Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where A. Lee Burns is active.

Publication


Featured researches published by A. Lee Burns.


Cell | 1999

Menin Interacts with the AP1 Transcription Factor JunD and Represses JunD-Activated Transcription

Sunita K. Agarwal; Siradanahalli C. Guru; Christina Heppner; Michael R. Erdos; Regina M. Collins; Sylvia Y. Park; Suraj Saggar; Settara C. Chandrasekharappa; Francis S. Collins; Allen M. Spiegel; Stephen J. Marx; A. Lee Burns

MEN1 is a tumor suppressor gene that encodes a 610 amino acid nuclear protein (menin) of previously unknown function. Using a yeast two-hybrid screen with menin as the bait, we have identified the transcription factor JunD as a direct menin-interacting partner. Menin did not interact directly with other Jun and Fos family members. The menin-JunD interaction was confirmed in vitro and in vivo. Menin repressed transcriptional activation mediated by JunD fused to the Gal4 DNA-binding domain from a Gal4 responsive reporter, or by JunD from an AP1-responsive reporter. Several naturally occurring and clustered MEN1 missense mutations disrupted menin interaction with JunD. These observations suggest that menins tumor suppressor function involves direct binding to JunD and inhibition of JunD activated transcription.


Oncogene | 2001

The tumor suppressor protein menin interacts with NF-κB proteins and inhibits NF-κB-mediated transactivation

Christina Heppner; Karl Y. Bilimoria; Sunita K. Agarwal; MaryBeth Kester; Leslie J. Whitty; Siradanahalli C. Guru; Settara C. Chandrasekharappa; Francis S. Collins; Allen M. Spiegel; Stephen J. Marx; A. Lee Burns

Multiple endocrine neoplasia type 1 is an autosomal dominant tumor syndrome. Manifestations include neoplasms of the parathyroid glands, enteropancreatic neuroendocrine cells, and the anterior pituitary gland. The MEN1 tumor suppressor gene encodes menin, a 610 amino acid nuclear protein without sequence homology to other proteins. To elucidate menin function, we used immunoprecipitation to identify interacting proteins. The NF-κB proteins p50, p52 and p65 were found to interact specifically and directly with menin in vitro and in vivo. The region of NF-κB proteins sufficient for binding to menin is the N-terminus. Furthermore, amino acids 305–381 of menin are essential for this binding. Menin represses p65-mediated transcriptional activation on NF-κB sites in a dose-dependent and specific manner. Also, PMA (phorbol 12-myristate 13-acetate)-stimulated NF-κB activation is suppressed by menin. These observations suggest that menins ability to interact with NF-κB proteins and its modulation of NF-κB transactivation contribute to menins tumor suppressor function.


Molecular and Cellular Biology | 2003

Of mice and MEN1: Insulinomas in a conditional mouse knockout.

Judy S. Crabtree; Peter C. Scacheri; Jerrold M. Ward; Sara McNally; Gary P. Swain; Cristina Montagna; Jeffrey H. Hager; Douglas Hanahan; Helena Edlund; Mark A. Magnuson; Lisa Garrett-Beal; A. Lee Burns; Thomas Ried; Settara C. Chandrasekharappa; Stephen J. Marx; Allen M. Spiegel; Francis S. Collins

ABSTRACT Patients with multiple endocrine neoplasia type 1 (MEN1) develop multiple endocrine tumors, primarily affecting the parathyroid, pituitary, and endocrine pancreas, due to the inactivation of the MEN1 gene. A conditional mouse model was developed to evaluate the loss of the mouse homolog, Men1, in the pancreatic beta cell. Men1 in these mice contains exons 3 to 8 flanked by loxP sites, such that, when the mice are crossed to transgenic mice expressing cre from the rat insulin promoter (RIP-cre), exons 3 to 8 are deleted in beta cells. By 60 weeks of age, >80% of mice homozygous for the floxed Men1 gene and expressing RIP-cre develop multiple pancreatic islet adenomas. The formation of adenomas results in elevated serum insulin levels and decreased blood glucose levels. The delay in tumor appearance, even with early loss of both copies of Men1, implies that additional somatic events are required for adenoma formation in beta cells. Comparative genomic hybridization of beta cell tumor DNA from these mice reveals duplication of chromosome 11, potentially revealing regions of interest with respect to tumorigenesis.


Trends in Endocrinology and Metabolism | 2001

Multiple endocrine neoplasia type 1: new clinical and basic findings

Debra H. Schussheim; Monica C. Skarulis; Sunita K. Agarwal; William F. Simonds; A. Lee Burns; Allen M. Spiegel; Stephen J. Marx

Multiple endocrine neoplasia type 1 (MEN1) provides a prime example of how a rare disease can advance our understanding of basic cell biology, neoplasia and common endocrine tumors. MEN1 is expressed mainly as parathyroid, enteropancreatic neuroendocrine, anterior pituitary and foregut carcinoid tumors. It is an autosomal dominant disease caused by mutation of the MEN1 gene. Since its identification, the MEN1 gene has been implicated in many common endocrine and non-endocrine tumors. This is a brief overview of recent scientific advances relating to MEN1, including newly recognized clinical features that are now better characterized by genetic analysis, insights into the function of the MEN1-encoded protein menin, and refined recommendations for mutation testing and tumor screening, which highlight our increasing understanding of this complex syndrome.


Annals of the New York Academy of Sciences | 2004

Molecular Pathology of the MEN1 Gene

Sunita K. Agarwal; A. Lee Burns; Karen E. Sukhodolets; Patricia A. Kennedy; Victor H. Obungu; Alison Burgess Hickman; Michael E. Mullendore; Ira Whitten; Monica C. Skarulis; William F. Simonds; Carmen M. Mateo; Judy S. Crabtree; Peter C. Scacheri; Youngmi Ji; Elizabeth A. Novotny; Lisa Garrett-Beal; Jerrold M. Ward; Steven K. Libutti; H. Richard Alexander; Aniello Cerrato; Michael Parisi; Sonia Santa Anna-A; Brian Oliver; Settara C. Chandrasekharappa; Francis S. Collins; Allen M. Spiegel; Stephen J. Marx

Abstract: Multiple endocrine neoplasia type 1 (MEN1), among all syndromes, causes tumors in the highest number of tissue types. Most of the tumors are hormone producing (e.g., parathyroid, enteropancreatic endocrine, anterior pituitary) but some are not (e.g., angiofibroma). MEN1 tumors are multiple for organ type, for regions of a discontinuous organ, and for subregions of a continuous organ. Cancer contributes to late mortality; there is no effective prevention or cure for MEN1 cancers. Morbidities are more frequent from benign than malignant tumor, and both are indicators for screening. Onset age is usually earlier in a tumor type of MEN1 than of nonhereditary cases. Broad trends contrast with those in nonneoplastic excess of hormones (e.g., persistent hyperinsulinemic hypoglycemia of infancy). Most germline or somatic mutations in the MEN1 gene predict truncation or absence of encoded menin. Similarly, 11q13 loss of heterozygosity in tumors predicts inactivation of the other MEN1 copy. MEN1 somatic mutation is prevalent in nonhereditary, MEN1‐like tumor types. Compiled germline and somatic mutations show almost no genotype/phenotype relation. Normal menin is 67 kDa, widespread, and mainly nuclear. It may partner with junD, NF‐kB, PEM, SMAD3, RPA2, FANCD2, NM23β, nonmuscle myosin heavy chain II‐A, GFAP, and/or vimentin. These partners have not clarified menins pathways in normal or tumor tissues. Animal models have opened approaches to menin pathways. Local overexpression of menin in Drosophila reveals its interaction with the jun‐kinase pathway. The Men1+/− mouse has robust MEN1; its most important difference from human MEN1 is marked hyperplasia of pancreatic islets, a tumor precursor stage.


Molecular and Cellular Biology | 2003

The 32-Kilodalton Subunit of Replication Protein A Interacts with Menin, the Product of the MEN1 Tumor Suppressor Gene

Karen E. Sukhodolets; Alison Burgess Hickman; Sunita K. Agarwal; Maxim V. Sukhodolets; Victor H. Obungu; Elizabeth A. Novotny; Judy S. Crabtree; Settara C. Chandrasekharappa; Francis S. Collins; Allen M. Spiegel; A. Lee Burns; Stephen J. Marx

ABSTRACT Menin is a 70-kDa protein encoded by MEN1, the tumor suppressor gene disrupted in multiple endocrine neoplasia type 1. In a yeast two-hybrid system based on reconstitution of Ras signaling, menin was found to interact with the 32-kDa subunit (RPA2) of replication protein A (RPA), a heterotrimeric protein required for DNA replication, recombination, and repair. The menin-RPA2 interaction was confirmed in a conventional yeast two-hybrid system and by direct interaction between purified proteins. Menin-RPA2 binding was inhibited by a number of menin missense mutations found in individuals with multiple endocrine neoplasia type 1, and the interacting regions were mapped to the N-terminal portion of menin and amino acids 43 to 171 of RPA2. This region of RPA2 contains a weak single-stranded DNA-binding domain, but menin had no detectable effect on RPA-DNA binding in vitro. Menin bound preferentially in vitro to free RPA2 rather than the RPA heterotrimer or a subcomplex consisting of RPA2 bound to the 14-kDa subunit (RPA3). However, the 70-kDa subunit (RPA1) was coprecipitated from HeLa cell extracts along with RPA2 by menin-specific antibodies, suggesting that menin binds to the RPA heterotrimer or a novel RPA1-RPA2-containing complex in vivo. This finding was consistent with the extensive overlap in the nuclear localization patterns of endogenous menin, RPA2, and RPA1 observed by immunofluorescence.


FEBS Letters | 1989

Cloning and sequencing of the human nucleolin cDNA.

Meera Srivastava; Patrick J. Fleming; Harvey B. Pollard; A. Lee Burns

A cDNA containing the entire coding region for human nucleolin has been isolated from a λ gt10 human retinal library using a bovine cDNA probe. The cDNA hybridized to a transcript of 3000 bases from fast‐dividing cells, as well as terminally differentiated tissues of several species. Translation of the nucleotide sequence revealed a long open reading frame which predicts a 707 amino acid protein with several distinct domains. These include repeating elements, four conserved RNA‐binding regions, a glycine‐rich carboxy‐terminal domain and sites for phosphorylation, glycosylation and dibasic cleavage. Human and bovine nucleolin exhibited more additions and/or substitutions of aspartate, glutamate and serine residues in the chromatin‐binding domains by comparison with the hamster and mouse nucleolins. These differences may be related to species‐specific functions in transcription.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Transcription factor JunD, deprived of menin, switches from growth suppressor to growth promoter

Sunita K. Agarwal; Elizabeth A. Novotny; Judy S. Crabtree; Jonathan Weitzman; Moshe Yaniv; A. Lee Burns; Settara C. Chandrasekharappa; Francis S. Collins; Allen M. Spiegel; Stephen J. Marx

Different components of the AP1 transcription factor complex appear to have distinct effects on cell proliferation and transformation. In contrast to other AP1 components, JunD has been shown to inhibit cell proliferation. Also, in prior studies, JunD alone bound menin, product of the MEN1 tumor suppressor gene, and JunDs transcriptional activity was inhibited by menin, suggesting that JunD might achieve all or most of its unique properties through binding to menin. Analyses of JunD and menin effects on proliferation, morphology, and cyclin D1 in stable cell lines unmasked an unexpected growth promoting activity of JunD. Whereas stable overexpression of wild-type (wt) mouse JunD in JunD–/– immortalized fibroblasts inhibited their proliferation and reverted their transformed-like phenotype, overexpression of a missense mouse JunD mutant (mJunDG42E) with disabled binding to menin showed opposite or growth promoting effects. Similarly, stable overexpression of wt mouse JunD in wt immortalized fibroblasts inhibited growth. In contrast, its overexpression in Men1–/– immortalized fibroblasts enhanced their already transformed-like characteristics. To conclude, JunD changed from growth suppressor to growth promoter when its binding to menin was prevented by a JunD mutant unable to bind menin or by Men1-null genetic background.


Human Mutation | 1998

Common ancestral mutations in the MEN1 gene is likely responsible for the prolactinoma variant of MEN1 (MEN1Burin) in four kindreds from Newfoundland

Shodimu Emmanuel Olufemi; Jane Green; Pachiappan Manickam; Siradanahalli C. Guru; Sunita K. Agarwal; Mary Beth Kester; Qihan Dong; A. Lee Burns; Allen M. Spiegel; Stephen J. Marx; Francis S. Collins; Settara C. Chandrasekharappa

Familial multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disorder with affected individuals developing parathyroid, gastrointestinal (GI) endocrine, and anterior pituitary tumors. Four large kindreds from the Burin peninsula/Fortune Bay area of Newfoundland with prominent features of prolactinomas, carcinoids, and parathyroid tumors (referred to as MEN1Burin) have been described, and they show linkage to 11q13, the same locus as that of MEN1. Haplotype analysis with 16 polymorphic markers now reveals that representative affected individuals from all four families share a common haplotype over a 2.5 Mb region. A nonsense mutation in the MEN1 gene has been found to be responsible for the disease in the affected members in all four of the MEN1Burin families, providing convincing evidence of a common founder. Hum Mutat 11:264–269, 1998. Published 1998 Wiley‐Liss, Inc.


Cancer Genetics and Cytogenetics | 1998

Comparative genomic hybridization analysis of human parathyroid tumors.

Sunita K. Agarwal; Evelin Schröck; Mary Beth Kester; A. Lee Burns; Clara S. Heffess; Thomas Ried; Stephen J. Marx

Primary hyperparathyroidism is characterized by hypercalcemia and elevated parathyroid hormone levels. It can be caused by overactivity of one (adenoma or carcinoma) or more (hyperplasia or multiple adenoma) parathyroid glands. Parathyroid adenoma and hyperplasia are usually mono- or oligoclonal neoplasms. To establish whether parathyroid cancer has a genetic composition distinct from parathyroid adenoma, we analyzed 10 adenoma and 10 carcinoma cases by comparative genomic hybridization (CGH). Results show clear differences between the constitution of adenoma and carcinoma genomic DNA. The most frequent genomic alterations in adenoma included deletions on chromosomes 11, 17 (5 of 10 cases), and 22 (7 of 10 cases). In parathyroid carcinoma, frequent chromosomal deletions were on chromosome arm 1p (4 of 10 cases) and chromosome 17 (3 of 10 cases), and gains were on chromosome 5 (3 of 10 cases). Our data indicate that different genetic changes could contribute to the development of parathyroid adenoma and carcinoma; genomic losses predominate in adenoma, and gains along with some losses are found in carcinoma. Furthermore, the CGH results implicate several chromosomal regions that may harbor genes that could be potentially involved in the development of parathyroid adenoma and carcinoma.

Collaboration


Dive into the A. Lee Burns's collaboration.

Top Co-Authors

Avatar

Stephen J. Marx

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Allen M. Spiegel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sunita K. Agarwal

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Siradanahalli C. Guru

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Pachiappan Manickam

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Judy S. Crabtree

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Irina A. Lubensky

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge