Stemness Markers and Biomarkers of Pituitary Cytogenesis: Current Armamentarium of Molecular Targets

Editorial

Austin Biomark Diagn. 2014;1(1): 3.

Stemness Markers and Biomarkers of Pituitary Cytogenesis: Current Armamentarium of Molecular Targets

Aydin Sav*

Department of Pathology, Acibadem University, School of Medicine, Turkey

*Corresponding author: Aydin Sav, Department of Pathology, Acibadem University, School of Medicine, Kerem Aydinlar Yerleskesi, Icerenkoy Mahallesi, Kayisdagi Caddesi, No: 32, Atasehir, Istanbul, Turkey.

Received: Oct 07, 2014; Accepted: Oct 09, 2014; Published: Oct 14, 2014

Editorial

This editorial emphasizes recent advances in neonatal and adult stem cells, stem cells of Anterior Pituitary (AP) and stemness markers and biomarkers of pituitary cytogenesis.

Therefore concise information for each specific stem cell biomarker is presented in the light of recent cellular and molecular research.

Stemness markers have diverse effects either in pituitary embryogenesis and tumor development. This puzzling group consists of several elements, i.e. nest in, Sca1, Sox2+/E- cadherin, S100 proteins, Prop-1, Oct-4, Bmi-1, CD133, beta-catenin, SOX2, Pax6, Notch2, Notch3, Pit1, p57. Each specific marker has an eloquent effect on intimate molecular pathways which take part in pituitary cytogenesis. Therefore their formal roles will be briefly mentioned herein [1,2].

Nestin is expressed in various stem (Sox2+) cells in situ (marginal zone) and in culture (pituispheres) [3-5]. It is not only expressed by pituitary stem cells but also pituitary’s non-hormonal cell population as well as in some pericytes [6-8].

Sca1– (Stem Cell Antigen 1) is more or less expressed in Stem Cell-enriching Side Population (SC-SP) fraction [5]. Additionally is expressed in pituispheres [3] and in some colony-forming cells [9]. The exact function of Sca1 expressivity in the pituitary needs further elucidation.

Sox2+/E-Cadherin+ Stem Cells

Tumor suppressor gene E-cadherin is a classical member of the cadherin super family [10,11]. As a family cadherins regulate signaling pathways that organize cell proliferation and motility controlled by process of Epithelial– Mesenchymal Transition (EMT) [12]. Rathke’s cleft overlying marginal cells and S100+ cells within AP lobe robustly express E-cadherin (Cdh1) [3].

S100 proteins are engaged in regulation of Ca++ homeostasis, protein phosphorylation, the dynamics of cytoskeleton constituents, cell growth and differentiation, transcription factors, inflammatory response and enzyme activities. Randomly distributed follicostellate cells of pituitary present distinct nuclear and cytoplasmic S100 reactivity hence these cells are believed to be the analogs of brain glial cell lineage [6,7,13,14].

Prop1 is a protein that in humans is encoded by PROP1 gene and also a homeobox protein prophet of PIT-1 [15]. Major two functions of PROP1 are its transcriptional activation ability and DNA binding. So its expressivity guides ontogenesis of pituitary gonadotropes, somatotropes, lactotropes, and thyrotropes. PROP1 inactivation results in deficits Luteinizing Hormone (LH), Follicle Stimulating Hormone (FSH), Growth Hormone (GH), Prolactin (PRL), and Thyroid-Stimulating Hormone (TSH) [15]. Its inactivation terminates in failure of embryonic progenitors to leave dorsal Proliferative Region of RP (Rathke’s pouch) for supplementary differentiation and colonization of development of AP [16].

Oct-4 (Octamer-binding transcription factor 4) is a homeodomain transcription factor of the POU family [17] that has a role in protracting self-renewal capacity of adult somatic stem cells (i.e. stem cells from epithelium, bone marrow, liver, pituitary, etc.) [18].

Bmi-1 is an oncogene activated in lymphoma [19], exclusively adjust Hematopoietic Stem Cells (HSC) [20] and Neural Stem Cells (NSC) [21]. In half of human pituitary adenomas Bmi-1 over expressivity is present which is interpreted as this stem cell marker might have profound role in pituitary tumorigenesis [22].

CD133 is also referred Prominent 1 (PROM1) which is a member of pentaspan transmembrane glycoproteins (5-transmembrane, 5-TM), which specifically localize to cellular protrusions [23]. In addition to its expressivity in hematopoietic stem cells [24], endothelial progenitor cells [25], glioblastoma, neuronal and glial stem cells [26], various pediatric brain tumors [27], adult kidney, mammary glands, trachea, salivary glands, placenta, digestive tract, testes, its presence was documented in isolated stem/progenitor- like cells from pituitary adenoma. Therefore expression of neuronal progenitor markersincluding CD133 supported CD133+ cells continuation in pituitary adenomas [28].

Beta-catenin (or β-catenin) is a protein responsible for regulating the coordination of cell - cell adhesion and gene transcription. Its extensive location in AP is also associated within somatotropes and cells in marginal zone [29].

Sox2 is SRY (sex determining region Y)-box 2, also known as Sox2, is a transcription factor in maintaining self renewal of undifferentiated embryonic stem cells. Co-expression of nest in and Sox2+ in some stem cells (marginal zone) and in culture (pituispheres) was documented [3-5]. Sox2 expressivity in pituisphere cultures is a marker for higher proliferation rate and rapid differentiation capacity [30].

Pax6 is a member of transcriptional regulators and signaling factors those of which has some critical roles in early embryogenesis of pituitary, (viz. Hesx1, Lhx4, Prop1, Pax6, Otx2, and specific Notch pathway components). Its expressivity culminates in non- Sca1high SP [31-36].

Notch 2 as a member of the notch signaling pathway family, it has been identified in embryonic periluminal progenitor zone [34,36]. In adult pituitary a wide spectrum of molecules SC-SP (in particular Jag1, Notch2, Notch3, Hes1 and Hey1) are present [5,37,38]. So these cells present important transcriptional, signaling and growthregulatory factors previously thought to be restricted to the pituitary embryonic phase (such as Hesx1, Lhx4, Pax6, Prop1, Otx2, Notch2, Notch3 [5,37]. Notch1 and Notch2, and legends Jag1 and Jag2 are not co localized with hormones but with S100 (except for Jag2) in abouthalf of the (S100+) marginal cells [39].

Notch 3 is Transgenically activated gene encodes notch pilots to amplify Sox2 immunoreactivity in embryonic pituitary [40]. Intensified Prop1 and Sox2 are mediated by Notch signaling system in adult pituitary stem cells. So Notch 3 is responsible for protracting self-renewal/proliferation of stem cells as in other adult tissues [41,42].

Pit-1 is a member of the POU family of transcription factors that regulate mammalian development. This pituitary-specific transcription factor is responsible for pituitary histogenesis and hormone expression in mammals. Pit1 is also important for regulation of the genes encoding prolactin and Thyroid-Stimulating Hormone beta subunit (TSH-beta) by Thyrotropin-Releasing Hormone (TRH) and cyclic AMP [43,44]. In embryonic rat pituitary Pit-1 and Prop1 are co-localized in pituitary in as small percentage of cells (1-10%) but in postnatal life Pit-1+ cells progressively diminish whereas no Prop1 cells subsist in adult pituitary [45]. Prop1 is probably down regulated before stem cell differentiation enhances [12]. By chemical composition Pit1 is consisted of 2 major protein domains, POUspecific and POU-homeo, both are necessary for high affinity DNA binding on genes encoding Growth Hormone (GH) and prolactin (PRL).

P57 a cyclin-dependent kinase inhibitor 1C (p57, Kip2, CDKN1C) which is encoded by the CDKN1C imprinted gene in humans. This cyclin-dependent kinase inhibitor 1C is a tight- binding inhibitor of several G1 cyclin/Cdk complexes and a negative regulator of cell proliferation. High levels of p57 (CDKN1C) mRNA in adult SC-SP may point to a similar role for p57 in stem cell compartment of adult pituitary gland [37].

Future Aspects

A wide spectrum of stem/progenitor cell biomarkers has recently been characterized in the last decade. Extrapolated information indisputably expands our awareness of pituitary physiology and pathology. Comprehension of ceaseless alterations of stem cells both in pituitary neoplastic, vascular, non-neoplastic and senescent conditions will enlighten pathogenetic mechanisms of pituitary. Stemness markers unhesitatingly shed light about mechanistic changes of underlying circumstances. Biomarker armamentarium will be an incentive in medicine for stratification and extraction of putative stem cells that will most likely lead allow clinicians in management of new treatment modalities of diverse pituitary pathologies.

References

  1. Sav A. Pituitary Stem/Progenitor Cells: Their Enigmatic Roles in Embryogenesis and Pituitary Neoplasia - A Review Article. J Neurol Disord. 2014; 2: 146.
  2. Sav A, Rotondo F, Syro LV, Scheithauer BW, Kovacs K. Biomarkers of pituitary neoplasms. Anticancer Res. 2012; 32: 4639-4654.
  3. Fauquier T, Rizzoti K, Dattani M, Lovell-Badge R, Robinson IC. SOX2-expressing progenitor cells generate all of the major cell types in the adult mouse pituitary gland. Proc Natl Acad Sci USA. 2008; 105: 2907-2912.
  4. Gleiberman AS, Michurina T, Encinas JM, Roig JL, Krasnov P, Balordi F, et al. Genetic approaches identify adult pituitary stem cells. Proc Natl Acad Sci U S A. 2008; 105: 6332-6337.
  5. Chen J, Gremeaux L, Fu Q, Liekens D, Van Laere S, Vankelecom H. Pituitary progenitor cells tracked down by side population dissection. Stem Cells. 2009; 27: 1182-1195.
  6. Vankelecom H. Stem cells in the postnatal pituitary? Neuroendocrinology. 2007; 85: 110-130.
  7. Vankelecom H. Non-hormonal cell types in the pituitary candidating for stem cell. Semin Cell Dev Biol. 2007; 18: 559-570.
  8. Krylyshkina O, Chen J, Mebis L, Denef C, Vankelecom H. Nestin-immunoreactive cells in rat pituitary are neither hormonal nor typical folliculo-stellate cells. Endocrinology. 2005; 146: 2376-2387.
  9. Lepore AC, Neuhuber B, Connors TM, Han SS, Liu Y, Daniels MP, et al. Long-term fate of neural precursor cells following transplantation into developing and adult CNS. Neuroscience. 2006; 139: 513-530.
  10. Semb H, Christofori G. The tumor-suppressor function of E-cadherin. Am J Hum Genet. 1998; 63: 1588-1593.
  11. Wong AS, Gumbiner BM. Adhesion-independent mechanism for suppression of tumor cell invasion by E-cadherin. J Cell Biol. 2003; 161: 1191-1203.
  12. Vankelecom H, Chen J. Pituitary stem cells: where do we stand? Mol Cell Endocrinol. 2014; 385: 2-17.
  13. Allaerts W, Vankelecom H. History and perspectives of pituitary folliculo-stellate cell research. Eur J Endocrinol. 2005; 153: 1-12.
  14. Morris J, Christian H. Folliculo-stellate cells: paracrine communicators in the anterior pituitary. Open Neuroendocrinol J. 2011; 4: 77-89.
  15. Wu W, Cogan JD, Pfaffle RW, Dasen JS, Frisch H, O'Connell SM, et al. Mutations in PROP1 cause familial combined pituitary hormone deficiency. Nat Genet. 1998; 18: 147-149.
  16. Ward RD, Raetzman LT, Suh H, Stone BM, Nasonkin IO, Camper SA. Role of PROP1 in pituitary gland growth. Mol Endocrinol. 2005; 19: 698-710.
  17. Takeda J, Seino S, Bell GI. Human Oct3 gene family: cDNA sequences, alternative splicing, gene organization, chromosomal location, and expression at low levels in adult tissues. Nucleic Acids Res. 1992; 20: 4613–4620.
  18. Tai MH, Chang CC, Kiupel M, Webster JD, Olson LK, Trosko JE. Oct4 expression in adult human stem cells: evidence in support of the stem cell theory of carcinogenesis. Carcinogenesis. 2005; 26: 495-502.
  19. Haupt Y, Bath ML, Harris AW, Adams JM. bmi-1 transgene induces lymphomas and collaborates with myc in tumorigenesis. Oncogene. 1993; 8: 3161-3164.
  20. Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL, et al. Bmi-1 is required for maintenance of adult self-renewing haematopoietic stem cells. Nature. 2003; 423: 302-305.
  21. Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Morrison SJ. Bmi-1 dependence distinguishes neural stem cell self-renewal from progenitor proliferation. Nature. 2003; 425: 962-967.
  22. Westerman BA, Blom M, Tanger E, van der Valk M, Song JY, van Santen M, et al. GFAP-Cre-mediated transgenic activation of Bmi1 results in pituitary tumors. PLoS One. 2012; 7: e35943.
  23. Bertolini G, Roz L, Perego P, Tortoreto M, Fontanella E, Gatti L, et al. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proc Natl Acad Sci USA. 2009; 106: 16281-16286.
  24. Salnikov AV, Gladkich J, Moldenhauer G, Volm M, Mattern J, Herr I. CD133 is indicative for a resistance phenotype but does not represent a prognostic marker for survival of non-small cell lung cancer patients. Int J Cancer. 2010; 126: 950-958.
  25. Corbeil D, Roper K, Hellwig A, Tavian M, Miraglia S, Watt SM, et al. The human AC133 hematopoietic stem cell antigen is also expressed in epithelial cells and targeted to plasma membrane protrusions. J Biol Chem. 2000; 275: 5512-5520.
  26. Sanai N, Alvarez-Buylla A, Berger MS. Neural stem cells and the origin of gliomas. N Engl J Med. 2005; 353: 811-822.
  27. Mizrak D, Brittan M, Alison M. CD133: molecule of the moment. J Pathol. 2008; 214: 3-9.
  28. Xu Q, Yuan X, Tunici P, Liu G, Fan X, Xu M, et al. Isolation of tumour stem-like cells from benign tumours. Br J Cancer. 2009; 101: 303-311.
  29. Garcia-Lavandeira M, Quereda V, Flores I, Saez C, Diaz-Rodriguez E, Japon MA, et al. A GRFa2/Prop1/stem (GPS) cell niche in the pituitary. PLoS One. 2009; 4: e4815.
  30. Gremeaux L, Fu Q, Chen J, Vankelecom H. Activated phenotype of the pituitary stem/progenitor cell compartment during the early-postnatal maturation phase of the gland. Stem Cells Dev. 2012; 21: 801-813.
  31. Cohen LE, Radovick S. Molecular basis of combined pituitary hormone deficiencies. Endocr Rev. 2002; 23: 431-442.
  32. Drouin J. Molecular mechanisms of pituitary differentiation and regulation: implications for hormone deficiencies and hormone resistance syndromes. Front Horm Res. 2006; 35: 74-87.
  33. Raetzman LT, Ward R, Camper SA. Lhx4 and Prop1 are required for cell survival and expansion of the pituitary primordia. Development. 2002; 129: 4229-4239.
  34. Raetzman LT, Ross SA, Cook S, Dunwoodie SL, Camper SA, Thomas PQ. Developmental regulation of Notch signaling genes in the embryonic pituitary: Prop1 deficiency affects Notch2 expression. Dev Biol. 2004; 265: 329-340.
  35. Zhu X, Lin CR, Prefontaine GG, Tollkuhn J, Rosenfeld MG. Genetic control of pituitary development and hypopituitarism. Curr Opin Genet Dev. 2005; 15: 332-340.
  36. Zhu X, Zhang J, Tollkuhn J, Ohsawa R, Bresnick EH, Guillemot F, et al. Sustained Notch signaling in progenitors is required for sequential emergence of distinct cell lineages during organogenesis. Genes Dev. 2006; 20: 2739-2753.
  37. Vankelecom H. Pituitary stem/progenitor cells: embryonic players in the adult gland? Eur J Neurosci. 2010; 32: 2063-2081.
  38. Chen J, Crabbe A, Van Duppen V, Vankelecom H. The notch signaling system is present in the postnatal pituitary: marked expression and regulatory activity in the newly discovered side population. Mol Endocrinol. 2006; 20: 3293-3307.
  39. Tando Y, Fujiwara K, Yashiro T, Kikuchi M. Localization of Notch signaling molecules and their effect on cellular proliferation in adult rat pituitary. Cell Tissue Res. 2013; 351: 511-519.
  40. Goldberg LB, Aujla PK, Raetzman LT. Persistent expression of activated Notch inhibits corticotrope and melanotrope differentiation and results in dysfunction of the HPA axis. Dev Biol. 2011; 358: 23-32.
  41. Basak O, Giachino C, Fiorini E, Macdonald HR, Taylor V. Neurogenic subventricular zone stem/progenitor cells are Notch1-dependent in their active but not quiescent state. J Neurosci. 2012; 32: 5654-5666.
  42. Conboy IM, Rando TA. The regulation of Notch signaling controls satellite cell activation and cell fate determination in postnatal myogenesis. Dev Cell. 2002; 3: 397-409.
  43. Dasen JS, O'Connell SM, Flynn SE, Treier M, Gleiberman AS, Szeto DP, et al. Reciprocal interactions of Pit1 and GATA2 mediate signaling gradient-induced determination of pituitary cell types. Cell. 1999; 97: 587-598.
  44. Szeto DP, Ryan AK, O'Connell SM, Rosenfeld MG. P-OTX: a PIT-1-interacting homeodomain factor expressed during anterior pituitary gland development. Proc Natl Acad Sci USA. 1996; 93: 7706-7710.
  45. Yoshida S, Kato T, Susa T, Cai LY, Nakayama M, Kato Y. PROP1 coexists with SOX2 and induces PIT1-commitment cells. Biochem Biophys Res Commun. 2009; 385: 11-15.

Download PDF

Citation: Sav A. Stemness Markers and Biomarkers of Pituitary Cytogenesis: Current Armamentarium of Molecular Targets. Austin Biomark Diagn. 2014;1(1): 3. ISSN: 2378-9867

Home
Journal Scope
Online First
Current Issue
Editorial Board
Instruction for Authors
Submit Your Article
Contact Us