Network


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

Hotspot


Dive into the research topics where Nirmitha I. Herath is active.

Publication


Featured researches published by Nirmitha I. Herath.


Journal of Gastroenterology and Hepatology | 2006

Review of genetic and epigenetic alterations in hepatocarcinogenesis

Nirmitha I. Herath; Barbara A. Leggett; Graeme A. Macdonald

Abstract  Hepatocellular carcinoma (HCC) is associated with multiple risk factors and is believed to arise from pre‐neoplastic lesions, usually in the background of cirrhosis. However, the genetic and epigenetic events of hepatocarcinogenesis are relatively poorly understood. HCC display gross genomic alterations, including chromosomal instability (CIN), CpG island methylation, DNA rearrangements associated with hepatitis B virus (HBV) DNA integration, DNA hypomethylation and, to a lesser degree, microsatellite instability. Various studies have reported CIN at chromosomal regions, 1p, 4q, 5q, 6q, 8p, 10q, 11p, 16p, 16q, 17p and 22q. Frequent promoter hypermethylation and subsequent loss of protein expression has also been demonstrated in HCC at tumor suppressor gene (TSG), p16, p14, p15, SOCS1, RIZ1, E‐cadherin and 14–3‐3 σ. An interesting observation emerging from these studies is the presence of a methylator phenotype in hepatocarcinogenesis, although it does not seem advantageous to have high levels of microsatellite instability. Methylation also appears to be an early event, suggesting that this may precede cirrhosis. However, these genes have been studied in isolation and global studies of methylator phenotype are required to assess the significance of epigenetic silencing in hepatocarcinogenesis. Based on previous data there are obvious fundamental differences in the mechanisms of hepatic carcinogenesis, with at least two distinct mechanisms of malignant transformation in the liver, related to CIN and CpG island methylation. The reason for these differences and the relative importance of these mechanisms are not clear but likely relate to the etiopathogenesis of HCC. Defining these broad mechanisms is a necessary prelude to determine the timing of events in malignant transformation of the liver and to investigate the role of known risk factors for HCC.


International Journal of Cancer | 2009

The role of Eph receptors and ephrin ligands in colorectal cancer

Nirmitha I. Herath; Andrew W. Boyd

Eph receptors and their ephrin ligands constitute the largest subfamily of receptor tyrosine kinases and are components of the cell signaling pathways involved during development. Eph and ephrin overexpression have been documented in a variety of human cancers including gastrointestinal malignancies and in particular colorectal malignancies. EphB and ephrin B proteins have been implicated in the homeostasis of the gastrointestinal tract where EphB2‐ and EphB3‐ephrin B signaling regulates cell sorting in the mature epithelium. These proteins are also reported to be upregulated in colon carcinomas. The EphA/ephrin A system has also been implicated in epithelial tissue structure and function. More recently, EphA receptors and their corresponding ligands have been implicated in numerous malignancies. Of these, EphA2 in particular has been intensively investigated and has been proposed as a therapeutic target. An interesting observation emerging from these studies is the role for Ephs and ephrins in critical aspects of cell adhesion, migration and positioning, and a crucial role in tumor progression and metastasis. However, the underlying role of Ephs and ephrins in these processes has generally been studied on individual Eph or ephrin genes. Given the multiplicity of Eph expression on gut epithelial cells, a more global approach is needed to define the precise role of Eph–ephrin interaction in malignant transformation. Here, we will review the recent advances on the role of Eph–ephrin signaling in colorectal malignancies.


BMC Cancer | 2006

Over-expression of Eph and ephrin genes in advanced ovarian cancer: ephrin gene expression correlates with shortened survival

Nirmitha I. Herath; Mark D. Spanevello; Sabe Sabesan; Tanya R. Newton; Margaret C. Cummings; Shannon Duffy; Douglas J. Lincoln; Glen M. Boyle; Peter G. Parsons; Andrew W. Boyd

BackgroundIncreased expression of Eph receptor tyrosine kinases and their ephrin ligands has been implicated in tumor progression in a number of malignancies. This report describes aberrant expression of these genes in ovarian cancer, the commonest cause of death amongst gynaecological malignancies.MethodsEph and ephrin expression was determined using quantitative real time RT-PCR. Correlation of gene expression was measured using Spearmans rho statistic. Survival was analysed using log-rank analysis and (was visualised by) Kaplan-Meier survival curves.ResultsGreater than 10 fold over-expression of EphA1 and a more modest over-expression of EphA2 were observed in partially overlapping subsets of tumors. Over-expression of EphA1 strongly correlated (r = 0.801; p < 0.01) with the high affinity ligand ephrin A1. A similar trend was observed between EphA2 and ephrin A1 (r = 0.387; p = 0.06). A striking correlation of both ephrin A1 and ephrin A5 expression with poor survival (r = -0.470; p = 0.02 and r = -0.562; p < 0.01) was observed. Intriguingly, there was no correlation between survival and other clinical parameters or Eph expression.ConclusionThese data imply that increased levels of ephrins A1 and A5 in the presence of high expression of Ephs A1 and A2 lead to a more aggressive tumor phenotype. The known functions of Eph/ephrin signalling in cell de-adhesion and movement may explain the observed correlation of ephrin expression with poor prognosis.


British Journal of Cancer | 2009

Epigenetic silencing of EphA1 expression in colorectal cancer is correlated with poor survival.

Nirmitha I. Herath; James D. Doecke; Mark D. Spanevello; Barbara A. Leggett; Andrew W. Boyd

Aberrant expression of Eph and ephrin proteins has well-established functions in oncogenesis and tumour progression. We describe EphA1 expression in 6 colorectal cancer (CRC) cell lines, 18 controls and 125 CRC specimens. In addition, a well-characterised cohort of 53 paired normal colon and CRCs was also assessed. Expression of EphA1 mRNA was assessed by quantitative real-time PCR and correlated with protein expression by flow cytometry, immunoprecipitation, western blotting and immunohistochemistry. Significant upregulation (2- to 10-fold) of EphA1 was seen in over 50% of cases (P=0.005) whereas many of the remainder showed downregulation of EphA1. Intriguingly, EphA1 over-expression was more prevalent in stage II compared to stage III CRCs (P=0.02). Low EphA1 expression significantly correlated with poor survival (P=0.02). Epigenetic silencing appeared to explain the loss of EphA1 expression as methylation of the EphA1 CpG island strongly correlated with low EphA1 expression (P<0.01). Furthermore, EphA1 re-expression could be induced by treatment with demethylating agents. Our findings identify EphA1 as a potential prognostic marker in CRC. Although therapies targeting high EphA1 expression seem plausible in CRC, the loss of expression in advanced disease suggests a potential risk that targeted therapy, by selecting for loss of expression, might contribute to disease progression.


European Journal of Cancer | 2012

Complex expression patterns of Eph receptor tyrosine kinases and their ephrin ligands in colorectal carcinogenesis

Nirmitha I. Herath; Mark D. Spanevello; James D. Doecke; Fiona M. Smith; Celio Pouponnot; Andrew W. Boyd

Aberrant expression of Eph and ephrin proteins in human cancers is extensively documented. However, data are frequently limited to one gene and therefore incomplete and in some instances conflicting. We analysed expression of all Eph and ephrin genes in colorectal cancer (CRC) cell lines and 153 clinical specimens, providing for the first time a comprehensive analysis of this system in CRC. Eph/ephrin mRNA expression was assessed by quantitative real-time PCR and correlated with protein expression (flow cytometry, Western blotting and immunocytochemistry). These data show that EphA1, EphA2, EphB2 and EphB4 were significantly over expressed in CRC. In all cases, at least one Eph gene was found in normal colon (EphA1, EphA2, EphB2, EphB4), where expression was observed at high levels in most CRCs. However, other Eph gene expression was lost in individual CRCs compared to the corresponding normal, EphA7 being a striking example. Loss of expression was more common in advanced disease and thus correlated with poor survival. This is consistent with the redundant functionality of Eph receptors, such that expression of a single Eph gene is sufficient for effector function. Overall, the data suggest a progressive loss of expression of individual Eph genes suggesting that individual CRCs need to be phenotyped to determine which Eph genes are highly expressed. Targeted therapies could then be selected from a group of specific antibodies, such as those developed for EphA1.


Genesis | 2008

Generation and characterization of EphA1 receptor tyrosine kinase reporter knockout mice

Shannon Duffy; Mark G. Coulthard; Mark D. Spanevello; Nirmitha I. Herath; Trina Yeadon; Jennifer K. McCarron; Jacinta C. Carter; Ian D. Tonks; Graham F. Kay; Gael E. Phillips; Andrew W. Boyd

Eph receptor tyrosine kinases (RTKs) are a highly conserved family of signaling proteins with functions in cellular migration, adhesion, apoptosis, and proliferation during both adult and embryonic life. Here, we describe a knock‐in mouse in which EphA1 expression is disrupted via the insertion of an internal ribosome entry site (IRES)‐human placental alkaline phosphatase (ALPP) reporter cassette into exon II of the EphA1 gene. This was shown to successfully knockout expression of endogenous EphA1 and enforce expression of the ALPP reporter by the EphA1 promoter. Staining for the ALPP reporter protein demonstrated an epithelially restricted expression pattern in mouse tissues. In EphA1 null mice, two separate phenotypes were identified: abnormal tail development manifesting as a kinky tail was found in ∼80% of homozygous adults. A second, distinct abnormality present in ∼18% of females was characterized by imperforate uterovaginal development with hydrometrocolpos and caused by a resistance of cells to apoptosis during reproductive tract canalization. These results indicate a possible role for EphA1 in tissue patterning and hormone‐induced apoptotic processes. genesis 46:553–561, 2008.


Journal of Gastroenterology and Hepatology | 2002

Reciprocal relationship between methylation status and loss of heterozygosity at the p14ARF locus in Australian and South African hepatocellular carcinomas

Nirmitha I. Herath; Michael C. Kew; Michael D. Walsh; Joanne Young; Lawrie W. Powell; Barbara A. Leggett; Graeme A. Macdonald

Abstract Chromosome 9p21, a locus comprising the tumor suppressor genes (TSG) p16INK4a and p14ARF, is a common region of loss of heterozygosity (LOH) in hepatocellular carcinoma (HCC). p14ARF shares exon 2 with p16 in a different reading frame. p14 binds to MDM2 resulting in a stabilization of functional p53. This study examined the roles of p14, p16 and p53 in hepatocarcinogenesis, in 37 Australian and 24 South African patients. LOH at 9p21 and 17p13.1, p14 and p16 mutation analysis, p14 and p16 promoter methylation and p14, p16 and p53 protein expression was examined. LOH at 9p21 was detected more frequently in South African HCC (P = 0.04). Comparable rates of p53 LOH were observed in Australian and South African HCC (10/22, 45%vs 13/22, 59%, respectively). Hypermethylation of the p14 promoter was more prevalent in Australian HCC than in South African HCC (17/37, 46%vs 7/24, 29%, respectively). In Australian HCC the prevalence of p14 methylation increased with age (P = 0.03). p16 promoter methylation was observed in 12/37 (32%) and 6/24 (25%) in Australian and South African HCC, respectively. Loss of p16 protein expression was detected in 14/36 Australian HCC whereas p53 protein expression was detected in 9/36. Significantly, a reciprocal relationship between 9p21 LOH and p14 promoter hypermethylation was observed (P≤0.05). No significant association between p14 and p53 was seen in this study. The reciprocal relationship identified indicates different pathways of tumorigenesis and likely reflects different etiologies of HCC in the two countries.


Protein and Peptide Letters | 2006

Eph/Ephrin membrane proteins: a mammalian expression vector pTIg-BOS-Fc allowing rapid protein purification.

Bryan W. Day; Fiona M. Smith; Ke Chen; Jennifer K. McCarron; Nirmitha I. Herath; Martin Lackmann; Andrew W. Boyd

There is an urgent need for high purity, single chain, fully functional Eph/ephrin membrane proteins. This report outlines the pTIg-BOS-Fc vector and purification approach resulting in rapid increased production of fully functional single chain extracellular proteins that were isolated with high purity and used in structure-function analysis and pre-clinical studies.


Journal of Gastroenterology and Hepatology | 2004

Cadherin/catenin complex appears to be intact in hepatocellular carcinomas from Australia and South Africa

Nirmitha I. Herath; Michael D. Walsh; Michael C. Kew; Joanne Young; Barbara A. Leggett; Graeme A. Macdonald

Background and Aim:  E‐cadherin binds to β‐catenin to form the cadherin/catenin complex required for strong cell adhesion. Inactivation of this complex in tumors facilitates invasion into surrounding tissues. Alterations of both proteins have been reported in hepatocellular carcinomas (HCC). However, the interactions between E‐cadherin and β‐catenin in HCC from different geographical groups have not been explored. The aim of the present study was to assess the role of E‐cadherin and β‐catenin in Australian and South African patients with HCC.


Atlas of genetics and cytogenetics in oncology and haematology | 2011

EPHA1 (EPH receptor A1)

Brett W. Stringer; Nirmitha I. Herath; Shannon Duffy; Mark G. Coulthard; Andrew W. Boyd

Review on EPHA1 (EPH receptor A1), with data on DNA, on the protein encoded, and where the gene is implicated.

Collaboration


Dive into the Nirmitha I. Herath's collaboration.

Top Co-Authors

Avatar

Barbara A. Leggett

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Joanne Young

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew W. Boyd

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Michael C. Kew

University of the Witwatersrand

View shared research outputs
Top Co-Authors

Avatar

Mark D. Spanevello

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

L. W. Powell

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Michael D. Walsh

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lisa A. Simms

QIMR Berghofer Medical Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge