Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Cell ; 173(4): 864-878.e29, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29681454

ABSTRACT

Diversity in the genetic lesions that cause cancer is extreme. In consequence, a pressing challenge is the development of drugs that target patient-specific disease mechanisms. To address this challenge, we employed a chemistry-first discovery paradigm for de novo identification of druggable targets linked to robust patient selection hypotheses. In particular, a 200,000 compound diversity-oriented chemical library was profiled across a heavily annotated test-bed of >100 cellular models representative of the diverse and characteristic somatic lesions for lung cancer. This approach led to the delineation of 171 chemical-genetic associations, shedding light on the targetability of mechanistic vulnerabilities corresponding to a range of oncogenotypes present in patient populations lacking effective therapy. Chemically addressable addictions to ciliogenesis in TTC21B mutants and GLUT8-dependent serine biosynthesis in KRAS/KEAP1 double mutants are prominent examples. These observations indicate a wealth of actionable opportunities within the complex molecular etiology of cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Lung Neoplasms/pathology , Small Molecule Libraries/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cytochrome P450 Family 4/deficiency , Cytochrome P450 Family 4/genetics , Drug Discovery , G1 Phase Cell Cycle Checkpoints/drug effects , Glucocorticoids/pharmacology , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
2.
Cell ; 155(3): 552-66, 2013 Oct 24.
Article in English | MEDLINE | ID: mdl-24243015

ABSTRACT

Context-specific molecular vulnerabilities that arise during tumor evolution represent an attractive intervention target class. However, the frequency and diversity of somatic lesions detected among lung tumors can confound efforts to identify these targets. To confront this challenge, we have applied parallel screening of chemical and genetic perturbations within a panel of molecularly annotated NSCLC lines to identify intervention opportunities tightly linked to molecular response indicators predictive of target sensitivity. Anchoring this analysis on a matched tumor/normal cell model from a lung adenocarcinoma patient identified three distinct target/response-indicator pairings that are represented with significant frequencies (6%-16%) in the patient population. These include NLRP3 mutation/inflammasome activation-dependent FLIP addiction, co-occurring KRAS and LKB1 mutation-driven COPI addiction, and selective sensitivity to a synthetic indolotriazine that is specified by a seven-gene expression signature. Target efficacies were validated in vivo, and mechanism-of-action studies informed generalizable principles underpinning cancer cell biology.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Drug Screening Assays, Antitumor , Indoles/pharmacology , Lung Neoplasms/metabolism , Triazines/pharmacology , Animals , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carrier Proteins , Cell Line, Tumor , Coatomer Protein/metabolism , Female , Genes, ras , Heterografts , Humans , Lung Neoplasms/pathology , Lysosomes/metabolism , Mice , Molecular Targeted Therapy , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasm Transplantation , Oxidative Phosphorylation
3.
Proc Natl Acad Sci U S A ; 114(25): 6611-6616, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28584109

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a class of antidiabetic drug used for the treatment of diabetes. These drugs are thought to lower blood glucose by blocking reabsorption of glucose by SGLT2 in the proximal convoluted tubules of the kidney. To investigate the effect of inhibiting SGLT2 on pancreatic hormones, we treated perfused pancreata from rats with chemically induced diabetes with dapagliflozin and measured the response of glucagon secretion by alpha cells in response to elevated glucose. In these type 1 diabetic rats, glucose stimulated glucagon secretion by alpha cells; this was prevented by dapagliflozin. Two models of type 2 diabetes, severely diabetic Zucker rats and db/db mice fed dapagliflozin, showed significant improvement of blood glucose levels and glucose disposal, with reduced evidence of glucagon signaling in the liver, as exemplified by reduced phosphorylation of hepatic cAMP-responsive element binding protein, reduced expression of phosphoenolpyruvate carboxykinase 2, increased hepatic glycogen, and reduced hepatic glucose production. Plasma glucagon levels did not change significantly. However, dapagliflozin treatment reduced the expression of the liver glucagon receptor. Dapagliflozin in rodents appears to lower blood glucose levels in part by suppressing hepatic glucagon signaling through down-regulation of the hepatic glucagon receptor.


Subject(s)
Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Glucagon/metabolism , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Signal Transduction/drug effects , Animals , Blood Glucose/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Down-Regulation/drug effects , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Mice , Rats , Rats, Sprague-Dawley , Rats, Zucker , Rodentia/metabolism , Sodium-Glucose Transporter 2/metabolism
4.
EMBO J ; 33(17): 1960-76, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25024437

ABSTRACT

The antimitotic anti-cancer drugs, including taxol, perturb spindle dynamics, and induce prolonged, spindle checkpoint-dependent mitotic arrest in cancer cells. These cells then either undergo apoptosis triggered by the intrinsic mitochondrial pathway or exit mitosis without proper cell division in an adaptation pathway. Using a genome-wide small interfering RNA (siRNA) screen in taxol-treated HeLa cells, we systematically identify components of the mitotic apoptosis and adaptation pathways. We show that the Mad2 inhibitor p31(comet) actively promotes mitotic adaptation through cyclin B1 degradation and has a minor separate function in suppressing apoptosis. Conversely, the pro-apoptotic Bcl2 family member, Noxa, is a critical initiator of mitotic cell death. Unexpectedly, the upstream components of the mitochondrial apoptosis pathway and the mitochondrial fission protein Drp1 contribute to mitotic adaption. Our results reveal crosstalk between the apoptosis and adaptation pathways during mitotic arrest.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Epithelial Cells/cytology , Epithelial Cells/drug effects , Mitosis , Paclitaxel/pharmacology , RNA, Small Interfering/analysis , Adaptation, Physiological , Gene Expression Profiling , HeLa Cells , Humans , RNA, Small Interfering/genetics
5.
Proc Natl Acad Sci U S A ; 112(8): 2503-8, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25675519

ABSTRACT

Insulin monotherapy can neither maintain normoglycemia in type 1 diabetes (T1D) nor prevent the long-term damage indicated by elevated glycation products in blood, such as glycated hemoglobin (HbA1c). Here we find that hyperglycemia, when unaccompanied by an acute increase in insulin, enhances itself by paradoxically stimulating hyperglucagonemia. Raising glucose from 5 to 25 mM without insulin enhanced glucagon secretion ∼two- to fivefold in InR1-G9 α cells and ∼18-fold in perfused pancreata from insulin-deficient rats with T1D. Mice with T1D receiving insulin treatment paradoxically exhibited threefold higher plasma glucagon during hyperglycemic surges than during normoglycemic intervals. Blockade of glucagon action with mAb Ac, a glucagon receptor (GCGR) antagonizing antibody, maintained glucose below 100 mg/dL and HbA1c levels below 4% in insulin-deficient mice with T1D. In rodents with T1D, hyperglycemia stimulates glucagon secretion, up-regulating phosphoenolpyruvate carboxykinase and enhancing hyperglycemia. GCGR antagonism in mice with T1D normalizes glucose and HbA1c, even without insulin.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/pathology , Insulin/therapeutic use , Receptors, Glucagon/immunology , Animals , Antibodies, Monoclonal/pharmacology , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 1/blood , Female , Glucagon/metabolism , Humans , Mice , Mice, Inbred NOD , Paracrine Communication/drug effects , Rats , Rats, Zucker
6.
Proc Natl Acad Sci U S A ; 111(36): 13217-22, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25157166

ABSTRACT

To determine the role of glucagon action in diet-induced and genetic type 2 diabetes (T2D), we studied high-fat-diet-induced obese (DIO) and leptin receptor-defective (LepR(-/-)) rodents with and without glucagon receptors (GcgRs). DIO and LepR(-/-),GcgR(+/+) mice both developed hyperinsulinemia, increased liver sterol response element binding protein 1c, and obesity. DIO GcgR(+/+) mice developed mild T2D, whereas LepR(-/-),GcgR(+/+) mice developed severe T2D. High-fat-fed (HFF) glucagon receptor-null mice did not develop hyperinsulinemia, increased liver sterol response element binding protein 1c mRNA, or obesity. Insulin treatment of HFF GcgR(-/-) to simulate HFF-induced hyperinsulinemia caused obesity and mild T2D. LepR(-/-),GcgR(-/-) did not develop hyperinsulinemia or hyperglycemia. Adenoviral delivery of GcgR to GcgR(-/-),LepR(-/-) mice caused the severe hyperinsulinemia and hyperglycemia of LepR(-/-) mice to appear. Spontaneous disappearance of the GcgR transgene abolished the hyperinsulinemia and hyperglycemia. In conclusion, T2D hyperglycemia requires unsuppressible hyperglucagonemia from insulin-resistant α cells and is prevented by glucagon suppression or blockade.


Subject(s)
Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Glucagon-Secreting Cells/pathology , Hyperglycemia/complications , Hyperglycemia/pathology , Insulin/pharmacology , Animals , Blood Glucose/metabolism , Body Temperature/drug effects , Body Weight/drug effects , Cell Line , Ceramides/pharmacology , Cricetinae , Diet , Disease Models, Animal , Feeding Behavior/drug effects , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Hyperglycemia/blood , Hyperinsulinism/blood , Hyperinsulinism/complications , Hyperinsulinism/pathology , Insulin/blood , Insulin/genetics , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Lipogenesis/drug effects , Male , Mice, Inbred C57BL , RNA, Messenger/blood , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Glucagon/metabolism
7.
Diabetes Metab Res Rev ; 32(7): 660-665, 2016 10.
Article in English | MEDLINE | ID: mdl-26729301

ABSTRACT

Although insulin monotherapy prevents death from ketoacidosis, it does not prevent either the hyperglycemic surges or the hypoglycemic plunges of glucose levels that plague the majority of patients with type 1 diabetes. However, significant improvements have occurred with the combination of continuous insulin delivery matched by continuous glucose monitoring, but the technology is not available for all patients, requires extensive education, is expensive and moreover, while much better than standard care, it almost never reduces haemoglobin A1c (HbA1c ) to below 6%. This may indicate that an improved diabetes therapy involving antagonism of glucagon action will for the first time control glucose levels to normal and eradicate the long-term complications of diabetes. Although one can never predict that results in animals will be reproduced in humans, the available evidence suggests that patients with type 1 and type 2 diabetes may expect far superior control of the metabolic abnormalities without the need for significant monitoring of glucose, a very important but expensive part of any insulin regimen.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Glucagon/antagonists & inhibitors , Hypoglycemic Agents/therapeutic use , Humans
8.
Nat Chem Biol ; 7(10): 712-9, 2011 Sep 11.
Article in English | MEDLINE | ID: mdl-21909097

ABSTRACT

A chemical genetics approach was taken to identify inhibitors of NS1, a major influenza A virus virulence factor that inhibits host gene expression. A high-throughput screen of 200,000 synthetic compounds identified small molecules that reversed NS1-mediated inhibition of host gene expression. A counterscreen for suppression of influenza virus cytotoxicity identified naphthalimides that inhibited replication of influenza virus and vesicular stomatitis virus (VSV). The mechanism of action occurs through activation of REDD1 expression and concomitant inhibition of mammalian target of rapamycin complex 1 (mTORC1) via TSC1-TSC2 complex. The antiviral activity of naphthalimides was abolished in REDD1(-/-) cells. Inhibition of REDD1 expression by viruses resulted in activation of the mTORC1 pathway. REDD1(-/-) cells prematurely upregulated viral proteins via mTORC1 activation and were permissive to virus replication. In contrast, cells conditionally expressing high concentrations of REDD1 downregulated the amount of viral protein. Thus, REDD1 is a new host defense factor, and chemical activation of REDD1 expression represents a potent antiviral intervention strategy.


Subject(s)
Antiviral Agents/pharmacology , Naphthalimides/pharmacology , Orthomyxoviridae/drug effects , Transcription Factors/metabolism , Vesiculovirus/drug effects , Animals , Antiviral Agents/chemistry , Cell Line , Dogs , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , High-Throughput Screening Assays , Humans , Mice , Microbial Sensitivity Tests , Molecular Structure , Naphthalimides/chemistry , Orthomyxoviridae/genetics , Orthomyxoviridae/metabolism , Structure-Activity Relationship , Transcription Factors/deficiency , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
9.
Nature ; 446(7137): 815-9, 2007 Apr 12.
Article in English | MEDLINE | ID: mdl-17429401

ABSTRACT

Abundant evidence suggests that a unifying principle governing the molecular pathology of cancer is the co-dependent aberrant regulation of core machinery driving proliferation and suppressing apoptosis. Anomalous proteins engaged in support of this tumorigenic regulatory environment most probably represent optimal intervention targets in a heterogeneous population of cancer cells. The advent of RNA-mediated interference (RNAi)-based functional genomics provides the opportunity to derive unbiased comprehensive collections of validated gene targets supporting critical biological systems outside the framework of preconceived notions of mechanistic relationships. We have combined a high-throughput cell-based one-well/one-gene screening platform with a genome-wide synthetic library of chemically synthesized small interfering RNAs for systematic interrogation of the molecular underpinnings of cancer cell chemoresponsiveness. NCI-H1155, a human non-small-cell lung cancer line, was employed in a paclitaxel-dependent synthetic lethal screen designed to identify gene targets that specifically reduce cell viability in the presence of otherwise sublethal concentrations of paclitaxel. Using a stringent objective statistical algorithm to reduce false discovery rates below 5%, we isolated a panel of 87 genes that represent major focal points of the autonomous response of cancer cells to the abrogation of microtubule dynamics. Here we show that several of these targets sensitize lung cancer cells to paclitaxel concentrations 1,000-fold lower than otherwise required for a significant response, and we identify mechanistic relationships between cancer-associated aberrant gene expression programmes and the basic cellular machinery required for robust mitotic progression.


Subject(s)
Genes, Lethal/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Paclitaxel/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Survival/drug effects , DNA Mutational Analysis , Dose-Response Relationship, Drug , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Neoplasms/drug therapy , Microtubules/drug effects , Microtubules/metabolism , Mitosis/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Spindle Apparatus/pathology
10.
Front Endocrinol (Lausanne) ; 14: 1114799, 2023.
Article in English | MEDLINE | ID: mdl-37152965

ABSTRACT

Purpose: Type 1 diabetes (T1D) accounts for an estimated 5% of all diabetes in the United States, afflicting over 1.25 million individuals. Maintaining long-term blood glucose control is the major goal for individuals with T1D. In T1D, insulin-secreting pancreatic islet ß-cells are destroyed by the immune system, but glucagon-secreting islet α-cells survive. These remaining α-cells no longer respond properly to fluctuating blood glucose concentrations. Dysregulated α-cell function contributes to hyper- and hypoglycemia which can lead to macrovascular and microvascular complications. To this end, we sought to discover small molecules that suppress α-cell function for their potential as preclinical candidate compounds. Prior high-throughput screening identified a set of glucagon-suppressing compounds using a rodent α-cell line model, but these compounds were not validated in human systems. Results: Here, we dissociated and replated primary human islet cells and exposed them to 24 h treatment with this set of candidate glucagon-suppressing compounds. Glucagon accumulation in the medium was measured and we determined that compounds SW049164 and SW088799 exhibited significant activity. Candidate compounds were also counter-screened in our InsGLuc-MIN6 ß-cell insulin secretion reporter assay. SW049164 and SW088799 had minimal impact on insulin release after a 24 h exposure. To further validate these hits, we treated intact human islets with a selection of the top candidates for 24 h. SW049164 and SW088799 significantly inhibited glucagon release into the medium without significantly altering whole islet glucagon or insulin content. In concentration-response curves SW088799 exhibited significant inhibition of glucagon release with an IC50 of 1.26 µM. Conclusion: Given the set of tested candidates were all top hits from the primary screen in rodent α-cells, this suggests some conservation of mechanism of action between human and rodents, at least for SW088799. Future structure-activity relationship studies of SW088799 may aid in elucidating its protein target(s) or enable its use as a tool compound to suppress α-cell activity in vitro.


Subject(s)
Diabetes Mellitus, Type 1 , Glucagon-Secreting Cells , Islets of Langerhans , Humans , Animals , Glucagon/metabolism , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Glucagon-Secreting Cells/metabolism
11.
J Am Chem Soc ; 134(46): 18998-9003, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23088155

ABSTRACT

Psymberin is the only member of the pederin natural product family that contains a dihydroisocoumarin side chain. Structural modifications of psymberin uncoupled inhibition of protein translation from cytotoxicity, suggesting that psymberin has more than one bioactivity. A forward genetic screen in Caenorhabditis elegans was conducted to identify the molecular target(s) of psymberin. Multiple independent psymberin-resistant mutants were isolated, each containing the same point mutation in a gene encoding a ribosomal protein. However, a psymberin-resistant mutant strain bearing this mutation was not cross-resistant to the pederin family member mycalamide A, which binds to the archaeal form of the same protein. Thus, two pederin family members likely differ in how they bind the same molecular target. The accumulation of psymberin in cells was sensitive to the stereochemistry of the amide side chain at C4 or C8 and the presence of the dihydroisocoumarin side chain. The observation that psymberin diastereomers or dihydroisocoumarin-truncated analogs lose all cytotoxic activity while retaining the ability to inhibit protein translation in a cell-free in vitro assay can be explained in the context of these differential cell uptake issues. Finally, we also demonstrate that the blistering activity associated with pederin and other members of the family is not due to their protein synthesis inhibiting activity. Unlike pederin and mycalamide, psymberin does not display irritant or blistering activity.


Subject(s)
Pyrones/chemistry , Pyrones/pharmacology , Coumarins , HeLa Cells , Humans , Structure-Activity Relationship
12.
Dev Cell ; 13(1): 3-4, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17609102

ABSTRACT

The extent to which the many different pathways of endocytosis share underlying molecular mechanisms is currently unknown. In this issue of Developmental Cell, Yarar et al. (2007) report that SNX9, a protein that binds phosphatidylinositides, dynamin, and N-WASP, coordinates actin assembly with several distinct endocytic processes.


Subject(s)
Actins/metabolism , Endocytosis/physiology , Microfilament Proteins/metabolism , Animals
13.
Nat Chem Biol ; 5(2): 100-7, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19125156

ABSTRACT

The pervasive influence of secreted Wnt signaling proteins in tissue homeostasis and tumorigenesis has galvanized efforts to identify small molecules that target Wnt-mediated cellular responses. By screening a diverse synthetic chemical library, we have discovered two new classes of small molecules that disrupt Wnt pathway responses; whereas one class inhibits the activity of Porcupine, a membrane-bound acyltransferase that is essential to the production of Wnt proteins, the other abrogates destruction of Axin proteins, which are suppressors of Wnt/beta-catenin pathway activity. With these small molecules, we establish a chemical genetic approach for studying Wnt pathway responses and stem cell function in adult tissue. We achieve transient, reversible suppression of Wnt/beta-catenin pathway response in vivo, and we establish a mechanism-based approach to target cancerous cell growth. The signal transduction mechanisms shown here to be chemically tractable additionally contribute to Wnt-independent signal transduction pathways and thus could be broadly exploited for chemical genetics and therapeutic goals.


Subject(s)
Neoplasms/metabolism , Regeneration , Signal Transduction/drug effects , Wnt Proteins/physiology , Axin Protein , Humans , Molecular Structure , Repressor Proteins/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism , beta Catenin/physiology
14.
PLoS Genet ; 4(12): e1000298, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19057677

ABSTRACT

Melanin protects the skin and eyes from the harmful effects of UV irradiation, protects neural cells from toxic insults, and is required for sound conduction in the inner ear. Aberrant regulation of melanogenesis underlies skin disorders (melasma and vitiligo), neurologic disorders (Parkinson's disease), auditory disorders (Waardenburg's syndrome), and opthalmologic disorders (age related macular degeneration). Much of the core synthetic machinery driving melanin production has been identified; however, the spectrum of gene products participating in melanogenesis in different physiological niches is poorly understood. Functional genomics based on RNA-mediated interference (RNAi) provides the opportunity to derive unbiased comprehensive collections of pharmaceutically tractable single gene targets supporting melanin production. In this study, we have combined a high-throughput, cell-based, one-well/one-gene screening platform with a genome-wide arrayed synthetic library of chemically synthesized, small interfering RNAs to identify novel biological pathways that govern melanin biogenesis in human melanocytes. Ninety-two novel genes that support pigment production were identified with a low false discovery rate. Secondary validation and preliminary mechanistic studies identified a large panel of targets that converge on tyrosinase expression and stability. Small molecule inhibition of a family of gene products in this class was sufficient to impair chronic tyrosinase expression in pigmented melanoma cells and UV-induced tyrosinase expression in primary melanocytes. Isolation of molecular machinery known to support autophagosome biosynthesis from this screen, together with in vitro and in vivo validation, exposed a close functional relationship between melanogenesis and autophagy. In summary, these studies illustrate the power of RNAi-based functional genomics to identify novel genes, pathways, and pharmacologic agents that impact a biological phenotype and operate outside of preconceived mechanistic relationships.


Subject(s)
Genomics , Melanocytes/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Skin Diseases/genetics , Skin Pigmentation/genetics , Animals , Cell Line , Genome, Human , Humans , Melanins/genetics , Melanins/metabolism , Mice , Mice, Inbred C57BL , RNA Interference , Skin Diseases/metabolism
15.
Cancer Res ; 81(2): 400-413, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33172929

ABSTRACT

Polyploid giant cancer cells (PGCC) are common in tumors and have been associated with resistance to cancer therapy, tumor relapse, malignancy, immunosuppression, metastasis, cancer stem cell production, and modulation of the tumor microenvironment. However, the molecular mechanisms that cause these cells to form are not yet known. In this study, we discover that Aurora kinases are synergistic determinants of a switch from the proliferative cell cycle to polyploid growth and multinucleation in lung cancer cell lines. When Aurora kinases were inhibited together, lung cancer cells uniformly grew into multinucleated PGCCs. These cells adopted an endoreplication in which the genome replicates, mitosis is omitted, and cells grow in size. Consequently, such cells continued to safely grow in the presence of antimitotic agents. These PGCC re-entered the proliferative cell cycle and grew in cell number when treatment was terminated. Thus, PGCC formation might represent a fundamental cellular response to Aurora kinase inhibitors and contributes to therapy resistance or tumor relapse. SIGNIFICANCE: These findings provide a novel insight about how cancer cells respond to Aurora kinase inhibitors and identify a new mechanism responsible for resistance to these agents and other antimitotic drugs.


Subject(s)
Antimitotic Agents/pharmacology , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase B/antagonists & inhibitors , Drug Resistance, Neoplasm , Giant Cells/drug effects , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Apoptosis , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Endoreduplication , Gene Expression Regulation, Neoplastic , Giant Cells/metabolism , Giant Cells/pathology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Tumor Microenvironment
16.
Traffic ; 9(8): 1233-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18422860

ABSTRACT

The two mammalian phosphatidylcholine (PC)-selective phospholipase D (PLD) enzymes remove the choline head group from PC to produce phosphatidic acid (PA). PA stimulates phosphatidylinositol(4)phosphate 5-kinases, can function as a binding site for membrane proteins, is required for certain membrane fusion or fission events and is an important precursor for the production of diacylglycerol (DAG). Both PA and DAG are lipids that favor negatively curved membranes rather than planar bilayers and can reduce the energetic barrier to membrane fission and fusion. Recent data provide a mechanistic explanation for the role PLDs play in some aspects of membrane traffic and provide an explanation for why some membrane fusion reactions require PA and some do not. PLDs also act as guanosine triphosphatase-activating proteins for dynamin and may participate with dynamin in the process of vesicle fission.


Subject(s)
Cell Membrane/enzymology , Phospholipase D/metabolism , Animals , Catalytic Domain , Cell Membrane/metabolism , Dynamins/metabolism , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Humans , Lipid Bilayers/chemistry , Lipids/chemistry , Models, Biological , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Transport , Saccharomyces cerevisiae/metabolism
17.
J Cell Biol ; 163(4): 879-88, 2003 Nov 24.
Article in English | MEDLINE | ID: mdl-14623870

ABSTRACT

Lipid rafts play important roles in cellular functions through concentrating or sequestering membrane proteins. This requires proteins to differ in the stability of their interactions with lipid rafts. However, knowledge of the dynamics of membrane protein-raft interactions is lacking. We employed FRAP to measure in live cells the lateral diffusion of influenza hemagglutinin (HA) proteins that differ in raft association. This approach can detect weak interactions with rafts not detectable by biochemical methods. Wild-type (wt) HA and glycosylphosphatidylinositol (GPI)-anchored HA (BHA-PI) diffused slower than a nonraft HA mutant, but became equal to the latter after cholesterol depletion. When antigenically distinct BHA-PI and wt HA were coexpressed, aggregation of BHA-PI into immobile patches reduced wt HA diffusion rate, suggesting transient interactions with BHA-PI raft patches. Conversely, patching wt HA reduced the mobile fraction of BHA-PI, indicating stable interactions with wt HA patches. Thus, the anchoring mode determines protein-raft interaction dynamics. GPI-anchored and transmembrane proteins can share the same rafts, and different proteins can interact stably or transiently with the same raft domains.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Animals , Binding Sites/physiology , Cell Line , Cholesterol/deficiency , Diffusion , Glycosylphosphatidylinositols/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Models, Molecular , Mutation/genetics , Protein Binding/physiology
18.
J Cell Biol ; 162(4): 693-701, 2003 Aug 18.
Article in English | MEDLINE | ID: mdl-12913109

ABSTRACT

Overexpression of phosphatidylinositol phosphate 5-kinase (PIP5KI) isoforms alpha, beta, or gamma in CV-1 cells increased phosphatidylinositol 4,5-bisphosphate (PIP2) levels by 35, 180, and 0%, respectively. Endocytosis of transferrin receptors, association of AP-2 proteins with membranes, and the number of clathrin-coated pits at the plasma membrane increased when PIP2 increased. When expression of PIP5KIbeta was inhibited with small interference RNA in HeLa cells, expression of PIP5KIalpha was also reduced slightly, but PIP5KIgamma expression was increased. PIP2 levels and internalization of transferrin receptors dropped 50% in these cells; thus, PIP5KIgamma could not compensate for loss of PIP5KIbeta. When expression of PIP5KIalpha was reduced, expression of both PIP5KIbeta and PIP5KIgamma increased and PIP2 levels did not change. A similar increase of PIP5KIalpha and PIP5KIbeta occurred when PIP5KIgamma was inhibited. These results indicate that constitutive endocytosis in CV-1 and HeLa cells requires (and may be regulated by) PIP2 produced primarily by PIP5KIbeta.


Subject(s)
Adaptor Protein Complex 2/metabolism , Cell Membrane/metabolism , Endocytosis/physiology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Transferrin/metabolism , Actins/metabolism , Animals , Coated Pits, Cell-Membrane/metabolism , Cytoskeleton/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans
20.
Mol Biol Cell ; 17(2): 598-606, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16291863

ABSTRACT

RNA interference-mediated depletion of phospholipase D2 (PLD2), but not PLD1, inhibited recycling of transferrin receptors in HeLa cells, whereas the internalization rate was unaffected by depletion of either PLD. Although reduction of both PLD isoforms inhibits PLD activity stimulated by phorbol 12-myristic 13-acetate, only depletion of PLD2 decreased nonstimulated activity. Cells with reduced PLD2 accumulated a greater fraction of transferrin receptors in a perinuclear compartment that was positive for Rab11, a marker of recycling endosomes. EFA6, an exchange factor for Arf6, has been proposed to stimulate the recycling of transferrin receptors. Thus, one consequence of EFA6 overexpression would be a reduction of the internal pool of receptors. We confirmed this observation in control HeLa cells; however, overexpression of EFA6 failed to decrease the internal pool of transferrin receptors that accumulate in cells previously depleted of PLD2. These observations suggest that either PLD2 is required for a constitutive Arf6-mediated recycling pathway or in the absence of PLD2 transferrin receptors accumulate in recycling endosomes that are not responsive to overexpression of EFA6.


Subject(s)
Endocytosis/physiology , Phospholipase D/physiology , Receptors, Transferrin/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/physiology , Cell Membrane/metabolism , Endosomes/metabolism , Gene Expression Regulation/drug effects , Guanine Nucleotide Exchange Factors , HeLa Cells , Humans , Nerve Tissue Proteins/metabolism , Phosphatidylinositol 4,5-Diphosphate/biosynthesis , Phospholipase D/antagonists & inhibitors , Phospholipase D/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RNA Interference , Tetradecanoylphorbol Acetate/pharmacology , rab GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL