18 results on '"Hewett, Duncan R."'
Search Results
2. Clodronate-Liposome Mediated Macrophage Depletion Abrogates Multiple Myeloma Tumor Establishment In Vivo
- Author
-
Opperman, Khatora S., Vandyke, Kate, Clark, Kimberley C., Coulter, Elizabeth A., Hewett, Duncan R., Mrozik, Krzysztof M., Schwarz, Nisha, Evdokiou, Andreas, Croucher, Peter I, Psaltis, Peter J, Noll, Jacqueline E, and Zannettino, Andrew CW
- Published
- 2019
- Full Text
- View/download PDF
3. A niche-dependent myeloid transcriptome signature defines dormant myeloma cells
- Author
-
Khoo, Weng Hua, Ledergor, Guy, Weiner, Assaf, Roden, Daniel L., Terry, Rachael L., McDonald, Michelle M., Chai, Ryan C., De Veirman, Kim, Owen, Katie L., Opperman, Khatora S., Vandyke, Kate, Clark, Justine R., Seckinger, Anja, Kovacic, Natasa, Nguyen, Akira, Mohanty, Sindhu T., Pettitt, Jessica A., Xiao, Ya, Corr, Alexander P., Seeliger, Christine, Novotny, Mark, Lasken, Roger S., Nguyen, Tuan V., Oyajobi, Babatunde O., Aftab, Dana, Swarbrick, Alexander, Parker, Belinda, Hewett, Duncan R., Hose, Dirk, Vanderkerken, Karin, Zannettino, Andrew C.W., Amit, Ido, Phan, Tri Giang, and Croucher, Peter I.
- Published
- 2019
- Full Text
- View/download PDF
4. Subclonal evolution in disease progression from MGUS/SMM to multiple myeloma is characterised by clonal stability
- Author
-
Dutta, Ankit K., Fink, J. Lynn, Grady, John P., Morgan, Gareth J., Mullighan, Charles G., To, Luen B., Hewett, Duncan R., and Zannettino, Andrew C. W.
- Published
- 2019
- Full Text
- View/download PDF
5. DNA Barcoding Reveals Habitual Clonal Dominance of Myeloma Plasma Cells in the Bone Marrow Microenvironment
- Author
-
Hewett, Duncan R., Vandyke, Kate, Lawrence, David M., Friend, Natasha, Noll, Jacqueline E., Geoghegan, Joel M., Croucher, Peter I., and Zannettino, Andrew C.W.
- Published
- 2017
- Full Text
- View/download PDF
6. Myeloperoxidase creates a permissive microenvironmental niche for the progression of multiple myeloma.
- Author
-
Williams, Connor M. D., Noll, Jacqueline E., Bradey, Alanah L., Duggan, Jvaughn, Wilczek, Vicki J., Masavuli, Makutiro G., Grubor‐Bauk, Branka, Panagopoulos, Romana A., Hewett, Duncan R., Mrozik, Krzysztof M., Zannettino, Andrew C. W., Vandyke, Kate, and Panagopoulos, Vasilios
- Subjects
MULTIPLE myeloma ,MYELOPEROXIDASE ,MYELOID-derived suppressor cells ,MONOCLONAL gammopathies ,MYELOID cells ,CELL populations ,BORTEZOMIB - Abstract
Summary: Expression of myeloperoxidase (MPO), a key inflammatory enzyme restricted to myeloid cells, is negatively associated with the development of solid tumours. Activated myeloid cell populations are increased in multiple myeloma (MM); however, the functional consequences of myeloid‐derived MPO within the myeloma microenvironment are unknown. Here, the role of MPO in MM pathogenesis was investigated, and the capacity for pharmacological inhibition of MPO to impede MM progression was evaluated. In the 5TGM1‐KaLwRij mouse model of myeloma, the early stages of tumour development were associated with an increase in CD11b+ myeloid cell populations and an increase in Mpo expression within the bone marrow (BM). Interestingly, MM tumour cell homing was increased towards sites of elevated myeloid cell numbers and MPO activity within the BM. Mechanistically, MPO induced the expression of key MM growth factors, resulting in tumour cell proliferation and suppressed cytotoxic T‐cell activity. Notably, tumour growth studies in mice treated with a small‐molecule irreversible inhibitor of MPO (4‐ABAH) demonstrated a significant reduction in overall MM tumour burden. Taken together, our data demonstrate that MPO contributes to MM tumour growth, and that MPO‐specific inhibitors may provide a new therapeutic strategy to limit MM disease progression. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. SAMSN1 Is a Tumor Suppressor Gene in Multiple Myeloma
- Author
-
Noll, Jacqueline E., Hewett, Duncan R., Williams, Sharon A., Vandyke, Kate, Kok, Chung, To, Luen B., and Zannettino, Andrew C.W.
- Published
- 2014
- Full Text
- View/download PDF
8. Tetraspanin 7 (TSPAN7) expression is upregulated in multiple myeloma patients and inhibits myeloma tumour development in vivo
- Author
-
Cheong, Chee Man, Chow, Annie W.S., Fitter, Stephen, Hewett, Duncan R., Martin, Sally K., Williams, Sharon A., To, Bik L., Zannettino, Andrew C.W., and Vandyke, Kate
- Published
- 2015
- Full Text
- View/download PDF
9. A p53-dependent mechanism underlies macrocytic anemia in a mouse model of human 5q-syndrome
- Author
-
Barlow, Jillian L., Drynan, Lesley F., Hewett, Duncan R., Holmes, Luke R., Lorenzo-Abalde, Silvia, Lane, Alison L., Jolin, Helen E., Pannell, Richard, Middleton, Angela J., Wong, See Heng, Warren, Alan J., Wainscoat, James S., Boultwood, Jacqueline, and McKenzie, Andrew N.J.
- Subjects
Myelodysplastic syndromes -- Research ,Bone marrow cells -- Research ,Human chromosome abnormalities -- Research ,Chromosome deletion -- Research ,Macrocytic anemia -- Research ,Biological sciences ,Health - Abstract
The identification of the genes associated with chromosomal translocation breakpoints has fundamentally changed understanding of the molecular basis of hematological malignancies. By contrast, the study of chromosomal deletions has been hampered by the large number of genes deleted and the complexity of their analysis. We report the generation of a mouse model for human 5q-syndrome using large-scale chromosomal engineering. Haploinsufficiency of the Cd74-Nid67 interval (containing Rps14, encoding the ribosomal protein S14) caused macrocytic anemia, prominent erythroid dysplasia and monolobulated megakaryocytes in the bone marrow. These effects were associated with defective bone marrow progenitor development, the appearance of bone marrow cells expressing high amounts of the tumor suppressor p53 and increased bone marrow cell apoptosis. Notably, intercrossing with p53-deficient mice completely rescued the progenitor cell defect, restoring common myeloid progenitor and megakaryocytic-erythroid progenitor, granulocyte-monocyte progenitor and hematopoietic stem cell bone marrow populations. This mouse model suggests that a p53-dependent mechanism underlies the pathophysiology of the 5q-syndrome., 5q-syndrome was first described in 1974 as a consistent association of the deletion of the long arm of chromosome 5 (del(5q)) with hematological abnormalities: macrocytosis, anemia, normal or high platelet [...]
- Published
- 2010
- Full Text
- View/download PDF
10. Lethal, neonatal ichthyosis with increased proteolytic processing of filaggrin in a mouse model of Netherton syndrome
- Author
-
Hewett, Duncan R., Simons, Alison L., Mangan, Niamh E., Jolin, Helen E., Green, Shelia M., Fallon, Padraic G., and McKenzie, Andrew N.J.
- Published
- 2005
11. Characterization of the role of Samsn1 loss in multiple myeloma development.
- Author
-
Friend, Natasha L., Hewett, Duncan R., Panagopoulos, Vasilios, Noll, Jacqueline E., Vandyke, Kate, Mrozik, Krzysztof M., Fitter, Stephen, and Zannettino, Andrew C.W.
- Abstract
The protein SAMSN1 was recently identified as a putative tumor suppressor in multiple myeloma, with re‐expression of Samsn1 in the 5TGM1/KaLwRij murine model of myeloma leading to a near complete abrogation of intramedullary tumor growth. Here, we sought to clarify the mechanism underlying this finding. Intratibial administration of 5TGM1 myeloma cells into KaLwRij mice revealed that Samsn1 had no effect on primary tumor growth, but that its expression significantly inhibited the metastasis of these primary tumors. Notably, neither in vitro nor in vivo migration was affected by Samsn1 expression. Both knocking‐out SAMSN1 in the RPMI‐8226 and JJN3 human myeloma cell lines, and retrovirally expressing SAMSN1 in the LP‐1 and OPM2 human myeloma cell lines had no effect on either cell proliferation or migration in vitro. Altering SAMSN1 expression in these human myeloma cells did not affect the capacity of the cells to establish either primary or metastatic intramedullary tumors when administered intratibially into immune deficient NSG mice. Unexpectedly, the tumor suppressive and anti‐metastatic activity of Samsn1 in 5TGM1 cells were not evidenced following cell administration either intratibially or intravenously to NSG mice. Crucially, the growth of Samsn1‐expressing 5TGM1 cells was limited in C57BL/6/Samsn1−/− mice but not in C57BL/6 Samsn1+/+ mice. We conclude that the reported potent in vivo tumor suppressor activity of Samsn1 can be attributed, in large part, to graft‐rejection from Samsn1−/− recipient mice. This has broad implications for the design and interpretation of experiments that utilize cancer cells and knockout mice that are mismatched for expression of specific proteins. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
12. LCRF‐0006, a small molecule mimetic of the N‐cadherin antagonist peptide ADH‐1, synergistically increases multiple myeloma response to bortezomib.
- Author
-
Mrozik, Krzysztof M., Cheong, Chee M., Hewett, Duncan R., Noll, Jacqueline E., Opperman, Khatora S., Adwal, Alaknanda, Russell, Darryl L., Blaschuk, Orest W., Vandyke, Kate, and Zannettino, Andrew C. W.
- Abstract
N‐cadherin is a homophilic cell‐cell adhesion molecule that plays a critical role in maintaining vascular stability and modulating endothelial barrier permeability. Pre‐clinical studies have shown that the N‐cadherin antagonist peptide, ADH‐1, increases the permeability of tumor‐associated vasculature thereby increasing anti‐cancer drug delivery to tumors and enhancing tumor response. Small molecule library screens have identified a novel compound, LCRF‐0006, that is a mimetic of the classical cadherin His‐Ala‐Val sequence‐containing region of ADH‐1. Here, we evaluated the vascular permeability‐enhancing and anti‐cancer properties of LCRF‐0006 using in vitro vascular disruption and cell apoptosis assays, and a well‐established pre‐clinical model (C57BL/KaLwRij/5TGM1) of the hematological cancer multiple myeloma (MM). We found that LCRF‐0006 disrupted endothelial cell junctions in a rapid, transient and reversible manner, and increased vascular permeability in vitro and at sites of MM tumor in vivo. Notably, LCRF‐0006 synergistically increased the in vivo anti‐MM tumor response to low‐dose bortezomib, a frontline anti‐MM agent, leading to regression of disease in 100% of mice. Moreover, LCRF‐0006 and bortezomib synergistically induced 5TGM1 MM tumor cell apoptosis in vitro. Our findings demonstrate the potential clinical utility of LCRF‐0006 to significantly increase bortezomib effectiveness and enhance the depth of tumor response in patients with MM. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
13. GLIPR1 expression is reduced in multiple myeloma but is not a tumour suppressor in mice.
- Author
-
Friend, Natasha, Noll, Jacqueline E., Opperman, Khatora S., Clark, Kimberley C., Mrozik, Krzysztof M., Vandyke, Kate, Hewett, Duncan R., and Zannettino, Andrew C. W.
- Subjects
MULTIPLE myeloma ,CARDIAC amyloidosis ,PLASMA cells ,TUMORS ,CELLULAR aging ,MICE - Abstract
Multiple myeloma, a plasma cell malignancy, is a genetically heterogeneous disease and the genetic factors that contribute to its development and progression remain to be fully elucidated. The tumour suppressor gene GLIPR1 has previously been shown to be deleted in approximately 10% of myeloma patients, to inhibit the development of plasma cell tumours in ageing mice and to have reduced expression levels in the plasma cells of patients with light-chain amyloidosis, a myeloma-related malignancy. Therefore, we hypothesised that GLIPR1 may have tumour suppressor activity in multiple myeloma. In this study, we demonstrate that plasma cell expression of GLIPR1 is reduced in the majority of myeloma patients and Glipr1 expression is lost in the 5TGM1 murine myeloma cell line. However, overexpression of GLIPR1 in a human myeloma cell line did not affect cell proliferation in vitro. Similarly, re-expression of Glipr1 in 5TGM1 cells did not significantly reduce their in vitro proliferation or in vivo growth in C57BL/KaLwRij mice. In addition, using CRISPR-Cas9 genome editing, we generated C57BL/Glipr1
-/- mice and showed that loss of Glipr1 in vivo did not affect normal haematopoiesis or the development of monoclonal plasma cell expansions in these mice up to one year of age. Taken together, our results suggest that GLIPR1 is unlikely to be a potent tumour suppressor in multiple myeloma. However, it remains possible that the down-regulation of GLIPR1 may cooperate with other genetic lesions to promote the development of myeloma. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
14. Cutting edge genomics reveal new insights into tumour development, disease progression and therapeutic impacts in multiple myeloma.
- Author
-
Dutta, Ankit K., Hewett, Duncan R., Fink, J. Lynn, Grady, John P., and Zannettino, Andrew C. W.
- Subjects
- *
MULTIPLE myeloma , *PLASMA cells , *BONE marrow , *MONOCLONAL gammopathies , *TUMORS - Abstract
Multiple Myeloma ( MM) is a haematological malignancy characterised by the clonal expansion of plasma cells ( PCs) within the bone marrow. Despite advances in therapy, MM remains a largely incurable disease with a median survival of 6 years. In almost all cases, the development of MM is preceded by the benign PC condition Monoclonal Gammopathy of Undetermined Significance ( MGUS). Recent studies show that the transformation of MGUS to MM is associated with complex genetic changes. Understanding how these changes contribute to evolution will present targets for clinical intervention. We discuss three models of MM evolution; the linear, the expansionist and the intraclonal heterogeneity models. Of particular interest is the intraclonal heterogeneity model. Here, distinct populations of MM PCs carry differing combinations of genetic mutations. Acquisition of additional mutations can contribute to subclonal lineages where 'driver' mutations may influence selective pressure and dominance, and 'passenger' mutations are neutral in their effects. Furthermore, studies show that clinical intervention introduces additional selective pressure on tumour cells and can influence subclone survival, leading to therapy resistance. This review discusses how Next Generation Sequencing approaches are revealing critical insights into the genetics of MM development, disease progression and treatment. MM disease progression will illuminate possible mechanisms underlying the tumour. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
15. PTTG1 expression is associated with hyperproliferative disease and poor prognosis in multiple myeloma.
- Author
-
Noll, Jacqueline E., Vandyke, Kate, Hewett, Duncan R., Mrozik, Krzysztof M., Bala, Rachel J., Williams, Sharon A., Kok, Chung H., and Zannettino, Andrew C. W.
- Subjects
MULTIPLE myeloma ,HEMATOLOGICAL oncology ,PLASMA cells ,PITUITARY tumors ,DISEASE progression ,BONE marrow diseases ,GENE expression - Abstract
Background: Multiple myeloma (MM) is an incurable haematological malignancy characterised by the clonal proliferation of malignant plasma cells within the bone marrow. We have previously identified pituitary tumour transforming gene 1 (Pttg1) as a gene that is significantly upregulated in the haematopoietic compartment of the myeloma-susceptible C57BL/KaLwRij mouse strain, when compared with the myeloma-resistant C57BL/6 mouse. Over-expression of PTTG1 has previously been associated with malignant progression and an enhanced proliferative capacity in solid tumours. Methods: In this study, we investigated PTTG1 gene and protein expression in MM plasma cells from newly diagnosed MM patients. Gene expression profiling was used to identify gene signatures associated with high PTTG1 expression in MM patients. Additionally, we investigated the effect of short hairpin ribonucleic acid (shRNA)-mediated PTTG1 knockdown on the proliferation of the murine myeloma plasma cell line 5TGM1 in vitro and in vivo. Results: PTTG1 was found to be over-expressed in 36-70 % of MM patients, relative to normal controls, with high PTTG1 expression being associated with poor patient outcomes (hazard ratio 2.49; 95 % CI 1.28 to 4.86; p = 0.0075; log-rank test). In addition, patients with high PTTG1 expression exhibited increased expression of cell proliferation-associated genes including CCNB1, CCNB2, CDK1, AURKA, BIRC5 and DEPDC1. Knockdown of Pttg1 in 5TGM1 cells decreased cellular proliferation, without affecting cell cycle distribution or viability, and decreased expression of Ccnb1, Birc5 and Depdc1 in vitro. Notably, Pttg1 knockdown significantly reduced MM tumour development in vivo, with an 83.2 % reduction in tumour burden at 4 weeks (p < 0.0001, two-way ANOVA). Conclusions: This study supports a role for increased PTTG1 expression in augmenting tumour development in a subset of MM patients. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
16. Identification of an interleukin (IL)-25--dependent cell population that provides IL-4, IL-5, and IL-13 at the onset of helminth expulsion.
- Author
-
Fallon, Padraic G., Ballantyne, Sarah J., Mangan, Niamh E., Barlow, Jillian L., Dasvarma, Ayan, Hewett, Duncan R., McIlgorm, Ann, Jolin, Helen E., and McKenzie, Andrew N. J.
- Subjects
INTERLEUKINS ,LYMPHOCYTES ,IMMUNITY ,NATURAL immunity ,CELL populations - Abstract
Type 2 immunity, which involves coordinated regulation of innate and adaptive immune responses, can protect against helminth parasite infection, but may lead to allergy and asthma after inappropriate activation. We demonstrate that il25
-/- mice display inefficient Nippostrongylus brasiliensis expulsion and delayed cytokine production by T helper 2 cells. We further establish a key role for interleukin (IL)-25 in regulating a novel population of IL-4-, IL-5-, IL-13-producing non-B/non-T (NBNT), c-kit+ , FeεR1- cells during helminth infection. A deficit in this population in il25-/- mice correlates with inefficient IV. brasiliensis expulsion. In contrast, administration of recombinant IL-25 in vivo induces the appearance of NBNT, c-kit+ , FcεR- cells and leads to rapid worm expulsion that is T and B cell independent, but type 2 cytokine dependent. We demonstrate that these IL-25-regulated cells appear rapidly in the draining lymph nodes, implicating them as a source of type 2 cytokines during initiation of worm expulsion. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
17. Targeted Disruption of Bone Marrow Stromal Cell-Derived Gremlin1 Limits Multiple Myeloma Disease Progression In Vivo.
- Author
-
Clark, Kimberley C., Hewett, Duncan R., Panagopoulos, Vasilios, Plakhova, Natalya, Opperman, Khatora S., Bradey, Alanah L., Mrozik, Krzysztof M., Vandyke, Kate, Mukherjee, Siddhartha, Davies, Gareth C.G., Worthley, Daniel L., and Zannettino, Andrew C.W.
- Subjects
- *
ANIMAL experimentation , *BONE marrow diseases , *GLYCOPROTEINS , *GROWTH factors , *MULTIPLE myeloma , *DISEASE progression , *IN vivo studies - Abstract
In most instances, multiple myeloma (MM) plasma cells (PCs) are reliant on factors made by cells of the bone marrow (BM) stroma for their survival and growth. To date, the nature and cellular composition of the BM tumor microenvironment and the critical factors which drive tumor progression remain imprecisely defined. Our studies show that Gremlin1 (Grem1), a highly conserved protein, which is abundantly secreted by a subset of BM mesenchymal stromal cells, plays a critical role in MM disease development. Analysis of human and mouse BM stromal samples by quantitative PCR showed that GREM1/Grem1 expression was significantly higher in the MM tumor-bearing cohorts compared to healthy controls (p < 0.05, Mann–Whitney test). Additionally, BM-stromal cells cultured with 5TGM1 MM PC line expressed significantly higher levels of Grem1, compared to stromal cells alone (p < 0.01, t-test), suggesting that MM PCs promote increased Grem1 expression in stromal cells. Furthermore, the proliferation of 5TGM1 MM PCs was found to be significantly increased when co-cultured with Grem1-overexpressing stromal cells (p < 0.01, t-test). To examine the role of Grem1 in MM disease in vivo, we utilized the 5TGM1/KaLwRij mouse model of MM. Our studies showed that, compared to immunoglobulin G (IgG) control antibody-treated mice, mice treated with an anti-Grem1 neutralizing antibody had a decrease in MM tumor burden of up to 81.2% (p < 0.05, two-way ANOVA). The studies presented here demonstrate, for the first time, a novel positive feedback loop between MM PCs and BM stroma, and that inhibiting this vicious cycle with a neutralizing antibody can dramatically reduce tumor burden in a preclinical mouse model of MM. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
18. Twist-1 is upregulated by NSD2 and contributes to tumour dissemination and an epithelial-mesenchymal transition-like gene expression signature in t(4;14)-positive multiple myeloma.
- Author
-
Cheong, Chee Man, Mrozik, Krzysztof M., Hewett, Duncan R., Bell, Elyse, Panagopoulos, Vasilios, Noll, Jacqueline E., Licht, Jonathan D., Gronthos, Stan, Zannettino, Andrew C.W., and Vandyke, Kate
- Subjects
- *
MULTIPLE myeloma , *GENE expression , *GENE expression profiling , *CHROMOSOMAL translocation , *TUMORS , *PROTEIN metabolism , *CHROMOSOMES , *PROTEINS , *CANCER cell culture , *RESEARCH , *NUCLEAR proteins , *ANIMAL experimentation , *RESEARCH methodology , *CELL physiology , *PROGNOSIS , *APOPTOSIS , *EVALUATION research , *MEDICAL cooperation , *COMPARATIVE studies , *TRANSFERASES , *CHROMOSOME abnormalities , *GENES , *RESEARCH funding , *MICE - Abstract
Approximately 15% of patients with multiple myeloma (MM) harbour the t(4;14) chromosomal translocation, leading to the overexpression of the histone methyltransferase NSD2. Patients with this translocation display increased tumour dissemination, accelerated disease progression and rapid relapse. Using publicly available gene expression profile data from NSD2high (n = 135) and NSD2low (n = 878) MM patients, we identified 39 epithelial-mesenchymal transition (EMT)-associated genes which are overexpressed in NSD2high MM plasma cells. In addition, our analyses identified Twist-1 as a key transcription factor upregulated in NSD2high MM patients and t(4;14)-positive cell lines. Overexpression and knockdown studies confirmed that Twist-1 is involved in driving the expression of EMT-associated genes in the human MM cell line KMS11 and promoted the migration of myeloma cell lines in vitro. Notably, Twist-1 overexpression in the mouse MM cell line 5TGM1 significantly increased tumour dissemination in an intratibial tumour model. These findings demonstrate that Twist-1, downstream of NSD2, contributes to the induction of an EMT-like signature in t(4;14)-positive MM and enhances the dissemination of MM plasma cells in vivo, which may, in part, explain the aggressive disease features associated with t(4;14)-positive MM. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.