Elsevier

Vitamins & Hormones

Volume 79, 2008, Pages 235-266
Vitamins & Hormones

Chapter 8 Folate Receptor Expression in Pituitary Adenomas: Cellular and Molecular Analysis

https://doi.org/10.1016/S0083-6729(08)00408-1Get rights and content

Abstract

Clinically nonfunctional pituitary adenomas cause hypopituitarism or compression of regional structures. Unlike functional tumors, there is no available medical treatment or specific imaging technique for these tumors. We have recently discovered that both folate receptor (FR)α mRNA and protein are uniquely overexpressed in nonfunctional pituitary tumors, but not in functional adenomas. We hypothesized that FRα may hold significant promise for medical treatment by enabling novel molecular imaging and targeted therapy. Here, we used murine pituitary tumor cell line αT3‐1 as a model to investigate the biological significance of FRα and its mutant FR67. We demonstrate that overexpression of FR facilitated tumor cell growth and anchorage‐independent growth in soft agar. More colonies were observed in FR overexpressing cells than in mutant FR67 clones in soft agar. Cell proliferation rate was increased, the percentage of cells in S‐phase was increased, and high PCNA staining was detected in cells overexpressing the receptor. In αT3‐1 cells transfected with mutant FR67, cell proliferation rate was reduced, the percentage of cells residing in S‐phase was slightly decreased, and low PCNA staining was observed. By real‐time quantitative PCR, the genes involved in NOTCH3 pathway including NOTCH3, HES‐1, and TLE2 were altered; the mRNA expression of FGFR1 was upregulated, and ERβ mRNA was downregulated in FR overexpressing cells. Our findings suggest that FRα plays a role in pituitary tumor formation, and this effect may in part be due to its regulation of the NOTCH3 pathway.

Introduction

Pituitary tumors are mostly benign adenomas arising from adenohypophyseal cells in the anterior pituitary. They comprise 10% of all brain tumors and occur in ∼20% of the population. They cause significant morbidity by compression of regional structures and the inappropriate expression of pituitary hormones (Asa, 1998, Greenman and Melmed, 1996). Functional tumors, such as GH and ACTH adenomas, give rise to severe life‐threatening clinical syndromes, such as Acromegaly or Cushing's disease, and PRL adenomas result in impaired reproduction. However, ∼30% of all anterior pituitary adenomas are termed nonfunctional (NF) pituitary adenomas due to their lack of clinical hormone hypersecretion (Asa and Kovacs, 1992). Clinically, NF tumors manifest as hypopituitarism or visual field defects due to regional compression of the optic chiasm (Asa and Ezzat, 1998, Asa and Kovacs, 1992, Black et al., 1987, Katznelson et al., 1993). The NF tumors are uniquely heterogeneous (Table 8.1). They typically are quite large and cause hypopituitarism or blindness from regional compression (Greenman and Melmed, 1996). Despite the lack of clinical hormone hypersecretion, immunocytochemical staining for hormones reveals evidence for hormone expression in ≤79% of these tumors, and we refer to these as immunohistochemically positive (NF+). The remainder is negative for hormone expression (Asa et al., 1992, Katznelson et al., 1993) and these are referred to as immunohistochemically negative (NF−).

In a previous study (Evans et al., 2001), we used cDNA microarray analysis and real‐time quantitative PCR (RT‐qPCR) to compare expression profiles of 7075 genes in the normal pituitary with that in different adenomas, including NF−, PRL‐producing adenomas, GH‐producing adenomas, and ACTH‐secreting adenomas. In those experiments, we found that the folate receptor (FR)α gene was significantly overexpressed in NF− adenomas. Next, we have discovered that both FRα mRNA and protein are uniquely overexpressed in NF tumors, but not in functional adenomas (Evans et al., 2003, Moreno et al., 2005). But whether or how FRα plays a role in pituitary tumorigenesis is unclear. The FR genes are located on chromosome 11q13.3–13.5, a region commonly amplified in carcinomas of the head and neck and breast (Rijnboutt et al., 1996). There are three isoforms of FR (FRα, FRβ, and FRγ) that vary in sequence, ligand preference, and tissue distribution (Miotti et al., 1987, Shen et al., 1994). FRα (GenBank U20391) is the major isoform mediating folate transport and is the subject of this study. The FRα receptor is absent or weakly expressed in most normal tissues. FRα is vastly overexpressed in tumors such as ovarian, renal, breast, and colorectal carcinomas, as well as anaplastic ependymomas, choroid plexus, and pituitary tumors (Evans et al., 2001, Kane et al., 1986, Mathias and Green, 1998, Mathias et al., 1998, Miotti et al., 1987).

It is suggested that elevated levels of FRα induce cell proliferation not only by mediating folate uptake but also by generating other regulatory signals. In vitro studies demonstrate that cellular overexpression of the FRα results in enhanced proliferation and survival by providing enhanced folic acid uptake (Antony, 1992, Chung et al., 1993, Kane et al., 1986, Luhrs et al., 1992, Ross et al., 1994).

Cell culture studies also demonstrate that some NF tumors secrete hormone in vitro (Yamada et al., 1989). Unlike functional tumors, currently, there is no available medical treatment or specific imaging technique for these NF tumors.

In order to study the possible influence of FRα overexpression in NF pituitary tumors cell growth, we analyzed the biochemical and the biological characteristics of the murine αT3‐1 cell transfected with FRα cDNA and a mutant FR67 cDNA. We took advantage of this mutant FR67 in our current study because it significantly inhibited folate binding and uptake in one tumor cell line in other studies (Orr and Kamen, 1994, Orr and Kamen, 1995). We also examined molecules that might physically and functionally associate with FRα in order to elucidate the involvement of the receptor in signal transduction of pituitary adenoma pathogenesis.

Section snippets

Cell culture

Cells (αT3‐1) were maintained in monolayer culture in high‐glucose DMEM (Invitrogen, Carlsbad, California) with 10% FBS and were grown in folate‐free RPMI with 5% FBS for all the experiments in humidified 5% CO2 at 37 °C. Cells were routinely passaged with 0.5 mmol/liter EDTA in phosphate‐buffered saline. Cell numbers were determined by hemocytometer.

Stable transfection

The cells were plated in 35‐mm culture wells at 5 × 105 cell/ml and cultured for 1 day. The following day, the transfections were performed by a

Result

In a previous study (Evans et al., 2001), we used cDNA microarray analysis and RT‐qPCR to compare expression profiles of 7075 genes in the normal pituitary with that in different adenomas, including NF−, PRL‐producing adenomas, GH‐producing adenomas, and ACTH‐secreting adenomas. In those experiments, we found that the FRα gene was significantly overexpressed in NF− adenomas. Next, we characterized the expression of FRα in NF+ and NF−, PRL, GH, and ACTH pituitary adenomas (Evans et al., 2003).

Discussion

Our result demonstrates that FRα overexpression induces growth in the NF pituitary tumor cell line αT3‐1 in physiological folic acid culture condition and soft agar. The increased cell growth in FRwt cells were further confirmed by BrdU incorporation assay (BrdU is only incorporated into the DNA of proliferating cells) and PCNA staining (PCNA is a nuclear antigen that is only expressed in proliferating cells and absent in resting cells). These results indicate that FRα overexpression may confer

Acknowledgments

We gratefully acknowledge financial assistance to Nelson M. Oyesiku, MD, PhD, FACS, from the National Institutes of Health (R01‐NS5143901). We thank the Department of Neuropathology, Emory University Hospital, for the histology and IHC analysis.

References (53)

  • S.L. Asa et al.

    Human pituitary null cell adenomas and oncocytomas in vitro: Effects of adenohypophysiotropic hormones and gonadal steroids on hormone secretion and tumor cell morphology

    J. Clin. Endocrinol. Metab.

    (1992)
  • M. Bagnoli et al.

    Downmodulation of caveolin‐1 expression in human ovarian carcinoma is directly related to alpha‐folate receptor over‐expression

    Oncogene

    (2000)
  • D. Bellavia et al.

    Combined expression of pTalpha and Notch3 in T cell leukemia identifies the requirement of preTCR for leukemogenesis

    Proc. Natl. Acad. Sci. USA

    (2002)
  • P.M. Black et al.

    Hormone production in clinically nonfunctioning pituitary adenomas

    J. Neurosurg.

    (1987)
  • F. Bottero et al.

    Gene transfection and expression of the ovarian carcinoma marker folate binding protein on NIH/3T3 cells increases cell growth in vitro and in vivo

    Cancer Res.

    (1993)
  • C. Bradshaw et al.

    Pituitary tumor transforming gene: An important gene in normal cellular functions and tumorigenesis

    Histol. Histopathol.

    (2007)
  • S.S. Chaidarun et al.

    Expression of epidermal growth factor (EGF), its receptor, and related oncoprotein (erbB‐2) in human pituitary tumors and response to EGF in vitro

    Endocrinology

    (1994)
  • K.N. Chung et al.

    Stable transfectants of human MCF‐7 breast cancer cells with increased levels of the human folate receptor exhibit an increased sensitivity to antifolates

    J. Clin. Invest.

    (1993)
  • L. Dang et al.

    Notch3 signaling initiates choroid plexus tumor formation

    Oncogene

    (2006)
  • T.P. Dang et al.

    Constitutive activation of Notch3 inhibits terminal epithelial differentiation in lungs of transgenic mice

    Oncogene

    (2003)
  • C.O. Evans et al.

    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.

    (2001)
  • C.O. Evans et al.

    Differential expression of folate receptor in pituitary adenomas

    Cancer Res.

    (2003)
  • M. Figini et al.

    Reversion of transformed phenotype in ovarian cancer cells by intracellular expression of anti folate receptor antibodies

    Gene Ther.

    (2003)
  • S. Fukui et al.

    Subcellular localization of basic fibroblast growth factor and fibroblast growth factor receptor 1 in pituitary adenomas

    Brain Tumor Pathol.

    (2002)
  • Y. Gao et al.

    Estrogen prevents bone loss through transforming growth factor beta signaling in T cells

    Proc. Natl. Acad. Sci. USA

    (2004)
  • N.J. Gittoes

    Current perspectives on the pathogenesis of clinically non‐functioning pituitary tumours

    J. Endocrinol.

    (1998)
  • Cited by (19)

    • Intraoperative Fluorescent Visualization of Pituitary Adenomas

      2019, Neurosurgery Clinics of North America
      Citation Excerpt :

      Postoperative immunohistochemistry confirmed that none of the functional adenomas overexpressed folate receptor alpha, which is the target binding site for OTL38. This was consistent with prior studies looking at folate receptor expression.23–26 In contrast, nonfunctional adenomas demonstrated significantly stronger fluorescence (SBR = 2.6 ± 0.91) and folate receptor overexpression was seen in 64% (9/14) of cases, again consistent with prior studies.

    • Technological and Ideological Innovations in Endoscopic Skull Base Surgery

      2019, World Neurosurgery
      Citation Excerpt :

      This might have future clinical significance for intraoperative use of 5-ALA in patients with chordomas, although additional study is required. Nonfunctioning pituitary adenomas overexpress folate receptor alpha.29-31 Lee et al.32 used OTL38 (On Target Laboratories, West Lafayette, Indiana, USA), a folate analog conjugated to a cyanine dye that specifically binds folate receptors to image folate, in a series of functioning and nonfunctioning adenomas.

    • Endocrinologically Silent Pituitary Tumors

      2012, Schmidek and Sweet Operative Neurosurgical Techniques: Indications, Methods, and Results: Sixth Edition
    • Intraoperative Tumor Detection Using Pafolacianine

      2022, International Journal of Molecular Sciences
    View all citing articles on Scopus
    View full text