ENDOCRINE GENETICS
     

Constantine Stratakis, M.D., D.Sc., Principal Investigator
Lawrence S. Kirschner, M.D., Ph.D., Senior Staff Fellow
Thalia Bei, Ph.D., Guest Researcher
Isabelle Bourdeau, M.D., Guest Researcher
Ludmila Matyakhina, Ph.D., Visiting Fellow
Fabiano Sandrini, M.D., Visiting Fellow
Constantine Farmakidis, B.S., Predoctoral Fellow
Laura Gosselink, B.A., Predoctoral Fellow
Sara M. Lenherr, B.S., Predoctoral Fellow
Audrey Robinson-White, Ph.D., Volunteer

For More Information

Constantine Stratakis
 

The goal of our work is to understand the genetic and molecular mechanisms leading to disorders that affect the adrenal cortex, with emphasis on disorders that are developmental, hereditary, and associated with adrenal hypoplasia or hyperplasia, multiple tumors, and abnormalities in other endocrine glands (i.e., the pituitary gland). In this context, our laboratory has studied congenital adrenal hypoplasia caused by Allgrove syndrome (or triple A syndrome) and multiple endocrine deficiencies (APECED syndrome), familial hyperaldosteronism, adrenocortical and thyroid cancer, pituitary tumors and multiple endocrine neoplasia syndromes (MEN 1, Cowden disease) affecting the pituitary, thyroid, and adrenal glands, and Carney complex, an autosomal dominant disease that affects the adrenal cortex. Carney complex is the only inherited form of adrenal gland-dependent Cushing’s syndrome; it is also a mutliple endocrine neoplasia affecting the pituitary and thyroid glands and the gonads and is associated with a variety of other tumors, including myxomas (heart and other myxomas) and schwannomas (it is one of three conditions associated with inherited schwannomas; the other two are neurofibromatosis and isolated schwannomatosis), and skin pigmentation defects (lentigines, café-au-lait spots, and nevi). Recently, our laboratory identified the regulatory subunit type 1-_ (Ri_) __of protein kinase A (PKA), which is coded by the PRKAR1A gene, the gene responsible for almost half the cases of Carney complex. Since then, our laboratory has largely focused on PKA-stimulated signaling pathways, PKA effects on tumor suppression and/or development, the cell cycle and chromosomal stability, and PKA-dependent hormonal interactions.

Carney Complex Genetics

Kirschner, Bei, Bourdeau, Matyakhina, Sandrini, Stratakis in collaboration with J.A. Carney (Mayo Clinic)
Families with Carney complex (CNC) and related syndromes from a number of collaborating institutions worldwide have been accessed. Genetic linkage analysis located two loci harboring genes for CNC on chromosomes 2 (2p16) and 17 (17q22-24) while a possible third locus for this genetically heterogeneous condition is currently under investigation. With the application of state-of-the-art molecular cytogenetic techniques, the participation of the two genomic loci on chromosomes 2 and 17 in the expression of the disease is undergoing study. A comprehensive genetic and physical map of the 2p16 chromosomal region was constructed for the cloning of the CNC-associated sequences from this region. Studies in cultured primary tumor cell lines (established from our patients) identified a region of genomic instability in the center of the map. Tumor studies (Figure 1) led to the identification of the PRKAR1A gene on 17q22-24 as the gene responsible for CNC in approximately 40 percent of cases of the disease. PRKAR1A was identified as a novel tumor suppressor gene in CNC-associated tumors; it is also the main regulatory subunit of protein kinase A (PKA), a central signaling pathway for many cellular functions and hormonal responses. With more patients with CNC now participating in genotype-phenotype correlation studies, the studies are expected to shed light on the complex biochemical and molecular pathways regulated by PRKAR1A and PKA.

Figure 3

Figure 3

Mapping of the PRKAR1A gene on chromosome 17 and identification of genetic markers of the PRKAR1A locus that were used for genetic studies in CNC families (left panel). A probe containing the PRKAR1A gene from a chromosome 17 human genome library was then used in an ovarian tumor cell line from a patient with CNC in a fluorescent in situ hybridization experiment (right panel); the cells demonstrate allelic loss of the PRKAR1A-containing probe (one copy per cell instead of two), suggesting that PRKAR1A is a tumor suppressor gene.

PRKAR1A Effects on Protein Kinase A Activity and Endocrine Tumor Development
Kirschner, Matyakhina, Sandrini, Lenherr, Gosselink, Robinson-White, Stratakis in collaboration with Y. Cho-Chung (NCI)
The functional consequences of PRKAR1A mutations are being investigated in cell lines established from CNC patients and their tumors. Both cAMP and PKA activity are measured in the cell lines, along with the expression of the other subunits of the PKA tetramer. We have established stable transfectants of antisense PRKAR1A constructs in mouse endocrine and other commercially available cell lines and have studied the cell lines for the effects of PRKAR1A silencing on cell growth, differentiation, and proliferation. It is hypothesized that the tumorigenicity of PRKAR1A-inactivating mutations relies on the switch from type-I PKA (based almost exclusively on endocrine cells on PRKAR1A) to type-II PKA activity; cell lines with an antisense PRKAR1A construct are believed to be a representative model of the in vivo situation in CNC patients. In addition, we are looking for mutations of the PRKAR1A gene that would further establish its role as a general tumor suppressor in sporadic endocrine and nonendocrine tumors (thyroid adenomas and carcinomas, adrenocortical adenomas and carcinomas, ovarian carcinomas, melanomas and benign and malignant pigmented lesions, and heart myxomas); a variety of investigators within the NIH and around the world provide specimens on a collaborative basis.

PRKAR1A Animal Models

Kirschner, Gosselink, Lenherr, Stratakis in collaboration with H. Westphal (LMGD, NICHD)
Given that the PRKAR1A-knockout (KO) (-/-) mouse (created several years ago by S. McKnight of the University of Washington, Seattle) dies on day nine of embryonic development of heart and central nervous system abnormalities and PRKAR1A’s involvement in CNC, our laboratory became interested in developing alternative animal models that would address the complex in vivo effects of PRKAR1A inactivation. The several animal models under development in our laboratory fall into two categories: conditional PRKAR1A KOs in endocrine tissues (adrenal cortex, anterior lobe of the pituitary and the thyroid gland) and (2) transgenic antisense PRKAR1A expression.

PRKAR1A, the Cell Cycle, Chromosomal Stability, and Other Signaling Pathways

Matyakhina, Robinson-White, Stratakis in collaboration with V. Papadopoulos (Georgetown University, Washington DC)
Genes implicated in cyclic nucleotide-dependent signaling have long been considered likely candidates for endocrine tumorigenesis. Somatic activating mutations in a number of G-protein coupled receptors (GPCRs) and the gene coding for a subunit of the stimulatory G protein (GNAS1) lead to increased cAMP production and are responsible for a number of endocrine tumors of various types. To date, however, there is no convincing evidence that GNAS1 or GPCR activation in the absence of additional genetic abnormalities is involved in cancer. Individuals with McCune-Albright syndrome (MAS, a disease similar to CNC) who bear somatic GNAS1 mutations in their endocrine glands may be predisposed to some cancers. However, activation of additional pathways and/or other changes appear to be required for the in vitro transformation of 3T3 or FRTL5 cells by constitutively active GPCR transgenes or in other settings of increased cAMP signaling that lead to malignant transformation. Thus, other genes that regulate PKA function and increase cAMP-dependent proliferation and related signals may be altered in the process of endocrine tumorigenesis initiated by a mutant PRKAR1A, a gene with important functions in the cell cycle as well as in chromosomal stability. Our work aims to identify the interactions of PRKAR1A by studying mitogenic and other growth signaling pathways in cell lines that express the antisense PRKAR1A contructs referred to above. In addition, chromosomal stability in both human and mouse cell lines in which PRKAR1A has been inactivated is under investigation with the techniques of classic and molecular cytogenetics, including fluorescent in situ hybridization (FISH), spectral karyotyping (SKY), and comparative genomic hybridization (CGH). Finally, more recently, we are investigating proteins that are directly bound to PRKAR1A (RI_) and regulate its function, including a novel protein, PAP7 that a collaborating laboratory identified. Compartmentalization of PKA function is supposed to be mediated by anchoring proteins; almost all the PKA anchoring proteins known to date, however, are bound to type II-PKA. PAP7 may be the first PKA type-I-specific anchoring protein.

Genetic Investigations on Other Adrenocortical Diseases and Tumors

Bourdeau, Sandrini, Farmakidis, Stratakis in collaboration with S. Libutti (NCI), W.Y. Chan (NICHD), J.A. Carney (Mayo Clinic), M Stowasser and D Torpy (University of Queensland, Australia), A. Lacroix (University of Montreal, Canada), and J. Bertherat (Hospital Cochin, France)
Our work aims at using general and pathway-specific microarrays on a variety of adrenocortical tumors to identify genes with important functions in adrenal oncogenetics; examining specific candidate genes (such as TP53 and other tumor suppressors and oncogenes) for their roles in adrenocortical tumors and development; and identifying by positional cloning additional genes with a role in inherited adrenocortical diseases. Our laboratory engages in collaborations for a large part of its work, as is evident from the number of collaborating investigators and institutions. As part of this work, we and our collaborators have recently completed the following: identification of a novel TP53 mutation in a cohort of adrenocortical tumors from southern Brazil with important implications for p53’s function in adrenocortical tumor suppression; development of a genomewide screen for the identification of gene(s) responsible for inherited adrenocortical aldosteronomas (familial hyperaldosteronism type II - FH-II) and that identified a locus for FH-II on chromosome 7 (7p22); ongoing development of a genomewide screen for the identification of a syndrome composed of familial paragangliomas and adrenal, gastric stromal. and pulmonary tumors; and the identification of new mutations in the APECED and Allgrove syndrome genes leading to congenital adrenal hypoplasia.

Genetic Investigations on Other Endocrine Neoplasias and Related Syndromes

Bourdeau, Sandrini, Stratakis
Additional work in our laboratory is exploring the genetics of CNC- and adrenal-related endocrine tumors, including childhood adrenocortical cancer and thyroid and pituitary tumors. As part of this work, we described a novel form of acromegaloid syndrome in which the responsible genetic defect was associated with a chromosome 11 abnormality (see figure). Finally, we are identifying the genetic defects in patients with CNC-related syndromes (the lentigenoses, i.e., Peutz-Jeghers syndrome and others) largely in collaboration with several other investigators at the NIH and elsewhere.

Figure 4

 

Figure 4

A) G-banding high resolution karyotype of a patient with pseudoacromegaly; the arrows point to a newly described chromosome 11 abnormality [inv(11)(p15.3;q23.3)], which is undetectable without proper analysis of band density. B) Fluorescent in situ hybridization on cultured lymphocytes from the proband with two probes that hybridize to the 11p telomere (11pter) and to the MLL gene on 11q23, respectively, showing that, on one chromosome 11, the two probes were positioned proximal to each other (arrow), whereas they hybridized to their expected positions on the other chromosome.

Clinical Investigations in the Diagnosis and Treatment of Adrenal and Pituitary Tumors
Kirschner, Stratakis, Keil
Patients with adrenal tumors and other types of Cushing’s syndrome (and occasionally other pituitary tumors) come to the NIH Clinical Center for diagnosis and treatment. Ongoing investigations include the prevalence of ectopic hormone receptor expression in adrenal adenomas and massive macronodular adrenocortical disease; the diagnostic use of high- sensitivity magnetic resonance imaging for the earlier detection of pituitary tumors; the diagnosis, management, and postoperative care of children with Cushing’s disease and other pituitary tumors.

 

 

PUBLICATIONS

  1. Balemans W, Ebeling M, Patel N, Hul EV, Olson P, Dioszegl, Lacza C, Wuyts W, Van den Ende J, Willems P, Paes-Alves AF, Hill S, Bueno M, Ramos FJ, Tacconi P, Dikkers FG, _Stratakis CA, Lindpaintner K, Vickery B, Foernzler D, Van Hul W. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST). Hum Mol Genet 2001;10:537-543.
  2. Boardman LA, Couch FJ, Burgart LJ, Schwartz D, Berry R, McDonnell SK, Schaid DJ, Hartmann LC, Schroeder JJ, Stratakis CA, Thibodeau SN. Genetic heterogeneity in Peutz-Jeghers syndrome. Hum Mutat 2000;16:23-30.
  3. Carney JA, Boccon-Gibod L, Jarka D, Tanaka Y, Swee RG, Unni KK, Stratakis CA. Osteochondromyxoma of bone: a congenital tumor associated with lentigines and other unusual disorders. Am J Surg Pathol 2001;25:164-176.
  4. Doppman JL, Chrousos GP, Papanicolaou DA, Stratakis CA, Bartlet DL, Nieman LK. Adrenococrticotropin (ACTH)-independent macronodular adrenal hyperplasia; an uncommon cause of primary adrenal hypercortisolism. Radiology 2000;216:797-802.
  5. Egan CA, Stratakis CA, Turner ML. Multiple lentigines associated with cutaneous myxomas. Am J Acad Dermatol 2001;44:282-284.
  6. Glasow A, Horn L-C, Taymans SE, Stratakis CA, Kelly PA, Kohler U, Gillespie J, Vonderhaar BK, Bornstein SR. Mutational analysis of the prolactin receptor (PRLR) gene in human breast tumors with differential PRLR protein expression. J Clin Encocrinol Metab 2001;86:3826-3832.
  7. Kirschner LS, Carney JA, Pack SD, Taymans SE, Giatzakis C, Cho YS, Cho-Chung YS, Stratakis CA. Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit in patients with the Carney complex. Nat Genet 2000;26:89-92.
  8. Kirschner LS, Sandrini F, Monbo J, Lin JP, Carney JA, Stratakis CA. Genetic heterogeneity and spectrum of mutations of the PRKAR1A gene in patients with Carney complex. Hum Mol Genet 2000;9:3037-3046.
  9. Kirschner LS, Stratakis CA. Gene structure of the human ubiquitin fusion gene Uba80 (RPS27a) and one of its pseudogenes. Biochem Biophys Res Commun 2000;270:1106-1110.
  10. Kirschner LS, Stratakis CA. Isolated familial somatotropinomas: does the disease map to 11q13 or to 2p16? (Letter) J Clin Endocrinol Metab 2000;85:4920-4924.
  11. Lafferty AR, Torpy DJ, Stowasser M, Taymans SE, Lin JP, Huggard P, Gordon RD, Stratakis CA. A novel genetic locus for low renin hypertension: familial hyperaldosteronism type II maps to chromosome 7 (7p22). J Med Genet 2000;37:831-835.
  12. Marsh DJ, Stratakis CA. Hamartoma and lentiginosis syndromes: clinical and molecular aspects. In: Dahia PLM, Eng, C, eds. Genetic disorders of endocrine neoplasia. Front Horm Res 2001;28:167-213.
  13. Ng D, Stratakis CA. Premature adrenal cortical dysfunction in mandibuloacral dysplasia, a progeroid-like syndrome. Am J Med Genet 2000;95:293-295.
  14. Pack S, Kirshner LS, Pak E, Carney JA, Zhuang Z, Stratakis CA. Pituitary tumors in patients with the complex of spotty skin pigmentation, myxomas, endocrine overactivity and schwannomas (Carney complex): evidence for progression from somatomammotroph hyperplasia to adenoma. J Clin Encocrinol Metab 2000;85:3860-3865.
  15. Raff SB, Carney JA, Krugman D, Doppman JL, Stratakis CA. Prolactin secretion abnormalities in patients with the "syndrome of spotty skin pigmentation, myxomas, endocrine overactivity and schwannomas." J Pediatr Endocrinol Metab 2000;13:373-379.
  16. Ribeiro RC, Sandrini F, Figueiredo B, Zambetti GP, Michalkiewicz E, Lafferty AR, DeLacerda L, Rabin M, Cadwell C, Sampaio G, Cat I, Stratakis CA, Sandrini R. An inherited p53 mutation that contributes in a tissue-specific manner to pediatric adrenal cortical carcinoma. Proc Natl Acad Sci USA 2001;98:9330-9335.
  17. Sandrini F, Farmakidis C, Kirschner LS, Wu S-M, Tullio-Pelet A, Lyonnet S, Metzger DL, Bourdony CJ, Tiosano D, Chan WY, Stratakis CA. Spectrum of mutations of the AAAS Gene in Allgrove Syndrome: lack of mutations in six kindreds with isolated resistance to corticotropin. J Clin Endocrinol Metab 2001;86:5433-5437.
  18. Sprunger LK, Meisler MH, Stratakis CA. Recombination between the sodium channel SCN8A and the Allgrove syndrome gene in a Puerto Rican kindred. J Endocr Genet 2000;1:165-169.
  19. Stratakis CA. Clinical genetics of multiple endocrine neoplasias, Carney complex and related syndromes. J Endocr Invest 2001;24:370-383.
  20. Stratakis CA. Cushing syndrome and Addison disease. In: Hughes IA, Clark AJL, eds. Endocrine development: adrenal diseases in childhood, clinical and molecular aspects. Basel: Karger AG, 2000;150-173.
  21. Stratakis CA. Genetics of adrenocortical tumors: Carney complex. Ann Endocrinol (Paris) 2001;62:180-184.
  22. Stratakis CA. Genetics of Peutz-Jeghers syndrome, Carney complex and other familial lentiginoses. Horm Res 2000;54:334-343.
  23. Stratakis CA, Ball DW. Multiple endocrine neoplasia and related syndromes. J Pediatr Endocrinol Metab 2000;13:457-465.
  24. Stratakis CA, Kirschner LS, Carney JA. Clinical and molecular features of the Carney complex: diagnostic criteria and recommendations for patient evaluation. J Clin Endocrinol Metab 2001;86:4041-4046.
  25. Stratakis CA, Lafferty A, Taymans SE, Gafni RI, Meck JM, Blancato J. Anisomastia associated with interstitial duplication of chromosome 16, mental retardation, obesity, dysmorphic facies, and digital anomalies: molecular mapping of a new syndrome by fluorescent in situ hybridization and microsatellites to 16q13 (D16S419-D16S503). J Clin Endocrinol Metab 2000;85:3396-3401.
  26. Stratakis CA, Papageorgiou T, Premkumar N, Kirschner LS, Taymans SE, Pack S, Zhuang Z, Oelkers WH, Carney JA. Ovarian lesions in Carney complex: clinical genetics studies and possible predisposition to malignancy. J Clin Encocrinol Metab 2000;85:4359-4366 [editorial in J Clin Endocrinol Metab 2000;85:4010-4012].
  27. Stratakis CA, Russovici D, Kulin HE, Finkelstein JW. ACTH and cortisol responses to L-DOPA and insulin-induced hypoglycemia in children with short stature. J Pediatr Endocrinol Metab 2000;13:1095-1100.
  28. Stratakis CA, Schussheim DH, Freedman SM, Keil MF, Pack SF, Agarwal SK, Skarulis MC, Weil RJ, Lubensky LA, Zhuang Z, Oldfield EH, Marx SJ. Pituitary macroadenoma in a 5-year-old: an early expression of Multiple Endocrine Neoplasia type 1. J Clin Encocrinol Metab 2000;85:4776-4780.
  29. Stratakis CA, Taymans SE, Daruwala R, Song J, Levine M. Mapping of the human genes (SLC23A2 and SLC23A1) coding for vitamin C transporters 1 and 2 (SVCT1 and SVCT2) to 5q23 and 20p12, respectively. J Med Genet 2000;37:E20-E23.
  30. Stratakis CA, Taymans SE, Schteingart D, Haddad BR. Segmental uniparental isodisomy (UPD) for 2p16 without clinical symptoms: implications for UPD and other genetic studies of chromosome 2 [Brief Report]. J Med Genet 2001;38:106-109.
  31. Stratakis CA, Turner ML, Lafferty A, Toro JR, Hill S, Meck JM, Blancato J. A syndrome of overgrowth and acromegaloidism with normal growth hormone secretion is associated with chromosome 11 pericentric inversion [Brief Report]. J Med Genet 2001;38:338-343.
  32. Torpy DJ, Stratakis CA, Chrousos GP. Familial hyperaldosteronism. Braz J Med Biol Res 2000;33:1149-1155.
  33. Tsilou E, Stratakis CA, Rubin CA, Hay BN, Patronas N, Kaiser-Kupfer MI. Ophthalmic manifestations of Allgrove syndrome: report of a case. Clin Dysmorphol 2001;10:231-233.
  34. Watson JC, Stratakis CA, Bryant-Greenwood PK, Koch CA, Kirschner LS, Ngyen T, Carney JA, Oldfield EH. Neurosurgical implications of Carney complex. J Neurosurg 2000;92:413-418.
  35. Zouboulis CC, Stratakis CA, Gollnick HP, Orfanos CE. Keratosis pilaris/ulerythema ophryogenes and 18p deletion: is it possible that the LAMA1 gene is involved? [Brief Report]. J Med Genet 2001;38:127-128.