Network


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

Hotspot


Dive into the research topics where Akira Nguyen is active.

Publication


Featured researches published by Akira Nguyen.


Nature Medicine | 2010

miR-380-5p represses p53 to control cellular survival and is associated with poor outcome in MYCN amplified neuroblastoma

Alexander Swarbrick; Susan L. Woods; Alex D. Shaw; Asha Balakrishnan; Yuwei Phua; Akira Nguyen; Yvan Chanthery; Lionel Lim; Lesley J. Ashton; Robert L. Judson; Noelle E. Huskey; Robert Blelloch; Michelle Haber; Murray D. Norris; Peter Lengyel; Christopher S. Hackett; Thomas Preiss; Albert Chetcuti; Christopher S. Sullivan; Eric G. Marcusson; William A. Weiss; Noelle D. L'Etoile; Andrei Goga

Inactivation of the p53 tumor suppressor pathway allows cell survival in times of stress and occurs in many human cancers; however, normal embryonic stem cells and some cancers such as neuroblastoma maintain wild-type human TP53 and mouse Trp53 (referred to collectively as p53 herein). Here we describe a miRNA, miR-380-5p, that represses p53 expression via a conserved sequence in the p53 3′ untranslated region (UTR). miR-380-5p is highly expressed in mouse embryonic stem cells and neuroblastomas, and high expression correlates with poor outcome in neuroblastomas with neuroblastoma derived v-myc myelocytomatosis viral-related oncogene (MYCN) amplification. miR-380 overexpression cooperates with activated HRAS oncoprotein to transform primary cells, block oncogene-induced senescence and form tumors in mice. Conversely, inhibition of endogenous miR-380-5p in embryonic stem or neuroblastoma cells results in induction of p53, and extensive apoptotic cell death. In vivo delivery of a miR-380-5p antagonist decreases tumor size in an orthotopic mouse model of neuroblastoma. We demonstrate a new mechanism of p53 regulation in cancer and stem cells and uncover a potential therapeutic target for neuroblastoma.


Cancer Research | 2011

Hedgehog Overexpression Is Associated with Stromal Interactions and Predicts for Poor Outcome in Breast Cancer

Sandra A O'Toole; Dorothy A Machalek; Robert F. Shearer; Ewan K.A. Millar; Radhika Nair; Peter R. Schofield; Duncan McLeod; Caroline Cooper; Catriona M. McNeil; Andrea McFarland; Akira Nguyen; Christopher J. Ormandy; Min Qiu; Brian Rabinovich; Luciano G. Martelotto; Duc Vu; Gregory E. Hannigan; Elizabeth A. Musgrove; Daniel Christ; Robert L. Sutherland; David Watkins; Alexander Swarbrick

Hedgehog (Hh) signaling plays an important role in several malignancies but its clinical significance in breast cancer is unclear. In a cohort of 279 patients with invasive ductal carcinoma of the breast, expression of Hh ligand was significantly associated with increased risk of metastasis, breast cancer-specific death, and a basal-like phenotype. A paracrine signature, encompassing high epithelial Hh ligand and high stromal Gli1, was an independent predictor for overall survival in multivariate analysis. In 2 independent histological progression series (n = 301), Hh expression increased with atypia. Hh ligand overexpression in a mouse model of basal breast cancer increased growth, induced a poorly differentiated phenotype, accelerated metastasis, and reduced survival. A stromal requirement for these effects was supported by the lack of similar Hh-mediated changes in vitro, and by stromal-specific expression of Hh target genes in vivo. Furthermore, inhibition of Hh ligand with a monoclonal antibody (5E1) inhibited tumor growth and metastasis. These data suggest that epithelial-stromal Hh signaling, driven by ligand expression in carcinoma cells, promotes breast cancer growth and metastasis. Blockade of Hh signaling to peritumoral stromal cells may represent a novel therapeutic approach in some basal-like breast cancers.


Immunity | 2015

T Follicular Helper Cells Have Distinct Modes of Migration and Molecular Signatures in Naive and Memory Immune Responses

Dan Suan; Akira Nguyen; Imogen Moran; Katherine Bourne; Jana R. Hermes; Mehreen Arshi; Henry R. Hampton; Michio Tomura; Yoshihiro Miwa; Anthony D. Kelleher; Warren Kaplan; Elissa K. Deenick; Stuart G. Tangye; Robert Brink; Tatyana Chtanova; Tri Giang Phan

B helper follicular T (Tfh) cells are critical for long-term humoral immunity. However, it remains unclear how these cells are recruited and contribute to secondary immune responses. Here we show that primary Tfh cells segregate into follicular mantle (FM) and germinal center (GC) subpopulations that display distinct gene expression signatures. Restriction of the primary Tfh cell subpopulation in the GC was mediated by downregulation of chemotactic receptor EBI2. Following collapse of the GC, memory T cells persisted in the outer follicle where they scanned CD169(+) subcapsular sinus macrophages. Reactivation and intrafollicular expansion of these follicular memory T cells in the subcapsular region was followed by their extrafollicular dissemination via the lymphatic flow. These data suggest that Tfh cells integrate their antigen-experience history to focus T cell help within the GC during primary responses but act rapidly to provide systemic T cell help after re-exposure to the antigen.


Cancer Discovery | 2015

Real-Time Intravital Imaging Establishes Tumor-Associated Macrophages as the Extraskeletal Target of Bisphosphonate Action in Cancer

Simon Junankar; Gemma Shay; Julie Jurczyluk; Naveid Ali; Jenny Down; Nicholas Pocock; Andrew Parker; Akira Nguyen; Shuting Sun; Boris A. Kashemirov; Charles E. McKenna; Peter I. Croucher; Alexander Swarbrick; Katherine N. Weilbaecher; Tri Giang Phan; Michael J. Rogers

UNLABELLED Recent clinical trials have shown that bisphosphonate drugs improve breast cancer patient survival independent of their antiresorptive effects on the skeleton. However, because bisphosphonates bind rapidly to bone mineral, the exact mechanisms of their antitumor action, particularly on cells outside of bone, remain unknown. Here, we used real-time intravital two-photon microscopy to show extensive leakage of fluorescent bisphosphonate from the vasculature in 4T1 mouse mammary tumors, where it initially binds to areas of small, granular microcalcifications that are engulfed by tumor-associated macrophages (TAM), but not tumor cells. Importantly, we also observed uptake of radiolabeled bisphosphonate in the primary breast tumor of a patient and showed the resected tumor to be infiltrated with TAMs and to contain similar granular microcalcifications. These data represent the first compelling in vivo evidence that bisphosphonates can target cells in tumors outside the skeleton and that their antitumor activity is likely to be mediated via TAMs. SIGNIFICANCE Bisphosphonates are assumed to act solely in bone. However, mouse models and clinical trials show that they have surprising antitumor effects outside bone. We provide unequivocal evidence that bisphosphonates target TAMs, but not tumor cells, to exert their extraskeletal effects, offering a rationale for use in patients with early disease.


Oncogene | 2014

c-Myc and Her2 cooperate to drive a stem-like phenotype with poor prognosis in breast cancer

Radhika Nair; Daniel Roden; Wee Siang Teo; Andrea McFarland; Simon Junankar; S Ye; Akira Nguyen; Jessica Yang; Iva Nikolic; M. Hui; Adrienne Morey; J Shah; Adam D. Pfefferle; Jerry Usary; Cristina Selinger; Laura A Baker; Nicola J. Armstrong; Mark J. Cowley; Matthew J. Naylor; Christopher J. Ormandy; Sunil R. Lakhani; J I Herschkowitz; C M Perou; Warren Kaplan; Sandra A O'Toole; Alexander Swarbrick

The HER2 (ERBB2) and MYC genes are commonly amplified in breast cancer, yet little is known about their molecular and clinical interaction. Using a novel chimeric mammary transgenic approach and in vitro models, we demonstrate markedly increased self-renewal and tumour-propagating capability of cells transformed with Her2 and c-Myc. Coexpression of both oncoproteins in cultured cells led to the activation of a c-Myc transcriptional signature and acquisition of a self-renewing phenotype independent of an epithelial–mesenchymal transition programme or regulation of conventional cancer stem cell markers. Instead, Her2 and c-Myc cooperated to induce the expression of lipoprotein lipase, which was required for proliferation and self-renewal in vitro. HER2 and MYC were frequently coamplified in breast cancer, associated with aggressive clinical behaviour and poor outcome. Lastly, we show that in HER2+ breast cancer patients receiving adjuvant chemotherapy (but not targeted anti-Her2 therapy), MYC amplification is associated with a poor outcome. These findings demonstrate the importance of molecular and cellular context in oncogenic transformation and acquisition of a malignant stem-like phenotype and have diagnostic and therapeutic consequences for the clinical management of HER2+ breast cancer.


Journal of Experimental Medicine | 2016

Unique and shared signaling pathways cooperate to regulate the differentiation of human CD4+ T cells into distinct effector subsets.

Cindy S. Ma; Natalie Wong; Geetha Rao; Akira Nguyen; Danielle T. Avery; Kathryn Payne; James Torpy; Patrick O’Young; Elissa K. Deenick; Jacinta Bustamante; Anne Puel; Satoshi Okada; Masao Kobayashi; Rubén Martínez-Barricarte; Michael Elliott; Sara Sebnem Kilic; Jamila El Baghdadi; Yoshiyuki Minegishi; Aziz Bousfiha; Nic Robertson; Sophie Hambleton; Peter D. Arkwright; Martyn A. French; Annaliesse K. Blincoe; Peter Hsu; Dianne E. Campbell; Michael Stormon; Melanie Wong; Stephen Adelstein; David A. Fulcher

Tangye and collaborators use a series of mutants to elucidate the pathways required to generate distinct subsets of human effector CD4+ T cells.


Breast Cancer Research | 2015

MicroRNA profiling of the pubertal mouse mammary gland identifies miR-184 as a candidate breast tumour suppressor gene

Yu Wei Phua; Akira Nguyen; Daniel Roden; Benjamin Elsworth; Niantao Deng; Iva Nikolic; Jessica Yang; Andrea McFarland; Roslin Russell; Warren Kaplan; Mark J. Cowley; Radhika Nair; Elena Zotenko; Sandra A. O’Toole; Shi-Xiong Tan; David E. James; Susan J. Clark; Hosein Kouros-Mehr; Alexander Swarbrick

IntroductionThe study of mammalian development has offered many insights into the molecular aetiology of cancer. We previously used analysis of mammary morphogenesis to discover a critical role for GATA-3 in mammary developmental and carcinogenesis. In recent years an important role for microRNAs (miRNAs) in a myriad of cellular processes in development and in oncogenesis has emerged.MethodsmicroRNA profiling was conducted on stromal and epithelial cellular subsets microdissected from the pubertal mouse mammary gland. miR-184 was reactivated by transient or stable overexpression in breast cancer cell lines and examined using a series of in vitro (proliferation, tumour-sphere and protein synthesis) assays. Orthotopic xenografts of breast cancer cells were used to assess the effect of miR-184 on tumourigenesis as well as distant metastasis. Interactions between miR-184 and its putative targets were assessed by quantitative PCR, microarray, bioinformatics and 3′ untranslated region Luciferase reporter assay. The methylation status of primary patient samples was determined by MBD-Cap sequencing. Lastly, the clinical prognostic significance of miR-184 putative targets was assessed using publicly available datasets.ResultsA large number of microRNA were restricted in their expression to specific tissue subsets. MicroRNA-184 (miR-184) was exclusively expressed in epithelial cells and markedly upregulated during differentiation of the proliferative, invasive cells of the pubertal terminal end bud (TEB) into ductal epithelial cells in vivo. miR-184 expression was silenced in mouse tumour models compared to non-transformed epithelium and in a majority of breast cancer cell line models. Ectopic reactivation of miR-184 inhibited the proliferation and self-renewal of triple negative breast cancer (TNBC) cell lines in vitro and delayed primary tumour formation and reduced metastatic burden in vivo. Gene expression studies uncovered multi-factorial regulation of genes in the AKT/mTORC1 pathway by miR-184. In clinical breast cancer tissues, expression of miR-184 is lost in primary TNBCs while the miR-184 promoter is methylated in a subset of lymph node metastases from TNBC patients.ConclusionsThese studies elucidate a new layer of regulation in the PI3K/AKT/mTOR pathway with relevance to mammary development and tumour progression and identify miR-184 as a putative breast tumour suppressor.


Journal of Experimental Medicine | 2018

Germline-activating mutations in PIK3CD compromise B cell development and function

Danielle T. Avery; Alisa Kane; Tina Nguyen; Anthony Lau; Akira Nguyen; Helen Lenthall; Kathryn Payne; Wei Shi; Henry Brigden; Elise French; Julia Bier; Jana R. Hermes; David Zahra; William A. Sewell; Danyal Butt; Michael Elliott; Kaan Boztug; Isabelle Meyts; Sharon Choo; Peter Hsu; Melanie Wong; Lucinda J. Berglund; Paul Gray; Michael O’Sullivan; Theresa Cole; Steven M. Holland; Cindy S. Ma; Christoph Burkhart; Lynn M. Corcoran; Tri Giang Phan

Gain-of-function (GOF) mutations in PIK3CD, encoding the p110&dgr; subunit of phosphatidylinositide 3-kinase (PI3K), cause a primary immunodeficiency. Affected individuals display impaired humoral immune responses following infection or immunization. To establish mechanisms underlying these immune defects, we studied a large cohort of patients with PIK3CD GOF mutations and established a novel mouse model using CRISPR/Cas9-mediated gene editing to introduce a common pathogenic mutation in Pik3cd. In both species, hyperactive PI3K severely affected B cell development and differentiation in the bone marrow and the periphery. Furthermore, PI3K GOF B cells exhibited intrinsic defects in class-switch recombination (CSR) due to impaired induction of activation-induced cytidine deaminase (AID) and failure to acquire a plasmablast gene signature and phenotype. Importantly, defects in CSR, AID expression, and Ig secretion were restored by leniolisib, a specific p110&dgr; inhibitor. Our findings reveal key roles for balanced PI3K signaling in B cell development and long-lived humoral immunity and memory and establish the validity of treating affected individuals with p110&dgr; inhibitors.


Frontiers in Immunology | 2018

Single Cell RNA Sequencing of Rare Immune Cell Populations

Akira Nguyen; Weng Hua Khoo; Imogen Moran; Peter I. Croucher; Tri Giang Phan

Single-cell RNA sequencing (scRNA-Seq) is transforming our ability to characterize cells, particularly rare cells that are often overlooked in bulk population analytical approaches. This has lead to the discovery of new cell types and cellular states that echo the underlying heterogeneity and plasticity in the immune system. Technologies for the capture, sequencing, and bioinformatic analysis of single cells are rapidly improving, and scRNA-Seq is now becoming much more accessible to non-specialized laboratories. Here, we describe our experiences in adopting scRNA-Seq to the study of rare immune cells in their microanatomical niches.


Nature Communications | 2018

Memory B cells are reactivated in subcapsular proliferative foci of lymph nodes

Imogen Moran; Akira Nguyen; Weng Hua Khoo; Danyal Butt; Katherine Bourne; Clara Young; Jana R. Hermes; Maté Biro; Gary Gracie; Cindy S. Ma; C. Mee Ling Munier; Fabio Luciani; John Zaunders; Andrew Parker; Anthony D. Kelleher; Stuart G. Tangye; Peter I. Croucher; Robert Brink; Mark Read; Tri Giang Phan

Vaccine-induced immunity depends on the generation of memory B cells (MBC). However, where and how MBCs are reactivated to make neutralising antibodies remain unknown. Here we show that MBCs are prepositioned in a subcapsular niche in lymph nodes where, upon reactivation by antigen, they rapidly proliferate and differentiate into antibody-secreting plasma cells in the subcapsular proliferative foci (SPF). This novel structure is enriched for signals provided by T follicular helper cells and antigen-presenting subcapsular sinus macrophages. Compared with contemporaneous secondary germinal centres, SPF have distinct single-cell molecular signature, cell migration pattern and plasma cell output. Moreover, SPF are found both in human and mouse lymph nodes, suggesting that they are conserved throughout mammalian evolution. Our data thus reveal that SPF is a seat of immunological memory that may be exploited to rapidly mobilise secondary antibody responses and improve vaccine efficacy.Memory B cells need to be reactivated to produce high affinity antibody responses on subsequent antigen encounters. Here the authors show that memory B cells localise to lymph node subcapsular proliferative foci (SPF), which have distinct properties from the germinal centre, for rapid expansion and the induction of B memory responses.

Collaboration


Dive into the Akira Nguyen's collaboration.

Top Co-Authors

Avatar

Tri Giang Phan

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Alexander Swarbrick

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Cindy S. Ma

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Jana R. Hermes

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Andrea McFarland

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Danyal Butt

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Imogen Moran

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Katherine Bourne

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Peter I. Croucher

Garvan Institute of Medical Research

View shared research outputs
Top Co-Authors

Avatar

Radhika Nair

Garvan Institute of Medical Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge