lesions

All the melanocytic nevi (12) were positive for both Glut-1 and Glut-3. Glut-1 immunoreactivity was weak in five cases, moderate in six cases [Figure 1], and intense in one case. The immunoreactivity for Glut-3 was weak in one case, moderate in five cases, and intense in six cases [Figure 2].

Figure 1
figure 1

Melanocytic nevus with moderate staining for Glut-1.

Figure 2
figure 2

Strong immunoreactivity for Glut-3 in a melanocytic nevus.

Among the twelve cases of Spitz nevus, positive immunostaining for Glut-1 was observed in nine cases. Eight of these showed weak immunoreactivity [Figure 3], and one showed moderate immunoreactivity. Only three cases of Spitz nevi did not show Glut-1 immunoreactivity. Glut-3 expression was evident in all cases, and ten showed strong expression. Only two cases were weakly positive. Therefore, nine cases were positive for both Glut-1 and Glut-3.

Figure 3
figure 3

Spitz nevus showing weak immunoreactivity for Glut-1.

Overall, among the 24 benign melanocytic lesions, Glut-1 was expressed in 21 cases (87.5%), and Glut-3 in all cases (100%).

Malignant melanomas

Of the ten tumours in the group with thickness ≤ 1 mm, five were positive for the expression of Glut-1, and immunoreactivity was not or barely detected in the remaining five specimens. All ten tumours were positive for Glut-3. The intensity of staining for Glut-3 was weak in two cases, moderate in one case, and intense in seven cases. Therefore, five cases showed the concurrent expression of Glut-1 and Glut-3.

Of the ten cases in the group with thickness > 1 mm, Glut-1 immunoreactivity was present in four tumours, with weak immunoreactivity in three cases and moderate immunoreactivity in one case. Six tumours did not show Glut-1 immunoreactivity [Figure 4]. Glut-3 expression was evident in all tumours, with intense immunoreactivity in nine cases [Figure 5] and weak immunoreactivity in one case. Four cases were positive for both Glut-1 and Glut-3.

Figure 4
figure 4

Glut-1 is not expressed in a malignant melanoma > 1 mm thick.

Figure 5
figure 5

Strong immunoreactivity for Glut-3 in a malignant melanoma > 1 mm thick.

Statistical analysis

For statistical purposes, the 44 specimens were categorized in two groups: benign melanocytic lesions, which comprised melanocytic nevi and Spitz nevi, and malignant melanomas. The results of the statistical analysis showed that Glut-1 was significantly expressed in benign lesions as compared to malignant melanomas (p = 0.0118). Conversely, Glut-3 was expressed in all benign and malignant melanocytic lesions, with no significant differences. With regard to the malignant melanomas, no significant correlation was found between Glut-1 expression and the thickness of the tumour.

Discussion

Reported data indicate that glucose transporters, in particular Glut-1, play an important role, although not completely yet understood, in tumour progression. Glut-1, whose expression can be induced in cancer cells by oncogenes, growth factors, interleukin-1, local hypoxia and inflammatory changes, enhances the activity of the matrix metalloproteinases, whose activities have been directly related to tumour invasiveness and metastasis [2, 6, 9, 19]. Several reports indicate that the expression of the glucose transporters Glut-1 and/or Glut-3 is upregulated in many neoplasms and correlates with the uptake of the glucose analogue 18FDG in a variety of human tumours [20–24].

To the best of our knowledge, there have not been any previous immunohistochemical studies that have successfully detected Glut-1 and Glut-3 in melanocytic lesions. In our study, immunoreactivity of Glut-1 was detected in 100% of the melanocytic nevi, in 75% of the Spitz nevi and in 45% of the melanomas, whereas Glut-3 was expressed in all the melanocytic lesions, both benign and malignant. The proportion of the benign tumours which expressed Glut-1 was significantly higher than that of the malignant tumours which were positive for Glut-1. We also observed that the intensity of immunoreactivity for Glut-1 was higher in benign lesions than in malignant ones. These findings suggest that Glut-1 and Glut-3 play a role in the regulation of glucose metabolism in human melanocytic lesions.

Although Breslow's thickness remains the most important prognostic factor for cutaneous melanoma, our study has shown no significant correlation between Glut-1 expression and the thickness of the tumour [25]. Additional studies seem to be necessary to confirm the data.

The variation in the immunoreactivity that we obtained in melanomas is in agreement with the results of the study by Wachsberger et al, in which a wide variability in Glut-1 levels as detected by western immunoblotting on human melanomas was reported [26]. Also, our findings agree with a number of reports that indicate that metabolic imaging with 18FDG is not a sensitive indicator of occult metastases in patients with malignant melanoma [27–30].

The results of our study have significant differences when compared with those of previous studies, which demonstrated that Glut-1 is overexpressed in human tumours. Firstly, most of the published reports studied epithelial neoplasms, whereas nevi and malignant melanomas arise from melanocytes, which originate from the neural crest. It is well known that melanocytes and derived cells express antigens that are not found in epithelial elements. Secondly, the tumours previously described in the medical literature showed variable intratumoural expression of Glut-1 with an intense immunoreactivity in aggressive regions of the tumour, such as the poorly differentiated and central hypoxic areas [9]. However, we observed homogeneous Glut-1 expression throughout the tumours examined. In our study, all positive samples (benign and malignant) showed a membranous and cytoplasmic immunoreactivity for both Glut-1 and Glut-3, independent of staining intensity, whereas other researchers have demonstrated a cytoplasmic and/or membranous staining pattern [7, 11, 21]. It should be noted that an increase in the immunoreactivity of Glut-1 and the membranous staining pattern have been reported as indicators of hypoxia [11, 31–34]. Finally, the melanomas were much smaller in volume and had less cellularity than the carcinomas, and did not contain any cystic, necrotic or haemorrhagic components.

In the medical literature, we have found only one study that investigated Glut-1 in nevi and melanomas, and the authors reported that all the melanocytic lesions examined were negative for Glut-1 expression [18]. These differing results could be due to the use of a different antibody. Nevertheless, we are unaware of any previous study that examined Glut-3 expression in benign and/or malignant melanocytic lesions.

Conclusion

Our study clearly shows for the first time that Glut-1 is expressed in all melanocytic nevi and the majority of Spitz nevi, whereas Glut-1 expression is downregulated in 55% of malignant melanomas. However, Glut-3 is present in malignant melanomas as well as in benign lesions. Our results suggest that human malignant melanomas and benign nevi show a differential expression of the glucose transporter Glut-1. Also, these data support the idea that, in a number of melanomas, other key factors, i.e. other glucose transporter isoforms, rather Glut-1, or other processes may contribute to glucose transport across the cell membrane. However, additional studies needed to further explore glucose metabolism of melanocytic lesions.

References

  1. Olson AL, Pessin JE: Structure, function, and regulation of the mammalian facilitative glucose transporter gene family. Annu Rev Nutr. 1996, 16: 235-256. 10.1146/annurev.nu.16.070196.001315.

    Article  CAS  Google Scholar 

  2. Ito S, Fukusato T, Nemoto T, Sekihara H, Seyama Y, Kubota S: Coexpression of glucose transporter 1 and matrix metalloproteinase-2 in human cancers. J Natl Cancer Inst. 2002, 94: 1080-1091.

    Article  CAS  Google Scholar 

  3. Chang S, Lee S, Lee C, Kim JI, Kim Y: Expression of the human erythrocyte glucose transporter in transitional cell carcinoma of the bladder. Urol. 2000, 55: 448-452. 10.1016/S0090-4295(99)00474-4.

    Article  CAS  Google Scholar 

  4. Wang D, Pascual JM, Yang H, Engelstad K, Mao X, Cheng J, Yoo J, Noebels JL, De Vivo DC: A mouse model for Glut-1 haploinsufficiency. Hum Molecular Genet. 2006, 15: 1169-1179. 10.1093/hmg/ddl032.

    Article  CAS  Google Scholar 

  5. Grover-McKay M, Walsh SA, Seftor EA, Thomas PA, Hendrix MJ: Role for glucose transporter 1 protein in human breast cancer. Pathol Oncol Res. 1998, 4: 115-120.

    Article  CAS  Google Scholar 

  6. Ito H, Duxbury M, Zinner MJ, Ashley SW, Whang EE: Glucose transporter-1 gene expression is associated with pancreatic cancer invasiveness and MMP-2 activity. Surgery. 2004, 136: 548-556. 10.1016/j.surg.2004.05.032.

    Article  Google Scholar 

  7. Airley R, Loncaster J, Davidson S, Bromley M, Roberts S, Patterson A, Hunter R, Stratford I, West C: Glucose transporter glut-1 expression correlates with tumor hypoxia and predicts metastasis-free survival in advanced carcinoma of the cervix. Clin Cancer Res. 2001, 7 (4): 928-934.

    CAS  Google Scholar 

  8. Ashton-Sager A, Paulino AF, Afify AM: GLUT-1 is preferentially expressed in atypical endometrial hyperplasia and endometrial adenocarcinoma. Appl Immunohistochem Mol Morphol. 2006, 14: 187-192. 10.1097/01.pai.0000162003.43334.c7.

    Article  CAS  Google Scholar 

  9. Ito T, Noguchi Y, Satoh S, Hayashi H, Inayama Y, Kitamura H: Expression of facilitative glucose transporter isoforms in lung carcinomas: its relation to histologic type, differentiation grade, and tumor stage. Mod Pathol. 1998, 11: 437-443.

    CAS  Google Scholar 

  10. Kato Y, Tsuta K, Seki K, Maeshima AM, Watanabe S, Suzuki K, Asamura H, Tsuchiya R, Matsuno Y: Immunohistochemical detection of GLUT-1 can discriminate between reactive mesothelium and malignant mesothelioma. Mod Pathol. 2007, 20: 215-220. 10.1038/modpathol.3800732.

    Article  CAS  Google Scholar 

  11. Wincewicz A, Sulkowska M, Koda M, Sulkowski S: Clinicopathological significance and linkage of the distribution of HIF-1alpha and GLUT-1 in human primary colorectal cancer. Pathol Oncol Res. 2007, 13: 15-20.

    Article  CAS  Google Scholar 

  12. Yasuda M, Ogane N, Hayashi H, Kameda Y, Miyagi Y, Iida T, Mori Y, Tsukinoki K, Minematsu T, Osamura Y: Glucose transporter-1 expression in the thyroid gland: clinicopathological significance for papillary carcinoma. Oncol Rep. 2005, 14: 1499-1504.

    CAS  Google Scholar 

  13. Tateishi U, Yamaguchi U, Seki K, Terauchi T, Arai Y, Hasegawa T: Glut-1 expression and enhanced glucose metabolism are associated with tumour grade in bone and soft tissue sarcomas: a prospective evaluation by [18F]fluorodeoxyglucose positron emission tomography. Eur J Nucl Med Mol Imaging. 2006, 33: 683-691. 10.1007/s00259-005-0044-8.

    Article  CAS  Google Scholar 

  14. Kunkel M, Moergel M, Stockinger M, Jeong JH, Fritz G, Lehr HA, Whiteside TL: Overexpression of GLUT-1 is associated with resistance to radiotherapy and adverse prognosis in squamous cell carcinoma of the oral cavity. Oral Oncol. 2007, 43: 796-803. 10.1016/j.oraloncology.2006.10.009.

    Article  CAS  Google Scholar 

  15. Sakashita M, Aoyama N, Minami R, Maekawa S, Kuroda K, Shirasaka D, Ichihara T, Kuroda Y, Maeda S, Kasuga M: Glut1 expression in T1 and T2 stage colorectal carcinomas: its relationship to clinicopathological features. Eur J Cancer. 2001, 37: 204-209. 10.1016/S0959-8049(00)00371-3.

    Article  CAS  Google Scholar 

  16. Brown RS, Wahl RL: Overexpression of Glut-1 glucose transporter in human breast cancer. An immunohistochemical study. Cancer. 1993, 72: 2979-2985. 10.1002/1097-0142(19931115)72:10<2979::AID-CNCR2820721020>3.0.CO;2-X.

    Article  CAS  Google Scholar 

  17. Markert JM, Fuller CM, Gillespie GY, Bubien JK, McLean LA, Hong RL, Lee K, Gullans SR, Mapstone TB, Benos DJ: Differential gene expression profiling in human brain tumors. Physiol Genomics. 2001, 5: 21-33.

    CAS  Google Scholar 

  18. Baer SC, Casaubon L, Younes M: Expression of the human erythrocyte glucose transporter Glut1 in cutaneous neoplasia. J Am Acad Dermatol. 1997, 37: 575-577. 10.1016/S0190-9622(97)70174-9.

    Article  CAS  Google Scholar 

  19. Bird TA, Davies A, Baldwin SA, Saklatvala J: Interleukin 1 stimulates hexose transport in fibroblasts by increasing the expression of glucose transporters. J Biol Chem. 1990, 265: 13578-13583.

    CAS  Google Scholar 

  20. Higashi T, Tamaki N, Honda T, Torizuka T, Kimura T, Inokuma T, Ohshio G, Hosotani R, Imamura M, Konishi J: Expression of glucose transporters in human pancreatic tumors compared with increased FDG accumulation in PET study. J Nucl Med. 1997, 38: 1337-1344.

    CAS  Google Scholar 

  21. de Geus-Oei LF, van Krieken JH, Aliredjo RP, Krabbe PF, Frielink C, Verhagen AF, Boerman OC, Oyen WJ: Biological correlates of FDG uptake in non-small cell lung cancer. Lung Cancer. 2007, 55: 79-87. 10.1016/j.lungcan.2006.08.018.

    Article  Google Scholar 

  22. Chung JK, Lee YJ, Kim C, Choi SR, Kim M, Lee K, Jeong JM, Lee DS, Jang JJ, Lee MC: Mechanisms related to [18F]fluorodeoxyglucose uptake of human colon cancers transplanted in nude mice. J Nucl Med. 1999, 40: 339-346.

    CAS  Google Scholar 

  23. Yen TC, See LC, Lai CH, Yah-Huei CW, Ng KK, Ma SY, Lin WJ, Chen JT, Chen WJ, Lai CR, Hsueh S: 18F-FDG uptake in squamous cell carcinoma of the cervix is correlated with glucose transporter 1 expression. J Nucl Med. 2004, 45: 22-29.

    CAS  Google Scholar 

  24. Gu J, Yamamoto H, Fukunaga H, Danno K, Takemasa I, Ikeda M, Tatsumi M, Sekimoto M, Hatazawa J, Nishimura T, Monden M: Correlation of GLUT-1 overexpression, tumor size, and depth of invasion with 18F-2-fluoro-2-deoxy-D-glucose uptake by positron emission tomography in colorectal cancer. Dig Dis Sci. 2006, 51: 2198-2205. 10.1007/s10620-006-9428-2.

    Article  CAS  Google Scholar 

  25. Minutilli E, Giannarelli D, Anzà M, Garinei R, Perri P, Botti C, Piarulli L, Bruno P, Rea S, Visca P, Mottolese M, Di Filippo S, Di Filippo F: Sentinel node biopsy in cutaneous melanoma: correlations between melanoma prognostic factors and sentinel node status. J Exp Clin Cancer Res. 2007, 26: 71-76.

    CAS  Google Scholar 

  26. Wachsberger PR, Gressen EL, Bhala A, Bobyock SB, Storck C, Coss RA, Berd D, Leeper DB: Variability in glucose transporter-1 levels and hexokinase activity in human melanoma. Melanoma Res. 2002, 12: 35-43. 10.1097/00008390-200202000-00006.

    Article  CAS  Google Scholar 

  27. Wagner JD, Schauwecker D, Davidson D, Coleman JJ, Saxman S, Hutchins G, Love C, Hayes JT: Prospective study of fluorodeoxyglucose-positron emission tomography imaging of lymph node basins in melanoma patients undergoing sentinel node biopsy. J Clin Oncol. 1999, 17: 1508-1515.

    CAS  Google Scholar 

  28. Fink AM, Holle-Robatsch S, Herzog N, Mirzaei S, Rappersberger K, Lilgenau N, Jurecka W, Steiner A: Positron emission tomography is not useful in detecting metastasis in the sentinel lymph node in patients with primary malignant melanoma stage I and II. Melanoma Res. 2004, 14: 141-145. 10.1097/00008390-200404000-00011.

    Article  Google Scholar 

  29. Belhocine T, Pierard G, De Labrassinne M, Lahaye T, Rigo P: Staging of regional nodes in AJCC stage I and II melanoma: 18FDG PET imaging versus sentinel node detection. Oncologist. 2002, 7: 271-278. 10.1634/theoncologist.7-4-271.

    Article  Google Scholar 

  30. Maubec E, Lumbroso J, Masson F, Suciu V, Kolb F, Mamelle G, Cavalcanti A, Boitier F, Spatz A, Aupérin A, Leboulleux S, Avril MF: F-18 fluorodeoxy-D-glucose positron emission tomography scan in the initial evaluation of patients with a primary melanoma thicker than 4 mm. Melanoma Res. 2007, 17: 147-154. 10.1097/CMR.0b013e32815c10b0.

    Article  Google Scholar 

  31. Davies CA, Jeziorska M, Freemont AJ, Herrick AL: The differential expression of VEGF, VEGFR-2, and GLUT-1 proteins in disease subtypes of systemic sclerosis. Hum Pathol. 2006, 37: 190-197. 10.1016/j.humpath.2005.10.007.

    Article  CAS  Google Scholar 

  32. Airley RE, Loncaster J, Raleigh JA, Harris AL, Davidson SE, Hunter RD, West CM, Stratford IJ: GLUT-1 and CAIX as intrinsic markers of hypoxia in carcinoma of the cervix: relationship to pimonidazole binding. Int J Cancer. 2003, 104: 85-91. 10.1002/ijc.10904.

    Article  CAS  Google Scholar 

  33. Vleugel MM, Shvarts D, van der Wall E, van Diest PJ: p300 and p53 levels determine activation of HIF-1 downstream targets in invasive breast cancer. Hum Pathol. 2006, 37: 1085-1092. 10.1016/j.humpath.2006.03.015.

    Article  CAS  Google Scholar 

  34. Elson DA, Ryan HE, Snow JW, Johnson R, Arbeit JM: Coordinate up-regulation of hypoxia inducible factor (HIF)-1α and HIF-1 target genes during multi-stage epidermal carcinogenesis and wound healing. Cancer Res. 2000, 60: 6189-6195.

    CAS  Google Scholar 

Download references

Acknowledgements

We thank F. Federico and F. Castri for help in revising the histological slides, and F. Ranelletti for his expert contribution to the statistical analyses.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Antonella Coli.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

PP conceived of the study, collected the data and drafted the manuscript. AC designed this study and modified the manuscript. GM procured reagents, supervised the experimental work and performed microphotography. AM and MMF carried out the immunohistochemical analyses. GB participated in the designed of the study, and has been involved in revising the manuscript. All authors read and approved the final manuscript.

Authors’ original submitted files for images

Rights and permissions

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.

Reprints and permissions

About this article

Cite this article

Parente, P., Coli, A., Massi, G. et al. Immunohistochemical expression of the glucose transporters Glut-1 and Glut-3 in human malignant melanomas and benign melanocytic lesions. J Exp Clin Cancer Res 27, 34 (2008). https://doi.org/10.1186/1756-9966-27-34

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1756-9966-27-34

Keywords