Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Dev Cell ; 59(7): 898-910.e6, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38366599

ABSTRACT

The liver exhibits a remarkable capacity to regenerate following injury. Despite this unique attribute, toxic injury is a leading cause of liver failure. The temporal processes by which the liver senses injury and initiates regeneration remain unclear. Here, we developed a transgenic zebrafish model wherein hepatocyte-specific expression of uracil phosphoribosyltransferase (UPRT) enabled the implementation of SLAM-ITseq to investigate the nascent transcriptome during initiation of liver injury and regeneration. Using this approach, we identified a rapid metabolic transition from the fed to the fasted state that was followed by induction of the nuclear erythroid 2-related factor (Nrf2) antioxidant program. We find that activation of Nrf2 in hepatocytes is required to induce the pentose phosphate pathway (PPP) and improve survival following liver injury. Mechanistically, we demonstrate that inhibition of the PPP disrupts nucleotide biosynthesis to prevent liver regeneration. Together, these studies provide fundamental insights into the mechanism by which early metabolic adaptation to injury facilitates tissue regeneration.


Subject(s)
Liver Regeneration , Pentose Phosphate Pathway , Animals , Pentose Phosphate Pathway/genetics , Liver Regeneration/genetics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Liver/metabolism
2.
Proc Natl Acad Sci U S A ; 120(22): e2217425120, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37216554

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.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , NF-E2-Related Factor 2 , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lysosomes/metabolism , NF-E2-Related Factor 2/metabolism , Animals
3.
Oncogene ; 41(34): 4066-4078, 2022 08.
Article in English | MEDLINE | ID: mdl-35851845

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.


Subject(s)
Glutamine , Triple Negative Breast Neoplasms , Cell Line, Tumor , Citric Acid Cycle , Glucose/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism
4.
Dev Cell ; 57(6): 719-731.e8, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35216681

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.


Subject(s)
Lipogenesis , Sterol Regulatory Element Binding Proteins , Cell Proliferation , Lipogenesis/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
5.
Cancer Discov ; 12(3): 774-791, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34862195

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.


Subject(s)
Iron , bcl-2 Homologous Antagonist-Killer Protein , Apoptosis , Cell Death , Humans , Iron/metabolism , Mitochondria/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
6.
EMBO J ; 40(21): e109575, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34582060

ABSTRACT

AMP-activated protein kinase (AMPK) is recognized as a critical regulator of cellular energy metabolism impacted by AMP/ATP and ADP/ATP ratios, or glucose- and fatty acid-derived metabolites. However, its ability to sense alterations in amino acid levels is poorly understood. Recent work by Yuan et al (2021) identifies a novel mechanism of AMPK regulation responsive to changes in availability of the sulfur-containing amino acid cysteine.


Subject(s)
AMP-Activated Protein Kinases , Amino Acids , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Energy Metabolism , Fatty Acids , Glucose
7.
Cancer Discov ; 11(6): 1582-1599, 2021 06.
Article in English | MEDLINE | ID: mdl-33436370

ABSTRACT

Internal tandem duplication of the FMS-like tyrosine kinase 3 gene (FLT3-ITD) occurs in 30% of all acute myeloid leukemias (AML). Limited clinical efficacy of FLT3 inhibitors highlights the need for alternative therapeutic modalities in this subset of disease. Using human and murine models of FLT3-ITD-driven AML, we demonstrate that FLT3-ITD promotes serine synthesis and uptake via ATF4-dependent transcriptional regulation of genes in the de novo serine biosynthesis pathway and neutral amino acid transport. Genetic or pharmacologic inhibition of PHGDH, the rate-limiting enzyme of de novo serine biosynthesis, selectively inhibited proliferation of FLT3-ITD AMLs in vitro and in vivo. Moreover, pharmacologic inhibition of PHGDH sensitized FLT3-ITD AMLs to the standard-of-care chemotherapeutic cytarabine. Collectively, these data reveal novel insights into FLT3-ITD-induced metabolic reprogramming and reveal a targetable vulnerability in FLT3-ITD AML. SIGNIFICANCE: FLT3-ITD mutations are common in AML and are associated with poor prognosis. We show that FLT3-ITD stimulates serine biosynthesis, thereby rendering FLT3-ITD-driven leukemias dependent upon serine for proliferation and survival. This metabolic dependency can be exploited pharmacologically to sensitize FLT3-ITD-driven AMLs to chemotherapy.This article is highlighted in the In This Issue feature, p. 1307.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Serine/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Line, Tumor/metabolism , Disease Models, Animal , Humans , Mice , Protein Kinase Inhibitors
8.
Nat Commun ; 10(1): 296, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30655532

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.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Triple Negative Breast Neoplasms/genetics , src-Family Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Immediate-Early Proteins/antagonists & inhibitors , Mass Spectrometry/methods , Mice, Inbred BALB C , Mice, Nude , Oncogenes/genetics , Protein Interaction Mapping/methods , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proteomics/methods , Quinazolines/pharmacology , Quinazolines/therapeutic use , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , src-Family Kinases/antagonists & inhibitors
9.
EMBO J ; 37(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30348863

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.


Subject(s)
Glucose/metabolism , Liver/embryology , Nucleotides/biosynthesis , Signal Transduction/physiology , Trans-Activators/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Glucose/genetics , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Mice , Nucleotides/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Serine-Threonine Kinase 3 , Trans-Activators/genetics , YAP-Signaling Proteins , Zebrafish/genetics , Zebrafish Proteins/genetics
10.
Medchemcomm ; 9(4): 606-613, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-30108951

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.

11.
Angew Chem Int Ed Engl ; 57(42): 13821-13824, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30138551

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 through 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.


Subject(s)
Amino Acids/chemistry , Biocatalysis , Alkylation , Amino Acids/metabolism , Animals , Chromatography, High Pressure Liquid , Humans , Ketones/chemistry , Pseudomonas/enzymology , Stereoisomerism
12.
Sci Immunol ; 3(23)2018 05 18.
Article in English | MEDLINE | ID: mdl-29776993

ABSTRACT

Immunotherapy has revolutionized outcomes for cancer patients, but the mechanisms of resistance remain poorly defined. We used a series of whole-genome clustered regularly interspaced short palindromic repeat (CRISPR)-based screens performed in vitro and in vivo to identify mechanisms of tumor immune evasion from cytotoxic lymphocytes [CD8+ T cells and natural killer (NK) cells]. Deletion of key genes within the tumor necrosis factor (TNF) signaling, interferon-γ (IFN-γ) signaling, and antigen presentation pathways provided protection of tumor cells from CD8+ T cell-mediated killing and blunted antitumor immune responses in vivo. Deletion of a number of genes in the TNF pathway also emerged as the key mechanism of immune evasion from primary NK cells. Our screens also identified that the metabolic protein 2-aminoethanethiol dioxygenase (Ado) modulates sensitivity to TNF-mediated killing by cytotoxic lymphocytes and is required for optimal control of tumors in vivo. Remarkably, we found that tumors delete the same genes when exposed to perforin-deficient CD8+ T cells, demonstrating that the dominant immune evasion strategy used by tumor cells is acquired resistance to T cell-derived cytokine-mediated antitumor effects. We demonstrate that TNF-mediated bystander killing is a potent T cell effector mechanism capable of killing antigen-negative tumor cells. In addition to highlighting the importance of TNF in CD8+ T cell- and NK cell-mediated killing of tumor cells, our study also provides a comprehensive picture of the roles of the TNF, IFN, and antigen presentation pathways in immune-mediated tumor surveillance.


Subject(s)
Interferon-gamma/immunology , Tumor Escape/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Killer Cells, Natural/immunology , Mice
13.
Cancer Discov ; 7(4): 391-399, 2017 04.
Article in English | MEDLINE | ID: mdl-28255083

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. 380This article is highlighted in the In This Issue feature, p. 339.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Resistance, Neoplasm/drug effects , Pyrimidines/biosynthesis , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cellular Reprogramming/drug effects , DNA Damage/drug effects , Doxorubicin/administration & dosage , Female , Humans , Isoxazoles/administration & dosage , Leflunomide , Mice , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
14.
Proc Natl Acad Sci U S A ; 113(38): E5562-71, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27588899

ABSTRACT

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.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/genetics , Gastrointestinal Neoplasms/genetics , Selenoproteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Female , Gastrointestinal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Homeostasis , Humans , Male , Oxidation-Reduction , Oxidative Stress/genetics , Selenium/metabolism , Selenoproteins/metabolism , Transcriptome/genetics , Zebrafish/genetics
15.
Nat Cell Biol ; 18(8): 886-896, 2016 08.
Article in English | MEDLINE | ID: mdl-27428308

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.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Transformation, Neoplastic/genetics , Liver/growth & development , Phosphoproteins/genetics , Trans-Activators/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Glutamine/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Phosphoproteins/metabolism , Transcription Factors , YAP-Signaling Proteins , Zebrafish
16.
Cell Rep ; 11(9): 1358-66, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26027929

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms , DNA Damage/drug effects , Drug Resistance, Neoplasm/physiology , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Repair/drug effects , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Female , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoprecipitation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Tissue Array Analysis
17.
F1000Prime Rep ; 7: 13, 2015.
Article in English | MEDLINE | ID: mdl-25750731

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.

18.
Nat Chem Biol ; 10(9): 774-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25086508

ABSTRACT

Human fatty acid synthase (hFAS) is a complex, multifunctional enzyme that is solely responsible for the de novo synthesis of long chain fatty acids. hFAS is highly expressed in a number of cancers, with low expression observed in most normal tissues. Although normal tissues tend to obtain fatty acids from the diet, tumor tissues rely on de novo fatty acid synthesis, making hFAS an attractive metabolic target for the treatment of cancer. We describe here the identification of GSK2194069, a potent and specific inhibitor of the ß-ketoacyl reductase (KR) activity of hFAS; the characterization of its enzymatic and cellular mechanism of action; and its inhibition of human tumor cell growth. We also present the design of a new protein construct suitable for crystallography, which resulted in what is to our knowledge the first co-crystal structure of the human KR domain and includes a bound inhibitor.


Subject(s)
3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/metabolism , Enzyme Inhibitors/metabolism , Fatty Acid Synthases/antagonists & inhibitors , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Triazoles/metabolism , Triazoles/pharmacology , 3-Oxoacyl-(Acyl-Carrier-Protein) Reductase/chemistry , Catalytic Domain , Cell Line, Tumor , Fatty Acid Synthases/chemistry , Humans , Models, Molecular , Protein Conformation , X-Ray Diffraction
19.
Cell Rep ; 6(1): 56-69, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24388745

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.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Liver/drug effects , Nitric Oxide Donors/pharmacology , S-Nitrosothiols/pharmacology , Acetaminophen/toxicity , Aldehyde Oxidoreductases/metabolism , Animals , Liver/growth & development , Liver/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Nitric Oxide Donors/therapeutic use , S-Nitrosothiols/therapeutic use , Zebrafish , Zebrafish Proteins/metabolism
20.
Dev Cell ; 23(3): 560-72, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22975325

ABSTRACT

Integrin recycling is critical for cell migration. Protein kinase D (PKD) mediates signals from the platelet-derived growth factor receptor (PDGF-R) to control αvß3 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 αvß3, which in turn inhibits α5ß1 integrin recycling. Rab4, but not Rab5, interacts with phosphorylated Rabaptin-5 toward the front of migrating cells to promote delivery of αvß3 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 αvß3-dependent invasion. Conversely, invasive migration that is dependent on α5ß1 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.


Subject(s)
Cell Movement , Integrin alphaVbeta3/metabolism , Neoplasm Invasiveness , Protein Kinase C/metabolism , Vesicular Transport Proteins/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...