Skip to main content
Top
Published in: Pituitary 3/2008

01-09-2008

Molecular pathogenesis of human prolactinomas identified by gene expression profiling, RT-qPCR, and proteomic analyses

Authors: Chheng-Orn Evans, Carlos S. Moreno, Xianquan Zhan, Michael T. McCabe, Paula M. Vertino, Dominic M. Desiderio, Nelson M. Oyesiku

Published in: Pituitary | Issue 3/2008

Login to get access

Abstract

The molecular pathogenesis of prolactinomas has resisted elucidation; with the exception of a RAS mutation in a single aggressive prolactinoma, no mutational changes have been identified. In prolactinomas, a further obstacle has been the paucity of surgical specimens suitable for molecular analysis since prolactionomas are infrequently removed due to the availability and effectiveness of medical therapy. In the absence of mutational events, gene expression changes have been sought and detected. Using high-throughput analysis from a large bank of human pituitary adenomas, we examined these tumors according to their molecular profiles rather than traditional immunohistochemistry. We examined six prolactinomas and eight normal pituitary glands using oligonucleotide GeneChip microarrays, reverse transcription-real time quantitative polymerase chain reaction using 10 prolactinomas, and proteomic analysis to examine protein expression in four prolactinomas. Microarray analyses identified 726 unique genes that were statistically significantly different between prolactinomas and normal glands, whereas proteomic analysis identified four differently up-regulated and 19 down-regulated proteins. Several components of the Notch pathway were altered in prolactinomas, and there was an increased expression of the Pit-1 transcription factor, and the survival factor BAG1 but decreased E-cadherin and N-cadherin expression. Taken together, expression profiling and proteomic analyses have identified molecular features unique to prolactinomas that may contribute to their pathogenesis. In the current era of molecular medicine, these findings greatly enhance our understanding and supercede immunohistochemical diagnosis.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Forsyth IA, Wallis M (2002) Growth hormone and prolactin-molecular and function evolution. J Mammary Gland Biol Neoplasia 7:291–312PubMedCrossRef Forsyth IA, Wallis M (2002) Growth hormone and prolactin-molecular and function evolution. J Mammary Gland Biol Neoplasia 7:291–312PubMedCrossRef
3.
go back to reference Ben-Jonathan N et al (1996) Extrapituitary prolactin: distribution, regulation and clinical aspects. Endocr Rev 17(6):639–669PubMedCrossRef Ben-Jonathan N et al (1996) Extrapituitary prolactin: distribution, regulation and clinical aspects. Endocr Rev 17(6):639–669PubMedCrossRef
4.
go back to reference Berwaer M, Martial JA, Davis JR (1994) Characterization of an up-stream promoter directing extrapituitary expression of the human prolactin gene. Mol Endocrinol 8(5):635–642PubMedCrossRef Berwaer M, Martial JA, Davis JR (1994) Characterization of an up-stream promoter directing extrapituitary expression of the human prolactin gene. Mol Endocrinol 8(5):635–642PubMedCrossRef
5.
go back to reference Gellersen B et al (1994) Nonpituitary human prolactin gene transcription is independent of Pit-1 and differentially controlled in lymphocytes and in endometrial stroma. Mol Endocrinol 8(3):356–373PubMedCrossRef Gellersen B et al (1994) Nonpituitary human prolactin gene transcription is independent of Pit-1 and differentially controlled in lymphocytes and in endometrial stroma. Mol Endocrinol 8(3):356–373PubMedCrossRef
6.
go back to reference Anderson B, Rosenfeld MG (2001) POU domain factors in the neuroendocrine system: lesson from developmental biology provide insights into human disease. Endocr Rev 22:2–35CrossRef Anderson B, Rosenfeld MG (2001) POU domain factors in the neuroendocrine system: lesson from developmental biology provide insights into human disease. Endocr Rev 22:2–35CrossRef
7.
go back to reference Verhelst J, Abs R (2003) Hyperprolactinemia: pathophysiology and management. Treat Endocrinol 2(1):23–32PubMedCrossRef Verhelst J, Abs R (2003) Hyperprolactinemia: pathophysiology and management. Treat Endocrinol 2(1):23–32PubMedCrossRef
8.
go back to reference Herman V et al (1990) Clonal origin of pituitary adenomas. J Clin Endocrinol Metab 71(6):1427–1433PubMedCrossRef Herman V et al (1990) Clonal origin of pituitary adenomas. J Clin Endocrinol Metab 71(6):1427–1433PubMedCrossRef
9.
go back to reference Boikos SA, Stratakis CA (2007) Molecular genetics of the cAMP-dependent protein kinase pathway and of sporadic pituitary tumorigenesis. Hum Mol Genet 16 Spec No 1:R80–R87 Boikos SA, Stratakis CA (2007) Molecular genetics of the cAMP-dependent protein kinase pathway and of sporadic pituitary tumorigenesis. Hum Mol Genet 16 Spec No 1:R80–R87
10.
go back to reference Herman V et al (1993) Molecular screening of pituitary adenomas for gene mutations and rearrangements. J Clin Endocrinol Metab 77(1):50–55PubMedCrossRef Herman V et al (1993) Molecular screening of pituitary adenomas for gene mutations and rearrangements. J Clin Endocrinol Metab 77(1):50–55PubMedCrossRef
11.
go back to reference Evans CO et al (2000) Screening for MEN1 tumor suppressor gene mutations in sporadic pituitary tumors. J Endocrinol Invest 23(5):304–309PubMed Evans CO et al (2000) Screening for MEN1 tumor suppressor gene mutations in sporadic pituitary tumors. J Endocrinol Invest 23(5):304–309PubMed
12.
go back to reference Asa SL, Somers K, Ezzat S (1998) The MEN-1 gene is rarely down-regulated in pituitary adenomas. J Clin Endocrinol Metab 83(9):3210–3212PubMedCrossRef Asa SL, Somers K, Ezzat S (1998) The MEN-1 gene is rarely down-regulated in pituitary adenomas. J Clin Endocrinol Metab 83(9):3210–3212PubMedCrossRef
13.
go back to reference Satta MA et al (1999) Expression of menin gene mRNA in pituitary tumours. Eur J Endocrinol 140(4):358–361PubMedCrossRef Satta MA et al (1999) Expression of menin gene mRNA in pituitary tumours. Eur J Endocrinol 140(4):358–361PubMedCrossRef
14.
go back to reference Farrell WE et al (1999) Sequence analysis and transcript expression of the MEN1 gene in sporadic pituitary tumours. Br J Cancer 80(1–2):44–50PubMedCrossRef Farrell WE et al (1999) Sequence analysis and transcript expression of the MEN1 gene in sporadic pituitary tumours. Br J Cancer 80(1–2):44–50PubMedCrossRef
15.
go back to reference Orr RB, Kreisler AR, Kamen BA (1995) Similarity of folate receptor expression in UMSCC 38 cells to squamous cell carcinoma differentiation markers. J Natl Cancer Inst 87(4):299–303PubMedCrossRef Orr RB, Kreisler AR, Kamen BA (1995) Similarity of folate receptor expression in UMSCC 38 cells to squamous cell carcinoma differentiation markers. J Natl Cancer Inst 87(4):299–303PubMedCrossRef
16.
go back to reference Evans CO et al (2001) Novel patterns of gene expression in pituitary adenomas identified by complementary deoxyribonucleic acid microarrays and quantitative reverse transcription-polymerase chain reaction. J Clin Endocrinol Metab 86(7):3097–3107PubMedCrossRef Evans CO et al (2001) Novel patterns of gene expression in pituitary adenomas identified by complementary deoxyribonucleic acid microarrays and quantitative reverse transcription-polymerase chain reaction. J Clin Endocrinol Metab 86(7):3097–3107PubMedCrossRef
17.
go back to reference Evans CO et al (2003) Differential expression of folate receptor in pituitary adenomas. Cancer Res 63(41):4218–4224PubMed Evans CO et al (2003) Differential expression of folate receptor in pituitary adenomas. Cancer Res 63(41):4218–4224PubMed
18.
go back to reference Moreno CS et al (2005) Novel molecular signaling and classification of human clinically nonfunctional pituitary adenomas identified by gene expression profiling and proteomic analyses. Cancer Res 65(22):10214–10222PubMedCrossRef Moreno CS et al (2005) Novel molecular signaling and classification of human clinically nonfunctional pituitary adenomas identified by gene expression profiling and proteomic analyses. Cancer Res 65(22):10214–10222PubMedCrossRef
19.
20.
go back to reference Genkai N et al (2006) Increased expression of pituitary tumor-transforming gene (PTTG)-1 is correlated with poor prognosis in glioma patients. Oncol Rep 15(6):1569–1574PubMed Genkai N et al (2006) Increased expression of pituitary tumor-transforming gene (PTTG)-1 is correlated with poor prognosis in glioma patients. Oncol Rep 15(6):1569–1574PubMed
21.
go back to reference Stoika R, Melmed S (2002) Expression and function of pituitary tumour transforming gene for T-lymphocyte activation. Br J Haematol 119(4):1070–1074PubMedCrossRef Stoika R, Melmed S (2002) Expression and function of pituitary tumour transforming gene for T-lymphocyte activation. Br J Haematol 119(4):1070–1074PubMedCrossRef
22.
go back to reference Rehfeld N et al (2006) The influence of the pituitary tumor transforming gene-1 (PTTG-1) on survival of patients with small cell lung cancer and non-small cell lung cancer. J Carcinog 5:4PubMedCrossRef Rehfeld N et al (2006) The influence of the pituitary tumor transforming gene-1 (PTTG-1) on survival of patients with small cell lung cancer and non-small cell lung cancer. J Carcinog 5:4PubMedCrossRef
23.
go back to reference Tsai SJ et al (2005) Expression and functional analysis of pituitary tumor transforming gene-1 [corrected] in uterine leiomyomas. J Clin Endocrinol Metab 90(6):3715–3723PubMedCrossRef Tsai SJ et al (2005) Expression and functional analysis of pituitary tumor transforming gene-1 [corrected] in uterine leiomyomas. J Clin Endocrinol Metab 90(6):3715–3723PubMedCrossRef
24.
go back to reference Asa SL, Ezzat S (1998) The cytogenesis and pathogenesis of pituitary adenomas. Endocr Rev 19(6):798–827PubMedCrossRef Asa SL, Ezzat S (1998) The cytogenesis and pathogenesis of pituitary adenomas. Endocr Rev 19(6):798–827PubMedCrossRef
25.
go back to reference Shimon I, Melmed S (1997) Genetic basis of endocrine disease: pituitary tumor pathogenesis. J Clin Endocrinol Metab 82(6):1675–1681PubMedCrossRef Shimon I, Melmed S (1997) Genetic basis of endocrine disease: pituitary tumor pathogenesis. J Clin Endocrinol Metab 82(6):1675–1681PubMedCrossRef
26.
go back to reference Spada A, Faglia G (1996) G-protein dysfunction in pituitary tumors. In: Oncogenesis and molecular biology of pituitary tumors. M. S, Editor. Karger, Basel, pp 108–121 Spada A, Faglia G (1996) G-protein dysfunction in pituitary tumors. In: Oncogenesis and molecular biology of pituitary tumors. M. S, Editor. Karger, Basel, pp 108–121
27.
go back to reference Irizarry RA et al (2003) Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4(2):249–264PubMedCrossRef Irizarry RA et al (2003) Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4(2):249–264PubMedCrossRef
28.
go back to reference Tusher VG, Tibshirani R, Chu G (2001) Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA 98(9):5116–5121PubMedCrossRef Tusher VG, Tibshirani R, Chu G (2001) Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci USA 98(9):5116–5121PubMedCrossRef
29.
go back to reference Zhan X, Desiderio DM (2003) A reference map of a human pituitary adenoma proteome. Proteomics 3:699–713PubMedCrossRef Zhan X, Desiderio DM (2003) A reference map of a human pituitary adenoma proteome. Proteomics 3:699–713PubMedCrossRef
30.
go back to reference Zhan X, Desiderio DM (2003) Spot volume vs. amount of protein loaded onto a gel: a detailed, statistical comparison of two gel electrophoresis systems. Electrophoresis 24:1818–1833PubMedCrossRef Zhan X, Desiderio DM (2003) Spot volume vs. amount of protein loaded onto a gel: a detailed, statistical comparison of two gel electrophoresis systems. Electrophoresis 24:1818–1833PubMedCrossRef
31.
go back to reference Zhan X, Desiderio DM (2003) Differences in the spatial and quantitative reproducibility between two second-dimensional gel electrophoresis systems. Electrophoresis 24:1834–1846PubMedCrossRef Zhan X, Desiderio DM (2003) Differences in the spatial and quantitative reproducibility between two second-dimensional gel electrophoresis systems. Electrophoresis 24:1834–1846PubMedCrossRef
32.
go back to reference Zhan X, Desiderio DM (2003) Heterogeneity analysis of the human pituitary proteome. Clin Chem 49(10):1740–1751PubMedCrossRef Zhan X, Desiderio DM (2003) Heterogeneity analysis of the human pituitary proteome. Clin Chem 49(10):1740–1751PubMedCrossRef
33.
go back to reference Herman JG et al (1996) Methylation-specific PCR: a noval PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 93(18):9821–9826PubMedCrossRef Herman JG et al (1996) Methylation-specific PCR: a noval PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 93(18):9821–9826PubMedCrossRef
34.
go back to reference Conway KE et al (2000) TMS1, a novel proapoptotic caspase recruitment domain protein, is a target of methylation-induced gene silencing in hunan breast cancers. Cancer Res 60(22):6236–6242PubMed Conway KE et al (2000) TMS1, a novel proapoptotic caspase recruitment domain protein, is a target of methylation-induced gene silencing in hunan breast cancers. Cancer Res 60(22):6236–6242PubMed
35.
go back to reference Esteller M et al (1998) Inactivation of glutathione S-transferase P1 gene by promotor hypermethylation in human neoplasia. Cancer Res 58(20):4515–4518PubMed Esteller M et al (1998) Inactivation of glutathione S-transferase P1 gene by promotor hypermethylation in human neoplasia. Cancer Res 58(20):4515–4518PubMed
36.
go back to reference Karavitaki N et al (2006) Do the limits of serum prolactin in disconnection hyperprolactinaemia need re-definition? A study of 226 patients with histologically verified non-functioning pituitary macroadenoma. Clin Endocrinol (Oxf) 65(4):524–529CrossRef Karavitaki N et al (2006) Do the limits of serum prolactin in disconnection hyperprolactinaemia need re-definition? A study of 226 patients with histologically verified non-functioning pituitary macroadenoma. Clin Endocrinol (Oxf) 65(4):524–529CrossRef
37.
go back to reference Garcia de la Torre N, Turner HE, Wass JAH (2005) Angiogenesis in prolactinomas: regulation and relationship with tumor behaviour. Pituitary 8:17–23PubMedCrossRef Garcia de la Torre N, Turner HE, Wass JAH (2005) Angiogenesis in prolactinomas: regulation and relationship with tumor behaviour. Pituitary 8:17–23PubMedCrossRef
38.
go back to reference Picard C et al (2007) Gs alpha overexpression and loss of Gs alpha imprinting in human somatotroph adenomas: association with tumor size and response to pharmacologic treatment. Int J Cancer 121(6):1245–1252PubMedCrossRef Picard C et al (2007) Gs alpha overexpression and loss of Gs alpha imprinting in human somatotroph adenomas: association with tumor size and response to pharmacologic treatment. Int J Cancer 121(6):1245–1252PubMedCrossRef
39.
go back to reference Ruebel KH et al (2006) Patterns of gene expression in pituitary carcinomas and adenomas analyzed by high-density oligonucleotide arrays, reverse-transcriptase-quantitative PCR, and protein expression. Endocrine 29:435–444PubMedCrossRef Ruebel KH et al (2006) Patterns of gene expression in pituitary carcinomas and adenomas analyzed by high-density oligonucleotide arrays, reverse-transcriptase-quantitative PCR, and protein expression. Endocrine 29:435–444PubMedCrossRef
40.
go back to reference Chen G, Courey A (2000) Groucho/TLE family proteins and transcriptional repression. Gene 249:1–16, [Review]PubMedCrossRef Chen G, Courey A (2000) Groucho/TLE family proteins and transcriptional repression. Gene 249:1–16, [Review]PubMedCrossRef
41.
go back to reference Plank MJ, Sleeman BD, Jones PF (2004) The role of the angiopoietins in tumour angiogenesis. Growth Factors 22:1–11PubMedCrossRef Plank MJ, Sleeman BD, Jones PF (2004) The role of the angiopoietins in tumour angiogenesis. Growth Factors 22:1–11PubMedCrossRef
42.
go back to reference Ishitani A et al (2003) Protein expression and peptide binding suggest unique and interacting functional roles for HLA-E, F, and G in maternal-placental immune recognition. J Immunol 171(3):1376–1384PubMed Ishitani A et al (2003) Protein expression and peptide binding suggest unique and interacting functional roles for HLA-E, F, and G in maternal-placental immune recognition. J Immunol 171(3):1376–1384PubMed
43.
go back to reference Barczyski M et al (2007) Posterior retroperitoneoscopic adrenalectomy: a comparison between the initial experience in the invention phase and introductory phase of the new surgical technique. World J Surg 31(1):65–71CrossRef Barczyski M et al (2007) Posterior retroperitoneoscopic adrenalectomy: a comparison between the initial experience in the invention phase and introductory phase of the new surgical technique. World J Surg 31(1):65–71CrossRef
44.
go back to reference Gotz R et al (2005) Bag1 is essential for differentiation and survival of hematopoietic and neuronal cells. Nat Neurosci 8(9):1169–1178PubMedCrossRef Gotz R et al (2005) Bag1 is essential for differentiation and survival of hematopoietic and neuronal cells. Nat Neurosci 8(9):1169–1178PubMedCrossRef
45.
go back to reference Morris DG et al (2005) Differential gene expression in pituitary adenomas by oligonucleotide array analysis. Eur J Endocrinol 153:143–151PubMedCrossRef Morris DG et al (2005) Differential gene expression in pituitary adenomas by oligonucleotide array analysis. Eur J Endocrinol 153:143–151PubMedCrossRef
46.
go back to reference Schade R et al (2007) Dopamine agonists and the risk of cardiac-valve regurgitation. N Engl J Med 356:29–38PubMedCrossRef Schade R et al (2007) Dopamine agonists and the risk of cardiac-valve regurgitation. N Engl J Med 356:29–38PubMedCrossRef
47.
go back to reference Sarkar DK, Kim KH, Minami S (1992) Transforming growth factor-β1 mRNA and protein expression in the pituitary gland and its action on PRL secretion and lactropic growth. Mol Endocirinol 6:1825–1833CrossRef Sarkar DK, Kim KH, Minami S (1992) Transforming growth factor-β1 mRNA and protein expression in the pituitary gland and its action on PRL secretion and lactropic growth. Mol Endocirinol 6:1825–1833CrossRef
48.
go back to reference Minami S, Sarkar DK (1997) Transforming growth factor-B1 inhibits prolactin secretion and lactotropic cell proliferation in the pituitarty in the pituitary of estrogen-treated Fischer 344 rats. Neurochem Int 30:499–506PubMedCrossRef Minami S, Sarkar DK (1997) Transforming growth factor-B1 inhibits prolactin secretion and lactotropic cell proliferation in the pituitarty in the pituitary of estrogen-treated Fischer 344 rats. Neurochem Int 30:499–506PubMedCrossRef
49.
go back to reference Mochizuki S, Okada Y (2007) ADAMs in cancer cell proliferation and progression. Cancer Sci 98:621–628PubMedCrossRef Mochizuki S, Okada Y (2007) ADAMs in cancer cell proliferation and progression. Cancer Sci 98:621–628PubMedCrossRef
50.
go back to reference Ohtsuka T et al (2006) ADAM28 is overexpressed in human non-small cell lung carcinomas and correlates with cell proliferation and lymph node metastasis. Int J Cancer 118:263–273PubMedCrossRef Ohtsuka T et al (2006) ADAM28 is overexpressed in human non-small cell lung carcinomas and correlates with cell proliferation and lymph node metastasis. Int J Cancer 118:263–273PubMedCrossRef
Metadata
Title
Molecular pathogenesis of human prolactinomas identified by gene expression profiling, RT-qPCR, and proteomic analyses
Authors
Chheng-Orn Evans
Carlos S. Moreno
Xianquan Zhan
Michael T. McCabe
Paula M. Vertino
Dominic M. Desiderio
Nelson M. Oyesiku
Publication date
01-09-2008
Publisher
Springer US
Published in
Pituitary / Issue 3/2008
Print ISSN: 1386-341X
Electronic ISSN: 1573-7403
DOI
https://doi.org/10.1007/s11102-007-0082-2

Other articles of this Issue 3/2008

Pituitary 3/2008 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.