84 results on '"Kristin K. Brown"'
Search Results
2. MERIT40 Is an Akt Substrate that Promotes Resolution of DNA Damage Induced by Chemotherapy
- Author
-
Kristin K. Brown, Laleh Montaser-Kouhsari, Andrew H. Beck, and Alex Toker
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Resistance to cytotoxic chemotherapy drugs, including doxorubicin, is a significant obstacle to the effective treatment of breast cancer. Here, we have identified a mechanism by which the PI3K/Akt pathway mediates resistance to doxorubicin. In addition to inducing DNA damage, doxorubicin triggers sustained activation of Akt signaling in breast cancer cells. We show that Akt contributes to chemotherapy resistance such that PI3K or Akt inhibitors sensitize cells to doxorubicin. We identify MERIT40, a component of the BRCA1-A DNA damage repair complex, as an Akt substrate that is phosphorylated following doxorubicin treatment. MERIT40 phosphorylation facilitates assembly of the BRCA1-A complex in response to DNA damage and contributes to DNA repair and cell survival following doxorubicin treatment. Finally, MERIT40 phosphorylation in human breast cancers is associated with estrogen receptor positivity. Our findings suggest that combination therapy with PI3K or Akt inhibitors and doxorubicin may constitute a successful strategy for overcoming chemotherapy resistance.
- Published
- 2015
- Full Text
- View/download PDF
3. S-Nitrosothiol Signaling Regulates Liver Development and Improves Outcome following Toxic Liver Injury
- Author
-
Andrew G. Cox, Diane C. Saunders, Peter B. Kelsey Jr., Allie A. Conway, Yevgenia Tesmenitsky, Julio F. Marchini, Kristin K. Brown, Jonathan S. Stamler, Dorothy B. Colagiovanni, Gary J. Rosenthal, Kevin J. Croce, Trista E. North, and Wolfram Goessling
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Toxic liver injury is a leading cause of liver failure and death because of the organ’s inability to regenerate amidst massive cell death, and few therapeutic options exist. The mechanisms coordinating damage protection and repair are poorly understood. Here, we show that S-nitrosothiols regulate liver growth during development and after injury in vivo; in zebrafish, nitric-oxide (NO) enhanced liver formation independently of cGMP-mediated vasoactive effects. After acetaminophen (APAP) exposure, inhibition of the enzymatic regulator S-nitrosoglutathione reductase (GSNOR) minimized toxic liver damage, increased cell proliferation, and improved survival through sustained activation of the cytoprotective Nrf2 pathway. Preclinical studies of APAP injury in GSNOR-deficient mice confirmed conservation of hepatoprotective properties of S-nitrosothiol signaling across vertebrates; a GSNOR-specific inhibitor improved liver histology and acted with the approved therapy N-acetylcysteine to expand the therapeutic time window and improve outcome. These studies demonstrate that GSNOR inhibitors will be beneficial therapeutic candidates for treating liver injury.
- Published
- 2014
- Full Text
- View/download PDF
4. The KEAP1–NRF2 pathway regulates TFEB/TFE3-dependent lysosomal biogenesis
- Author
-
Athena Jessica S. Ong, Cerys E. Bladen, Tara A. Tigani, Anthony P. Karamalakis, Kimberley J. Evason, Kristin K. Brown, and Andrew G. Cox
- Subjects
Multidisciplinary - Abstract
The maintenance of redox and metabolic homeostasis is integral to embryonic development. Nuclear factor erythroid 2-related factor 2 (NRF2) is a stress-induced transcription factor that plays a central role in the regulation of redox balance and cellular metabolism. Under homeostatic conditions, NRF2 is repressed by Kelch-like ECH-associated protein 1 (KEAP1). Here, we demonstrate that Keap1 deficiency induces Nrf2 activation and postdevelopmental lethality. Loss of viability is preceded by severe liver abnormalities characterized by an accumulation of lysosomes. Mechanistically, we demonstrate that loss of Keap1 promotes aberrant activation of transcription factor EB (TFEB)/transcription factor binding to IGHM Enhancer 3 (TFE3)-dependent lysosomal biogenesis. Importantly, we find that NRF2-dependent regulation of lysosomal biogenesis is cell autonomous and evolutionarily conserved. These studies identify a role for the KEAP1–NRF2 pathway in the regulation of lysosomal biogenesis and suggest that maintenance of lysosomal homeostasis is required during embryonic development.
- Published
- 2023
5. Data from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Author
-
Mark A. Dawson, Raphaël Rodriguez, Kate McArthur, Andrew H. Wei, David C.S. Huang, Kristin K. Brown, Andrew G. Cox, Georg Ramm, Sarah-Jane Dawson, Estelle Duprez, Mathilde Poplineau, Giovanna Pomilio, Véronique Litalien, Marian L. Burr, Tatiana Cañeque, Enid Y.N. Lam, Brian Liddicoat, Laura MacPherson, Lorey Smith, Kevin Tran, Kah Lok Chan, James A. Kuzich, Caitlin L. Rowe, Ali Motazedian, Yih-Chih Chan, Fiona C. Brown, Sebastian Müller, Andrew A. Guirguis, and Sylvain Garciaz
- Abstract
Cancer cell metabolism is increasingly recognized as providing an exciting therapeutic opportunity. However, a drug that directly couples targeting of a metabolic dependency with the induction of cell death in cancer cells has largely remained elusive. Here we report that the drug-like small-molecule ironomycin reduces the mitochondrial iron load, resulting in the potent disruption of mitochondrial metabolism. Ironomycin promotes the recruitment and activation of BAX/BAK, but the resulting mitochondrial outer membrane permeabilization (MOMP) does not lead to potent activation of the apoptotic caspases, nor is the ensuing cell death prevented by inhibiting the previously established pathways of programmed cell death. Consistent with the fact that ironomycin and BH3 mimetics induce MOMP through independent nonredundant pathways, we find that ironomycin exhibits marked in vitro and in vivo synergy with venetoclax and overcomes venetoclax resistance in primary patient samples.Significance:Ironomycin couples targeting of cellular metabolism with cell death by reducing mitochondrial iron, resulting in the alteration of mitochondrial metabolism and the activation of BAX/BAK. Ironomycin induces MOMP through a different mechanism to BH3 mimetics, and consequently combination therapy has marked synergy in cancers such as acute myeloid leukemia.This article is highlighted in the In This Issue feature, p. 587
- Published
- 2023
6. Supplementary movie from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Author
-
Mark A. Dawson, Raphaël Rodriguez, Kate McArthur, Andrew H. Wei, David C.S. Huang, Kristin K. Brown, Andrew G. Cox, Georg Ramm, Sarah-Jane Dawson, Estelle Duprez, Mathilde Poplineau, Giovanna Pomilio, Véronique Litalien, Marian L. Burr, Tatiana Cañeque, Enid Y.N. Lam, Brian Liddicoat, Laura MacPherson, Lorey Smith, Kevin Tran, Kah Lok Chan, James A. Kuzich, Caitlin L. Rowe, Ali Motazedian, Yih-Chih Chan, Fiona C. Brown, Sebastian Müller, Andrew A. Guirguis, and Sylvain Garciaz
- Abstract
Supplementary movie from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Published
- 2023
7. Supplementary table from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Author
-
Mark A. Dawson, Raphaël Rodriguez, Kate McArthur, Andrew H. Wei, David C.S. Huang, Kristin K. Brown, Andrew G. Cox, Georg Ramm, Sarah-Jane Dawson, Estelle Duprez, Mathilde Poplineau, Giovanna Pomilio, Véronique Litalien, Marian L. Burr, Tatiana Cañeque, Enid Y.N. Lam, Brian Liddicoat, Laura MacPherson, Lorey Smith, Kevin Tran, Kah Lok Chan, James A. Kuzich, Caitlin L. Rowe, Ali Motazedian, Yih-Chih Chan, Fiona C. Brown, Sebastian Müller, Andrew A. Guirguis, and Sylvain Garciaz
- Abstract
Supplementary table from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Published
- 2023
8. Supplementary Data from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Author
-
Mark A. Dawson, Raphaël Rodriguez, Kate McArthur, Andrew H. Wei, David C.S. Huang, Kristin K. Brown, Andrew G. Cox, Georg Ramm, Sarah-Jane Dawson, Estelle Duprez, Mathilde Poplineau, Giovanna Pomilio, Véronique Litalien, Marian L. Burr, Tatiana Cañeque, Enid Y.N. Lam, Brian Liddicoat, Laura MacPherson, Lorey Smith, Kevin Tran, Kah Lok Chan, James A. Kuzich, Caitlin L. Rowe, Ali Motazedian, Yih-Chih Chan, Fiona C. Brown, Sebastian Müller, Andrew A. Guirguis, and Sylvain Garciaz
- Abstract
Supplementary Data from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Published
- 2023
9. Supplementary Figure from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Author
-
Mark A. Dawson, Raphaël Rodriguez, Kate McArthur, Andrew H. Wei, David C.S. Huang, Kristin K. Brown, Andrew G. Cox, Georg Ramm, Sarah-Jane Dawson, Estelle Duprez, Mathilde Poplineau, Giovanna Pomilio, Véronique Litalien, Marian L. Burr, Tatiana Cañeque, Enid Y.N. Lam, Brian Liddicoat, Laura MacPherson, Lorey Smith, Kevin Tran, Kah Lok Chan, James A. Kuzich, Caitlin L. Rowe, Ali Motazedian, Yih-Chih Chan, Fiona C. Brown, Sebastian Müller, Andrew A. Guirguis, and Sylvain Garciaz
- Abstract
Supplementary Figure from Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Published
- 2023
10. Supplementary Figure S1 from Adaptive Reprogramming of De Novo Pyrimidine Synthesis Is a Metabolic Vulnerability in Triple-Negative Breast Cancer
- Author
-
Alex Toker, John M. Asara, Jessica B. Spinelli, and Kristin K. Brown
- Abstract
Supplementary Figure S1. Chemotherapy exposure stimulates an increase in pyrimidine nucleotides in TNBC cells.
- Published
- 2023
11. Pharmacologic Reduction of Mitochondrial Iron Triggers a Noncanonical BAX/BAK-Dependent Cell Death
- Author
-
Andrew H. Wei, Kevin Tran, Kristin K. Brown, Fiona C. Brown, Caitlin L. Rowe, Tatiana Cañeque, Ali Motazedian, Sebastian Müller, Giovanna Pomilio, Raphaël Rodriguez, Andrew G. Cox, Kate McArthur, Mathilde Poplineau, Marian L. Burr, Enid Y.N. Lam, Sylvain Garciaz, Mark A. Dawson, Lorey K. Smith, Brian Liddicoat, Georg Ramm, Estelle Duprez, Kah Lok Chan, Yih-Chih Chan, Sarah-Jane Dawson, Andrew A Guirguis, James Anton Kuzich, Veronique Litalien, David C.S. Huang, Laura MacPherson, University of Melbourne, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC), Chimie biologique des membranes et ciblage thérapeutique (CBMCT - UMR 3666 / U1143), Institut Curie [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), Monash University [Melbourne], The Walter and Eliza Hall Institute of Medical Research (WEHI), and Duprez, Estelle
- Subjects
Programmed cell death ,Iron ,Apoptosis ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,[SDV.BBM.BM] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,In vivo ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Humans ,Caspase ,bcl-2-Associated X Protein ,030304 developmental biology ,0303 health sciences ,Cell Death ,biology ,Chemistry ,Venetoclax ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,Metabolism ,In vitro ,Mitochondria ,3. Good health ,Cell biology ,bcl-2 Homologous Antagonist-Killer Protein ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] - Abstract
Cancer cell metabolism is increasingly recognized as providing an exciting therapeutic opportunity. However, a drug that directly couples targeting of a metabolic dependency with the induction of cell death in cancer cells has largely remained elusive. Here we report that the drug-like small-molecule ironomycin reduces the mitochondrial iron load, resulting in the potent disruption of mitochondrial metabolism. Ironomycin promotes the recruitment and activation of BAX/BAK, but the resulting mitochondrial outer membrane permeabilization (MOMP) does not lead to potent activation of the apoptotic caspases, nor is the ensuing cell death prevented by inhibiting the previously established pathways of programmed cell death. Consistent with the fact that ironomycin and BH3 mimetics induce MOMP through independent nonredundant pathways, we find that ironomycin exhibits marked in vitro and in vivo synergy with venetoclax and overcomes venetoclax resistance in primary patient samples. Significance: Ironomycin couples targeting of cellular metabolism with cell death by reducing mitochondrial iron, resulting in the alteration of mitochondrial metabolism and the activation of BAX/BAK. Ironomycin induces MOMP through a different mechanism to BH3 mimetics, and consequently combination therapy has marked synergy in cancers such as acute myeloid leukemia. This article is highlighted in the In This Issue feature, p. 587
- Published
- 2022
12. Glutamine addiction promotes glucose oxidation in triple-negative breast cancer
- Author
-
Lake-Ee Quek, Michelle van Geldermalsen, Yi Fang Guan, Kanu Wahi, Chelsea Mayoh, Seher Balaban, Angel Pang, Qian Wang, Mark J. Cowley, Kristin K. Brown, Nigel Turner, Andrew J. Hoy, and Jeff Holst
- Subjects
Cancer Research ,Glucose ,Cell Line, Tumor ,Glutamine ,Citric Acid Cycle ,Genetics ,Glutamic Acid ,Humans ,Triple Negative Breast Neoplasms ,Molecular Biology - Abstract
Glutamine is a conditionally essential nutrient for many cancer cells, but it remains unclear how consuming glutamine in excess of growth requirements confers greater fitness to glutamine-addicted cancers. By contrasting two breast cancer subtypes with distinct glutamine dependencies, we show that glutamine-indispensable triple-negative breast cancer (TNBC) cells rely on a non-canonical glutamine-to-glutamate overflow, with glutamine carbon routed once through the TCA cycle. Importantly, this single-pass glutaminolysis increases TCA cycle fluxes and replenishes TCA cycle intermediates in TNBC cells, a process that achieves net oxidation of glucose but not glutamine. The coupling of glucose and glutamine catabolism appears hard-wired via a distinct TNBC gene expression profile biased to strip and then sequester glutamine nitrogen, but hampers the ability of TNBC cells to oxidise glucose when glutamine is limiting. Our results provide a new understanding of how metabolically rigid TNBC cells are sensitive to glutamine deprivation and a way to select vulnerable TNBC subtypes that may be responsive to metabolic-targeted therapies.
- Published
- 2022
13. AMPK CA(R)Sts a new light on amino acid sensing
- Author
-
Kristin K. Brown
- Subjects
chemistry.chemical_classification ,General Immunology and Microbiology ,General Neuroscience ,Fatty Acids ,Regulator ,AMPK ,Articles ,Metabolism ,AMP-Activated Protein Kinases ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Amino acid ,Glucose ,chemistry ,Biochemistry ,Sense (molecular biology) ,Amino Acids ,Cellular energy ,Energy Metabolism ,Protein kinase A ,Molecular Biology ,Cysteine - Abstract
Adenosine 5'‐monophosphate (AMP)‐activated protein kinase (AMPK) is an important cellular metabolite‐sensing enzyme that can directly sense changes not only in ATP but also in metabolites associated with carbohydrates and fatty acids. However, less is known about whether and how AMPK senses variations in cellular amino acids. Here, we show that cysteine deficiency significantly triggers calcium/calmodulin‐dependent protein kinase kinase 2 (CaMKK2)‐mediated activation of AMPK. In addition, we found that CaMKK2 directly associates with cysteinyl‐tRNA synthetase (CARS), which then binds to AMPKγ2 under cysteine deficiency to activate AMPK. Interestingly, we discovered that cysteine inhibits the binding of CARS to AMPKγ2, and thus, under cysteine deficiency conditions wherein the inhibitory effect of cysteine is abrogated, CARS mediates the binding of AMPK to CaMKK2, resulting in the phosphorylation and activation of AMPK by CaMKK2. Importantly, we demonstrate that blocking AMPK activation leads to cell death under cysteine‐deficient conditions. In summary, our study is the first to show that CARS senses the absence of cysteine and activates AMPK through the cysteine–CARS–CaMKK2–AMPKγ2 axis, a novel adaptation strategy for cell survival under nutrient deprivation conditions.
- Published
- 2021
14. YAP regulates an SGK1/mTORC1/SREBP-dependent lipogenic program to support proliferation and tissue growth
- Author
-
Srimayee Vaidyanathan, Talhah M. Salmi, Rasan M. Sathiqu, Malcolm J. McConville, Andrew G. Cox, and Kristin K. Brown
- Subjects
Sterol Regulatory Element Binding Proteins ,Lipogenesis ,Cell Biology ,Mechanistic Target of Rapamycin Complex 1 ,Sterol Regulatory Element Binding Protein 1 ,Molecular Biology ,General Biochemistry, Genetics and Molecular Biology ,Developmental Biology ,Cell Proliferation - Abstract
The coordinated regulation of growth control and metabolic pathways is required to meet the energetic and biosynthetic demands associated with proliferation. Emerging evidence suggests that the Hippo pathway effector Yes-associated protein 1 (YAP) reprograms cellular metabolism to meet the anabolic demands of growth, although the mechanisms involved are poorly understood. Here, we demonstrate that YAP co-opts the sterol regulatory element-binding protein (SREBP)-dependent lipogenic program to facilitate proliferation and tissue growth. Mechanistically, YAP stimulates de novo lipogenesis via mechanistic target of rapamcyin (mTOR) complex 1 (mTORC1) signaling and subsequent activation of SREBP. Importantly, YAP-dependent regulation of serum- and glucocorticoid-regulated kinase 1 (SGK1) is required to activate mTORC1/SREBP and stimulate de novo lipogenesis. We also find that the SREBP target genes fatty acid synthase (FASN) and stearoyl-CoA desaturase (SCD) are conditionally required to support YAP-dependent proliferation and tissue growth. These studies reveal that de novo lipogenesis is a metabolic vulnerability that can be targeted to disrupt YAP-dependent proliferation and tissue growth.
- Published
- 2021
15. Characterization of the Src-regulated kinome identifies SGK1 as a key mediator of Src-induced transformation
- Author
-
Elizabeth V. Nguyen, Christina Anne Mitchell, Cheng Huang, Jiangning Song, Rachel S. Lee, Howard Chan, Luxi Zhang, Ralf B. Schittenhelm, Kristin K. Brown, Samuel J. Rodgers, Roger J. Daly, Xiuquan Ma, Kaylene J. Simpson, Jianmin Wu, Chanly Chheang, and Fuyi Li
- Subjects
0301 basic medicine ,Proteomics ,Cell signaling ,Science ,General Physics and Astronomy ,Mice, Nude ,Antineoplastic Agents ,Triple Negative Breast Neoplasms ,02 engineering and technology ,Biology ,Protein Serine-Threonine Kinases ,General Biochemistry, Genetics and Molecular Biology ,Mass Spectrometry ,Article ,Immediate-Early Proteins ,03 medical and health sciences ,Cell Line, Tumor ,Protein Interaction Mapping ,Animals ,Humans ,Kinome ,Benzodioxoles ,RNA, Small Interfering ,lcsh:Science ,Mice, Inbred BALB C ,Multidisciplinary ,Oncogene ,Kinase ,HEK 293 cells ,General Chemistry ,Oncogenes ,021001 nanoscience & nanotechnology ,Xenograft Model Antitumor Assays ,Cell biology ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cell Transformation, Neoplastic ,HEK293 Cells ,src-Family Kinases ,Gene Knockdown Techniques ,Quinazolines ,Phosphorylation ,Female ,lcsh:Q ,0210 nano-technology ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
Despite significant progress, our understanding of how specific oncogenes transform cells is still limited and likely underestimates the complexity of downstream signalling events. To address this gap, we use mass spectrometry-based chemical proteomics to characterize the global impact of an oncogene on the expressed kinome, and then functionally annotate the regulated kinases. As an example, we identify 63 protein kinases exhibiting altered expression and/or phosphorylation in Src-transformed mammary epithelial cells. An integrated siRNA screen identifies nine kinases, including SGK1, as being essential for Src-induced transformation. Accordingly, we find that Src positively regulates SGK1 expression in triple negative breast cancer cells, which exhibit a prominent signalling network governed by Src family kinases. Furthermore, combined inhibition of Src and SGK1 reduces colony formation and xenograft growth more effectively than either treatment alone. Therefore, this approach not only provides mechanistic insights into oncogenic transformation but also aids the design of improved therapeutic strategies., The systemic understanding of oncogenic kinase signalling is still limited. Here, the authors combine chemical proteomics with functional screens to assess the impact of oncogenic Src on the expressed kinome and identify SGK1 as a critical mediator of Src-induced cell transformation.
- Published
- 2019
16. Reprogramming of serine metabolism is an actionable vulnerability in FLT3-ITD driven acute myeloid leukaemia
- Author
-
Lev Kats, Thomas Abrehart, Giovanna Pomilio, Carolyn Shembrey, Ricky W. Johnstone, Stephin J. Vervoort, Gareth P. Gregory, Andrew H. Wei, Izabela Todorovski, Stefan Bjelosevic, Andrea Newbold, Kristin K. Brown, and Emily Gruber
- Subjects
FLT3 Internal Tandem Duplication ,Purine ,Myeloid ,DNA damage ,hemic and immune systems ,Biology ,Pediatric cancer ,Serine ,chemistry.chemical_compound ,fluids and secretions ,medicine.anatomical_structure ,chemistry ,hemic and lymphatic diseases ,embryonic structures ,Cancer research ,Cytarabine ,medicine ,Phosphoglycerate dehydrogenase ,medicine.drug - Abstract
Activating FMS-like tyrosine kinase 3 (FLT3) mutations occur in approximately 30% of all acute myeloid leukaemias (AMLs) and are associated with poor prognosis. The limited clinical efficacy of FLT3 inhibitor monotherapy has highlighted the need for alternative therapeutic targets and treatments for FLT3-mutant AML. Using human and murine models of MLL-rearranged AML harbouring FLT3 internal tandem duplication (FLT3-ITD) and primary patient samples, we have demonstrated that FLT3-ITD promotes serine uptake and serine synthesis via transcriptional regulation of neutral amino acid transporters (SLC1A4 and SLC1A5) and genes in the de novo serine synthesis pathway (PHGDH and PSAT1). Mechanistically, dysregulation of serine metabolism in FLT3-mutant AML is dependent on the mTORC1-ATF4 axis, that drives RNA-Pol II occupancy at PHGDH, PSAT1, SLC1A4 and SLC1A5. Genetic or pharmacological inhibition of the de novo serine synthesis pathway selectively inhibited the proliferation of FLT3-ITD AML cells, and this was potentiated by withdrawal of exogenous serine. Purine supplementation effectively rescued the antiproliferative effect of inhibiting de novo serine synthesis, consistent with the idea that serine fuels purine nucleotide synthesis in FLT3-mutant AML. Pharmacological inhibition of the de novo serine synthesis pathway, using the PHGDH inhibitor WQ-2101, sensitises FLT3-mutant AML cells to the standard of care chemotherapy agent cytarabine via exacerbation of DNA damage. Collectively, these data reveal new insights as to how FLT3 mutations reprogram metabolism in AML, and reveal a combination therapy strategy to improve the treatment of FLT3-mutant AML.Statement of SignificanceFLT3 mutations are common in AML and are associated with poor prognosis. We show that FLT3-ITD stimulates serine metabolism, thereby rendering FLT3-ITD leukemias dependent on serine for proliferation and survival. This metabolic dependency can be exploited pharmacologically to sensitize FLT3-mutant AML to chemotherapy.
- Published
- 2020
17. Biocatalytic Synthesis of Chiral N‐Functionalized Amino Acids
- Author
-
Sarah L. Lovelock, Peter W. Sutton, Kristin K. Brown, Gheorghe-Doru Roiban, Julia F. Hyslop, Allan J. B. Watson, and University of St Andrews. School of Chemistry
- Subjects
biocatalysis ,Chemistry(all) ,Alkylation ,Bioactive molecules ,NDAS ,010402 general chemistry ,reductive amination ,01 natural sciences ,Reductive amination ,Catalysis ,α-keto amino acids ,Pseudomonas ,Animals ,Humans ,QD ,ketimine reductases ,Amino Acids ,Chromatography, High Pressure Liquid ,chemistry.chemical_classification ,N-methyl amino acid dehydrogenases ,010405 organic chemistry ,Chemistry ,N-methyl amino acid deyhydrogenases ,Sustainable manufacturing ,Enantioselective synthesis ,Stereoisomerism ,General Chemistry ,Ketones ,QD Chemistry ,Combinatorial chemistry ,0104 chemical sciences ,Amino acid ,Biocatalysis ,Yield (chemistry) ,SDG 9 - Industry, Innovation, and Infrastructure - Abstract
N-functionalized amino acids are important building blocks for the preparation of diverse bioactive molecules including peptides. The development of sustainable manufacturing routes to chiral N-alkylated amino acids remains a significant challenge in the pharmaceutical and fine chemical industries. Herein we report the discovery of a structurally diverse panel of biocatalysts which catalyze the asymmetric synthesis of N-alkyl amino acids via the reductive coupling of ketones and amines. Reactions have been performed on a gram scale to yield optically pure N-alkyl functionalized products in high yields. Postprint
- Published
- 2018
18. Biocatalytic Synthesis of Chiral N‐Functionalized Amino Acids
- Author
-
Julia F. Hyslop, Sarah L. Lovelock, Peter W. Sutton, Kristin K. Brown, Allan J. B. Watson, and Gheorghe‐Doru Roiban
- Subjects
010405 organic chemistry ,General Medicine ,010402 general chemistry ,01 natural sciences ,0104 chemical sciences - Published
- 2018
19. Pharmacological Reduction of Mitochondrial Iron in AML Triggers a BAX/BAK Dependent Non-Canonical Cell Death Synergistic with Venetoclax
- Author
-
Kevin Tran, Tatiana Cañeque, Giovanna Pomilio, Kristin K. Brown, Yih-Chih Chan, Andrew G. Cox, Kate McArthur, Veronique Litalien, Sebastian Müller, Estelle Duprez, Mark A. Dawson, Sylvain Garciaz, Brian Liddicoat, Georg Ramm, Kah-Lok Chan, Raphaël Rodriguez, Marian L. Burr, Enid Y.N. Lam, Laura MacPherson, Fiona C. Brown, Sarah-Jane Dawson, Andrew A Guirguis, David C.S. Huang, Mathilde Poplineau, and Andrew H. Wei
- Subjects
Reduction (complexity) ,Programmed cell death ,chemistry.chemical_compound ,Non canonical ,Chemistry ,Venetoclax ,Immunology ,Cancer research ,Cell Biology ,Hematology ,Biochemistry - Abstract
Although the BCL2 inhibitor venetoclax have been transformative in the management of AML, therapeutic resistance and relapse are frequently observed. In light of the urgent need to uncover novel therapeutic options in AML, we sought to study the potential role of ironomycin (AM5), a recently described small molecule that induces cell death through the sequestration of lysosomal iron. To evaluate the effects of ironomycin in AML, we chose a diverse panel of AML cell lines. These data showed a potent and dose-dependent effect, on proliferation, cell cycle progression and survival at a nanomolar range. In contrast to venetoclax, the cell death induced by ironomycin did not result in potent caspase activation or PARP1 cleavage. Neither the caspase inhibitor Z-VAD-fmk nor the necroptosis inhibitor necrostatin-1 did prevent cell death. Consistent with previous observations, we found that ironomycin accumulates in the lysosomes of AML cells leading to a sequestration of iron in this organelle but inhibitors of canonical ferroptosis, including ferrostatin-1 and liproxstatin-1 failed to prevent the activity of ironomycin. To gain greater insight into the molecular mechanism of ironomycin in AML cells, we performed a genome-wide positive-selection resistance screen under ironomycin selection pressure and collected several samples for sequencing. We found nine genes whose knock out conferred resistance to the drug. Interestingly, these data implicated key components of mitochondrial metabolic pathways, including phosphoglycolate phosphatase (PGP), a central phosphatase involved in glycolysis and pentose phosphate pathway (PPP) regulation and Hexokinase 2 (HK2), the first enzyme of glycolysis. Mass-spectrometry metabolomics analyses highlighted that ironomycin treatment significantly reduced key components of the TCA cycle and consequently the reducing agent nicotinamide adenine dinucleotide (NADH) and increased the intracellular concentration of amino acids. These data were corroborated with RNAseq showing a mitochondrial stress response mediated through the Activating Transcription Factor 4 (ATF4) and its paralog Activating Transcription Factor 5 (ATF5). As mitochondria are major hubs of iron utilization for oxidative respiration, we used Mass-spectrometry to measure mitochondrial iron load. We observed a rapid and dose-dependent decrease in mitochondrial iron after treatment mirroring the iron sequestration into the lysosomes and inducing the mitochondrial dysfunction. We next examined the ultrastructural appearance of mitochondria after ironomycin using transmission electron microscopy and observed a dramatic alteration of the structural integrity of mitochondria resulting in abnormal cristae, matrix density changes and mitochondrial membrane blebbing. In cells lacking BAX and BAK, the two main effectors of mitochondrial membrane permeabilization, structural changes and cell death were almost completely rescued but cell proliferation was still markedly affected, consistent with a BAX/BAK dependent cell death following mitochondrial iron deprivation. In vivo imaging confirmed that BAX activation occurred after 30 hours of treatment and preceded cell death, but we observed some major differences with canonical apoptosis induced by venetoclax. First, the structural alterations were clearly distinct. Next, delay between MOMP and cell death was significantly longer and caspase inhibitors weakly delayed cell death. Finally, BCL2 overexpression and P53 deletion did not rescue ironomycin cell death. These non-canonical features prompted us to assess the efficacy of the combination between ironomycin and venetoclax. In vitro experiment on AML cell lines found a high synergy between the two drugs. In vivo experiments on xenotransplanted mice confirmed the efficacy of the combination, which was associated with a significant increase in mice survival in comparison with the controls (Figure). Finally, primary AML samples from patients clinically resistant or refractory to venetoclax were sensitive to ironomycin in monotherapy and even more in combination with venetoclax. These results demonstrate that the novel mechanism of ironomycin action can be leveraged to resensitize AML cells to venetoclax and substitute for cytotoxic drugs as a more effective therapeutic combination in the salvage setting. Figure 1 Figure 1. Disclosures Huang: The Walter and Eliza Hall Institute of Medical Research: Patents & Royalties: Employee of the Walter and Eliza Hall Institute and eligilble for payments in relation to venetoclax. Wei: Novartis, Celgene, AbbVie, Servier, AstraZeneca, and Amgen: Research Funding; Novartis, Janssen, Amgen, Roche, Pfizer, Abbvie, Servier, BMS, Macrogenics, Agios, Gilead: Membership on an entity's Board of Directors or advisory committees; Astellas: Honoraria.
- Published
- 2021
20. Adaptive Reprogramming of De Novo Pyrimidine Synthesis Is a Metabolic Vulnerability in Triple-Negative Breast Cancer
- Author
-
John M. Asara, Kristin K. Brown, Jessica B. Spinelli, and Alex Toker
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Chemotherapy ,DNA damage ,medicine.medical_treatment ,Pharmacology ,Biology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,Oncology ,chemistry ,Pyrimidine metabolism ,medicine ,Neoplasm ,Doxorubicin ,Nucleotide ,Triple-negative breast cancer ,medicine.drug ,Leflunomide - Abstract
Chemotherapy resistance is a major barrier to the treatment of triple-negative breast cancer (TNBC), and strategies to circumvent resistance are required. Using in vitro and in vivo metabolic profiling of TNBC cells, we show that an increase in the abundance of pyrimidine nucleotides occurs in response to chemotherapy exposure. Mechanistically, elevation of pyrimidine nucleotides induced by chemotherapy is dependent on increased activity of the de novo pyrimidine synthesis pathway. Pharmacologic inhibition of de novo pyrimidine synthesis sensitizes TNBC cells to genotoxic chemotherapy agents by exacerbating DNA damage. Moreover, combined treatment with doxorubicin and leflunomide, a clinically approved inhibitor of the de novo pyrimidine synthesis pathway, induces regression of TNBC xenografts. Thus, the increase in pyrimidine nucleotide levels observed following chemotherapy exposure represents a metabolic vulnerability that can be exploited to enhance the efficacy of chemotherapy for the treatment of TNBC. Significance: The prognosis for patients with TNBC with residual disease after chemotherapy is poor. We find that chemotherapy agents induce adaptive reprogramming of de novo pyrimidine synthesis and show that this response can be exploited pharmacologically, using clinically approved inhibitors of de novo pyrimidine synthesis, to sensitize TNBC cells to chemotherapy. Cancer Discov; 7(4); 391–9. ©2017 AACR. See related article by Mathur et al., p. 380. This article is highlighted in the In This Issue feature, p. 339
- Published
- 2017
21. 3002 – REPROGRAMMING OF SERINE METABOLISM IS A METABOLIC VULNERABILITY IN FMS-LIKE TYROSINE KINASE 3 (FLT3) MUTANT ACUTE MYELOID LEUKAEMIA
- Author
-
Gareth P. Gregory, Carolyn Shembrey, Izabela Todorovski, Ricky W. Johnstone, Thomas Abrehart, Giovanna Pomilio, Kristin K. Brown, Emily Gruber, Andrew H. Wei, Stefan Bjelosevic, Andrea Newbold, and Stephin J. Vervoort
- Subjects
FLT3 Internal Tandem Duplication ,Cancer Research ,Myeloid ,DNA damage ,hemic and immune systems ,Context (language use) ,Cell Biology ,Hematology ,Biology ,Serine ,fluids and secretions ,medicine.anatomical_structure ,hemic and lymphatic diseases ,embryonic structures ,Fms-Like Tyrosine Kinase 3 ,Genetics ,Cancer research ,Cytarabine ,medicine ,Molecular Biology ,Reprogramming ,psychological phenomena and processes ,medicine.drug - Abstract
Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene occur in approximately 30% of all acute myeloid leukaemias (AMLs) and are associated with poor prognosis. The clinical utility of FLT3 inhibitor monotherapy has been limited by the rapid development of resistance, highlighting the need for identification of alternative therapeutic targets for the treatment of FLT3-mutant AML. Using a syngeneic murine model of AML harbouring FLT3 internal tandem duplication (FLT3-ITD), we demonstrate that FLT3-ITD promotes serine uptake and serine synthesis via transcriptional regulation of neutral amino acid transporters (SLC1A4 and SLC1A5) and genes in the de novo serine synthesis pathway (PHGDH and PSAT1). Mechanistically, dysregulation of serine metabolism was dependent on the mTORC1-ATF4 axis, which promoted RNA-Pol II occupancy at PHGDH, PSAT1, SLC1A4 and SLC1A5 in AML cells harbouring FLT3-ITD mutations. Genetic or pharmacological inhibition of the de novo serine synthesis pathway selectively inhibited the proliferation and viability of FLT3-ITD AML cells, and purine supplementation effectively rescued the apoptotic effects of inhibiting FLT3-ITD, consistent with the hypothesis that serine fuels purine nucleotide synthesis in FLT3-ITD AML cells. To exploit these findings in the context of standard-of-care therapy in AML, we show that pharmacological inhibition of the de novo serine synthesis pathway, using the PHGDH inhibitor WQ-2101, sensitises FLT3-ITD AML cells to the chemotherapy agent cytarabine by greatly exacerbating the DNA damage response in AML cell lines harbouring FLT3-ITD mutations, primary AML patient samples, and an aggressive PDX mouse model of FLT3-ITD-driven AML. Collectively, these data reveal new insights into FLT3-ITD-induced metabolic reprogramming in AML, and suggest a novel combinatorial therapeutic strategy for the treatment of FLT3-mutant AML.
- Published
- 2020
22. Abstract LB-015: Yap reprograms de novo lipogenesis to fuel liver cancer
- Author
-
Andrew G. Cox, Srimayee Vaidyanathan, Talhah M. Salmi, Kristin K. Brown, and Malcolm J. McConville
- Subjects
Cancer Research ,Hippo signaling pathway ,Cancer ,Lipid metabolism ,Biology ,medicine.disease ,medicine.disease_cause ,Oncology ,Lipid droplet ,Lipogenesis ,medicine ,Cancer research ,Carcinogenesis ,Liver cancer ,Transcription factor - Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer, originating from liver cells known as hepatocytes. At the molecular level, HCC is driven by transcription factors which are able to reprogram metabolism to support tumorigenesis that are still poorly understood. The Yes-associated protein (Yap) is the nuclear effector of the Hippo pathway, responsible in regulating organ size control and metabolism. Metabolic reprogramming has recently emerged as a fundamental hallmark of cancer. In our previous studies, we found that Yap integrates the anabolic demands of tumour growth by reprogramming glutamine and glucose metabolism to support nucleotide biosynthesis. The central aim of this study was to determine the role that Yap plays in regulating lipid metabolism in the context of liver cancer. We took advantage of a larval zebrafish model in which a hyperactivated form of Yap is specifically expressed in hepatocytes (lf:YapS87A;lf:NLS-mcherry). We found that the expression of Yap was sufficient to stimulate de novo lipogenesis (DNL) and induce lipid droplet formation in hepatocytes (steatosis). To determine whether the stimulation of DNL was required for oncogenic growth, we exposed larvae to pharmacological inhibitors of DNL and examined the impact on growth at the cellular level by multiphoton microscopy. Strikingly, we identified that fatty acid synthase (FAS) and stearoyl-CoA desaturase (SCD) inhibitors suppressed Yap-driven growth. To complement these studies, we took a genetic approach using CRISPR to generate FAS and SCD KO zebrafish larvae and we found that Yap-driven growth required both FAS and SCD. Importantly both chemical and genetic suppression of FAS and SCD activity had no effect on normal liver growth. Together, these findings suggest that oncogenic Yap-driven growth is conditionally dependent upon the stimulation of DNL. Consequently, these studies provide a rationale for examining the clinical efficacy of DNL inhibitors to combat liver cancer. Citation Format: Talhah M. Salmi, Srimayee Vaidyanathan, Malcolm J. McConville, Kristin K. Brown, Andrew G. Cox. Yap reprograms de novo lipogenesis to fuel liver cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr LB-015.
- Published
- 2020
23. Yap regulates glucose utilization and sustains nucleotide synthesis to enable organ growth
- Author
-
Matthew L. Steinhauser, Dean Yimlamai, Wolfram Goessling, Evan C. Lien, Mark R. Sullivan, Allison Tsomides, Joel B. Miesfeld, Brian A. Link, Erin Snay, John M. Asara, Katie L. Hwang, Michael Fort, Sagar Chhangawala, Matthew G. Vander Heiden, Kimberly Y Ma, Fernando D. Camargo, Yariv Houvras, Aaron M. Hosios, Min Yuan, Andrew G. Cox, Kristin K. Brown, Giorgio G. Galli, Brendan H. Fowl, and Koch Institute for Integrative Cancer Research at MIT
- Subjects
0301 basic medicine ,Glucose uptake ,Carbohydrate metabolism ,Protein Serine-Threonine Kinases ,Serine-Threonine Kinase 3 ,General Biochemistry, Genetics and Molecular Biology ,Impaired glucose tolerance ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Molecular Biology ,Zebrafish ,Tissue homeostasis ,030304 developmental biology ,0303 health sciences ,Hippo signaling pathway ,Glucose Transporter Type 1 ,General Immunology and Microbiology ,biology ,Effector ,Nucleotides ,General Neuroscience ,Glucose transporter ,YAP-Signaling Proteins ,Articles ,Zebrafish Proteins ,biology.organism_classification ,medicine.disease ,3. Good health ,Cell biology ,030104 developmental biology ,Glucose ,Liver ,Hippo signaling ,030220 oncology & carcinogenesis ,biology.protein ,Trans-Activators ,GLUT1 ,Signal Transduction - Abstract
The Hippo pathway and its nuclear effector Yap regulate organ size and cancer formation. While many modulators of Hippo activity have been identified, little is known about the Yap target genes that mediate these growth effects. Here, we show that yap−/− mutant zebrafish exhibit defects in hepatic progenitor potential and liver growth due to impaired glucose transport and nucleotide biosynthesis. Transcriptomic and metabolomic analyses reveal that Yap regulates expression of glucose transporter glut1, causing decreased glucose uptake and use for nucleotide biosynthesis in yap−/− mutants, and impaired glucose tolerance in adults. Nucleotide supplementation improves Yap deficiency phenotypes, indicating functional importance of glucose-fueled nucleotide biosynthesis. Yap-regulated glut1 expression and glucose uptake are conserved in mammals, suggesting that stimulation of anabolic glucose metabolism is an evolutionarily conserved mechanism by which the Hippo pathway controls organ growth. Together, our results reveal a central role for Hippo signaling in glucose metabolic homeostasis., National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (Grant P30DK034854), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (Grant 1R01DK090311), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (Grant R24OD017870), National Institutes of Health (U.S.) (Grant P30DK034854), National Institutes of Health (U.S.) (Grant 1R01DK090311), National Institutes of Health (U.S.) (Grant 1R01DK105198), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (Grant 1R01DK105198), National Institutes of Health (U.S.) (Grant R24OD017870), National Institute of General Medical Sciences (NIGMS) (Grant T32GM007753), NHMRC (Grant 1146558), National Cancer Institute (U.S.) (Grant 5P01CA120964), National Cancer Institute (U.S.) (Grant 5P30CA006516)
- Published
- 2018
24. Approaches to target tractability assessment - a practical perspective
- Author
-
Michael M. Hann, Rita Santos, Pamela Thomas, Kristin K. Brown, Kieran Todd, and Ami S. Lakdawala
- Subjects
0301 basic medicine ,Pharmacology ,Modalities ,Computer science ,Drug discovery ,In silico ,Organic Chemistry ,Perspective (graphical) ,MEDLINE ,Pharmaceutical Science ,Biochemistry ,Data science ,03 medical and health sciences ,Chemistry ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Intervention (counseling) ,Drug Discovery ,Molecular Medicine - Abstract
The assessment of the suitability of novel targets to intervention by different modalities, e.g. small molecules or antibodies, is increasingly seen as important in helping to select the most progressable targets at the outset of a drug discovery project. This perspective considers differing aspects of tractability and how it can be assessed using in silico and experimental approaches. We also share some of our experiences in using these approaches.
- Published
- 2017
25. S-Nitrosothiol Signaling Regulates Liver Development and Improves Outcome following Toxic Liver Injury
- Author
-
Yevgenia Tesmenitsky, Kristin K. Brown, Gary J. Rosenthal, Dorothy B. Colagiovanni, Jonathan S. Stamler, Trista E. North, Kevin Croce, Andrew G. Cox, Diane C. Saunders, Allie A. Conway, Julio Flávio Meirelles Marchini, Peter B. Kelsey, and Wolfram Goessling
- Subjects
Liver injury ,Programmed cell death ,Cell growth ,Pharmacology ,Biology ,biology.organism_classification ,medicine.disease ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Acetaminophen ,Nitric oxide ,Toxicology ,Nitric oxide synthase ,chemistry.chemical_compound ,lcsh:Biology (General) ,chemistry ,medicine ,biology.protein ,lcsh:QH301-705.5 ,Zebrafish ,Oxidative stress ,medicine.drug - Abstract
Summary Toxic liver injury is a leading cause of liver failure and death because of the organ's inability to regenerate amidst massive cell death, and few therapeutic options exist. The mechanisms coordinating damage protection and repair are poorly understood. Here, we show that S-nitrosothiols regulate liver growth during development and after injury in vivo; in zebrafish, nitric-oxide (NO) enhanced liver formation independently of cGMP-mediated vasoactive effects. After acetaminophen (APAP) exposure, inhibition of the enzymatic regulator S-nitrosoglutathione reductase (GSNOR) minimized toxic liver damage, increased cell proliferation, and improved survival through sustained activation of the cytoprotective Nrf2 pathway. Preclinical studies of APAP injury in GSNOR-deficient mice confirmed conservation of hepatoprotective properties of S-nitrosothiol signaling across vertebrates; a GSNOR-specific inhibitor improved liver histology and acted with the approved therapy N-acetylcysteine to expand the therapeutic time window and improve outcome. These studies demonstrate that GSNOR inhibitors will be beneficial therapeutic candidates for treating liver injury.
- Published
- 2014
26. Selenoprotein H is an essential regulator of redox homeostasis that cooperates with p53 in development and tumorigenesis
- Author
-
John M. Asara, Byung Cheon Lee, Andrew G. Cox, Kimberley J. Evason, Evan C. Lien, Sagar Chhangawala, Jerry R. Heidel, Bryan C. Dickinson, Yariv Houvras, Diane C. Saunders, Christopher J. Chang, Kristin K. Brown, Min Yuan, Allison Tsomides, Wolfram Goessling, Katie L. Hwang, Vadim N. Gladyshev, Andrew J. Kim, and Sahar Nissim
- Subjects
0301 basic medicine ,Male ,p53 ,DNA damage ,Carcinogenesis ,Biology ,medicine.disease_cause ,Transcriptome ,liver cancer ,03 medical and health sciences ,chemistry.chemical_compound ,Selenium ,Genetics ,medicine ,2.1 Biological and endogenous factors ,Animals ,Humans ,Homeostasis ,Aetiology ,Selenoproteins ,Zebrafish ,Cancer ,Nutrition ,Gastrointestinal Neoplasms ,Regulation of gene expression ,Neoplastic ,Multidisciplinary ,Methionine ,endoderm development ,Methionine sulfoxide ,Prevention ,Gene Expression Regulation, Neoplastic ,DNA-Binding Proteins ,Oxidative Stress ,030104 developmental biology ,PNAS Plus ,Mitochondrial biogenesis ,chemistry ,Biochemistry ,Gene Expression Regulation ,selenoproteins ,Female ,Tumor Suppressor Protein p53 ,Oxidation-Reduction ,Oxidative stress ,DNA Damage - Abstract
© 2016, National Academy of Sciences. All rights reserved. Selenium, an essential micronutrient known for its cancer prevention properties, is incorporated into a class of selenocysteine-containing proteins (selenoproteins). Selenoprotein H (SepH) is a recently identified nucleolar oxidoreductase whose function is not well understood. Here we report that seph is an essential gene regulating organ development in zebrafish. Metabolite profiling by targeted LC-MS/MS demonstrated that SepH deficiency impairs redox balance by reducing the levels of ascorbate and methionine, while increasing methionine sulfoxide. Transcriptome analysis revealed that SepH deficiency induces an inflammatory response and activates the p53 pathway. Consequently, loss of seph renders larvae susceptible to oxidative stress and DNA damage. Finally, we demonstrate that seph interacts with p53 deficiency in adulthood to accelerate gastrointestinal tumor development. Overall, our findings establish that seph regulates redox homeostasis and suppresses DNA damage. We hypothesize that SepH deficiency may contribute to the increased cancer risk observed in cohorts with low selenium levels.
- Published
- 2016
27. Sequence analysis of mutations and translocations across breast cancer subtypes
- Author
-
Alex Toker, Abbie M. Frederick, Alex H. Ramos, Kristian Cibulskis, Nam Pho, Verónica Bautista-Piña, Valeria Quintanar-Jurado, Eric S. Lander, Kornelia Polyak, Claudia Rangel-Escareño, Sergio Rodriguez-Cuevas, José Baselga, Kristin K. Brown, Sandra Romero-Cordoba, Antonio Maffuz-Aziz, Shouyong Peng, Jorge Melendez-Zajgla, Rameen Beroukhim, Michael S. Lawrence, Alfredo Hidalgo-Miranda, Stacey Gabriel, Dennis C. Sgroi, Gerardo Jimenez-Sanchez, Andrea L. Richardson, Daniel Auclair, Gad Getz, Nicolas Stransky, Andrey Sivachenko, Rosa Rebollar-Vega, Fujiko Duke, Melissa Parkin, Todd R. Golub, Levi A. Garraway, Juan Carlos Fernández-López, Maria L. Cortes, Lihua Zou, Joonil Jung, Robert C. Onofrio, Laura Uribe-Figueroa, Kristin G. Ardlie, Kristin Thompson, Shantanu Banerji, Scott L. Carter, Joshua M. Francis, Matthew Meyerson, Carrie Sougnez, and Steven E. Schumacher
- Subjects
DNA Copy Number Variations ,DNA Mutational Analysis ,Breast Neoplasms ,MAP3K1 ,Biology ,medicine.disease_cause ,Core Binding Factor beta Subunit ,Article ,Translocation, Genetic ,Evolution, Molecular ,Fusion gene ,Aromatase ,Breast cancer ,medicine ,Humans ,Exome ,skin and connective tissue diseases ,Mexico ,Gene ,Genetics ,Mutation ,Multidisciplinary ,Aromatase Inhibitors ,Genome, Human ,Gene Expression Profiling ,Membrane Proteins ,Cancer ,Oncogenes ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Cell Transformation, Neoplastic ,Vietnam ,Mutagenesis ,Core Binding Factor Alpha 2 Subunit ,Female ,Gene Fusion ,Breast carcinoma ,Proto-Oncogene Proteins c-akt ,Algorithms - Abstract
Breast carcinoma is the leading cause of cancer-related mortality in women worldwide with an estimated 1.38 million new cases and 458,000 deaths in 2008 alone1. This malignancy represents a heterogeneous group of tumours with characteristic molecular features, prognosis, and responses to available therapy2–4. Recurrent somatic alterations in breast cancer have been described including mutations and copy number alterations, notably ERBB2 amplifications, the first successful therapy target defined by a genomic aberration5. Prior DNA sequencing studies of breast cancer genomes have revealed additional candidate mutations and gene rearrangements 6–10. Here we report the whole-exome sequences of DNA from 103 human breast cancers of diverse subtypes from patients in Mexico and Vietnam compared to matched-normal DNA, together with whole-genome sequences of 22 breast cancer/normal pairs. Beyond confirming recurrent somatic mutations in PIK3CA11, TP536, AKT112, GATA313, and MAP3K110, we discovered recurrent mutations in the CBFB transcription factor gene and deletions of its partner RUNX1. Furthermore, we have identified a recurrent MAGI3-AKT3 fusion enriched in triple-negative breast cancer lacking estrogen and progesterone receptors and ERBB2 expression. The Magi3-Akt3 fusion leads to constitutive activation of Akt kinase, which is abolished by treatment with an ATP-competitive Akt small-molecule inhibitor.
- Published
- 2012
28. Direct Modification of the Proinflammatory Cytokine Macrophage Migration Inhibitory Factor by Dietary Isothiocyanates
- Author
-
Mark B. Hampton, Jürgen Bernhagen, Kristin K. Brown, Christine C. Winterbourn, Frances H. Blaikie, Joel D. A. Tyndall, Hongqi Lue, and Robin A.J. Smith
- Subjects
Phenethyl isothiocyanate ,Protein Conformation ,medicine.drug_class ,medicine.medical_treatment ,Molecular Sequence Data ,Biology ,Monoclonal antibody ,Models, Biological ,Biochemistry ,Proinflammatory cytokine ,Jurkat Cells ,chemistry.chemical_compound ,Affinity chromatography ,Isothiocyanates ,medicine ,Humans ,Amino Acid Sequence ,Receptor ,Macrophage Migration-Inhibitory Factors ,Molecular Biology ,Inflammation ,Dose-Response Relationship, Drug ,Protein Synthesis, Post-Translational Modification, and Degradation ,Cell Membrane ,Antibodies, Monoclonal ,Cell Biology ,In vitro ,Cytokine ,Models, Chemical ,chemistry ,Mutagenesis, Site-Directed ,Cytokines ,Macrophage migration inhibitory factor - Abstract
Isothiocyanates are a class of phytochemicals with widely reported anti-cancer and anti-inflammatory activity. However, knowledge of their activity at a molecular level is limited. The objective of this study was to identify biological targets of phenethyl isothiocyanate (PEITC) using an affinity purification approach. An analogue of PEITC was synthesized to enable conjugation to a solid-phase resin. The pleiotropic cytokine macrophage migration inhibitory factor (MIF) was the major protein captured from cell lysates. Site-directed mutagenesis and mass spectrometry showed that PEITC covalently modified the N-terminal proline residue of MIF. This resulted in complete loss of catalytic tautomerase activity and disruption of protein conformation, as determined by impaired recognition by a monoclonal antibody directed to the region that receptors and interacting proteins bind to MIF. The conformational change was supported by in silico modeling. Monoclonal antibody binding to plasma MIF was disrupted in humans consuming watercress, a major dietary source of PEITC. The isothiocyanates have significant potential for development as MIF inhibitors, and this activity may contribute to the biological properties of these phytochemicals.
- Published
- 2009
29. Yap reprograms glutamine metabolism to increase nucleotide biosynthesis and enable liver growth
- Author
-
Sagar Chhangawala, Dean Yimlamai, Min Yuan, Sebastian Beltz, John M. Asara, Sahar Nissim, Akihiro Minami, Fernando D. Camargo, Julia Wucherpfennig, Giorgio G. Galli, Yariv Houvras, Wolfram Goessling, Kimberley J. Evason, Andrew G. Cox, David E. Cohen, Evan C. Lien, Didier Y.R. Stainier, Katie L. Hwang, Kristin K. Brown, Keelin O’Connor, and Allison Tsomides
- Subjects
0301 basic medicine ,Carcinoma, Hepatocellular ,Glutamine ,Biology ,Article ,Transcriptome ,Animals, Genetically Modified ,03 medical and health sciences ,Animals ,Humans ,Nucleotide ,Zebrafish ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,YAP1 ,chemistry.chemical_classification ,Hippo signaling pathway ,Effector ,Liver Neoplasms ,YAP-Signaling Proteins ,Cell Biology ,Zebrafish Proteins ,biology.organism_classification ,Phosphoproteins ,Cell biology ,030104 developmental biology ,Cell Transformation, Neoplastic ,Biochemistry ,chemistry ,Liver ,Pyrimidine metabolism ,Trans-Activators ,Transcription Factors - Abstract
The Hippo pathway is an important regulator of organ size and tumorigenesis. It is unclear, however, how Hippo signalling provides the cellular building blocks required for rapid growth. Here, we demonstrate that transgenic zebrafish expressing an activated form of the Hippo pathway effector Yap1 (also known as YAP) develop enlarged livers and are prone to liver tumour formation. Transcriptomic and metabolomic profiling identify that Yap1 reprograms glutamine metabolism. Yap1 directly enhances glutamine synthetase (glul) expression and activity, elevating steady-state levels of glutamine and enhancing the relative isotopic enrichment of nitrogen during de novo purine and pyrimidine biosynthesis. Genetic or pharmacological inhibition of GLUL diminishes the isotopic enrichment of nitrogen into nucleotides, suppressing hepatomegaly and the growth of liver cancer cells. Consequently, Yap-driven liver growth is susceptible to nucleotide inhibition. Together, our findings demonstrate that Yap1 integrates the anabolic demands of tissue growth during development and tumorigenesis by reprogramming nitrogen metabolism to stimulate nucleotide biosynthesis.
- Published
- 2015
30. The phosphoinositide 3-kinase pathway and therapy resistance in cancer
- Author
-
Kristin K. Brown and Alex Toker
- Subjects
Phosphoinositide 3-kinase ,biology ,Combination therapy ,business.industry ,Cancer ,General Medicine ,Review Article ,medicine.disease ,Bioinformatics ,medicine.disease_cause ,Review article ,biology.protein ,Cancer research ,Medicine ,business ,Carcinogenesis ,Protein kinase B ,Mechanistic target of rapamycin ,PI3K/AKT/mTOR pathway - Abstract
The phosphoinositide 3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) signaling network is a master regulator of processes that contribute to tumorigenesis and tumor maintenance. The PI3K pathway also plays a critical role in driving resistance to diverse anti-cancer therapies. This review article focuses on mechanisms by which the PI3K pathway contributes to therapy resistance in cancer, and highlights potential combination therapy strategies to circumvent resistance driven by PI3K signaling. In addition, resistance mechanisms that limit the clinical efficacy of small molecule inhibitors of the PI3K pathway are discussed.
- Published
- 2015
31. Autolytic proteolysis within the function to find domain (FIIND) is required for NLRP1 inflammasome activity
- Author
-
Peter J. Gough, Michael Cook, John D. Lich, Joshua N. Finger, Chaya Duraiswami, Kristin K. Brown, Lauren Dare, and John Bertin
- Subjects
Cell signaling ,Inflammasomes ,Proteolysis ,Immunology ,Regulator ,NLR Proteins ,macromolecular substances ,Biology ,Biochemistry ,Polymorphism, Single Nucleotide ,Protein structure ,medicine ,Humans ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Genetics ,medicine.diagnostic_test ,C-terminus ,HEK 293 cells ,Signal transducing adaptor protein ,Inflammasome ,Cell Biology ,Immunity, Innate ,Cell biology ,Protein Structure, Tertiary ,HEK293 Cells ,Additions and Corrections ,Apoptosis Regulatory Proteins ,Protein Processing, Post-Translational ,medicine.drug - Abstract
Nucleotide-binding domain leucine-rich repeat proteins (NLRs) play a key role in immunity and disease through their ability to modulate inflammation in response to pathogen-derived and endogenous danger signals. Here, we identify the requirements for activation of NLRP1, an NLR protein associated with a number of human pathologies, including vitiligo, rheumatoid arthritis, and Crohn disease. We demonstrate that NLRP1 activity is dependent upon ASC, which associates with the C-terminal CARD domain of NLRP1. In addition, we show that NLRP1 activity is dependent upon autolytic cleavage at Ser(1213) within the FIIND. Importantly, this post translational event is dependent upon the highly conserved distal residue His(1186). A disease-associated single nucleotide polymorphism near His(1186) and a naturally occurring mRNA splice variant lacking exon 14 differentially affect this autolytic processing and subsequent NLRP1 activity. These results describe key molecular pathways that regulate NLRP1 activity and offer insight on how small sequence variations in NLR genes may influence human disease pathogenesis.
- Published
- 2012
32. PKD controls αvβ3 integrin recycling and tumor cell invasive migration through its substrate Rabaptin-5
- Author
-
Kristin K. Brown, Jim C. Norman, Claudine Christoforides, Alex Toker, and Elena Rainero
- Subjects
Integrin ,Vesicular Transport Proteins ,CD49c ,General Biochemistry, Genetics and Molecular Biology ,Receptor tyrosine kinase ,Article ,Collagen receptor ,Mice ,Cell Movement ,Animals ,Humans ,Neoplasm Invasiveness ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Protein Kinase C ,Integrin alphaVbeta3 ,biology ,Rabaptin ,Cell Biology ,Cell biology ,HEK293 Cells ,Integrin alpha M ,biology.protein ,NIH 3T3 Cells ,Integrin, beta 6 ,Developmental Biology - Abstract
SummaryIntegrin recycling is critical for cell migration. Protein kinase D (PKD) mediates signals from the platelet-derived growth factor receptor (PDGF-R) to control αvβ3 integrin recycling. We now show that Rabaptin-5, a Rab5 effector in endosomal membrane fusion, is a PKD substrate. PKD phosphorylates Rabaptin-5 at Ser407, and this is both necessary and sufficient for PDGF-dependent short-loop recycling of αvβ3, which in turn inhibits α5β1 integrin recycling. Rab4, but not Rab5, interacts with phosphorylated Rabaptin-5 toward the front of migrating cells to promote delivery of αvβ3 to the leading edge, thereby driving persistent cell motility and invasion that is dependent on this integrin. Consistently, disruption of Rabaptin-5 Ser407 phosphorylation reduces persistent cell migration in 2D and αvβ3-dependent invasion. Conversely, invasive migration that is dependent on α5β1 integrin is promoted by disrupting Rabaptin phosphorylation. These findings demonstrate that the PKD pathway couples receptor tyrosine kinase signaling to an integrin switch via Rabaptin-5 phosphorylation.
- Published
- 2011
33. Abstract 5151: Yap reprograms glutamine metabolism and supports growth during liver development and tumorigenesis
- Author
-
Dean Yimlamai, Giorgio G. Galli, Wolfram Goessling, Andrew G. Cox, Sebastian Beltz, John M. Asara, Didier Y.R. Stainier, Sagar Chhangawala, Katie L. Hwang, Kimberley J. Evason, Keelin O’Connor, Evan C. Lien, Fernando D. Camargo, Yariv Houvras, and Kristin K. Brown
- Subjects
Cancer Research ,medicine.medical_specialty ,Hippo signaling pathway ,Effector ,Cell growth ,Cancer ,Biology ,medicine.disease_cause ,medicine.disease ,Glutamine ,Transcriptome ,Endocrinology ,Oncology ,Internal medicine ,medicine ,Cancer research ,Carcinogenesis ,Flux (metabolism) - Abstract
Hepatocellular carcinoma is a global health problem with poor prognosis and limited therapeutic options. Recently, the Hippo pathway has emerged as a master regulator of organ size control and tumorigenesis. However, the metabolic impact of the pathway is poorly understood. Using a transgenic zebrafish model with liver-specific activation of the Hippo pathway effector Yap, we have shown that Yap promotes hepatomegaly and liver dysplasia. In addition, we demonstrate that the Yap transgenics are highly susceptible to chemically-induced hepatocarcinogenesis. Transcriptomic and metabolomic profiling reveals that Yap enhances glutamine synthase (GS) expression and elevates steady-state levels of glutamine, respectively. Intervention studies with the GS inhibitor methionine sulfoximine established that elevated GS activity contributes to the rapid liver growth observed during Yap-driven hepatomegaly. Finally, studies in cultured human cancer cells identify GS as a bone-fide Yap target gene, confirming that the GS regulation by Yap is evolutionarily conserved. We conclude that Yap regulates GS expression and reprograms nitrogen metabolism, which contributes to liver growth during development and tumorigenesis. We hypothesize that Yap integrates the anabolic demands of rapid cell proliferation by increasing the flux of glutamine into nucleotide biosynthesis. Citation Format: Andrew G. Cox, Katie L. Hwang, Kimberley Evason, Kristin K. Brown, Sebastian Beltz, Keelin O'Connor, Giorgio G. Galli, Dean Yimlamai, Sagar Chhangawala, Evan Lien, Fernando D. Camargo, John Asara, Yariv Houvras, Didier Y. Stainier, Wolfram Goessling. Yap reprograms glutamine metabolism and supports growth during liver development and tumorigenesis. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5151. doi:10.1158/1538-7445.AM2015-5151
- Published
- 2015
34. Abstract 5477: MERIT40 is an Akt substrate that promotes resolution of DNA damage induced by chemotherapy
- Author
-
Kristin K. Brown, Laleh Montaser-Kouhsari, Alex Toker, and Andrew H. Beck
- Subjects
Cancer Research ,Chemotherapy ,Oncology ,Chemistry ,DNA damage ,medicine.medical_treatment ,medicine ,Cancer research ,Substrate (chemistry) ,Protein kinase B ,Molecular biology - Abstract
Resistance to cytotoxic chemotherapy agents is a common phenomenon in breast cancer and has a drastic impact on patient survival. Characterization of the mechanisms that contribute to chemotherapy resistance, and identification of strategies to circumvent resistance, would provide more effective therapies for the management of breast cancer. In this study we show that in addition to inflicting DNA damage, the cytotoxic chemotherapy agent doxorubicin triggers rapid and sustained activation of Akt in breast cancer cells. Pharmacological inhibitors of PI 3-K or Akt effectively block Akt signaling downstream of DNA damage and render cells markedly more sensitive to doxorubicin. We sought to identify mechanisms by which Akt promotes resistance to doxorubicin and have identified MERIT40, a component of the nuclear BRCA1-A DNA damage repair complex, as a novel Akt substrate that is phosphorylated when cells are exposed to doxorubicin. We have demonstrated that MERIT40 phosphorylation is necessary for BRCA1-A complex assembly and is required for resolution of DNA damage. We propose that inhibition of MERIT40 phosphorylation contributes to the efficacy of combination therapy with PI 3-K or Akt inhibitors and doxorubicin. We also show that MERIT40 phosphorylation is associated with ER status in breast tumor tissue samples, suggesting that MERIT40 phosphorylation could be used as a biomarker in ER-positive breast cancer patients to predict sensitivity to cytotoxic chemotherapy agents. Our data also indicates that patients with high levels of phosphorylated MERIT40 would likely benefit from combination therapy with Akt inhibitors and doxorubicin. We are currently investigating additional mechanisms by which Akt activation downstream of DNA damage promotes chemotherapy resistance with the aim of identifying novel combination therapy regimens to circumvent chemotherapy resistance. Citation Format: Kristin K. Brown, Laleh Montaser-Kouhsari, Andrew H. Beck, Alex Toker. MERIT40 is an Akt substrate that promotes resolution of DNA damage induced by chemotherapy. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5477. doi:10.1158/1538-7445.AM2015-5477
- Published
- 2015
35. Phenethyl isothiocyanate triggers apoptosis in Jurkat cells made resistant by the overexpression of Bcl-2
- Author
-
Mark B. Hampton, Kristin K. Brown, Juliet M. Pullar, and Susan Thomson
- Subjects
Cancer Research ,Phenethyl isothiocyanate ,biology ,Phenyl isothiocyanate ,T-Lymphocytes ,Apoptosis ,Phosphatidylserines ,Allyl isothiocyanate ,Molecular biology ,Jurkat cells ,Enzyme Activation ,chemistry.chemical_compound ,Enzyme activator ,Jurkat Cells ,Oncology ,Biochemistry ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,Isothiocyanates ,Caspases ,biology.protein ,Humans ,Cytotoxicity ,Caspase - Abstract
Isothiocyanates are a class of naturally occuring chemopreventive agents known to be effective at triggering apoptosis. In this study, we show that whereas overexpression of the oncoprotein Bcl-2 renders Jurkat T-lymphoma cells resistant to a range of cytotoxic agents, phenethyl isothiocyanate is able to overcome the inhibitory action of Bcl-2 and trigger apoptosis. A 50-fold increase in Bcl-2 expression shifted the dose-response curve, with an increase in the phenethyl isothiocyanate LD50 from 7 to 15 μmol/L, but there was still a complete loss in cell viability at doses in excess of 20 μmol/L. At these concentrations, cytotoxicity was strongly associated with caspase activation, phosphatidylserine exposure, and morphologic changes characteristic of apoptosis. Cytotoxicity was inhibited by treatment of the cells with a broad-spectrum caspase inhibitor. A structure-activity analysis showed that the phenethyl and benzyl isothiocyanates were most effective at triggering apoptosis in cells overexpressing Bcl-2 whereas phenyl isothiocyanate and benzyl thiocyanate had no proapoptotic activity. Allyl isothiocyanate also had limited efficacy despite its ability to trigger apoptosis in the parental Jurkat cell line. From this information, we propose that isothiocyanates modify a key cysteine residue in an apoptosis regulatory protein and that the aromatic side chain facilitates access to the target site. An in-depth investigation of the cellular targets of the aromatic isothiocyanates is warranted.(Cancer Res 2006; 66(13): 6772-7)
- Published
- 2006
36. Abstract B195: MAGI3-Akt3, a novel fusion protein in the PI3K pathway in cancer
- Author
-
Alex Toker and Kristin K. Brown
- Subjects
Cancer Research ,Cancer ,Biology ,medicine.disease ,Fusion protein ,AKT3 ,Oncology ,biology.protein ,Akt Inhibitor MK2206 ,Cancer research ,medicine ,PTEN ,Kinase activity ,Protein kinase B ,PI3K/AKT/mTOR pathway - Abstract
Aberrant activation of the PI3K/Akt pathway is found in approximately 70% of breast cancers. We recently identified a recurrent rearrangement that produces an in-frame fusion protein containing Akt3 and the scaffolding protein MAGI3. MAGI3-Akt3 is the first fusion protein that has been identified in the PI3K pathway. We propose that MAGI3-Akt3 is a novel oncogene in breast cancer and that the oncogenic activity of the fusion protein results from disruption of the PI3K/Akt pathway. Within the fusion, Akt3 has a disrupted PH domain prior to glutamate 17 (E17). As a result the fusion protein is constitutively phosphorylated in the Akt3 kinase domain and expression of MAGI3-Akt3 in breast cancer cells enhances Akt substrate phosphorylation. Constitutive MAGI3-Akt3 kinase activity can be inhibited by ATP-competitive small molecule Akt inhibitors but is resistant to the allosteric Akt inhibitor MK2206 which is currently in phase II clinical trials. Our results indicate that ATP competitive Akt inhibitors should be evaluated in clinical trials for the treatment of fusion-positive breast cancers. The fusion protein also contains a severely truncated portion of the scaffolding protein MAGI3. In particular, the fusion lacks the second PDZ domain of MAGI3 that has been reported to bind to PTEN and be required for the inhibitory effect of PTEN on PI3K pathway activity. We have demonstrated that disruption of MAGI3 in breast cancer cell lines does indeed promote PI3K pathway hyperactivity in a PTEN-dependent manner. In addition, the fusion lacks the fifth PDZ domain of MAGI3 and our studies indicate that this disrupts the interaction of MAGI3 with PHLPP2, a phosphatase that directly dephosphorylates Akt at pS473. Our studies therefore demonstrate that the MAGI3-Akt3 translocation significantly perturbs PI3K pathway activity via activation of an oncogene (Akt3) and dysregulation of two tumor suppressors (PTEN and PHLPP2). We are currently continuing studies to elucidate the oncogenic mechanism(s) of MAGI3-Akt3 in breast cancer. Citation Information: Mol Cancer Ther 2013;12(11 Suppl):B195. Citation Format: Kristin Brown, Alex Toker. MAGI3-Akt3, a novel fusion protein in the PI3K pathway in cancer. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2013 Oct 19-23; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2013;12(11 Suppl):Abstract nr B195.
- Published
- 2013
37. Abstract 5418: Rapid LDH5 inhibition reverses malignant metabolic phenotype and impairs survival of hepatocellular carcinoma cells
- Author
-
Richard Wooster, Kristin K. Brown, Deping Chai, Mariela Colón, Jennifer L. Ariazi, Chad Quinn, Sharon Sweitzer, Nino Campobasso, Subhas J. Chakravorty, Gregory M. Waitt, Tony Shaw, Kevin J. Duffy, Roland S. Annan, Jacques Briand, Nathan Gaul, Christian S. Sherk, Elizabeth A. Davenport, Jeanelle McSurdy-Freed, Kelvin Nurse, Anthony J. Jurewicz, Dean E. McNulty, Gilbert F. Scott, Angela Smallwood, James P. Villa, Hong Lu, Paru Nuthulaganti, Julia Billiard, Christopher S Dodson, Jessica L. Schneck, Lisa Miller, and Seth A. Gilbert
- Subjects
Cancer Research ,Metabolism ,Pentose phosphate pathway ,Biology ,PKM2 ,Molecular biology ,Citric acid cycle ,chemistry.chemical_compound ,Oncology ,chemistry ,Biochemistry ,Anaerobic glycolysis ,Lactate dehydrogenase ,Cancer cell ,Glycolysis - Abstract
Many cancer cells generate energy by rapidly converting glucose to lactate in the cytosol, a process termed aerobic glycolysis. This metabolic phenotype is recognized as one of the hallmarks of cancer and is enabled by lactate dehydrogenase (LDH), which catalyzes pyruvate to lactate inter-conversion. We find that hepatocellular carcinoma cells express micromolar quantities of LDH5 and that LDH5 protein down-regulation takes about 5 days allowing time for the cells to adapt their metabolism. Since metabolic processes happen in minutes, addressing consequences of LDH5 inhibition by protein down-regulation is inadequate. We screened the GSK compound library and identified a series of quinoline acids as NADH-competitive LDH5 inhibitors. Subsequent lead optimization yielded molecules with LDH5 inhibitory potencies as low as 2-3 nM and selectivity over LDH1 on the order of 10-100-fold. These molecules were cell-permeable and did not have any appreciable activity against a panel of approximately fifty common enzymes, receptors and ion channels, making them the most potent and selective LDH5 inhibitors identified to date. Using these tool inhibitors, we find that rapid chemical inhibition of LDH5 in Snu398 hepatocellular carcinoma cells results in profound inhibition of lactate production and increase in pyruvate as measured by mass spectrometric analysis. Real-time analysis by NMR spectroscopy of live Snu398 cells fed with 13C-labeled glucose demonstrated that chemical LDH5 inhibition led to a rapid decrease in glucose uptake and concomitant slow-down of lactate production. Comprehensive analysis of more than 500 metabolites upon LDH5 inhibition in Snu398 cells revealed that the cytosolic glycolysis pathway was significantly impeded with some up-stream intermediates increasing as much as 40-fold. As the cell lost its ability for cytosolic glucose processing, the TCA cycle activity increased indicating that pyruvate entered the mitochondria and restored their activity resulting in increased oxygen consumption upon LDH5 inhibition. Several pathways that rely on glycolytic and TCA intermediates were also upregulated, including fatty acid metabolism and pentose phosphate pathway. LDH5 inhibition also strongly potentiated PKM2 activity. These profound metabolic alterations greatly impaired cell survival and induced cell death in Snu398 cells. In summary, we have shown that rapid chemical inhibition of LDH5 leads to profound metabolic alterations and impairs cell survival in hepatocellular carcinoma cells making it a compelling strategy for treating solid tumors relying on aerobic glycolysis. Citation Format: Julia Billiard, Roland Annan, Jennifer Ariazi, Jacques Briand, Kristin Brown, Nino Campobasso, Subhas Chakravorty, Deping Chai, Mariela Colón, Elizabeth Davenport, Christopher Dodson, Nathan Gaul, Seth Gilbert, Anthony Jurewicz, Hong Lu, Dean McNulty, Jeanelle McSurdy-Freed, Lisa Miller, Kelvin Nurse, Paru Rao Nuthulaganti, Chad Quinn, Jessica Schneck, Gilbert Scott, Tony Shaw, Christian Sherk, Angela Smallwood, Sharon Sweitzer, James Villa, Gregory Waitt, Richard Wooster, Kevin Duffy. Rapid LDH5 inhibition reverses malignant metabolic phenotype and impairs survival of hepatocellular carcinoma cells . [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5418. doi:10.1158/1538-7445.AM2013-5418
- Published
- 2013
38. Abstract 549: MAGI3-Akt3, a novel fusion protein in the PI 3-K pathway in cancer
- Author
-
Kristin K. Brown and Alex Toker
- Subjects
Cancer Research ,Oncology ,Akt/PKB signaling pathway ,Akt Inhibitor MK2206 ,biology.protein ,Cancer research ,PTEN ,Biology ,Kinase activity ,Protein kinase B ,Fusion protein ,AKT3 ,PI3K/AKT/mTOR pathway - Abstract
Aberrant activation of the PI3-K/Akt pathway is found in approximately 70% of breast cancers. We recently identified a recurrent rearrangement that produces an in-frame fusion protein containing Akt3 and the scaffolding protein MAGI3 (1). MAGI3-Akt3 is the first fusion protein that has been identified in the PI 3-K pathway. We propose that MAGI3-Akt3 is a novel oncogene in breast cancer and that the oncogenic activity of the fusion protein results from disruption of the PI 3-K/Akt pathway. Within the fusion, Akt3 has a disrupted PH domain prior to glutamate 17 (E17). As a result the fusion protein is constitutively phosphorylated in the Akt3 kinase domain and expression of MAGI3-Akt3 in breast cancer cells enhances Akt substrate phosphorylation. Constitutive MAGI3-Akt3 kinase activity can be inhibited by ATP-competitive small molecule Akt inhibitors but is resistant to the allosteric Akt inhibitor MK2206 which is currently in phase II clinical trials. Our results indicate that ATP competitive Akt inhibitors should be evaluated in clinical trials for the treatment of fusion-positive breast cancers. The fusion protein also contains a severely truncated portion of the scaffolding protein MAGI3. In particular, the fusion lacks the second PDZ domain of MAGI3 that has been reported to bind to PTEN and be required for the inhibitory effect of PTEN on PI 3-K pathway activity. We have demonstrated that disruption of MAGI3 in breast cancer cell lines does indeed promote PI 3-K pathway hyperactivity in a PTEN-dependent manner. In addition, the fusion lacks the fifth PDZ domain of MAGI3 and our studies indicate that this disrupts the interaction of MAGI3 with PHLPP2, a phosphatase that directly dephosphorylates Akt at pS473. Our studies therefore demonstrate that the MAGI3-Akt3 translocation significantly perturbs PI 3-K pathway activity via activation of an oncogene (Akt3) and dysregulation of two tumor suppressors (PTEN and PHLPP2). We are currently continuing studies to elucidate the oncogenic mechanism(s) of MAGI3-Akt3 in breast cancer. Citation Format: Kristin Brown, Alex Toker. MAGI3-Akt3, a novel fusion protein in the PI 3-K pathway in cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 549. doi:10.1158/1538-7445.AM2013-549
- Published
- 2013
39. Abstract PL07-01: Molecular profiling of breast cancer in Mexico: Identification of novel therapeutic targets through whole genome sequencing analysis
- Author
-
Andrey Sivachenko, Juan Carlos Fernández-López, Carrie Sougnez, Antonio Maffuz-Aziz, Jorge Melendez-Zajgla, Nam Pho, Rosa Rebollar-Vega, Lihua Zou, Valeria Quintanar-Jurado, Eric S. Lander, Kristin G. Ardlie, Joonil Jung, Verónica Bautista-Piña, Fujiko Duke, Shantanu Banerji, Sergio Rodriguez-Cuevas, Andrea L. Richardson, Stacey Gabriel, Kornelia Polyak, Gad Getz, Matthew Meyerson, Melissa Parkin, Kristin K. Brown, Sandra Romero-Cordoba, Kristian Cibulskis, Robert C. Onofrio, Shouyong Peng, Abbie M. Frederick, Kristin Thompson, Scott L. Carter, Dennis C. Sgroi, Joshua M. Francis, Nicolas Stransky, Daniel Auclair, Claudia Rangel-Escareño, José Baselga, Laura Uribe-Figueroa, Steven E. Schumacher, Alex H. Ramos, Alex Toker, Rameen Beroukhim, Michael S. Lawrence, Alfredo Hidalgo-Miranda, Gerardo Jimenez-Sanchez, Levi A. Garraway, Todd R. Golub, and Maria L. Cortes
- Subjects
Oncology ,Whole genome sequencing ,Genetics ,medicine.medical_specialty ,Epidemiology ,Sequence analysis ,Biology ,medicine.disease ,Genome ,Breast cancer ,Internal medicine ,microRNA ,medicine ,Ectopic expression ,Exome sequencing ,Cause of death - Abstract
Today, more than 55% of the world's breast cancer cases are diagnosed in low and middle-income countries and in 2020, more that 70% of the cases will come from the developing nations. In Mexico, breast cancer-specific mortality doubled during the past 20 years, representing the second-leading cause of death in women between 30 and 59 years and the leading cause of cancer related death in the female population. According to statistics, in Mexico a woman dies due to breast cancer every two hours. Even though breast cancer represents a major public health problem in the developing world, knowledge about the genetic and genomic structure of breast tumors in Mexican or Latin American populations is very limited. In the past four years, we have participated in the Slim Initiative of Genomic Medicine (SIGMA) Project, a collaboration between the Carlos Slim Institute of Health, the Broad Institute, and the National Institute of Genomic Medicine in Mexico city. The goal of the SIGMA project is to characterize the genomic basis of common diseases, including several types of cancer. This effort has focused on the application of whole genome and whole exome sequencing of human tumors. In the case of breast cancer, we have analyzed the whole genomes of 22 tumor/normal tissue pairs and the whole exomes of 103 tumor/normal tissues from Mexican and Vietnamese patients. Sequence analysis led to the novel identification of potential loss of function mutations of the CBFB transcription factor, and deletions of its partner RUNX1, an event which has never been previously reported in breast tumors or in any other epithelial tumor. Of clinical relevance, we also identified a somatic translocation involving MAGI3 and AKT3 in a triple negative breast tumor. Ectopic expression of the fusion transcrip leads to constitutive phosphorylation of downstream GSK and loss of contact inhibition. Most importantly, the activity of the fusion protein can be abrogated by an ATP-competitive small molecule inhibitor of AKT, potentially representing a new therapeutic avenue for these patients. In parallel with sequencing, we have also been working on the analysis of somatic DNA copy number aberrations, messenger RNA expression, and microRNA expression patterns in tumors from Mexican patients. Intrinsic breast cancer sub-typing in 125 tumors from Mexican patients showed that 13.6% of the tumors were basal-like, 16.8% were Her2-enriched, 24.8% Luminal A, 34.4% Luminal B and 10.4 normal-like. With microRNA expression, we have identified a group of microRNAs whose role in breast cancer has not been previously described and are currently analyzing differential microRNA expression across tumor sub-types, in particular triple negative tumors, where we have been able to identify at least three different tumor sub-groups based on microRNA expression patterns. Citation Format: Shantanu Banerji, Kristian Cibulskis, Claudia Rangel-Escareño, Kristin K. Brown, Scott L. Carter, Abbie M. Frederick, Michael S. Lawrence, Andrey Y. Sivachenko, Carrie Sougnez, Lihua Zou, Maria L. Cortes, Juan C. Fernandez-Lopez, Shouyong Peng, Kristin G. Ardlie, Daniel Auclair, Veronica Bautista-Piña, Fujiko Duke, Joshua Francis, Joonil Jung, Antonio Maffuz-Aziz, Robert C. Onofrio, Melissa Parkin, Nam H. Pho, Valeria Quintanar-Jurado, Alex H. Ramos, Rosa Rebollar-Vega, Sergio A. Rodríguez-Cuevas, Sandra L. Romero-Cordoba, Steven E. Schumacher, Nicolas Stransky, Kristin M. Thompson, Laura Uribe-Figueroa, Jose Baselga, Rameen Beroukhim, Kornelia Polyak, Dennis C. Sgroi, Andrea L. Richardson, Gerardo Jimenez-Sánchez, Eric S. Lander, Stacey B. Gabriel, Levi A. Garraway, Todd R. Golub, Jorge Meléndez-Zajgla, Alex Toker, Gad Getz, Matthew Meyerson, Alfredo Hidalgo-Miranda. Molecular profiling of breast cancer in Mexico: Identification of novel therapeutic targets through whole genome sequencing analysis. [abstract]. In: Proceedings of the Fifth AACR Conference on the Science of Cancer Health Disparities in Racial/Ethnic Minorities and the Medically Underserved; 2012 Oct 27-30; San Diego, CA. Philadelphia (PA): AACR; Cancer Epidemiol Biomarkers Prev 2012;21(10 Suppl):Abstract nr PL07-01.
- Published
- 2012
40. Mitochondrial respiratory chain involvement in peroxiredoxin 3 oxidation by phenethyl isothiocyanate and auranofin
- Author
-
Andrew G. Cox, Mark B. Hampton, and Kristin K. Brown
- Subjects
Peroxiredoxin III ,Thioredoxin-Disulfide Reductase ,Phenethyl isothiocyanate ,Auranofin ,Redox signaling ,Thioredoxin reductase ,Biophysics ,Respiratory chain ,Antimycin A ,HL-60 Cells ,Biology ,Biochemistry ,Electron Transport ,chemistry.chemical_compound ,Isothiocyanates ,Structural Biology ,Genetics ,medicine ,Humans ,Enzyme Inhibitors ,Thioredoxin ,Molecular Biology ,Dose-Response Relationship, Drug ,Peroxiredoxins ,Cell Biology ,respiratory system ,Hydrogen peroxide ,Molecular biology ,Mitochondria ,Mitochondrial respiratory chain ,chemistry ,Oxidative stress ,Protein Multimerization ,Peroxiredoxin ,Oxidation-Reduction ,medicine.drug - Abstract
Mitochondrial peroxiredoxin 3 (Prx 3) is rapidly oxidized in cells exposed to phenethyl isothiocyanate (PEITC) and auranofin (AFN), but the mechanism of oxidation is unclear. Using HL-60 cells deplete of mitochondrial DNA we show that peroxiredoxin 3 oxidation and cytotoxicity requires a functional respiratory chain. Thioredoxin reductase (TrxR) could be inhibited by up to 90% by auranofin without direct oxidation of peroxiredoxin 3. However, inhibition of thioredoxin reductase promoted peroxiredoxin 3 oxidation and cytotoxicity in combination with phenethyl isothiocyanate or antimycin A. We conclude that rapid peroxiredoxin 3 oxidation occurs as a consequence of increased oxidant production from the mitochondrial respiratory chain.
- Full Text
- View/download PDF
41. MERIT40 Is an Akt Substrate that Promotes Resolution of DNA Damage Induced by Chemotherapy
- Author
-
Andrew H. Beck, Kristin K. Brown, Laleh Montaser-Kouhsari, and Alex Toker
- Subjects
DNA Repair ,DNA repair ,DNA damage ,medicine.medical_treatment ,Immunoblotting ,Fluorescent Antibody Technique ,Estrogen receptor ,Antineoplastic Agents ,Breast Neoplasms ,Pharmacology ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,medicine ,Humans ,Immunoprecipitation ,Doxorubicin ,Enzyme Inhibitors ,Phosphorylation ,lcsh:QH301-705.5 ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Adaptor Proteins, Signal Transducing ,Chemotherapy ,3. Good health ,lcsh:Biology (General) ,Drug Resistance, Neoplasm ,Tissue Array Analysis ,Female ,Proto-Oncogene Proteins c-akt ,DNA Damage ,medicine.drug - Abstract
SummaryResistance to cytotoxic chemotherapy drugs, including doxorubicin, is a significant obstacle to the effective treatment of breast cancer. Here, we have identified a mechanism by which the PI3K/Akt pathway mediates resistance to doxorubicin. In addition to inducing DNA damage, doxorubicin triggers sustained activation of Akt signaling in breast cancer cells. We show that Akt contributes to chemotherapy resistance such that PI3K or Akt inhibitors sensitize cells to doxorubicin. We identify MERIT40, a component of the BRCA1-A DNA damage repair complex, as an Akt substrate that is phosphorylated following doxorubicin treatment. MERIT40 phosphorylation facilitates assembly of the BRCA1-A complex in response to DNA damage and contributes to DNA repair and cell survival following doxorubicin treatment. Finally, MERIT40 phosphorylation in human breast cancers is associated with estrogen receptor positivity. Our findings suggest that combination therapy with PI3K or Akt inhibitors and doxorubicin may constitute a successful strategy for overcoming chemotherapy resistance.
- Full Text
- View/download PDF
42. Risk Factors for Recent HIV Infections among Adults in 14 Countries in Africa Identified by Population-Based HIV Impact Assessment Surveys, 2015-2019.
- Author
-
Currie DW, West CA, Patel HK, Favaloro J, Asiimwe F, Ndagije F, Silver R, Mugurungi O, Shang J, Ndongmo CB, Williams DB, Dzinotyiweyi E, Waruru A, Pasipamire M, Nuwagaba-Biribonwoha H, Dlamini S, McLeod N, Kayirangwa E, Rwibasira G, Minchella PA, Auld AF, Nyirenda R, Getaneh Y, Hailemariam AH, Tondoh-Koui I, Kohemun N, Mgomella GS, Njau PF, Kirungi WL, Dalhatu I, Stafford KA, Bodika SM, Ussery F, McCracken S, Stupp P, Brown K, Duong YT, Parekh BS, and Voetsch AC
- Subjects
- Humans, Adult, Female, Male, Africa epidemiology, Risk Factors, Sexual Partners, Data Collection, HIV Infections diagnosis, HIV Infections epidemiology
- Abstract
Identifying persons who have newly acquired HIV infections is critical for characterizing the HIV epidemic direction. We analyzed pooled data from nationally representative Population-Based HIV Impact Assessment surveys conducted across 14 countries in Africa for recent infection risk factors. We included adults 15-49 years of age who had sex during the previous year and used a recent infection testing algorithm to distinguish recent from long-term infections. We collected risk factor information via participant interviews and assessed correlates of recent infection using multinomial logistic regression, incorporating each survey's complex sampling design. Compared with HIV-negative persons, persons with higher odds of recent HIV infection were women, were divorced/separated/widowed, had multiple recent sex partners, had a recent HIV-positive sex partner or one with unknown status, and lived in communities with higher HIV viremia prevalence. Prevention programs focusing on persons at higher risk for HIV and their sexual partners will contribute to reducing HIV incidence.
- Published
- 2023
- Full Text
- View/download PDF
43. Awareness of and willingness to use oral pre-exposure prophylaxis (PrEP) for HIV prevention among sexually active adults in Malawi: results from the 2020 Malawi population-based HIV impact assessment.
- Author
-
Kabaghe AN, Singano V, Payne D, Maida A, Nyirenda R, Mirkovic K, Jahn A, Patel P, Brown K, Farahani M, Kayigamba F, Tenthani L, Ogollah F, Auld A, Zulu F, Msungama W, and Wadonda-Kabondo N
- Subjects
- Male, Adult, Humans, Female, Homosexuality, Male, HIV, Cross-Sectional Studies, Malawi, Health Knowledge, Attitudes, Practice, HIV Infections prevention & control, Pre-Exposure Prophylaxis methods
- Abstract
Background: The World Health Organization recommends Pre-Exposure Prophylaxis (PrEP) for all populations at substantial risk of HIV infection. Understanding PrEP awareness and interest is crucial for designing PrEP programs; however, data are lacking in sub-Saharan Africa. In Malawi, oral PrEP was introduced in 2018. We analyzed data from the 2020 Malawi Population-based HIV Impact Assessment (MPHIA) to assess PrEP awareness and factors associated with PrEP interest in Malawi., Methods: MPHIA 2020 was a national cross-sectional household-based survey targeting adults aged 15 + years. Oral PrEP was first described to the survey participants as taking a daily pill to reduce the chance of getting HIV. To assess awareness, participants were asked if they had ever heard of PrEP and to assess interest, were asked if they would take PrEP to prevent HIV, regardless of previous PrEP knowledge. Only sexually active HIV-negative participants are included in this analysis. We used multivariable logistic regression to assess sociodemographic factors and behaviors associated with PrEP interest. All results were weighted., Results: We included 13,995 HIV-negative sexually active participants; median age was 29 years old. Overall, 15.0%, 95% confidence interval (CI): 14.2-15.9% of participants were aware of PrEP. More males (adjusted odds ratio (aOR): 1.3, 95% CI: 1.2-1.5), those with secondary (aOR: 1.5, 95% CI: 1.2-2.0) or post-secondary (aOR: 3.4, 95% CI: 2.4-4.9) education and the wealthiest (aOR: 1.6, 95% CI: 1.2-2.0) were aware of PrEP than female, those without education and least wealthy participants, respectively. Overall, 73.0% (95% CI: 71.8-74.1%) of participants were willing to use PrEP. Being male (aOR: 1.2; 95% CI: 1.1-1.3) and having more than one sexual partner (aOR: 1.7 95% CI: 1.4-1.9), were associated higher willingness to use PrEP., Conclusions: In this survey, prior PrEP knowledge and use were low while PrEP interest was high. High risk sexual behavior was associated with willingness to use PrEP. Strategies to increase PrEP awareness and universal access, may reduce HIV transmission., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
44. The epidemiology of HIV population viral load in twelve sub-Saharan African countries.
- Author
-
Hladik W, Stupp P, McCracken SD, Justman J, Ndongmo C, Shang J, Dokubo EK, Gummerson E, Koui I, Bodika S, Lobognon R, Brou H, Ryan C, Brown K, Nuwagaba-Biribonwoha H, Kingwara L, Young P, Bronson M, Chege D, Malewo O, Mengistu Y, Koen F, Jahn A, Auld A, Jonnalagadda S, Radin E, Hamunime N, Williams DB, Kayirangwa E, Mugisha V, Mdodo R, Delgado S, Kirungi W, Nelson L, West C, Biraro S, Dzekedzeke K, Barradas D, Mugurungi O, Balachandra S, Kilmarx PH, Musuka G, Patel H, Parekh B, Sleeman K, Domaoal RA, Rutherford G, Motsoane T, Bissek AZ, Farahani M, and Voetsch AC
- Subjects
- Adult, Humans, Male, Female, Viremia drug therapy, Viral Load, Seroepidemiologic Studies, Lesotho, Zimbabwe, HIV Infections drug therapy, Anti-HIV Agents therapeutic use
- Abstract
Background: We examined the epidemiology and transmission potential of HIV population viral load (VL) in 12 sub-Saharan African countries., Methods: We analyzed data from Population-based HIV Impact Assessments (PHIAs), large national household-based surveys conducted between 2015 and 2019 in Cameroon, Cote d'Ivoire, Eswatini, Kenya, Lesotho, Malawi, Namibia, Rwanda, Tanzania, Uganda, Zambia, and Zimbabwe. Blood-based biomarkers included HIV serology, recency of HIV infection, and VL. We estimated the number of people living with HIV (PLHIV) with suppressed viral load (<1,000 HIV-1 RNA copies/mL) and with unsuppressed viral load (viremic), the prevalence of unsuppressed HIV (population viremia), sex-specific HIV transmission ratios (number female incident HIV-1 infections/number unsuppressed male PLHIV per 100 persons-years [PY] and vice versa) and examined correlations between a variety of VL metrics and incident HIV. Country sample sizes ranged from 10,016 (Eswatini) to 30,637 (Rwanda); estimates were weighted and restricted to participants 15 years and older., Results: The proportion of female PLHIV with viral suppression was higher than that among males in all countries, however, the number of unsuppressed females outnumbered that of unsuppressed males in all countries due to higher overall female HIV prevalence, with ratios ranging from 1.08 to 2.10 (median: 1.43). The spatial distribution of HIV seroprevalence, viremia prevalence, and number of unsuppressed adults often differed substantially within the same countries. The 1% and 5% of PLHIV with the highest VL on average accounted for 34% and 66%, respectively, of countries' total VL. HIV transmission ratios varied widely across countries and were higher for male-to-female (range: 2.3-28.3/100 PY) than for female-to-male transmission (range: 1.5-10.6/100 PY). In all countries mean log10 VL among unsuppressed males was higher than that among females. Correlations between VL measures and incident HIV varied, were weaker for VL metrics among females compared to males and were strongest for the number of unsuppressed PLHIV per 100 HIV-negative adults (R2 = 0.92)., Conclusions: Despite higher proportions of viral suppression, female unsuppressed PLHIV outnumbered males in all countries examined. Unsuppressed male PLHIV have consistently higher VL and a higher risk of transmitting HIV than females. Just 5% of PLHIV account for almost two-thirds of countries' total VL. Population-level VL metrics help monitor the epidemic and highlight key programmatic gaps in these African countries., Competing Interests: The authors have declared that no competing interests exist., (Copyright: This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.)
- Published
- 2023
- Full Text
- View/download PDF
45. Point of Care CD4 Testing in National Household Surveys - Results and Quality Indicators from Eleven Population-Based HIV Impact Assessment (PHIA) Surveys.
- Author
-
Birhanu S, Winterhalter FS, Stupp P, Cates M, Rottinghaus E, Yavo D, Wray-Gordon F, Lupoli K, Ndongmo CB, Longwe H, Reid GA, Metz M, Saito S, McCracken S, Brown K, Voetsch AC, Duong YT, Parekh BS, and Patel HK
- Subjects
- Humans, Anti-Retroviral Agents therapeutic use, CD4 Lymphocyte Count, HIV, Point-of-Care Testing, Quality Indicators, Health Care, HIV Infections diagnosis, HIV Infections drug therapy, HIV Infections epidemiology, Point-of-Care Systems
- Abstract
Population-based HIV Impact Assessments (PHIAs) are national household (HH) surveys that provide HIV diagnosis and CD4 testing with an immediate return of results. Accurate CD4 results improve HIV-positive participants' clinical care and inform the effectiveness of HIV programs. Here, we present CD4 results from the PHIA surveys that were conducted in 11 countries in sub-Saharan Africa between 2015 and 2018. All of the HIV-positive participants and 2 to 5% of the HIV-negative participants were offered Pima CD4 (Abbott, IL, USA) point-of-care (POC) tests. The quality of the CD4 test was ensured by conducting instrument verification, comprehensive training, quality control, a review of testing errors and an analysis of unweighted CD4 data by HIV status, age, gender, and antiretroviral (ARV) treatment status. Overall, CD4 testing was completed for 23,085 (99.5%) of the 23,209 HIV-positive and 7,329 (2.7%) of the 270,741 negative participants in 11 surveys. The instrument error rate was 11.3% (range, 4.4% to 15.7%). The median CD4 values among HIV-positive and HIV-negative participants (aged 15+) were 468 cells/mm
3 (interquartile range [IQR], 307 to 654) and 811 cells/mm3 (IQR, 647 to 1,013), respectively. Among the HIV-positive participants (aged 15+), those with detectable ARVs had higher CD4 values (508 cells/mm3 ) than those with undetectable ARVs (385.5 cells/mm3 ). Among the HIV-positive participants (aged 15+), 11.4% (2,528/22,253) had a CD4 value of less than 200 cells/mm3 , and approximately half of them (1,225/2,528 = 48.5%) had detectable ARVs, whereas 51.5% (1,303/2,528) had no detectable ARVs ( P < 0.0001). We successfully implemented high quality POC CD4 testing using Pima instruments. Our data come from nationally representative surveys in 11 countries and provide unique insights regarding the CD4 distribution among HIV-positive individuals as well as the baseline CD4 values among HIV-negative individuals. IMPORTANCE The manuscript describes CD4 levels among HIV-positive individuals and baseline CD4 levels among HIV-negative individuals from 11 sub-Saharan countries, thereby highlighting the importance of CD4 markers in the context of the HIV epidemic. Despite increased ARV access in each country, advanced HIV disease (CD4 < 200 cells/mm3 ) persists among approximately 11% of HIV-positive individuals. Therefore, it is important that our findings are shared with the scientific community to assist with similar implementations of point-of-care testing and to conduct a review of HIV programmatic gaps., Competing Interests: The authors declare no conflict of interest.- Published
- 2023
- Full Text
- View/download PDF
46. Multi-substrate Metabolic Tracing Reveals Marked Heterogeneity and Dependency on Fatty Acid Metabolism in Human Prostate Cancer.
- Author
-
Fidelito G, De Souza DP, Niranjan B, De Nardo W, Keerthikumar S, Brown K, Taylor RA, and Watt MJ
- Subjects
- Male, Humans, Mice, Animals, Energy Metabolism, Fatty Acids metabolism, Disease Models, Animal, Glucose, Glutamine metabolism, Prostatic Neoplasms pathology
- Abstract
Cancer cells undergo metabolic reprogramming to meet increased bioenergetic demands. Studies in cells and mice have highlighted the importance of oxidative metabolism and lipogenesis in prostate cancer; however, the metabolic landscape of human prostate cancer remains unclear. To address this knowledge gap, we performed radiometric (14C) and stable (13C) isotope tracing assays in precision-cut slices of patient-derived xenografts (PDX). Glucose, glutamine, and fatty acid oxidation was variably upregulated in malignant PDXs compared with benign PDXs. De novo lipogenesis (DNL) and storage of free fatty acids into phospholipids and triacylglycerols were increased in malignant PDXs. There was no difference in substrate utilization between localized and metastatic PDXs and hierarchical clustering revealed marked metabolic heterogeneity across all PDXs. Mechanistically, glucose utilization was mediated by acetyl-CoA production rather than carboxylation of pyruvate, while glutamine entered the tricarboxylic acid cycle through transaminase reactions before being utilized via oxidative or reductive pathways. Blocking fatty acid uptake or fatty acid oxidation with pharmacologic inhibitors was sufficient to reduce cell viability in PDX-derived organoids, whereas blockade of DNL, or glucose or glutamine oxidation induced variable and limited therapeutic efficacy. These findings demonstrate that human prostate cancer, irrespective of disease stage, can effectively utilize all metabolic substrates, albeit with marked heterogeneity across tumors. We also confirm that fatty acid uptake and oxidation are targetable metabolic dependencies in human prostate cancer., Implications: Prostate cancer utilizes multiple substrates to fuel energy requirements, yet pharmacologic targeting of fatty acid uptake and oxidation reveals metabolic dependencies in localized and metastatic tumors., (©2022 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
47. Male partner age, viral load, and HIV infection in adolescent girls and young women: evidence from eight sub-Saharan African countries.
- Author
-
Ayton S, Schwitters A, Mantell JE, Nuwagaba-Biribonwoha H, Hakim A, Hoffman S, Biraro S, Philip N, Wiesner L, Gummerson E, Brown K, Nyogea D, Barradas D, Nzima M, Fischer-Walker C, Payne D, Mulenga L, Mgomella G, Kirungi WL, Maile L, Aibo D, Musuka G, Mugurungi O, and Low A
- Subjects
- Adolescent, Female, Male, Humans, Aged, Viral Load, Eswatini, Lesotho, Sub-Saharan African People, HIV Infections epidemiology
- Abstract
Objective: We aimed to elucidate the role of partnerships with older men in the HIV epidemic among adolescent girls and young women (AGYW) aged 15-24 years in sub-Saharan Africa., Design: Analysis of Population-based HIV Impact Assessments in Eswatini, Lesotho, Malawi, Namibia, Tanzania, Uganda, Zambia, and Zimbabwe., Methods: We examined associations between reported partner age and recent HIV infection among AGYW, incorporating male population-level HIV characteristics by age-band. Recent HIV infection was defined using the LAg avidity assay algorithm. Viremia was defined as a viral load of more than 1000 copies/ml, regardless of serostatus. Logistic regression compared recent infection in AGYW with older male partners to those reporting younger partners. Dyadic analysis examined cohabitating male partner age, HIV status, and viremia to assess associations with AGYW infection., Results: Among 17 813 AGYW, increasing partner age was associated with higher odds of recent infection, peaking for partners aged 35-44 (adjusted odds ratio = 8.94, 95% confidence interval: 2.63-30.37) compared with partners aged 15-24. Population-level viremia was highest in this male age-band. Dyadic analyses of 5432 partnerships confirmed the association between partner age-band and prevalent HIV infection (male spousal age 35-44-adjusted odds ratio = 3.82, 95% confidence interval: 2.17-6.75). Most new infections were in AGYW with partners aged 25-34, as most AGYW had partners in this age-band., Conclusion: These results provide evidence that men aged 25-34 drive most AGYW infections, but partners over 9 years older than AGYW in the 35-44 age-band confer greater risk. Population-level infectiousness and male age group should be incorporated into identifying high-risk typologies in AGYW., (Copyright © 2022 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
48. Progress towards the UNAIDS 90-90-90 targets among persons aged 50 and older living with HIV in 13 African countries.
- Author
-
Farley SM, Wang C, Bray RM, Low AJ, Delgado S, Hoos D, Kakishozi AN, Harris TG, Nyirenda R, Wadonda N, Li M, Amuri M, Juma J, Kancheya N, Pietersen I, Mutenda N, Natanael S, Aoko A, Ngugi EW, Asiimwe F, Lecher S, Ward J, Chikwanda P, Mugurungi O, Moyo B, Nkurunziza P, Aibo D, Kabala A, Biraro S, Ndagije F, Musuka G, Ndongmo C, Shang J, Dokubo EK, Dimite LE, McCullough-Sanden R, Bissek AC, Getaneh Y, Eshetu F, Nkumbula T, Tenthani L, Kayigamba FR, Kirungi W, Musinguzi J, Balachandra S, Kayirangwa E, Ayite A, West CA, Bodika S, Sleeman K, Patel HK, Brown K, Voetsch AC, El-Sadr WM, and Justman JJ
- Subjects
- Adolescent, Adult, Aged, Female, Humans, Malawi, Male, Middle Aged, Serologic Tests, Surveys and Questionnaires, Viral Load, Young Adult, HIV Infections drug therapy, HIV Infections epidemiology, HIV Infections prevention & control
- Abstract
Introduction: Achieving optimal HIV outcomes, as measured by global 90-90-90 targets, that is awareness of HIV-positive status, receipt of antiretroviral (ARV) therapy among aware and viral load (VL) suppression among those on ARVs, respectively, is critical. However, few data from sub-Saharan Africa (SSA) are available on older people (50+) living with HIV (OPLWH). We examined 90-90-90 progress by age, 15-49 (as a comparison) and 50+ years, with further analyses among 50+ (55-59, 60-64, 65+ vs. 50-54), in 13 countries (Cameroon, Cote d'Ivoire, Eswatini, Ethiopia, Kenya, Lesotho, Malawi, Namibia, Rwanda, Tanzania, Uganda, Zambia and Zimbabwe)., Methods: Using data from nationally representative Population-based HIV Impact Assessments, conducted between 2015and 2019, participants from randomly selected households provided demographic and clinical information and whole blood specimens for HIV serology, VL and ARV testing. Survey weighted outcomes were estimated for 90-90-90 targets. Country-specific Poisson regression models examined 90-90-90 variation among OPLWH age strata., Results: Analyses included 24,826 HIV-positive individuals (15-49 years: 20,170; 50+ years: 4656). The first, second and third 90 outcomes were achieved in 1, 10 and 5 countries, respectively, by those aged 15-49, while OPLWH achieved outcomes in 3, 13 and 12 countries, respectively. Among those aged 15-49, women were more likely to achieve 90-90-90 targets than men; however, among OPLWH, men were more likely to achieve first and third 90 targets than women, with second 90 achievement being equivalent. Country-specific 90-90-90 regression models among OPLWH demonstrated minimal variation by age stratum across 13 countries. Among OLPWH, no first 90 target differences were noted by age strata; three countries varied in the second 90 by older age strata but not in a consistent direction; one country showed higher achievement of the third 90 in an older age stratum., Conclusions: While OPLWH in these 13 countries were slightly more likely than younger people to be aware of their HIV-positive status (first 90), this target was not achieved in most countries. However, OPLWH achieved treatment (second 90) and VL suppression (third 90) targets in more countries than PLWH <50. Findings support expanded HIV testing, prevention and treatment services to meet ongoing OPLWH health needs in SSA., (© 2022 The Authors. Journal of the International AIDS Society published by John Wiley & Sons Ltd on behalf of the International AIDS Society.)
- Published
- 2022
- Full Text
- View/download PDF
49. HIV incidence, viremia, and the national response in Eswatini: Two sequential population-based surveys.
- Author
-
Nkambule R, Philip NM, Reid G, Mnisi Z, Nuwagaba-Biribonwoha H, Ao TT, Ginindza C, Duong YT, Patel H, Saito S, Solmo C, Brown K, Moore CS, Voetsch AC, Bicego G, Bock N, Mhlanga F, Dlamini T, Mabuza K, Zwane A, Sahabo R, Dobbs T, Parekh BS, El-Sadr W, Ryan C, and Justman J
- Subjects
- Adolescent, Adult, Cross-Sectional Studies, Eswatini epidemiology, Female, HIV Infections virology, Humans, Incidence, Male, Middle Aged, Surveys and Questionnaires, Viremia virology, Young Adult, HIV Infections epidemiology, HIV-1 isolation & purification, Viral Load, Viremia epidemiology
- Abstract
With the highest HIV incidence and prevalence globally, the government of Eswatini started a substantial scale-up of HIV treatment and prevention services in 2011. Two sequential large population-based surveys were conducted before and after service expansion to assess the impact of the national response. Cross-sectional, household-based, nationally representative samples of adults, ages 18 to 49 years, were sampled in 2011 and 2016. We measured HIV prevalence, incidence (recent infection based on limiting antigen ≤1.5 optical density units and HIV RNA ≥1000 copies/mL), viral load suppression (HIV RNA <1000 copies/mL among all seropositive adults) and unsuppressed viremia (HIV RNA ≥1000 copies/mL among all, regardless of HIV status) and assessed for temporal changes by conducting a trend analysis of the log ratio of proportions, using a Z statistic distribution. HIV prevalence remained stable from 2011 to 2016 [32% versus 30%, p = 0.10]. HIV incidence significantly declined 48% [2.48% versus 1.30%, p = 0.01]. Incidence remained higher among women than men [2011: 3.16% versus 1.83%; 2016: 1.76% versus 0.86%], with a smaller but significant relative reduction among women [44%; p = 0.04] than men [53%; p = 0.09]. The proportion of seropositive adults with viral load suppression significantly increased from 35% to 71% [p < .001]. The proportion of the total adult population with unsuppressed viremia decreased from 21% to 9% [p < .001]. National HIV incidence in Eswatini decreased by nearly half and viral load suppression doubled over a five-year period. Unsuppressed viremia in the total population decreased 58%. These population-based findings demonstrate the national impact of expanded HIV services in a hyperendemic country., Competing Interests: As an inventor of LAg-Avidity EIA, BSP receives royalties from the sale of test kits sold by the manufacturer per US government policy. The other authors have no conflicts of interest to disclose.
- Published
- 2021
- Full Text
- View/download PDF
50. Optimizing community linkage to care and antiretroviral therapy Initiation: Lessons from the Nigeria HIV/AIDS Indicator and Impact Survey (NAIIS) and their adaptation in Nigeria ART Surge.
- Author
-
Jahun I, Said I, El-Imam I, Ehoche A, Dalhatu I, Yakubu A, Greby S, Bronson M, Brown K, Bamidele M, Boyd AT, Bachanas P, Dirlikov E, Agbakwuru C, Abutu A, Williams-Sherlock M, Onotu D, Odafe S, Williams DB, Bassey O, Ogbanufe O, Onyenuobi C, Adeola A, Meribe C, Efuntoye T, Fagbamigbe OJ, Fagbemi A, Ene U, Nguhemen T, Mgbakor I, Alagi M, Asaolu O, Oladipo A, Amafah J, Nzelu C, Dakum P, Mensah C, Aliyu A, Okonkwo P, Oyeledun B, Oko J, Ikpeazu A, Gambo A, Charurat M, Ellerbrock T, Aliyu S, and Swaminathan M
- Subjects
- Adolescent, Adult, Anti-Retroviral Agents therapeutic use, Cross-Sectional Studies, Delivery of Health Care organization & administration, Female, HIV Infections diagnosis, HIV Infections drug therapy, Humans, Male, Nigeria, Self Report, Surveys and Questionnaires, Young Adult, Delivery of Health Care methods, HIV Infections psychology, Telemedicine
- Abstract
Background: Ineffective linkage to care (LTC) is a known challenge for community HIV testing. To overcome this challenge, a robust linkage to care strategy was adopted by the 2018 Nigeria HIV/AIDS Indicator and Impact Survey (NAIIS). The NAIIS linkage to care strategy was further adapted to improve Nigeria's programmatic efforts to achieve the 1st 90 as part of the Nigeria Antiretroviral Therapy (ART) Surge initiative, which also included targeted community testing. In this paper we provide an overview of the NAIIS LTC strategy and describe the impact of this strategy on both the NAIIS and the Surge initiatives., Methods: The NAIIS collaborated with community-based organizations (CBOs) and deployed mobile health (mHealth) technology with real-time dashboards to manage and optimize community LTC for people living with HIV (PLHIV) diagnosed during the survey. In NAIIS, CBOs' role was to facilitate linkage of identified PLHIV in community to facility of their choice. For the ART Surge, we modified the NAIIS LTC strategy by empowering both CBOs and mobile community teams as responsible for not only active LTC but also for community testing, ART initiation, and retention in care., Results: Of the 2,739 PLHIV 15 years and above identified in NAIIS, 1,975 (72.1%) were either unaware of their HIV-positive status (N = 1890) or were aware of their HIV-positive status but not receiving treatment (N = 85). Of these, 1,342 (67.9%) were linked to care, of which 952 (70.9%) were initiated on ART. Among 1,890 newly diagnosed PLHIV, 1,278 (67.6%) were linked to care, 33.7% self-linked and 66.3% were linked by CBOs. Among 85 known PLHIV not on treatment, 64 (75.3%) were linked; 32.8% self-linked and 67.2% were linked by a CBO. In the ART Surge, LTC and treatment initiation rates were 98% and 100%, respectively. Three-month retention for monthly treatment initiation cohorts improved from 76% to 90% over 6 months., Conclusions: Active LTC strategies by local CBOs and mobile community teams improved LTC and ART initiation in the ART Surge initiative. The use of mHealth technology resulted in timely and accurate documentation of results in NAIIS. By deploying mHealth in addition to active LTC, CBOs and mobile community teams could effectively scale up ART with real-time documentation of client-level outcomes., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.