Network


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

Hotspot


Dive into the research topics where Pachiappan Manickam is active.

Publication


Featured researches published by Pachiappan Manickam.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Memory-specific temporal profiles of gene expression in the hippocampus

Sebastiano Cavallaro; Velia D'Agata; Pachiappan Manickam; Franck Dufour; Daniel L. Alkon

Many experiments in the past have demonstrated the requirement of de novo gene expression during the long-term retention of learning and memory. Although previous studies implicated individual genes or genetic pathways in learning and memory, they did not uncover the collective behaviors or patterns of the genes. We have used genome-scale screening to analyze gene expression during spatial learning of rats in the Morris water maze. Our results show distinct temporal gene expression profiles associated with learning and memory. Exogenous administration of one peptide whose sustained increase during memory retention was implicated by microarray analysis, fibroblast growth factor (FGF)-18, improved spatial learning behavior, suggesting that pharmacological modulation of pathways and targets identified may allow new therapeutic approaches for improving learning and memory. Results of this study also suggest that while learning and physical activity involve common groups of genes, the behavior of learning and memory emerges from unique patterns of gene expression across time.


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.


Genes, Chromosomes and Cancer | 2000

MEN1 gene mutation analysis of high‐grade neuroendocrine lung carcinoma

Larisa V. Debelenko; Jennifer I. Swalwell; Michael J. Kelley; Elisabeth Brambilla; Pachiappan Manickam; Galina Baibakov; Sunita K. Agarwal; Allen M. Spiegel; Stephen J. Marx; Settara C. Chandrasekharappa; Francis S. Collins; William D. Travis; Michael R. Emmert-Buck

Neuroendocrine tumors of the lung consist of a spectrum of neoplasms, including typical carcinoids, atypical carcinoids, large‐cell neuroendocrine carcinomas (LCNEC), and small‐cell lung carcinomas (SCLC). We previously reported frequent inactivation of the gene responsible for multiple endocrine neoplasia type 1 (MEN1) in both typical and atypical carcinoid tumors. In the present study, we extend the analysis of human NE lung tumors to include 9 primary SCLCs, 36 SCLC cell lines, and 13 primary LCNECs for MEN1 gene inactivation. In SCLC, loss of heterozygosity (LOH) at the MEN1 gene on chromosome band 11q13 was detected in one primary tumor and two cell lines. The coding sequence and splice junctions of the MEN1 gene were screened for mutations in all 44 tumors and cell lines, and no mutations were detected. Northern blot analysis of 13 SCLC cell lines showed the MEN1 transcript to be present and of normal size. In LCNECs, a somatic frameshift in the MEN1 gene (1226delC) was found in one of 13 tumors, representing the first mutation observed outside the spectrum of neoplasms associated with MEN1. Interestingly, neither a deletion nor a mutation was detected in the other allele, and wild‐type mRNA sequence was expressed in the tumor, suggesting that the MEN1 gene was not inactivated by a conventional two‐hit mechanism. The data support the hypothesis that SCLC and lung carcinoids develop via distinct molecular pathways; however, further investigation is necessary to determine the significance of the MEN1 gene mutation observed in a single case of LCNEC. Genes Chromosomes Cancer 28:58–65, 2000. Published 2000 Wiley‐Liss, Inc.


Mammalian Genome | 1999

Isolation, genomic organization, and expression analysis of Men1, the murine homolog of the MEN1 gene

Siradanahalli C. Guru; Judy S. Crabtree; Kevin D. Brown; Karen J. Dunn; Pachiappan Manickam; Nijaguna B. Prasad; Danny Wangsa; A. Lee Burns; Allen M. Spiegel; Stephen J. Marx; William J. Pavan; Francis S. Collins; Settara C. Chandrasekharappa

The mouse homolog of the human MEN1 gene, which is defective in a dominant familial cancer syndrome, multiple endocrine neoplasia type 1 (MEN1), has been identified and characterized. The mouse Men1 transcript contains an open reading frame encoding a protein of 611 amino acids which has 97% identity and 98% similarity to human menin. Sequence of the entire Men1 gene (9.3 kb) was assembled, revealing 10 exons, with exon 1 being non-coding; a polymorphic tetranucleotide repeat was located in the 5′- flanking region. The exon-intron organization and the size of the coding exons 2–9 were well conserved between the human and mouse genes. Fluorescence in situ hybridization localized the Men1 gene to mouse Chromosome (Chr) 19, a region known to be syntenic to human Chr 11q13, the locus for the MEN1 gene. Northern analysis indicated two messages—2.7 kb and 3.1 kb—expressed in all stages of the embryo analyzed and in all eight adult tissues tested. The larger transcript differs from the smaller by the inclusion of an unspliced intron 1. Whole-mount in situ hybridization of 10.5-day and 11.5-day embryos showed ubiquitous expression of Men1 RNA. Western analysis with antibodies raised against a conserved C-terminal peptide identified an approximately 67-kDa protein in the lysates of adult mouse brain, kidney, liver, pancreas, and spleen tissues, consistent with the size of human menin. The levels of mouse menin do not appear to fluctuate during the cell cycle.


Human Genetics | 1997

Eighteen new polymorphic markers in the multiple endocrine neoplasia type 1 (MEN1) region

Pachiappan Manickam; Siradanahalli C. Guru; Larisa V. Debelenko; Sunita K. Agarwal; Shodimu-Emmanuel Olufemi; Jane M. Weisemann; Mark S. Boguski; Judy S. Crabtree; Yingping Wang; Bruce A. Roe; Irina A. Lubensky; Zhengping Zhuang; Mary Beth Kester; A. Lee Burns; Allen M. Spiegel; Stephen J. Marx; Lance A. Liotta; Michael R. Emmert-Buck; Francis S. Collins; Settara C. Chandrasekharappa

Abstract Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disorder in which affected individuals develop tumors primarily in the parathyroids, anterior pituitary, endocrine pancreas, and duodenum. The locus for MEN1 is tightly linked to the marker PYGM on chromosome 11q13, and linkage analysis has previously placed the MEN1 gene within a 2-Mb interval flanked by markers D11S1883 and D11S449. Loss of heterozygosity (LOH) studies in MEN1 and sporadic tumors have helped narrow the location of the gene to a 600-kb interval between PYGM and D11S449. Eighteen new polymerase chain reaction (PCR)-based polymorphic markers were generated for the MEN1 region, with ten mapping to the PYGM-D11S449 interval. These new markers, along with 14 previously known polymorphic markers, were precisely mapped on a 2.8-Mb (D11S480–D11S913) high-density clone contig-based, physical map generated for the MEN1 region.


Journal of Internal Medicine | 1998

Germline and somatic mutation of the gene for multiple endocrine neoplasia type 1 (MEN1)

Stephen J. Marx; Sunita K. Agarwal; MaryBeth Kester; Christina Heppner; Yeonjung Kim; Michael R. Emmert-Buck; Larisa V. Debelenko; Irina A. Lubensky; Zhengping Zhuang; Siradanahalli C. Guru; Pachiappan Manickam; Shodimu-Emmanuel Olufemi; Monica C. Skarulis; John L. Doppman; R. H. Alexander; Lance A. Liotta; Francis S. Collins; Settara C. Chandrasekharappa; Allen M. Spiegel; Burns Al

Marx SJ, Agarwal SK, Kester MB, Heppner C, Kim YS, Emmert‐Buck MR, Debelenko LV, Lubensky IA, Zhuang Z, Guru SC, Manickam P, Olufemi SE, Skarulis MC, Doppman JL, Alexander RH, Liotta LA, Collins FS, Chandrasekharappa SC, Spiegel AM, Burns AL (National Institutes of Health, Bethesda, USA). Germline and somatic mutation of the gene for multiple endocrine neoplasia type 1 (MEN1) (Minisymposium: MEN & VHL). J Intern Med 1998; 243: 447–53.


Mammalian Genome | 2000

Isolation, characterization, expression and functional analysis of the zebrafish ortholog of MEN1

Pachiappan Manickam; Andreas M. Vogel; Sunita K. Agarwal; Takaya Oda; Allen M. Spiegel; Stephen J. Marx; Francis S. Collins; Brant M. Weinstein; Settara C. Chandrasekharappa

Abstract. Mutations in the MEN1 gene lead to an autosomal dominant disorder, multiple endocrine neoplasia type 1 (MEN1), which is characterized by tumors of the parathyroid, entero-pancreatic neuroendocrine, and pituitary tissues. The protein encoded by MEN1, 610-amino acid menin, resides primarily in the nucleus and binds to the transcription factor JunD, resulting in the repression of JunD-induced transcription. We report here a detailed characterization of the zebrafish men1 gene and its full-length (2551 nt) transcript, encoding a 617-amino acid protein with 67% identity and 80% similarity to human menin. Of the 81 missense mutations and in-frame deletions reported in MEN1 patients, 72 occur in residues that are identical in zebrafish, suggesting the importance of the conserved regions. The zebrafish men1 gene maps 61 cM from the top of linkage group 7 (LG7), a region that appears to show conserved synteny to the MEN1 loci at human 11q13. A 2.7-kb men1 message is detected at all stages of zebrafish development analyzed, from one-cell embryos to adult fish. Whole-mount in situ hybridization showed ubiquitous distribution of men1 message in zebrafish embryos at cleavage, blastula, gastrula, and early segmentation stages, with relatively abundant expression in blood cell progenitors (24 h post fertilization) and mesenchymal tissues (48 h post fertilization) at later stages. Zebrafish menin binds both human and mouse JunD, and represses JunD-induced transcription, indicating that the JunD-binding ability of menin is evolutionarily conserved.


Bone | 1999

The gene for multiple endocrine neoplasia type 1: recent findings

Stephen J. Marx; Sunita K. Agarwal; Christina Heppner; Young S. Kim; MaryBeth Kester; P.K. Goldsmith; Monica C. Skarulis; Allen M. Spiegel; Burns Al; Larisa V. Debelenko; Zhengping Zhuang; Irina A. Lubensky; Lance A. Liotta; Michael R. Emmert-Buck; Siradanahalli C. Guru; Pachiappan Manickam; Judy S. Crabtree; Francis S. Collins; Settara C. Chandrasekharappa

Multiple endocrine neoplasia type 1 (MENI) is a promising model to understand endocrine and other tumors. Its most common endocrine expressions are tumors of parathyroids, entero-pancreatic neuro-endocrine tissue, and anterior pituitary. Recently, collagenomas and multiple angiofibromas of the dermis also have been recognized as very common. MEN1 can be characterized from different perspectives: (a) as a hormone (parathyroid hormone, gastrin, prolactin, etc.) excess syndrome with excellent therapeutic options; (b) as a syndrome with sometimes lethal outcomes from malignancy of entero-pancreatic neuro-endocrine or foregut carcinoid tissues; or (c) as a disorder than can give insight about cell regulation in the endocrine, the dermal, and perhaps other tissue systems. The MEN1 gene was identified recently by positional cloning, a comprehensive strategy of narrowing the candidate interval and evaluating all or most genes in that interval. This discovery has opened new approaches to basic and clinical issues. Germline MEN1 mutations have been identified in most MEN1 families. Germline MENI mutations were generally not found in families with isolated hyperparathyroidism or with isolated pituitary tumor. Thus, studies with the MENI gene helped establish that mutation of other gene(s) is likely causative of these two MEN1 phenocopies. MEN1 proved to be the gene most frequent L4 mutated in common-variety, nonhereditary parathyroid tumor, gastrinoma, insulinoma, or bronchial carcinoid. For example, in common-variety parathyroid tumors, mutation of several other genes (such as cyclin D1 and P53) has been found, but much less frequently than MEN1 mutation. The majority of germline and somatic MEN1 mutations predicted truncation of the encoded protein (menin). Such inactivating mutations strongly supported prior predictions that MEN1 is a tumor suppressor gene insofar as stepwise mutational inactivation of both copies can release a cell from normal growth suppression. Menin is principally a nuclear protein; menin interacts with junD. Future studies, such as discovery of menins metabolic pathway, could lead to new opportunities in cell biology and in tumor therapy.


Human Mutation | 1998

Analysis of recurrent germline mutations in the MEN1 gene encountered in apparently unrelated families.

Sunita K. Agarwal; Larisa V. Debelenko; Mary Beth Kester; Siradanahalli C. Guru; Pachiappan Manickam; Shodimu-Emmanuel Olufemi; Monica C. Skarulis; Christina Heppner; Judy S. Crabtree; Irina A. Lubensky; Zhengping Zhuang; Young S. Kim; Settara C. Chandrasekharappa; Francis S. Collins; Lance A. Liotta; Allen M. Spiegel; A. Lee Burns; Michael R. Emmert-Buck; Stephen J. Marx

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disorder that manifests as varying combinations of tumors of endocrine and other tissues (parathyroids, pancreatic islets, duodenal endocrine cells, the anterior pituitary and others). The MEN1 gene is on chromosome 11q13; it was recently identified by positional cloning. We previously reported 32 different germline mutations in 47 of the 50 familial MEN1 probands studied at the NIH. Eight different germline MEN1 mutations were encountered repeatedly in two or more apparently unrelated families. We analyzed the haplotypes of families with recurrent MEN1 mutations with seven polymorphic markers in the 11q13 region surrounding the MEN1 gene (from D11S1883 to D11S4908). Disease haplotypes were inferred from germline DNA and also from tumors with 11q13 loss of heterozygosity. Two different disease haplotype cores were shared by apparently unrelated families for two mutations in exon 2 (five families with 416delC and six families with 512delC). These two repeat mutations were associated with the two founder effects that we reported in a prior haplotype analysis. The disease haplotypes for each of the other six repeat mutations (seen twice each) were discordant, suggesting independent origins of these recurrent mutations. Most of the MEN1 germline mutations including all of those recurring independently occur in regions of CpG/CpNpG, short DNA repeats or single nucleotide repeat motifs. In conclusion, recurring germline mutations account for about half of the mutations in North American MEN1 families. They result from either founder effects or independent occurrence of one mutation more than one time. Hum Mutat 12:75–82, 1998. Published 1998 Wiley‐Liss, Inc. This article is a US Government work and, as such, is in the public domain in the United States of America.


Genes, Chromosomes and Cancer | 2000

Interstitial deletion of 11q13 sequences in HeLa cells.

Eri S. Srivatsan; Ulla Bengtsson; Pachiappan Manickam; Payam Benyamini; Settara C. Chandrasekharappa; C. Sun; Eric J. Stanbridge; J. Leslie Redpath

Previous cytogenetic and molecular genetic studies have shown that the HeLa (cervical carcinoma) cell line D98/AH‐2 contains two apparently normal copies of chromosome 11 and additional 11q13–25 material translocated onto a chromosome 3 marker. To determine the 11q13 breakpoint, we performed fluorescence in situ hybridization (FISH) using 18 different 11q13 specific BAC (bacterial artificial chromosome) and cosmid probes spanning a 5.6 Mb interval. Markers localized to the multiple endocrine neoplasia type 1 (MEN1) gene (menin) were also included in the analysis. The FISH study identified an interstitial deletion between markers D11S449 and GSTP1, an interval of 2.3 Mb, in the marker chromosome. This deletion did not include the MEN1 gene. Because point mutations and methylations can inactivate the MEN1 gene, single stranded conformational polymorphism (SSCP) and Northern and Western blot analyses were performed with MEN1 specific probes and antibody. SSCP did not reveal mutations of the MEN1 gene in HeLa or in seven other cervical cancer cell lines. Northern and Western blot studies revealed normal levels of expression of this gene in the cervical cancer cell lines as well as in HeLa cell derived tumorigenic hybrids. Because deletions of tumor suppressor genes often occur in cancer progression, we hypothesize that the inactivation of a tumor suppressor gene other than MEN1, localized to the 2.3 Mb interval on 11q13, might play a role in the abnormal growth behavior of HeLa cells in vitro or in vivo.

Collaboration


Dive into the Pachiappan Manickam's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Allen M. Spiegel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Francis S. Collins

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Stephen J. Marx

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Siradanahalli C. Guru

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

Larisa V. Debelenko

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