Rose, A.J.
Fleming, M.M.
Francis, J.C.
Ning, J.
Patrikeev, A.
Chauhan, R.
Harrington, K.J.
Swain, A.
(2024). Cell-type-specific tumour sensitivity identified with a bromodomain targeting PROTAC in adenoid cystic carcinoma. J pathol,
Vol.262
(1),
pp. 37-49.
show abstract
Salivary gland adenoid cystic carcinoma (ACC) is a rare malignancy with limited treatment options. The development of novel therapies is hindered by a lack of preclinical models. We have generated ACC patient-derived xenograft (PDX) lines that retain the physical and genetic properties of the original tumours, including the presence of the common MYB::NFIB or MYBL1::NFIB translocations. We have developed the conditions for the generation of both 2D and 3D tumour organoid patient-derived ACC models that retain MYB expression and can be used for drug studies. Using these models, we show in vitro and in vivo sensitivity of ACC cells to the bromodomain degrader, dBET6. Molecular studies show a decrease in BRD4 and MYB protein levels and target gene expression with treatment. The most prominent effect of dBET6 on tumours in vivo was a change in the relative composition of ACC cell types expressing either myoepithelial or ductal markers. We show that dBET6 inhibits the progenitor function of ACC cells, particularly in the myoepithelial marker-expressing population, revealing a cell-type-specific sensitivity. These studies uncover a novel mechanistic effect of bromodomain inhibitors on tumours and highlight the need to impact both cell-type populations for more effective treatments in ACC patients. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland..
Neeb, A.
Figueiredo, I.
Bogdan, D.
Cato, L.
Stober, J.
Jiménez-Vacas, J.M.
Gourain, V.
Lee, I.I.
Seeger, R.
Muhle-Goll, C.
Gurel, B.
Welti, J.
Nava Rodrigues, D.
Rekowski, J.
Qiu, X.
Jiang, Y.
Di Micco, P.
Mateos, B.
Bielskutė, S.
Riisnaes, R.
Ferreira, A.
Miranda, S.
Crespo, M.
Buroni, L.
Ning, J.
Carreira, S.
Bräse, S.
Jung, N.
Gräßle, S.
Swain, A.
Salvatella, X.
Plymate, S.R.
Al-Lazikani, B.
Long, H.W.
Yuan, W.
Brown, M.
Cato, A.C.
de Bono, J.S.
Sharp, A.
(2024). Thio-2 Inhibits Key Signaling Pathways Required for the Development and Progression of Castration-resistant Prostate Cancer. Mol cancer ther,
Vol.23
(6),
pp. 791-808.
show abstract
full text
Therapies that abrogate persistent androgen receptor (AR) signaling in castration-resistant prostate cancer (CRPC) remain an unmet clinical need. The N-terminal domain of the AR that drives transcriptional activity in CRPC remains a challenging therapeutic target. Herein we demonstrate that BCL-2-associated athanogene-1 (BAG-1) mRNA is highly expressed and associates with signaling pathways, including AR signaling, that are implicated in the development and progression of CRPC. In addition, interrogation of geometric and physiochemical properties of the BAG domain of BAG-1 isoforms identifies it to be a tractable but challenging drug target. Furthermore, through BAG-1 isoform mouse knockout studies, we confirm that BAG-1 isoforms regulate hormone physiology and that therapies targeting the BAG domain will be associated with limited "on-target" toxicity. Importantly, the postulated inhibitor of BAG-1 isoforms, Thio-2, suppressed AR signaling and other important pathways implicated in the development and progression of CRPC to reduce the growth of treatment-resistant prostate cancer cell lines and patient-derived models. However, the mechanism by which Thio-2 elicits the observed phenotype needs further elucidation as the genomic abrogation of BAG-1 isoforms was unable to recapitulate the Thio-2-mediated phenotype. Overall, these data support the interrogation of related compounds with improved drug-like properties as a novel therapeutic approach in CRPC, and further highlight the clinical potential of treatments that block persistent AR signaling which are currently undergoing clinical evaluation in CRPC..
Herrera-Abreu, M.T.
Guan, J.
Khalid, U.
Ning, J.
Costa, M.R.
Chan, J.
Li, Q.
Fortin, J.-.
Wong, W.R.
Perampalam, P.
Biton, A.
Sandoval, W.
Vijay, J.
Hafner, M.
Cutts, R.
Wilson, G.
Frankum, J.
Roumeliotis, T.I.
Alexander, J.
Hickman, O.
Brough, R.
Haider, S.
Choudhary, J.
Lord, C.J.
Swain, A.
Metcalfe, C.
Turner, N.C.
(2024). Inhibition of GPX4 enhances CDK4/6 inhibitor and endocrine therapy activity in breast cancer. Nat commun,
Vol.15
(1),
p. 9550.
show abstract
full text
CDK4/6 inhibition in combination with endocrine therapy is the standard of care for estrogen receptor (ER+) breast cancer, and although cytostasis is frequently observed, new treatment strategies that enhance efficacy are required. Here, we perform two independent genome-wide CRISPR screens to identify genetic determinants of CDK4/6 and endocrine therapy sensitivity. Genes involved in oxidative stress and ferroptosis modulate sensitivity, with GPX4 as the top sensitiser in both screens. Depletion or inhibition of GPX4 increases sensitivity to palbociclib and giredestrant, and their combination, in ER+ breast cancer models, with GPX4 null xenografts being highly sensitive to palbociclib. GPX4 perturbation additionally sensitises triple negative breast cancer (TNBC) models to palbociclib. Palbociclib and giredestrant induced oxidative stress and disordered lipid metabolism, leading to a ferroptosis-sensitive state. Lipid peroxidation is promoted by a peroxisome AGPAT3-dependent pathway in ER+ breast cancer models, rather than the classical ACSL4 pathway. Our data demonstrate that CDK4/6 and ER inhibition creates vulnerability to ferroptosis induction, that could be exploited through combination with GPX4 inhibitors, to enhance sensitivity to the current therapies in breast cancer..
Francis, J.C.
Capper, A.
Rust, A.G.
Ferro, K.
Ning, J.
Yuan, W.
de Bono, J.
Pettitt, S.J.
Swain, A.
(2024). Identification of genes that promote PI3K pathway activation and prostate tumour formation. Oncogene,
Vol.43
(24),
pp. 1824-1835.
show abstract
full text
We have performed a functional in vivo mutagenesis screen to identify genes that, when altered, cooperate with a heterozygous Pten mutation to promote prostate tumour formation. Two genes, Bzw2 and Eif5a2, which have been implicated in the process of protein translation, were selected for further validation. Using prostate organoid models, we show that either Bzw2 downregulation or EIF5A2 overexpression leads to increased organoid size and in vivo prostate growth. We show that both genes impact the PI3K pathway and drive a sustained increase in phospho-AKT expression, with PTEN protein levels reduced in both models. Mechanistic studies reveal that EIF5A2 is directly implicated in PTEN protein translation. Analysis of patient datasets identified EIF5A2 amplifications in many types of human cancer, including the prostate. Human prostate cancer samples in two independent cohorts showed a correlation between increased levels of EIF5A2 and upregulation of a PI3K pathway gene signature. Consistent with this, organoids with high levels of EIF5A2 were sensitive to AKT inhibitors. Our study identified novel genes that promote prostate cancer formation through upregulation of the PI3K pathway, predicting a strategy to treat patients with genetic aberrations in these genes particularly relevant for EIF5A2 amplified tumours..
Kerrison, W.G.
Ning, J.
Krasny, L.
Arthur, A.
Guljar, N.
Elms, M.L.
Swain, A.
Jones, R.L.
Thway, K.
Huang, P.H.
(2022). Characterisation of a Novel Cell Line (ICR-SS-1) Established from a Patient-Derived Xenograft of Synovial Sarcoma. Cells,
Vol.11
(15).
show abstract
full text
Synovial sarcoma is a rare translocation-driven cancer with poor survival outcomes, particularly in the advanced setting. Previous synovial sarcoma preclinical studies have relied on a small panel of cell lines which suffer from the limitation of genomic and phenotypic drift as a result of being grown in culture for decades. Patient-derived xenografts (PDX) are a valuable tool for preclinical research as they retain many histopathological features of their originating human tumour; however, this approach is expensive, slow, and resource intensive, which hinders their utility in large-scale functional genomic and drug screens. To address some of these limitations, in this study, we have established and characterised a novel synovial sarcoma cell line, ICR-SS-1, which is derived from a PDX model and is amenable to high-throughput drug screens. We show that ICR-SS-1 grows readily in culture, retains the pathognomonic SS18::SSX1 fusion gene, and recapitulates the molecular features of human synovial sarcoma tumours as shown by proteomic profiling. Comparative analysis of drug response profiles with two other established synovial sarcoma cell lines (SYO-1 and HS-SY-II) finds that ICR-SS-1 harbours intrinsic resistance to doxorubicin and is sensitive to targeted inhibition of several oncogenic pathways including the PI3K-mTOR pathway. Collectively, our studies show that the ICR-SS-1 cell line model may be a valuable preclinical tool for studying the biology of anthracycline-resistant synovial sarcoma and identifying new salvage therapies following failure of doxorubicin..
Sowalsky, A.G.
Figueiredo, I.
Lis, R.T.
Coleman, I.
Gurel, B.
Bogdan, D.
Yuan, W.
Russo, J.W.
Bright, J.R.
Whitlock, N.C.
Trostel, S.Y.
Ku, A.T.
Patel, R.A.
True, L.D.
Welti, J.
Jimenez-Vacas, J.M.
Rodrigues, D.N.
Riisnaes, R.
Neeb, A.
Sprenger, C.T.
Swain, A.
Wilkinson, S.
Karzai, F.
Dahut, W.L.
Balk, S.P.
Corey, E.
Nelson, P.S.
Haffner, M.C.
Plymate, S.R.
de Bono, J.S.
Sharp, A.
(2022). Assessment of Androgen Receptor Splice Variant-7 as a Biomarker of Clinical Response in Castration-Sensitive Prostate Cancer. Clin cancer res,
Vol.28
(16),
pp. 3509-3525.
show abstract
full text
PURPOSE: Therapies targeting the androgen receptor (AR) have improved the outcome for patients with castration-sensitive prostate cancer (CSPC). Expression of the constitutively active AR splice variant-7 (AR-V7) has shown clinical utility as a predictive biomarker of AR-targeted therapy resistance in castration-resistant prostate cancer (CRPC), but its importance in CSPC remains understudied. EXPERIMENTAL DESIGN: We assessed different approaches to quantify AR-V7 mRNA and protein in prostate cancer cell lines, patient-derived xenograft (PDX) models, publicly available cohorts, and independent institutional clinical cohorts, to identify reliable approaches for detecting AR-V7 mRNA and protein and its association with clinical outcome. RESULTS: In CSPC and CRPC cohorts, AR-V7 mRNA was much less abundant when detected using reads across splice boundaries than when considering isoform-specific exonic reads. The RM7 AR-V7 antibody had increased sensitivity and specificity for AR-V7 protein detection by immunohistochemistry (IHC) in CRPC cohorts but rarely identified AR-V7 protein reactivity in CSPC cohorts, when compared with the EPR15656 AR-V7 antibody. Using multiple CRPC PDX models, we demonstrated that AR-V7 expression was exquisitely sensitive to hormonal manipulation. In CSPC institutional cohorts, AR-V7 protein quantification by either assay was associated neither with time to development of castration resistance nor with overall survival, and intense neoadjuvant androgen-deprivation therapy did not lead to significant AR-V7 mRNA or staining following treatment. Neither pre- nor posttreatment AR-V7 levels were associated with volumes of residual disease after therapy. CONCLUSIONS: This study demonstrates that further analytical validation and clinical qualification are required before AR-V7 can be considered for clinical use in CSPC as a predictive biomarker..
Francis, J.C.
Gardiner, J.R.
Renaud, Y.
Chauhan, R.
Weinstein, Y.
Gomez-Sanchez, C.
Lefrançois-Martinez, A.-.
Bertherat, J.
Val, P.
Swain, A.
(2021). HOX genes promote cell proliferation and are potential therapeutic targets in adrenocortical tumours. Br j cancer,
Vol.124
(4),
pp. 805-816.
show abstract
full text
BACKGROUND: Understanding the pathways that drive adrenocortical carcinoma (ACC) is essential to the development of more effective therapies. This study investigates the role of the transcription factor HOXB9 and other HOX factors in ACC and its treatment. METHODS: We used transgenic mouse models to determine the role of Hoxb9 in adrenal tumour development. Patient transcriptomic data was analysed for the expression of HOX genes and their association with disease. Drug response studies on various adrenocortical models were done to establish novel therapeutic options. RESULTS: Our human ACC dataset analyses showed high expression of HOXB9, and other HOX factors, are associated with poorer prognosis. Transgenic overexpression of Hoxb9 in the adrenal cortex of mice with activated Ctnnb1 led to larger adrenal tumours. This phenotype was preferentially observed in male mice and was characterised by more proliferating cells and an increase in the expression of cell cycle genes, including Ccne1. Adrenal tumour cells were found to be dependent on HOX function for survival and were sensitive to a specific peptide inhibitor. CONCLUSIONS: These studies show Hoxb9 can promote adrenal tumour progression in a sex-dependent manner and have identified HOX factors as potential drug targets, leading to novel therapeutic approaches in ACC..
Neeb, A.
Herranz, N.
Arce-Gallego, S.
Miranda, S.
Buroni, L.
Yuan, W.
Athie, A.
Casals, T.
Carmichael, J.
Rodrigues, D.N.
Gurel, B.
Rescigno, P.
Rekowski, J.
Welti, J.
Riisnaes, R.
Gil, V.
Ning, J.
Wagner, V.
Casanova-Salas, I.
Cordoba, S.
Castro, N.
Fenor de la Maza, M.D.
Seed, G.
Chandran, K.
Ferreira, A.
Figueiredo, I.
Bertan, C.
Bianchini, D.
Aversa, C.
Paschalis, A.
Gonzalez, M.
Morales-Barrera, R.
Suarez, C.
Carles, J.
Swain, A.
Sharp, A.
Gil, J.
Serra, V.
Lord, C.
Carreira, S.
Mateo, J.
de Bono, J.S.
(2021). Advanced Prostate Cancer with ATM Loss: PARP and ATR Inhibitors. Eur urol,
Vol.79
(2),
pp. 200-211.
show abstract
full text
BACKGROUND: Deleterious ATM alterations are found in metastatic prostate cancer (PC); PARP inhibition has antitumour activity against this subset, but only some ATM loss PCs respond. OBJECTIVE: To characterise ATM-deficient lethal PC and to study synthetic lethal therapeutic strategies for this subset. DESIGN, SETTING, AND PARTICIPANTS: We studied advanced PC biopsies using validated immunohistochemical (IHC) and next-generation sequencing (NGS) assays. In vitro cell line models modified using CRISPR-Cas9 to impair ATM function were generated and used in drug-sensitivity and functional assays, with validation in a patient-derived model. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: ATM expression by IHC was correlated with clinical outcome using Kaplan-Meier curves and log-rank test; sensitivity to different drug combinations was assessed in the preclinical models. RESULTS AND LIMITATIONS: Overall, we detected ATM IHC loss in 68/631 (11%) PC patients in at least one biopsy, with synchronous and metachronous intrapatient heterogeneity; 46/71 (65%) biopsies with ATM loss had ATM mutations or deletions by NGS. ATM IHC loss was not associated with worse outcome from advanced disease, but ATM loss was associated with increased genomic instability (NtAI:number of subchromosomal regions with allelic imbalance extending to the telomere, p = 0.005; large-scale transitions, p = 0.05). In vitro, ATM loss PC models were sensitive to ATR inhibition, but had variable sensitivity to PARP inhibition; superior antitumour activity was seen with combined PARP and ATR inhibition in these models. CONCLUSIONS: ATM loss in PC is not always detected by targeted NGS, associates with genomic instability, and is most sensitive to combined ATR and PARP inhibition. PATIENT SUMMARY: Of aggressive prostate cancers, 10% lose the DNA repair gene ATM; this loss may identify a distinct prostate cancer subtype that is most sensitive to the combination of oral drugs targeting PARP and ATR..
Welti, J.
Sharp, A.
Brooks, N.
Yuan, W.
McNair, C.
Chand, S.N.
Pal, A.
Figueiredo, I.
Riisnaes, R.
Gurel, B.
Rekowski, J.
Bogdan, D.
West, W.
Young, B.
Raja, M.
Prosser, A.
Lane, J.
Thomson, S.
Worthington, J.
Onions, S.
Shannon, J.
Paoletta, S.
Brown, R.
Smyth, D.
Harbottle, G.W.
Gil, V.S.
Miranda, S.
Crespo, M.
Ferreira, A.
Pereira, R.
Tunariu, N.
Carreira, S.
Neeb, A.J.
Ning, J.
Swain, A.
Taddei, D.
SU2C/PCF International Prostate Cancer Dream Team,
Schiewer, M.J.
Knudsen, K.E.
Pegg, N.
de Bono, J.S.
(2021). Targeting the p300/CBP Axis in Lethal Prostate Cancer. Cancer discov,
Vol.11
(5),
pp. 1118-1137.
show abstract
full text
Resistance to androgen receptor (AR) blockade in castration-resistant prostate cancer (CRPC) is associated with sustained AR signaling, including through alternative splicing of AR (AR-SV). Inhibitors of transcriptional coactivators that regulate AR activity, including the paralog histone acetyltransferase proteins p300 and CBP, are attractive therapeutic targets for lethal prostate cancer. Herein, we validate targeting p300/CBP as a therapeutic strategy for lethal prostate cancer and describe CCS1477, a novel small-molecule inhibitor of the p300/CBP conserved bromodomain. We show that CCS1477 inhibits cell proliferation in prostate cancer cell lines and decreases AR- and C-MYC-regulated gene expression. In AR-SV-driven models, CCS1477 has antitumor activity, regulating AR and C-MYC signaling. Early clinical studies suggest that CCS1477 modulates KLK3 blood levels and regulates CRPC biopsy biomarker expression. Overall, CCS1477 shows promise for the treatment of patients with advanced prostate cancer. SIGNIFICANCE: Treating CRPC remains challenging due to persistent AR signaling. Inhibiting transcriptional AR coactivators is an attractive therapeutic strategy. CCS1477, an inhibitor of p300/CBP, inhibits growth and AR activity in CRPC models, and can affect metastatic CRPC target expression in serial clinical biopsies.See related commentary by Rasool et al., p. 1011.This article is highlighted in the In This Issue feature, p. 995..
Gil, V.
Miranda, S.
Riisnaes, R.
Gurel, B.
D'Ambrosio, M.
Vasciaveo, A.
Crespo, M.
Ferreira, A.
Brina, D.
Troiani, M.
Sharp, A.
Sheehan, B.
Christova, R.
Seed, G.
Figueiredo, I.
Lambros, M.
Dolling, D.
Rekowski, J.
Alajati, A.
Clarke, M.
Pereira, R.
Flohr, P.
Fowler, G.
Boysen, G.
Sumanasuriya, S.
Bianchini, D.
Rescigno, P.
Aversa, C.
Tunariu, N.
Guo, C.
Paschalis, A.
Bertan, C.
Buroni, L.
Ning, J.
Carreira, S.
Workman, P.
Swain, A.
Califano, A.
Shen, M.M.
Alimonti, A.
Neeb, A.
Welti, J.
Yuan, W.
de Bono, J.
PCF/SU2C International Prostate Cancer Dream Team,
(2021). HER3 Is an Actionable Target in Advanced Prostate Cancer. Cancer res,
Vol.81
(24),
pp. 6207-6218.
show abstract
full text
It has been recognized for decades that ERBB signaling is important in prostate cancer, but targeting ERBB receptors as a therapeutic strategy for prostate cancer has been ineffective clinically. However, we show here that membranous HER3 protein is commonly highly expressed in lethal prostate cancer, associating with reduced time to castration resistance (CR) and survival. Multiplex immunofluorescence indicated that the HER3 ligand NRG1 is detectable primarily in tumor-infiltrating myelomonocytic cells in human prostate cancer; this observation was confirmed using single-cell RNA sequencing of human prostate cancer biopsies and murine transgenic prostate cancer models. In castration-resistant prostate cancer (CRPC) patient-derived xenograft organoids with high HER3 expression as well as mouse prostate cancer organoids, recombinant NRG1 enhanced proliferation and survival. Supernatant from murine bone marrow-derived macrophages and myeloid-derived suppressor cells promoted murine prostate cancer organoid growth in vitro, which could be reversed by a neutralizing anti-NRG1 antibody and ERBB inhibition. Targeting HER3, especially with the HER3-directed antibody-drug conjugate U3-1402, exhibited antitumor activity against HER3-expressing prostate cancer. Overall, these data indicate that HER3 is commonly overexpressed in lethal prostate cancer and can be activated by NRG1 secreted by myelomonocytic cells in the tumor microenvironment, supporting HER3-targeted therapeutic strategies for treating HER3-expressing advanced CRPC. SIGNIFICANCE: HER3 is an actionable target in prostate cancer, especially with anti-HER3 immunoconjugates, and targeting HER3 warrants clinical evaluation in prospective trials..
Ermini, L.
Francis, J.C.
Rosa, G.S.
Rose, A.J.
Ning, J.
Greaves, M.
Swain, A.
(2021). Evolutionary selection of alleles in the melanophilin gene that impacts on prostate organ function and cancer risk. Evol med public health,
Vol.9
(1),
pp. 311-321.
show abstract
full text
BACKGROUND AND OBJECTIVES: Several hundred inherited genetic variants or SNPs that alter the risk of cancer have been identified through genome-wide association studies. In populations of European ancestry, these variants are mostly present at relatively high frequencies. To gain insight into evolutionary origins, we screened a series of genes and SNPs linked to breast or prostate cancer for signatures of historical positive selection. METHODOLOGY: We took advantage of the availability of the 1000 genome data and we performed genomic scans for positive selection in five different Caucasian populations as well as one African reference population. We then used prostate organoid cultures to provide a possible functional explanation for the interplay between the action of evolutionary forces and the disease risk association. RESULTS: Variants in only one gene showed genomic signatures of positive, evolutionary selection within Caucasian populations melanophilin (MLPH). Functional depletion of MLPH in prostate organoids, by CRISPR/Cas9 mutation, impacted lineage commitment of progenitor cells promoting luminal versus basal cell differentiation and on resistance to androgen deprivation. CONCLUSIONS AND IMPLICATIONS: The MLPH variants influencing prostate cancer risk may have been historically selected for their adaptive benefit on skin pigmentation but MLPH is highly expressed in the prostate and the derivative, positively selected, alleles decrease the risk of prostate cancer. Our study suggests a potential functional mechanism via which MLPH and its genetic variants could influence risk of prostate cancer, as a serendipitous consequence of prior evolutionary benefits to another tissue. LAY SUMMARY: We screened a limited series of genomic variants associated with breast and prostate cancer risk for signatures of historical positive selection. Variants within the melanophilin (MLPH) gene fell into this category. Depletion of MLPH in prostate organoid cultures, suggested a potential functional mechanism for impacting on cancer risk, as a serendipitous consequence of prior evolutionary benefits to another tissue..
Krasny, L.
Bland, P.
Burns, J.
Lima, N.C.
Harrison, P.T.
Pacini, L.
Elms, M.L.
Ning, J.
Garcia Martinez, V.
Yu, Y.-.
Acton, S.E.
Ho, P.-.
Calvo, F.
Swain, A.
Howard, B.A.
Natrajan, R.C.
Huang, P.H.
(2020). A mouse SWATH-MS reference spectral library enables deconvolution of species-specific proteomic alterations in human tumour xenografts. Dis model mech,
.
show abstract
SWATH-mass spectrometry (MS) enables accurate and reproducible proteomic profiling in multiple model organisms including the mouse. Here we present a comprehensive mouse reference spectral library (MouseRefSWATH) that permits quantification of up to 10,597 proteins (62.2% of the mouse proteome) by SWATH-MS. We exploit MouseRefSWATH to develop an analytical pipeline for species-specific deconvolution of proteomic alterations in human tumour xenografts (XenoSWATH). This method overcomes the challenge of high sequence similarity between mouse and human proteins, facilitating the study of host microenvironment-tumour interactions from 'bulk tumour' measurements. We apply the XenoSWATH pipeline to characterise an intraductal xenograft model of breast ductal carcinoma in-situ and uncover complex regulation consistent with stromal reprogramming, where the modulation of cell migration pathways is not restricted to tumour cells but also operate in the mouse stroma upon progression to invasive disease. MouseRefSWATH and XenoSWATH opens new opportunities for in-depth and reproducible proteomic assessment to address wide-ranging biological questions involving this important model organism..
Krasny, L.
Bland, P.
Burns, J.
Lima, N.C.
Harrison, P.T.
Pacini, L.
Elms, M.L.
Ning, J.
Martinez, V.G.
Yu, Y.-.
Acton, S.E.
Ho, P.-.
Calvo, F.
Swain, A.
Howard, B.A.
Natrajan, R.C.
Huang, P.H.
(2020). A mouse SWATH-mass spectrometry reference spectral library enables deconvolution of species-specific proteomic alterations in human tumour xenografts. Dis model mech,
Vol.13
(7).
show abstract
full text
SWATH-mass spectrometry (MS) enables accurate and reproducible proteomic profiling in multiple model organisms including the mouse. Here, we present a comprehensive mouse reference spectral library (MouseRefSWATH) that permits quantification of up to 10,597 proteins (62.2% of the mouse proteome) by SWATH-MS. We exploit MouseRefSWATH to develop an analytical pipeline for species-specific deconvolution of proteomic alterations in human tumour xenografts (XenoSWATH). This method overcomes the challenge of high sequence similarity between mouse and human proteins, facilitating the study of host microenvironment-tumour interactions from 'bulk tumour' measurements. We apply the XenoSWATH pipeline to characterize an intraductal xenograft model of breast ductal carcinoma in situ and uncover complex regulation consistent with stromal reprogramming, where the modulation of cell migration pathways is not restricted to tumour cells but also operates in the mouse stroma upon progression to invasive disease. MouseRefSWATH and XenoSWATH open new opportunities for in-depth and reproducible proteomic assessment to address wide-ranging biological questions involving this important model organism..
Fletcher, C.E.
Sulpice, E.
Combe, S.
Shibakawa, A.
Leach, D.A.
Hamilton, M.P.
Chrysostomou, S.L.
Sharp, A.
Welti, J.
Yuan, W.
Dart, D.A.
Knight, E.
Ning, J.
Francis, J.C.
Kounatidou, E.E.
Gaughan, L.
Swain, A.
Lupold, S.E.
de Bono, J.S.
McGuire, S.E.
Gidrol, X.
Bevan, C.L.
(2019). Androgen receptor-modulatory microRNAs provide insight into therapy resistance and therapeutic targets in advanced prostate cancer. Oncogene,
Vol.38
(28),
pp. 5700-5724.
show abstract
full text
Androgen receptor (AR) signalling is a key prostate cancer (PC) driver, even in advanced 'castrate-resistant' disease (CRPC). To systematically identify microRNAs (miRs) modulating AR activity in lethal disease, hormone-responsive and -resistant PC cells expressing a luciferase-based AR reporter were transfected with a miR inhibitor library; 78 inhibitors significantly altered AR activity. Upon validation, miR-346, miR-361-3p and miR-197 inhibitors markedly reduced AR transcriptional activity, mRNA and protein levels, increased apoptosis, reduced proliferation, repressed EMT, and inhibited PC migration and invasion, demonstrating additive effects with AR inhibition. Corresponding miRs increased AR activity through a novel and anti-dogmatic mechanism of direct association with AR 6.9 kb 3'UTR and transcript stabilisation. In addition, miR-346 and miR-361-3p modulation altered levels of constitutively active AR variants, and inhibited variant-driven PC cell proliferation, so may contribute to persistent AR signalling in CRPC in the absence of circulating androgens. Pathway analysis of AGO-PAR-CLIP-identified miR targets revealed roles in DNA replication and repair, cell cycle, signal transduction and immune function. Silencing these targets, including tumour suppressors ARHGDIA and TAGLN2, phenocopied miR effects, demonstrating physiological relevance. MiR-346 additionally upregulated the oncogene, YWHAZ, which correlated with grade, biochemical relapse and metastasis in patients. These AR-modulatory miRs and targets correlated with AR activity in patient biopsies, and were elevated in response to long-term enzalutamide treatment of patient-derived CRPC xenografts. In summary, we identified miRs that modulate AR activity in PC and CRPC, via novel mechanisms, and may represent novel PC therapeutic targets..
Gonzalez-Exposito, R.
Semiannikova, M.
Griffiths, B.
Khan, K.
Barber, L.J.
Woolston, A.
Spain, G.
von Loga, K.
Challoner, B.
Patel, R.
Ranes, M.
Swain, A.
Thomas, J.
Bryant, A.
Saffery, C.
Fotiadis, N.
Guettler, S.
Mansfield, D.
Melcher, A.
Powles, T.
Rao, S.
Watkins, D.
Chau, I.
Matthews, N.
Wallberg, F.
Starling, N.
Cunningham, D.
Gerlinger, M.
(2019). CEA expression heterogeneity and plasticity confer resistance to the CEA-targeting bispecific immunotherapy antibody cibisatamab (CEA-TCB) in patient-derived colorectal cancer organoids. J immunother cancer,
Vol.7
(1),
p. 101.
show abstract
full text
BACKGROUND: The T cell bispecific antibody cibisatamab (CEA-TCB) binds Carcino-Embryonic Antigen (CEA) on cancer cells and CD3 on T cells, which triggers T cell killing of cancer cell lines expressing moderate to high levels of CEA at the cell surface. Patient derived colorectal cancer organoids (PDOs) may more accurately represent patient tumors than established cell lines which potentially enables more detailed insights into mechanisms of cibisatamab resistance and sensitivity. METHODS: We established PDOs from multidrug-resistant metastatic CRCs. CEA expression of PDOs was determined by FACS and sensitivity to cibisatamab immunotherapy was assessed by co-culture of PDOs and allogeneic CD8 T cells. RESULTS: PDOs could be categorized into 3 groups based on CEA cell-surface expression: CEAhi (n = 3), CEAlo (n = 1) and CEAmixed PDOs (n = 4), that stably maintained populations of CEAhi and CEAlo cells, which has not previously been described in CRC cell lines. CEAhi PDOs were sensitive whereas CEAlo PDOs showed resistance to cibisatamab. PDOs with mixed expression showed low sensitivity to cibisatamab, suggesting that CEAlo cells maintain cancer cell growth. Culture of FACS-sorted CEAhi and CEAlo cells from PDOs with mixed CEA expression demonstrated high plasticity of CEA expression, contributing to resistance acquisition through CEA antigen loss. RNA-sequencing revealed increased WNT/β-catenin pathway activity in CEAlo cells. Cell surface CEA expression was up-regulated by inhibitors of the WNT/β-catenin pathway. CONCLUSIONS: Based on these preclinical findings, heterogeneity and plasticity of CEA expression appear to confer low cibisatamab sensitivity in PDOs, supporting further clinical evaluation of their predictive effect in CRC. Pharmacological inhibition of the WNT/β-catenin pathway may be a rational combination to sensitize CRCs to cibisatamab. Our novel PDO and T cell co-culture immunotherapy models enable pre-clinical discovery of candidate biomarkers and combination therapies that may inform and accelerate the development of immuno-oncology agents in the clinic..
Welti, J.
Sharp, A.
Yuan, W.
Dolling, D.
Nava Rodrigues, D.
Figueiredo, I.
Gil, V.
Neeb, A.
Clarke, M.
Seed, G.
Crespo, M.
Sumanasuriya, S.
Ning, J.
Knight, E.
Francis, J.C.
Hughes, A.
Halsey, W.S.
Paschalis, A.
Mani, R.S.
Raj, G.V.
Plymate, S.R.
Carreira, S.
Boysen, G.
Chinnaiyan, A.M.
Swain, A.
de Bono, J.S.
International SU2C/PCF Prostate Cancer Dream Team,
(2018). Targeting Bromodomain and Extra-Terminal (BET) Family Proteins in Castration-Resistant Prostate Cancer (CRPC). Clin cancer res,
Vol.24
(13),
pp. 3149-3162.
show abstract
full text
Purpose: Persistent androgen receptor (AR) signaling drives castration-resistant prostate cancer (CRPC) and confers resistance to AR-targeting therapies. Novel therapeutic strategies to overcome this are urgently required. We evaluated how bromodomain and extra-terminal (BET) protein inhibitors (BETi) abrogate aberrant AR signaling in CRPC.Experimental Design: We determined associations between BET expression, AR-driven transcription, and patient outcome; and the effect and mechanism by which chemical BETi (JQ1 and GSK1210151A; I-BET151) and BET family protein knockdown regulates AR-V7 expression and AR signaling in prostate cancer models.Results: Nuclear BRD4 protein expression increases significantly (P ≤ 0.01) with castration resistance in same patient treatment-naïve (median H-score; interquartile range: 100; 100-170) and CRPC (150; 110-200) biopsies, with higher expression at diagnosis associating with worse outcome (HR, 3.25; 95% CI, 1.50-7.01; P ≤ 0.001). BRD2, BRD3, and BRD4 RNA expression in CRPC biopsies correlates with AR-driven transcription (all P ≤ 0.001). Chemical BETi, and combined BET family protein knockdown, reduce AR-V7 expression and AR signaling. This was not recapitulated by C-MYC knockdown. In addition, we show that BETi regulates RNA processing thereby reducing alternative splicing and AR-V7 expression. Furthermore, BETi reduce growth of prostate cancer cells and patient-derived organoids with known AR mutations, AR amplification and AR-V7 expression. Finally, BETi, unlike enzalutamide, decreases persistent AR signaling and growth (P ≤ 0.001) of a patient-derived xenograft model of CRPC with AR amplification and AR-V7 expression.Conclusions: BETi merit clinical evaluation as inhibitors of AR splicing and function, with trials demonstrating their blockade in proof-of-mechanism pharmacodynamic studies. Clin Cancer Res; 24(13); 3149-62. ©2018 AACR..
Francis, J.C.
Swain, A.
(2018). Prostate Organogenesis. Cold spring harb perspect med,
Vol.8
(7).
show abstract
The prostate is a male exocrine gland that secretes components of the seminal fluid. In men, prostate tumors are one of the most prevalent cancers. Studies on the development of the prostate have given a better understanding of the processes and genes that are important in the formation of this organ and have provided insights into the mechanisms of prostate tumorigenesis. These developmental studies have provided evidence that some of the genes and signaling pathways involved in development are reactivated or deregulated during prostate cancer. The prostate goes through a number of different stages during organogenesis, which include organ specification, epithelial budding, branching morphogenesis, canalization, and cytodifferentiation. During development, these processes are tightly regulated, many of which are controlled by the male hormone androgens. The majority of prostate tumors remain hormone regulated, and antiandrogen therapy is a first-line therapy, highlighting the important link between prostate organogenesis and cancer. In this review, we describe some of the data on genes that have important roles during prostate development that also have strong evidence linking them to prostate cancer..
Pettitt, S.J.
Krastev, D.B.
Brandsma, I.
Dréan, A.
Song, F.
Aleksandrov, R.
Harrell, M.I.
Menon, M.
Brough, R.
Campbell, J.
Frankum, J.
Ranes, M.
Pemberton, H.N.
Rafiq, R.
Fenwick, K.
Swain, A.
Guettler, S.
Lee, J.-.
Swisher, E.M.
Stoynov, S.
Yusa, K.
Ashworth, A.
Lord, C.J.
(2018). Genome-wide and high-density CRISPR-Cas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance. Nat commun,
Vol.9
(1),
p. 1849.
show abstract
full text
Although PARP inhibitors (PARPi) target homologous recombination defective tumours, drug resistance frequently emerges, often via poorly understood mechanisms. Here, using genome-wide and high-density CRISPR-Cas9 "tag-mutate-enrich" mutagenesis screens, we identify close to full-length mutant forms of PARP1 that cause in vitro and in vivo PARPi resistance. Mutations both within and outside of the PARP1 DNA-binding zinc-finger domains cause PARPi resistance and alter PARP1 trapping, as does a PARP1 mutation found in a clinical case of PARPi resistance. This reinforces the importance of trapped PARP1 as a cytotoxic DNA lesion and suggests that PARP1 intramolecular interactions might influence PARPi-mediated cytotoxicity. PARP1 mutations are also tolerated in cells with a pathogenic BRCA1 mutation where they result in distinct sensitivities to chemotherapeutic drugs compared to other mechanisms of PARPi resistance (BRCA1 reversion, 53BP1, REV7 (MAD2L2) mutation), suggesting that the underlying mechanism of PARPi resistance that emerges could influence the success of subsequent therapies..
Francis, J.C.
Capper, A.
Ning, J.
Knight, E.
de Bono, J.
Swain, A.
(2018). SOX9 is a driver of aggressive prostate cancer by promoting invasion, cell fate and cytoskeleton alterations and epithelial to mesenchymal transition. Oncotarget,
Vol.9
(7),
pp. 7604-7615.
show abstract
full text
Aggressive lethal prostate cancer is characterised by tumour invasion, metastasis and androgen resistance. Understanding the mechanisms by which localised disease progresses to advanced lethal stages is key to the development of effective therapies. Here we have identified a novel role for the transcription factor, SOX9, as a driver of aggressive invasive prostate cancer. Using genetically modified mouse models, we show that increased Sox9 expression in the prostate epithelia of animals with Pten loss leads to a highly invasive phenotype and metastasis. In depth analysis of these mice and related in vitro models reveals that SOX9 acts a key regulator of various processes that together promote tumour progression. We show that this factor promotes cell lineage plasticity with cells acquiring properties of basal stem cells and an increase in proliferation. In addition, increased SOX9 leads to changes in cytoskeleton and adhesion, deposition of extracellular matrix and epithelia to mesenchyme transition, properties of highly invasive cells. Analysis of castrated mice showed that the invasive phenotype driven by SOX9 is independent of androgen levels. Our study has identified a novel driver of prostate cancer progression and highlighted the cellular and molecular processes that are regulated by Sox9 to achieve invasive disease..
Pettitt, S.J.
Krastev, D.B.
Pemberton, H.N.
Fontebasso, Y.
Frankum, J.
Rehman, F.L.
Brough, R.
Song, F.
Bajrami, I.
Rafiq, R.
Wallberg, F.
Kozarewa, I.
Fenwick, K.
Armisen-Garrido, J.
Swain, A.
Gulati, A.
Campbell, J.
Ashworth, A.
Lord, C.J.
(2017). Genome-wide barcoded transposon screen for cancer drug sensitivity in haploid mouse embryonic stem cells. Sci data,
Vol.4,
p. 170020.
show abstract
full text
We describe a screen for cellular response to drugs that makes use of haploid embryonic stem cells. We generated ten libraries of mutants with piggyBac gene trap transposon integrations, totalling approximately 100,000 mutant clones. Random barcode sequences were inserted into the transposon vector to allow the number of cells bearing each insertion to be measured by amplifying and sequencing the barcodes. These barcodes were associated with their integration sites by inverse PCR. We exposed these libraries to commonly used cancer drugs and profiled changes in barcode abundance by Ion Torrent sequencing in order to identify mutations that conferred sensitivity. Drugs tested included conventional chemotherapeutics as well as targeted inhibitors of topoisomerases, poly(ADP-ribose) polymerase (PARP), Hsp90 and WEE1..
Shenoy, T.R.
Boysen, G.
Wang, M.Y.
Xu, Q.Z.
Guo, W.
Koh, F.M.
Wang, C.
Zhang, L.Z.
Wang, Y.
Gil, V.
Aziz, S.
Christova, R.
Rodrigues, D.N.
Crespo, M.
Rescigno, P.
Tunariu, N.
Riisnaes, R.
Zafeiriou, Z.
Flohr, P.
Yuan, W.
Knight, E.
Swain, A.
Ramalho-Santos, M.
Xu, D.Y.
de Bono, J.
Wu, H.
(2017). CHD1 loss sensitizes prostate cancer to DNA damaging therapy by promoting error-prone double-strand break repair. Ann oncol,
Vol.28
(7),
pp. 1495-1507.
show abstract
full text
BACKGROUND: Deletion of the chromatin remodeler chromodomain helicase DNA-binding protein 1 (CHD1) is a common genomic alteration found in human prostate cancers (PCas). CHD1 loss represents a distinct PCa subtype characterized by SPOP mutation and higher genomic instability. However, the role of CHD1 in PCa development in vivo and its clinical utility remain unclear. PATIENTS AND METHODS: To study the role of CHD1 in PCa development and its loss in clinical management, we generated a genetically engineered mouse model with prostate-specific deletion of murine Chd1 as well as isogenic CHD1 wild-type and homozygous deleted human benign and PCa lines. We also developed patient-derived organoid cultures and screened patients with metastatic PCa for CHD1 loss. RESULTS: We demonstrate that CHD1 loss sensitizes cells to DNA damage and causes a synthetic lethal response to DNA damaging therapy in vitro, in vivo, ex vivo, in patient-derived organoid cultures and in a patient with metastatic PCa. Mechanistically, CHD1 regulates 53BP1 stability and CHD1 loss leads to decreased error-free homologous recombination (HR) repair, which is compensated by increased error-prone non-homologous end joining (NHEJ) repair for DNA double-strand break (DSB) repair. CONCLUSIONS: Our study provides the first in vivo and in patient evidence supporting the role of CHD1 in DSB repair and in response to DNA damaging therapy. We uncover mechanistic insights that CHD1 modulates the choice between HR and NHEJ DSB repair and suggest that CHD1 loss may contribute to the genomic instability seen in this subset of PCas..
Magness, A.J.
Squires, J.A.
Griffiths, B.
Khan, K.
Swain, A.
Willison, K.R.
Cunningham, D.
Gerlinger, M.
Klug, D.R.
(2017). Multiplexed single cell protein expression analysis in solid tumours using a miniaturised microfluidic assay. Convergent science physical oncology,
Vol.3
(2).
Swain, A.
(2017). Ductal sex determination. Science,
Vol.357
(6352),
p. 648.
Pearson, A.
Smyth, E.
Babina, I.S.
Herrera-Abreu, M.T.
Tarazona, N.
Peckitt, C.
Kilgour, E.
Smith, N.R.
Geh, C.
Rooney, C.
Cutts, R.
Campbell, J.
Ning, J.
Fenwick, K.
Swain, A.
Brown, G.
Chua, S.
Thomas, A.
Johnston, S.R.
Ajaz, M.
Sumpter, K.
Gillbanks, A.
Watkins, D.
Chau, I.
Popat, S.
Cunningham, D.
Turner, N.C.
(2016). High-Level Clonal FGFR Amplification and Response to FGFR Inhibition in a Translational Clinical Trial. Cancer discov,
Vol.6
(8),
pp. 838-851.
show abstract
full text
UNLABELLED: FGFR1 and FGFR2 are amplified in many tumor types, yet what determines response to FGFR inhibition in amplified cancers is unknown. In a translational clinical trial, we show that gastric cancers with high-level clonal FGFR2 amplification have a high response rate to the selective FGFR inhibitor AZD4547, whereas cancers with subclonal or low-level amplification did not respond. Using cell lines and patient-derived xenograft models, we show that high-level FGFR2 amplification initiates a distinct oncogene addiction phenotype, characterized by FGFR2-mediated transactivation of alternative receptor kinases, bringing PI3K/mTOR signaling under FGFR control. Signaling in low-level FGFR1-amplified cancers is more restricted to MAPK signaling, limiting sensitivity to FGFR inhibition. Finally, we show that circulating tumor DNA screening can identify high-level clonally amplified cancers. Our data provide a mechanistic understanding of the distinct pattern of oncogene addiction seen in highly amplified cancers and demonstrate the importance of clonality in predicting response to targeted therapy. SIGNIFICANCE: Robust single-agent response to FGFR inhibition is seen only in high-level FGFR-amplified cancers, with copy-number level dictating response to FGFR inhibition in vitro, in vivo, and in the clinic. High-level amplification of FGFR2 is relatively rare in gastric and breast cancers, and we show that screening for amplification in circulating tumor DNA may present a viable strategy to screen patients. Cancer Discov; 6(8); 838-51. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 803..
International Surgical Outcomes Study group,
(2016). Global patient outcomes after elective surgery: prospective cohort study in 27 low-, middle- and high-income countries. Br j anaesth,
Vol.117
(5),
pp. 601-609.
show abstract
BACKGROUND: As global initiatives increase patient access to surgical treatments, there remains a need to understand the adverse effects of surgery and define appropriate levels of perioperative care. METHODS: We designed a prospective international 7-day cohort study of outcomes following elective adult inpatient surgery in 27 countries. The primary outcome was in-hospital complications. Secondary outcomes were death following a complication (failure to rescue) and death in hospital. Process measures were admission to critical care immediately after surgery or to treat a complication and duration of hospital stay. A single definition of critical care was used for all countries. RESULTS: A total of 474 hospitals in 19 high-, 7 middle- and 1 low-income country were included in the primary analysis. Data included 44 814 patients with a median hospital stay of 4 (range 2-7) days. A total of 7508 patients (16.8%) developed one or more postoperative complication and 207 died (0.5%). The overall mortality among patients who developed complications was 2.8%. Mortality following complications ranged from 2.4% for pulmonary embolism to 43.9% for cardiac arrest. A total of 4360 (9.7%) patients were admitted to a critical care unit as routine immediately after surgery, of whom 2198 (50.4%) developed a complication, with 105 (2.4%) deaths. A total of 1233 patients (16.4%) were admitted to a critical care unit to treat complications, with 119 (9.7%) deaths. Despite lower baseline risk, outcomes were similar in low- and middle-income compared with high-income countries. CONCLUSIONS: Poor patient outcomes are common after inpatient surgery. Global initiatives to increase access to surgical treatments should also address the need for safe perioperative care. STUDY REGISTRATION: ISRCTN51817007.
Francis, J.C.
Melchor, L.
Campbell, J.
Kendrick, H.
Wei, W.
Armisen-Garrido, J.
Assiotis, I.
Chen, L.
Kozarewa, I.
Fenwick, K.
Swain, A.
Smalley, M.J.
Lord, C.J.
Ashworth, A.
(2015). Whole-exome DNA sequence analysis of Brca2- and Trp53-deficient mouse mammary gland tumours. J pathol,
Vol.236
(2),
pp. 186-200.
show abstract
Germline mutations in the tumour suppressor BRCA2 predispose to breast, ovarian and a number of other human cancers. Brca2-deficient mouse models are used for preclinical studies but the pattern of genomic alterations in these tumours has not yet been described in detail. We have performed whole-exome DNA sequencing analysis of mouse mammary tumours from Blg-Cre Brca2(f/f) Trp53(f/f) animals, a model of BRCA2-deficient human cancer. We also used the sequencing data to estimate DNA copy number alterations in these tumours and identified a recurrent copy number gain in Met, which has been found amplified in other mouse mammary cancer models. Through a comparative genomic analysis, we identified several mouse Blg-Cre Brca2(f/f) Trp53(f/f) mammary tumour somatic mutations in genes that are also mutated in human cancer, but few of these genes have been found frequently mutated in human breast cancer. A more detailed analysis of these somatic mutations revealed a set of genes that are mutated in human BRCA2 mutant breast and ovarian tumours and that are also mutated in mouse Brca2-null, Trp53-null mammary tumours. Finally, a DNA deletion surrounded by microhomology signature found in human BRCA1/2-deficient cancers was not common in the genome of these mouse tumours. Although a useful model, there are some differences in the genomic landscape of tumours arising in Blg-Cre Brca2(f/f) Trp53(f/f) mice compared to human BRCA-mutated breast cancers. Therefore, this needs to be taken into account in the use of this model..
Mateo, J.
Carreira, S.
Sandhu, S.
Miranda, S.
Mossop, H.
Perez-Lopez, R.
Nava Rodrigues, D.
Robinson, D.
Omlin, A.
Tunariu, N.
Boysen, G.
Porta, N.
Flohr, P.
Gillman, A.
Figueiredo, I.
Paulding, C.
Seed, G.
Jain, S.
Ralph, C.
Protheroe, A.
Hussain, S.
Jones, R.
Elliott, T.
McGovern, U.
Bianchini, D.
Goodall, J.
Zafeiriou, Z.
Williamson, C.T.
Ferraldeschi, R.
Riisnaes, R.
Ebbs, B.
Fowler, G.
Roda, D.
Yuan, W.
Wu, Y.-.
Cao, X.
Brough, R.
Pemberton, H.
A'Hern, R.
Swain, A.
Kunju, L.P.
Eeles, R.
Attard, G.
Lord, C.J.
Ashworth, A.
Rubin, M.A.
Knudsen, K.E.
Feng, F.Y.
Chinnaiyan, A.M.
Hall, E.
de Bono, J.S.
(2015). DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N engl j med,
Vol.373
(18),
pp. 1697-1708.
show abstract
full text
BACKGROUND: Prostate cancer is a heterogeneous disease, but current treatments are not based on molecular stratification. We hypothesized that metastatic, castration-resistant prostate cancers with DNA-repair defects would respond to poly(adenosine diphosphate [ADP]-ribose) polymerase (PARP) inhibition with olaparib. METHODS: We conducted a phase 2 trial in which patients with metastatic, castration-resistant prostate cancer were treated with olaparib tablets at a dose of 400 mg twice a day. The primary end point was the response rate, defined either as an objective response according to Response Evaluation Criteria in Solid Tumors, version 1.1, or as a reduction of at least 50% in the prostate-specific antigen level or a confirmed reduction in the circulating tumor-cell count from 5 or more cells per 7.5 ml of blood to less than 5 cells per 7.5 ml. Targeted next-generation sequencing, exome and transcriptome analysis, and digital polymerase-chain-reaction testing were performed on samples from mandated tumor biopsies. RESULTS: Overall, 50 patients were enrolled; all had received prior treatment with docetaxel, 49 (98%) had received abiraterone or enzalutamide, and 29 (58%) had received cabazitaxel. Sixteen of 49 patients who could be evaluated had a response (33%; 95% confidence interval, 20 to 48), with 12 patients receiving the study treatment for more than 6 months. Next-generation sequencing identified homozygous deletions, deleterious mutations, or both in DNA-repair genes--including BRCA1/2, ATM, Fanconi's anemia genes, and CHEK2--in 16 of 49 patients who could be evaluated (33%). Of these 16 patients, 14 (88%) had a response to olaparib, including all 7 patients with BRCA2 loss (4 with biallelic somatic loss, and 3 with germline mutations) and 4 of 5 with ATM aberrations. The specificity of the biomarker suite was 94%. Anemia (in 10 of the 50 patients [20%]) and fatigue (in 6 [12%]) were the most common grade 3 or 4 adverse events, findings that are consistent with previous studies of olaparib. CONCLUSIONS: Treatment with the PARP inhibitor olaparib in patients whose prostate cancers were no longer responding to standard treatments and who had defects in DNA-repair genes led to a high response rate. (Funded by Cancer Research UK and others; ClinicalTrials.gov number, NCT01682772; Cancer Research UK number, CRUK/11/029.)..
Lokody, I.B.
Francis, J.C.
Gardiner, J.R.
Erler, J.T.
Swain, A.
(2015). Pten Regulates Epithelial Cytodifferentiation during Prostate Development. Plos one,
Vol.10
(6),
p. e0129470.
show abstract
full text
Gene expression and functional studies have indicated that the molecular programmes involved in prostate development are also active in prostate cancer. PTEN has been implicated in human prostate cancer and is frequently mutated in this disease. Here, using the Nkx3.1:Cre mouse strain and a genetic deletion approach, we investigate the role of Pten specifically in the developing mouse prostate epithelia. In contrast to its role in other developing organs, this gene is dispensable for the initial developmental processes such as budding and branching. However, as cytodifferentiation progresses, abnormal luminal cells fill the ductal lumens together with augmented epithelial proliferation. This phenotype resembles the hyperplasia seen in postnatal Pten deletion models that develop neoplasia at later stages. Consistent with this, gene expression analysis showed a number of genes affected that are shared with Pten mutant prostate cancer models, including a decrease in androgen receptor regulated genes. In depth analysis of the phenotype of these mice during development revealed that loss of Pten leads to the precocious differentiation of epithelial cells towards a luminal cell fate. This study provides novel insight into the role of Pten in prostate development as part of the process of coordinating the differentiation and proliferation of cell types in time and space to form a functional organ..
Bryant, S.L.
Francis, J.C.
Lokody, I.B.
Wang, H.
Risbridger, G.P.
Loveland, K.L.
Swain, A.
(2014). Sex specific retinoic acid signaling is required for the initiation of urogenital sinus bud development. Dev biol,
Vol.395
(2),
pp. 209-217.
show abstract
full text
The mammalian urogenital sinus (UGS) develops in a sex specific manner, giving rise to the prostate in the male and the sinus vagina in the embryonic female. Androgens, produced by the embryonic testis, have been shown to be crucial to this process. In this study we show that retinoic acid signaling is required for the initial stages of bud development from the male UGS. Enzymes involved in retinoic acid synthesis are expressed in the UGS mesenchyme in a sex specific manner and addition of ligand to female tissue is able to induce prostate-like bud formation in the absence of androgens, albeit at reduced potency. Functional studies in mouse organ cultures that faithfully reproduce the initiation of prostate development indicate that one of the roles of retinoic acid signaling in the male is to inhibit the expression of Inhba, which encodes the βA subunit of Activin, in the UGS mesenchyme. Through in vivo genetic analysis and culture studies we show that inhibition of Activin signaling in the female UGS leads to a similar phenotype to that of retinoic acid treatment, namely bud formation in the absence of androgens. Our data also reveals that both androgens and retinoic acid have extra independent roles to that of repressing Activin signaling in the development of the prostate during fetal stages. This study identifies a novel role for retinoic acid as a mesenchymal factor that acts together with androgens to determine the position and initiation of bud development in the male UGS epithelia..
Francis, J.C.
Thomsen, M.K.
Taketo, M.M.
Swain, A.
(2013). beta-Catenin Is Required for Prostate Development and Cooperates with Pten Loss to Drive Invasive Carcinoma. Plos genetics,
Vol.9
(1).
Buaas, F.W.
Gardiner, J.R.
Clayton, S.
Val, P.
Swain, A.
(2012). In vivo evidence for the crucial role of SF1 in steroid-producing cells of the testis, ovary and adrenal gland. Development,
Vol.139
(24),
pp. 4561-4570.
show abstract
Adrenal and gonadal steroids are essential for life and reproduction. The orphan nuclear receptor SF1 (NR5A1) has been shown to regulate the expression of enzymes involved in steroid production in vitro. However, the in vivo role of this transcription factor in steroidogenesis has not been elucidated. In this study, we have generated steroidogenic-specific Cre-expressing mice to lineage mark and delete Sf1 in differentiated steroid-producing cells of the testis, the ovary and the adrenal gland. Our data show that SF1 is a regulator of the expression of steroidogenic genes in all three organs. In addition, Sf1 deletion leads to a radical change in cell morphology and loss of identity. Surprisingly, sexual development and reproduction in mutant animals were not compromised owing, in part, to the presence of a small proportion of SF1-positive cells. In contrast to the testis and ovary, the mutant adult adrenal gland showed a lack of Sf1-deleted cells and our studies suggest that steroidogenic adrenal cells during foetal stages require Sf1 to give rise to the adult adrenal population. This study is the first to show the in vivo requirements of SF1 in steroidogenesis and provides novel data on the cellular consequences of the loss of this protein specifically within steroid-producing cells..
Strochlic, L.
Falk, J.
Goillot, E.
Sigoillot, S.
Bourgeois, F.
Delers, P.
Rouviere, J.
Swain, A.
Castellani, V.
Schaeffer, L.
Legay, C.
(2012). Wnt4 Participates in the Formation of Vertebrate Neuromuscular Junction. Plos one,
Vol.7
(1).
Gardiner, J.R.
Shima, Y.
Morohashi, K.-.
Swain, A.
(2012). SF-1 expression during adrenal development and tumourigenesis. Molecular and cellular endocrinology,
Vol.351
(1),
pp. 12-18.
Tsaousi, A.
Williams, H.
Lyon, C.A.
Taylor, V.
Swain, A.
Johnson, J.L.
George, S.J.
(2011). Wnt4/β-Catenin Signaling Induces VSMC Proliferation and Is Associated With Intimal Thickening. Circulation research,
Vol.108
(4),
pp. 427-27.
Thomsen, M.K.
Ambroisine, L.
Wynn, S.
Cheah, K.S.
Foster, C.S.
Fisher, G.
Berney, D.M.
Møller, H.
Reuter, V.E.
Scardino, P.
Cuzick, J.
Ragavan, N.
Singh, P.B.
Martin, F.L.
Butler, C.M.
Cooper, C.S.
Swain, A.
Transatlantic Prostate Group,
(2010). SOX9 elevation in the prostate promotes proliferation and cooperates with PTEN loss to drive tumor formation. Cancer res,
Vol.70
(3),
pp. 979-987.
show abstract
Dysregulation of tissue development pathways can contribute to cancer initiation and progression. In murine embryonic prostate epithelia, the transcription factor SOX9 is required for proper prostate development. In this study, we examined a role for SOX9 in prostate cancer in mouse and human. In Pten and Nkx3.1 mutant mice, cells with increased levels of SOX9 appeared within prostate epithelia at early stages of neoplasia, and higher expression correlated with progression at all stages of disease. In transgenic mice, SOX9 overexpression in prostate epithelia increased cell proliferation without inducing hyperplasia. In transgenic mice that were also heterozygous for mutant Pten, SOX9 overexpression quickened the induction of high-grade prostate intraepithelial neoplasia. In contrast, Sox9 attenuation led to a decrease proliferating prostate epithelia cells in normal and homozygous Pten mutant mice with prostate neoplasia. Analysis of a cohort of 880 human prostate cancer samples showed that SOX9 expression was associated with increasing Gleason grades and higher Ki67 staining. Our findings identify SOX9 as part of a developmental pathway that is reactivated in prostate neoplasia where it promotes tumor cell proliferation..
Francis, J.C.
McCarthy, A.
Thomsen, M.K.
Ashworth, A.
Swain, A.
(2010). Brca2 and Trp53 Deficiency Cooperate in the Progression of Mouse Prostate Tumourigenesis. Plos genetics,
Vol.6
(6),
p. 9.
Val, P.
Swain, A.
(2010). Gene dosage effects and transcriptional regulation of early mammalian adrenal cortex development. Molecular and cellular endocrinology,
Vol.323
(1),
pp. 105-10.
Buaas, F.W.
Val, P.
Swain, A.
(2009). The transcription co-factor CITED2 functions during sex determination and early gonad development. Hum mol genet,
Vol.18
(16),
pp. 2989-3001.
show abstract
The early bi-potential mammalian gonad requires the expression of a Y-linked gene, Sry, during a brief window of time to ensure proper testis development. WT1 and its direct target gene Sf1 function during sex determination as well as in the specified testes and ovaries. We have previously shown that the transcription co-factor CITED2 interacts with WT1 to stimulate the expression of Sf1 in the adrenogonadal primordium to ensure adrenal development. We now show through genetic interactions and expression analyses that Cited2 acts in the gonad with Wt1 and Sf1 to increase the expression of Sry levels to attain a critical threshold to efficiently initiate testis development. Reducing the gene dosage of Wt1 or Sf1 in Cited2 mutant gonads was sufficient to produce partial XY sex reversal while full sex reversal was attained in mutants containing a hypomorphic Sry(POS) allele. A direct correlation was observed between XY sex reversal and reduced expression levels of Sry and Sf1 during sex determination, which indicated that Sry is a downstream target of the CITED2/WT1/SF1 regulatory pathway. Our results provide in vivo evidence for the identification of the first transcription co-factor to function during mammalian sex determination, as part of the WT1/SF1 regulatory mechanism. This highlights the gene dosage sensitivity of the pathway's effect on Sry levels and embryonic gonad development..
Thomsen, M.K.
Francis, J.C.
Swain, A.
(2008). The role of Sox9 in prostate development. Differentiation,
Vol.76
(6),
pp. 728-8.
Thomsen, M.K.
Butler, C.M.
Shen, M.M.
Swain, A.
(2008). Sox9 is required for prostate development. Dev biol,
Vol.316
(2),
pp. 302-311.
show abstract
The mammalian prostate arises from the urogenital sinus and few factors have been identified to be important in the early stages of prostate development. In this study we show that the transcription factor Sox9 is expressed in the epithelia of all mouse prostatic lobes from the initial stages of their development. We used a conditional approach with mice expressing Cre recombinase under the control of Nkx3.1 regulatory sequences to delete Sox9 from the developing prostate. Mice with a prostate specific deletion of Sox9 showed a lack of ventral prostate development and abnormal anterior prostate differentiation. Analysis of these mutant animals revealed an early loss of expression of genes specific to the prostate epithelia such as Nkx3.1 and Shh and a marked reduction in proliferation in the ventral prostate but not in other lobes. Fgf signalling, through the MAPK pathway, has been shown to be important in prostate development and a lobe specific phenotype was reported for a prostate specific Fgfr2 mutant mouse model. Here we show that the levels of Fgfr2 and Sprouty2, a downstream target of Fgf signalling, were severely reduced in the ventral prostate of Sox9 mutant animals but not in other lobes. Prostate organ culture studies with a Mek inhibitor, U0126, and a Fgf receptor inhibitor, SU5402, indicate that the timing of expression of Cre in the mutant animals could account for the lobe specific phenotype in the Sox9 and Fgfr2 mutants. These studies imply that Sox9 is required for the early differentiation of the prostate bud epithelia..
Val, P.
Martinez-Barbera, J.-.
Swain, A.
(2007). Adrenal development is initiated by Cited2 and Wt1 through modulation of Sf-1 dosage. Development,
Vol.134
(12),
pp. 2349-2358.
show abstract
It has been proposed that the mammalian adrenal cortex and gonad are derived from the same primordium present during early urogenital development. Molecular pathways involved in the differentiation of the adrenal cortex from the adrenogonadal primordium (AGP) have yet to be determined. Here we show in mice that the transcription co-factor Cited2 is required for the specification of the adrenal cortex from the AGP. We present genetic and molecular evidence demonstrating that Cited2 interacts with the transcription factor Wt1 to stimulate expression of the nuclear hormone receptor Sf-1 (Nr5a1) in the AGP prior to the separation between gonad and adrenal cortex. We show a direct correlation between the expression levels of Sf-1 in the AGP and the defects in adrenal development found in mice with different Cited2 and Wt1 mutant backgrounds. Analysis of embryos heterozygous for mutations in both Sf-1 and Cited2 confirmed that these genes act in the same pathway during adrenal development. Our studies reveal a regulatory mechanism in which Cited2 acts as a Wt1 co-factor to increase, at a critical time in embryogenesis, the levels of the essential transcription factor Sf-1 in the AGP above the threshold required to determine adrenal development. These results highlight the importance of transcription factor dosage in organogenesis and the role of transcription co-factors such as Cited2 in determining the levels of these factors..
Val, P.
Jeays-Ward, K.
Swain, A.
(2006). Identification of a novel population of adrenal-like cells in the mammalian testis. Dev biol,
Vol.299
(1),
pp. 250-256.
show abstract
Steroidogenic cells of the adrenal and gonad are thought to be derived from a common primordium that divides into separate tissues during embryogenesis. In this paper, we show that cells with mixed adrenal and Leydig cell properties are found dispersed in the insterstitium of the embryonic and adult mouse testis. They express the adrenal markers Cyp11b1 and Cyp21 and respond to ACTH. Consistent with these properties, we show that the embryonic testis produces the adrenal steroid corticosterone. These cells also express Cyp17 and respond to hCG stimulation but do not express the Leydig specific marker Insl3 showing that they are a population of steroidogenic cells distinct from Leydig cells. Based on their properties, we refer to these cells as adrenal-like cells of the testis and propose that they are the mouse equivalent of the precursors of human adrenal rests, tumors found primarily in male patients with congenital adrenal hyperplasia. Organ culture studies show that ACTH-responsive cells are present at the gonad/mesonephros border and seem to migrate into the XY but not the XX gonad during development. Consistent with this, using transgenic Cyp11a1 reporter mice, we definitively show that steroidogenic cells can migrate from the mesonephros into the XY gonad. We also show that the region between the mesonephros and the gonad harbors steroidogenic cell precursors that are repressed by the presence of the mesonephros. We propose that this region is the source of the adrenal-like cells that migrate into the testis as it develops and are activated when Leydig cells differentiate. These studies reveal the complex nature of steroidogenic cell differentiation during urogenital development..
Swain, A.
(2006). Sex determination: time for meiosis? The gonad decides. Curr biol,
Vol.16
(13),
pp. R507-R509.
show abstract
Germ cell sex determination is directed by the gonad and is characterized by a difference in the timing of entry into meiosis. Recent data show that retinoic acid signalling is responsible for the induction of germ cell meiosis in the developing ovary. In the fetal testis, this process is inhibited by a retinoic acid metabolizing enzyme..
Val, P.
Swain, A.
(2005). Mechanisms of disease: normal and abnormal gonadal development and sex determination in mammals. Nature clinical practice urology,
Vol.2
(12),
pp. 616-12.
Yao, H.H.
Matzuk, M.M.
Jorgez, C.J.
Menke, D.B.
Page, D.C.
Swain, A.
Capel, B.
(2004). Follistatin operates downstream of Wnt4 in mammalian ovary organogenesis. Developmental dynamics,
Vol.230
(2),
pp. 210-6.
full text
Jeays-Ward, K.
Dandonneau, M.
Swain, A.
(2004). Wnt4 is required for proper male as well as female sexual development. Dev biol,
Vol.276
(2),
pp. 431-440.
show abstract
Genes previously implicated in mammalian sexual development have either a male- or female-specific role. The signaling molecule WNT4 has been shown to be important in female sexual development. Lack of Wnt4 gives rise to masculinization of the XX gonad and we showed previously that the role of WNT4 was to inhibit endothelial and steroidogenic cell migration into the developing ovary. Here we show that Wnt4 also has a function in the male gonad. We find that Sertoli cell differentiation is compromised in Wnt4 mutant testes and that this defect occurs downstream of the testis-determining gene Sry but upstream of Sox9 and Dhh, two early Sertoli cell markers. Genetic analysis shows that this phenotype is primarily due to the action of WNT4 within the early genital ridge. Analysis of different markers identifies the most striking difference in the genital ridge at early stages of its development between wild-type and Wnt4 mutant embryos to be a significant increase of steroidogenic cells in the Wnt4 -/- gonad. These results identify WNT4 as a new factor involved in the mammalian testis determination pathway and show that genes can have a specific but distinct role in both male and female gonad development..
Mizusaki, H.
Kawabe, K.
Mukai, T.
Ariyoshi, E.
Kasahara, M.
Yoshioka, H.
Swain, A.
Morohashi, K.
(2003). Dax-1 (dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1) gene transcription is regulated by Wnt4 in the female developing gonad. Molecular endocrinology,
Vol.17
(4),
pp. 507-13.
Jeays-Ward, K.
Hoyle, C.
Brennan, J.
Dandonneau, M.
Alldus, G.
Capel, B.
Swain, A.
(2003). Endothelial and steroidogenic cell migration are regulated by WNT4 in the developing mammalian gonad. Development,
Vol.130
(16),
pp. 3663-3670.
show abstract
The signalling molecule WNT4 has been associated with sex reversal phenotypes in mammals. Here we show that the role of WNT4 in gonad development is to pattern the sex-specific vasculature and to regulate steroidogenic cell recruitment. Vascular formation and steroid production in the mammalian gonad occur in a sex-specific manner. During testis development, endothelial cells migrate from the mesonephros into the gonad to form a coelomic blood vessel. Leydig cells differentiate and produce steroid hormones a day later. Neither of these events occurs in the XX gonad. We show that WNT4 represses mesonephric endothelial and steroidogenic cell migration in the XX gonad, preventing the formation of a male-specific coelomic blood vessel and the production of steroids. In the XY gonad, Wnt4 expression is downregulated after sex determination. Transgenic misexpression of Wnt4 in the embryonic testis did not inhibit coelomic vessel formation but vascular pattern was affected. Leydig cell differentiation was not affected in these transgenic animals and our data implies that Wnt4 does not regulate steroidogenic cell differentiation but represses the migration of steroidogenic adrenal precursors into the gonad. These studies provide a model for understanding how the same signalling molecule can act on two different cell types to coordinate sex development..
Swain, A.
(2002). Vertebrate sex determination: a new player in the field. Curr biol,
Vol.12
(17),
pp. R602-R603.
show abstract
Sex-determining genes have been identified in flies, worms and mammals but not, until recently, in non-mammalian vertebrates. Now, a gene has been isolated from the Y chromosome of the teleost fish medaka that is functionally comparable to the mammalian testis-determining gene, Sry..
Hoyle, C.
Narvaez, V.
Alldus, G.
Lovell-Badge, R.
Swain, A.
(2002). Dax1 expression is dependent on steroidogenic factor 1 in the developing gonad. Mol endocrinol,
Vol.16
(4),
pp. 747-756.
show abstract
The nuclear hormone receptor DAX1 has been implicated in mammalian gonad development and sex determination. The expression of the gene in the gonad follows a dynamic pattern in time and place in the embryo and the adult. We have undertaken the first in vivo study of the regulation of Dax1 expression. Using a transgenic mouse approach we have identified a novel 500-bp region 4 kb upstream of the mouse Dax1 start codon that is essential for LacZ reporter gene expression in the embryonic gonad. Within this region, a highly conserved steroidogenic factor 1 (SF1) consensus-binding site is necessary to direct LacZ expression to the embryonic gonad implicating SF1 in the regulation of Dax1 in the developing gonad. Consistent with this, Dax1 is expressed at much reduced levels in gonads of embryos that are deficient in SF1. In addition, our results show that SF1 consensus-binding sites close to the start of Dax1 transcription are important in regulating levels of expression in the developing gonad. These studies have identified the critical in vivo regulatory region for expression of Dax1 in the early gonad and provide novel information on how a specific enhancer element acts in different cell types at different stages of development..
De Vries, G.J.
Rissman, E.F.
Simerly, R.B.
Yang, L.Y.
Scordalakes, E.M.
Auger, C.J.
Swain, A.
Lovell-Badge, R.
Burgoyne, P.S.
Arnold, A.P.
(2002). A model system for study of sex chromosome effects on sexually dimorphic neural and behavioral traits. Journal of neuroscience,
Vol.22
(20),
pp. 9005-10.
Jordan, B.K.
Mohammed, M.
Ching, S.T.
Délot, E.
Chen, X.-.
Dewing, P.
Swain, A.
Rao, P.N.
Elejalde, B.R.
Vilain, E.
(2001). Up-Regulation of WNT-4 Signaling and Dosage-Sensitive Sex Reversal in Humans. The american journal of human genetics,
Vol.68
(5),
pp. 1102-1109.
Swain, A.
(2000). Molecular mechanisms in sex determination and gonadal development. Biology of reproduction,
Vol.62,
pp. 83-1.
Swain, A.
Lovell-Badge, R.
(1999). Mammalian sex determination: a molecular drama. Genes & development,
Vol.13
(7),
pp. 755-13.
Swain, A.
Lovell-Badge, R.
(1998). Developmental genetics - Too much sex is bad for males. Nature,
Vol.396
(6707),
pp. 115-2.
Swain, A.
Narvaez, V.
Burgoyne, P.
Camerino, G.
Lovell-Badge, R.
(1998). Dax1 antagonizes Sry action in mammalian sex determination. Nature,
Vol.391
(6669),
pp. 761-767.
show abstract
DAX1, which encodes an unusual member of the nuclear hormone-receptor superfamily, is a gene that may be responsible for a sex-reversal syndrome in humans, referred to as dosage-sensitive sex reversal, in which XY individuals carrying duplications of Xp21, part of the small arm of the X chromosome, develop as females. XY mice carrying extra copies of mouse Dax1 as a transgene show delayed testis development when the gene is expressed at high levels, but do not normally show sex reversal. Complete sex reversal occurs, however, when the transgene is tested against weak alleles of the sex-determining Y-chromosome gene Sry. These results show that DAX1 is largely, if not solely, responsible for dosage-sensitive sex reversal and provide a model for early events in mammalian sex determination, when precise levels and timing of gene expression are critical..
Swain, A.
Lovell-Badge, R.
(1997). A molecular approach to sex determination in mammals. Acta paediatr suppl,
Vol.423,
pp. 46-49.
show abstract
Mammalian sex determination occurs in the gonad of the developing embryo. This process is dependent on the Y-chromosome-encoded Sry gene that acts in the somatic cells of the genital ridge. The transient nature of Sry gene expression suggests that it acts as a switch from one cell fate to another. One of the roles of Sry is to initiate the differentiation of Sertoli cells, which are the first cell type of the testis to be formed. Two genes are thought to be important in Sertoli cell differentiation and function, Sox9, an Sry-related gene, and SF-1, a nuclear hormone receptor. Sox9 is expressed in Sertoli cells throughout development of the mouse embryo, and inactivating mutations in this gene in humans give rise to XY females. SF-1 is also expressed in Sertoli cells and is thought to activate the AMH gene--an early marker of these cells. DAX-1, an X-linked member of the nuclear hormone superfamily, is a candidate for a human condition in which duplication of regions of the X chromosome results in XY females. Expression of this gene during mouse development is associated with ovary development and is down-regulated in the differentiating testis. Mutations in DAX-1 in humans have shown that this gene is not necessary for testis development. The properties of the DAX-1 gene suggest that it is important in ovary determination and might therefore be antagonistic to the action of the Sry gene..
Swain, A.
Zanaria, E.
Hacker, A.
Lovell-Badge, R.
Camerino, G.
(1996). Mouse Dax1 expression is consistent with a role in sex determination as well as in adrenal and hypothalamus function. Nat genet,
Vol.12
(4),
pp. 404-409.
show abstract
Duplications of a chromosome Xp21 locus DSS (Dosage Sensitive Sex reversal) are associated with male to female sex reversal. An unusual member of the nuclear hormone receptor superfamily, DAX1, maps to the DSS critical region and is responsible for X-linked adrenal hypoplasia congenita. Here we describe the isolation of the mouse Dax1 gene and its pattern of expression during development. Expression was detected in the first stages of gonadal and adrenal differentiation and in the developing hypothalamus. Moreover, Dax1 expression is down-regulated coincident with overt differentiation in the testis, but persists in the developing ovary. Comparison of the predicted protein products of the human and mouse genes show that specific domains are evolving rapidly. Our results suggest a basis for adrenal insufficiency and hypogonadotropic hypogonadism in males affected by adrenal hypoplasia congenita and are consistent with a role for DAX1 in gonadal sex determination..
Morais da Silva, S.
Hacker, A.
Harley, V.
Goodfellow, P.
Swain, A.
Lovell-Badge, R.
(1996). Sox9 expression during gonadal development implies a conserved role for the gene in testis differentiation in mammals and birds. Nat genet,
Vol.14
(1),
pp. 62-68.
show abstract
Heterozygous mutations in SOX9 lead to a human dwarfism syndrome, Campomelic dysplasia. Consistent with a role in sex determination, we find that Sox9 expression closely follows differentiation of Sertoli cells in the mouse testis, in experimental sex reversal when fetal ovaries are grafted to adult kidneys and in the chick where there is no evidence for a Sry gene. Our results imply that Sox9 plays an essential role in sex determination, possibly immediately downstream of Sry in mammals, and that it functions as a critical Sertoli cell differentiation factor, perhaps in all vertebrates..
Ikeda, Y.
Swain, A.
Weber, T.J.
Hentges, K.E.
Zanaria, E.
Lalli, E.
Tamai, K.T.
Sassone-Corsi, P.
Lovell-Badge, R.
Camerino, G.
Parker, K.L.
(1996). Steroidogenic factor 1 and Dax-1 colocalize in multiple cell lineages: potential links in endocrine development. Molecular endocrinology,
Vol.10
(10),
pp. 1261-1272.
Ikeda, Y.
Swain, A.
Weber, T.J.
Hentges, K.E.
Zanaria, E.
Lalli, E.
Tamai, K.T.
SassoneCorsi, P.
LovellBadge, R.
Camerino, G.
Parker, K.L.
(1996). Steroidogenic factor 1 and Dax-1 colocalize in multiple cell lineages: Potential links in endocrine development. Molecular endocrinology,
Vol.10
(10),
pp. 1261-12.
Capel, B.
Swain, A.
Nicolis, S.
Hacker, A.
Walter, M.
Koopman, P.
Goodfellow, P.
Lovell-Badge, R.
(1993). Circular transcripts of the testis-determining gene Sry in adult mouse testis. Cell,
Vol.73
(5),
pp. 1019-1030.
show abstract
Sry is expressed at higher levels in the adult testis, where no function has been determined, than in the genital ridge, its critical site of action. cDNA and 5' RACE clones isolated from testis or from Sry-transfected cell lines have an unusual structure, with 3' sequences located in a 5' position. RNAase protection assays and reverse transcription polymerase chain reactions confirmed that these unusual RNA molecules represent the most abundant transcript in testis. Furthermore, oligonucleotide hybridization and RNAase H digestion proved that these Sry RNA molecules are circular. Similar transcripts were detected in the testes of mice with Mus musculus musculus, Mus musculus domesticus, and Mus spretus Sry genes. The circular RNA is found in the cytoplasm but is not substantially bound to polysomes. We suggest that the circles arise from normal splicing processes as a consequence of the unusual genomic structure surrounding the Sry locus in the mouse..