Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Clin Lung Cancer ; 23(1): 21-33, 2022 01.
Article in English | MEDLINE | ID: mdl-34226144

ABSTRACT

BACKGROUND: This phase III OAK trial (NCT02008227) subgroup analysis (data cutoff, January 9, 2019) evaluated the predictive value of 2 PD-L1 IHC tests (VENTANA SP142 and Dako 22C3) for benefit from atezolizumab versus docetaxel by programmed death ligand 1 (PD-L1) status in patients with previously treated metastatic non-small cell lung cancer. METHODS: PD-L1 expression was assessed prospectively with SP142 on tumor cells (TC) and tumor-infiltrating immune cells (IC) and retrospectively with 22C3 using a tumor proportion score (TPS) based on TC membrane staining. Efficacy was assessed in the 22C3 biomarker-evaluable population (22C3-BEP) (n = 577; 47.1% of SP142-intention-to-treat population) and non-22C3-BEP (n = 648) in PD-L1 subgroups (high, low, and negative) and according to selection by 1 or both assays. RESULTS: In the 22C3-BEP, overall survival benefits with atezolizumab versus docetaxel were observed across PD-L1 subgroups; benefits were greatest in SP142-defined PD-L1-high (TC3 or IC3: hazard ratio [HR], 0.39 [95% confidence interval (CI), 0.25-0.63]) and 22C3-defined PD-L1-high (TPS ≥ 50%: HR, 0.56 [95% CI, 0.38-0.82]) and low (TPS, 1% to < 50%: HR, 0.55 [95% CI, 0.37-0.82]) groups. Progression-free survival improved with increasing PD-L1 expression for both assays. SP142 and 22C3 assays identified overlapping and unique patient populations in PD-L1-high, positive, and negative subgroups. Overall survival and progression-free survival benefits favored atezolizumab over docetaxel in double PD-L1-positive and negative groups; patients with both SP142- and 22C3-positive tumors derived the greatest benefit. CONCLUSIONS: Despite different scoring algorithms and differing sensitivity levels, the SP142 and 22C3 assays similarly predicted atezolizumab benefit at validated PD-L1 thresholds in patients with non-small cell lung cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunohistochemistry , Lung Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Docetaxel/therapeutic use , Humans , Retrospective Studies , Treatment Outcome
2.
J Thorac Oncol ; 16(12): 2040-2050, 2021 12.
Article in English | MEDLINE | ID: mdl-34311110

ABSTRACT

INTRODUCTION: The Blood First Assay Screening Trial is an ongoing open-label, multicohort study, prospectively evaluating the relationship between blood-based next-generation sequencing (NGS) detection of actionable genetic alterations and activity of targeted therapies or immunotherapy in treatment-naive advanced or metastatic NSCLC. We present data from the ALK-positive cohort. METHODS: Patients aged more than or equal to 18 years with stage IIIB or IV NSCLC and ALK rearrangements detected by blood-based NGS using hybrid capture technology (FoundationACT) received alectinib 600 mg twice daily. Asymptomatic or treated central nervous system (CNS) metastases were permitted. Primary end point was investigator-assessed objective response rate (ORR; Response Evaluation Criteria in Solid Tumors version 1.1). Secondary end points were independent review facility-assessed ORR, duration of response, progression-free survival (PFS), overall survival, and safety. Exploratory end points were investigator-assessed ORR in patients with baseline CNS metastases and relationship between circulating biomarkers and response. RESULTS: In total, 2219 patients were screened and blood-based NGS yielded results in 98.6% of the cases. Of these, 119 patients (5.4%) had ALK-positive disease; 87 were enrolled and received alectinib. Median follow-up was 12.6 months (range: 2.6-18.7). Confirmed ORR was 87.4% (95% confidence interval [CI]: 78.5-93.5) by investigator and 92.0% (95% CI: 84.1-96.7) by independent review facility. Investigator-confirmed 12-month duration of response was 75.9% (95% CI: 63.6-88.2). In 35 patients (40%) with baseline CNS disease, investigator-assessed ORR was 91.4% (95% CI: 76.9-98.2). Median PFS was not reached; 12-month investigator-assessed PFS was 78.4% (95% CI: 69.1-87.7). Safety data were consistent with the known tolerability profile of alectinib. CONCLUSIONS: These results reveal the clinical application of blood-based NGS as a method to inform clinical decision-making in ALK-positive NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Anaplastic Lymphoma Kinase/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cohort Studies , Crizotinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use
3.
Nat Med ; 24(9): 1441-1448, 2018 09.
Article in English | MEDLINE | ID: mdl-30082870

ABSTRACT

Although programmed death-ligand 1-programmed death 1 (PD-L1-PD-1) inhibitors are broadly efficacious, improved outcomes have been observed in patients with high PD-L1 expression or high tumor mutational burden (TMB). PD-L1 testing is required for checkpoint inhibitor monotherapy in front-line non-small-cell lung cancer (NSCLC). However, obtaining adequate tumor tissue for molecular testing in patients with advanced disease can be challenging. Thus, an unmet medical need exists for diagnostic approaches that do not require tissue to identify patients who may benefit from immunotherapy. Here, we describe a novel, technically robust, blood-based assay to measure TMB in plasma (bTMB) that is distinct from tissue-based approaches. Using a retrospective analysis of two large randomized trials as test and validation studies, we show that bTMB reproducibly identifies patients who derive clinically significant improvements in progression-free survival from atezolizumab (an anti-PD-L1) in second-line and higher NSCLC. Collectively, our data show that high bTMB is a clinically actionable biomarker for atezolizumab in NSCLC.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/blood , Lung Neoplasms/genetics , Mutation/genetics , Tumor Burden/genetics , Antibodies, Monoclonal, Humanized , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Humans , Immunotherapy , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Progression-Free Survival , Treatment Outcome
4.
Cell Rep ; 15(11): 2536-49, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27264172

ABSTRACT

Zfp516, a brown fat (BAT)-enriched and cold-inducible transcription factor, promotes transcription of UCP1 and other BAT-enriched genes for non-shivering thermogenesis. Here, we identify lysine-specific demethylase 1 (LSD1) as a direct binding partner of Zfp516. We show that, through interaction with Zfp516, LSD1 is recruited to UCP1 and other BAT-enriched genes, such as PGC1α, to function as a coactivator by demethylating H3K9. We also show that LSD1 is induced during brown adipogenesis and that LSD1 and its demethylase activity is required for the BAT program. Furthermore, we show that LSD1 ablation in mice using Myf5-Cre alters embryonic BAT development. Moreover, BAT-specific deletion of LSD1 via the use of UCP1-Cre impairs the BAT program and BAT development, making BAT resemble WAT, reducing thermogenic activity and promoting obesity. Finally, we demonstrate an in vivo requirement of the Zfp516-LSD1 interaction for LSD1 function in BAT gene activation.


Subject(s)
Adipose Tissue, Brown/metabolism , Histone Demethylases/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Uncoupling Protein 1/genetics , 3T3-L1 Cells , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/growth & development , Adipose Tissue, White/metabolism , Animals , Cell Differentiation/genetics , Cold Temperature , HEK293 Cells , Humans , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Protein Binding , Thermogenesis/genetics
5.
Mol Cell ; 57(2): 235-46, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25578880

ABSTRACT

Uncoupling protein 1 (UCP1) mediates nonshivering thermogenesis and, upon cold exposure, is induced in brown adipose tissue (BAT) and subcutaneous white adipose tissue (iWAT). Here, by high-throughput screening using the UCP1 promoter, we identify Zfp516 as a transcriptional activator of UCP1 as well as PGC1α, thereby promoting a BAT program. Zfp516 itself is induced by cold and sympathetic stimulation through the cAMP-CREB/ATF2 pathway. Zfp516 directly binds to the proximal region of the UCP1 promoter, not to the enhancer region where other transcription factors bind, and interacts with PRDM16 to activate the UCP1 promoter. Although ablation of Zfp516 causes embryonic lethality, knockout embryos still show drastically reduced BAT mass. Overexpression of Zfp516 in adipose tissue promotes browning of iWAT even at room temperature, increasing body temperature and energy expenditure and preventing diet-induced obesity. Zfp516 may represent a future target for obesity therapeutics.


Subject(s)
Adipose Tissue, Brown/physiology , Adipose Tissue, White/physiology , Ion Channels/genetics , Mitochondrial Proteins/genetics , Trans-Activators/physiology , Adipogenesis , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Animals , Cold-Shock Response , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Ion Channels/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins/metabolism , Muscle Development , Phenotype , Promoter Regions, Genetic , Protein Binding , Thermogenesis , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Uncoupling Protein 1
6.
Dev Dyn ; 239(9): 2509-19, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20730906

ABSTRACT

Morphogenesis of the Drosophila embryonic trachea involves a stereotyped pattern of epithelial tube branching and fusion. Here, we report unexpected phenotypes resulting from maternal and zygotic (M/Z) loss of the homophilic cell adhesion molecule Echinoid (Ed), as well as the subcellular localization of Ed in the trachea. ed(M/Z) embryos have convoluted trachea reminiscent of septate junction (SJ) and luminal matrix mutants. However, Ed does not localize to SJs, and ed(M/Z) embryos have intact SJs and show normal luminal accumulation of the matrix-modifying protein Vermiform. Surprisingly, tracheal length is not increased in ed(M/Z) mutants, but a previously undescribed combination of reduced intersegmental spacing and deep epidermal grooves produces a convoluted tracheal phenotype. In addition, ed(M/Z) mutants have unique fusion defects involving supernumerary fusion cells, ectopic fusion events and atypical branch breaks. Tracheal-specific expression of Ed rescues these fusion defects, indicating that Ed acts in trachea to control fusion cell fate.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Fusion , Drosophila Proteins/metabolism , Drosophila melanogaster , Morphogenesis/physiology , Repressor Proteins/metabolism , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Cell Adhesion Molecules/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/embryology , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/physiology , Phenotype , Repressor Proteins/genetics , Trachea/anatomy & histology , Trachea/embryology , Wnt1 Protein/metabolism
7.
Curr Biol ; 20(1): 55-61, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20022244

ABSTRACT

Regulation of epithelial tube size is critical for organ function. However, the mechanisms of tube size control remain poorly understood. In the Drosophila trachea, tube dimensions are regulated by a luminal extracellular matrix (ECM). ECM organization requires apical (luminal) secretion of the protein Vermiform (Verm), which depends on the basolateral septate junction (SJ). Here, we show that apical and basolateral epithelial polarity proteins interact to control tracheal tube size independently of the Verm pathway. Mutations in yurt (yrt) and scribble (scrib), which encode SJ-associated polarity proteins, cause an expansion of tracheal tubes but do not disrupt Verm secretion. Reducing activity of the apical polarity protein Crumbs (Crb) suppresses the length defects in yrt but not scrib mutants, suggesting that Yrt acts by negatively regulating Crb. Conversely, Crb overexpression increases tracheal tube dimensions. Reducing crb dosage also rescues tracheal size defects caused by mutations in coracle (cora), which encodes an SJ-associated polarity protein. In addition, crb mutations suppress cora length defects without restoring Verm secretion. Together, these data indicate that Yrt, Cora, Crb, and Scrib operate independently of the Verm pathway. Our data support a model in which Cora and Yrt act through Crb to regulate epithelial tube size.


Subject(s)
Drosophila Proteins/physiology , Drosophila/embryology , Drosophila/physiology , Trachea/embryology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Body Patterning/physiology , Drosophila/genetics , Drosophila Proteins/genetics , Extracellular Matrix/physiology , Genes, Insect , Membrane Proteins/genetics , Membrane Proteins/physiology , Models, Biological , Mutation , Organ Size , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology
8.
Nature ; 459(7250): 1141-5, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19553998

ABSTRACT

The integrity of polarized epithelia is critical for development and human health. Many questions remain concerning the full complement and the function of the proteins that regulate cell polarity. Here we report that the Drosophila FERM proteins Yurt (Yrt) and Coracle (Cora) and the membrane proteins Neurexin IV (Nrx-IV) and Na(+),K(+)-ATPase are a new group of functionally cooperating epithelial polarity proteins. This 'Yrt/Cora group' promotes basolateral membrane stability and shows negative regulatory interactions with the apical determinant Crumbs (Crb). Genetic analyses indicate that Nrx-IV and Na(+),K(+)-ATPase act together with Cora in one pathway, whereas Yrt acts in a second redundant pathway. Moreover, we show that the Yrt/Cora group is essential for epithelial polarity during organogenesis but not when epithelial polarity is first established or during terminal differentiation. This property of Yrt/Cora group proteins explains the recovery of polarity in embryos lacking the function of the Lethal giant larvae (Lgl) group of basolateral polarity proteins. We also find that the mammalian Yrt orthologue EPB41L5 (also known as YMO1 and Limulus) is required for lateral membrane formation, indicating a conserved function of Yrt proteins in epithelial polarity.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Epithelium/physiology , Membrane Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Cell Polarity , Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epithelium/embryology , Gene Knockdown Techniques , Membrane Proteins/genetics , Mutation , Phenotype , Sodium-Potassium-Exchanging ATPase/genetics
9.
Hum Genet ; 126(3): 431-47, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19455355

ABSTRACT

Mutations affecting the Na(+), K(+) ATPase alpha subunit have been implicated in at least two distinct human diseases, rapid-onset dystonia Parkinsonism (RDP), and familial hemiplegic migraine (FHM). Over 40 mutations have been mapped to the human ATP1A2 and ATP1A3 genes and are known to result in RDP, FHM or a variant of FHM with neurological complications. To develop a genetically tractable model system for investigating the role of the Na(+), K(+) ATPase in neural pathologies we performed genetic screens in Drosophila melanogaster to isolate loss-of-function alleles affecting the Na(+), K(+) ATPase alpha subunit. Flies heterozygous for these mutations all exhibit reduced respiration, consistent with a loss-of-function in the major ATPase. However, these mutations do not affect all functions of the Na(+), K(+) ATPase alpha protein since embryos homozygous for these mutations have normal septate junction paracellular barrier function and tracheal morphology. Importantly, all of these mutations cause neurological phenotypes and, akin to the mutations that cause RDP and FHM, these new alleles are missense mutations. All of these alleles exhibit progressive stress-induced locomotor impairment suggesting neuromuscular dysfunction, yet neurodegeneration is observed in an allele-specific manner. Surprisingly, studies of longevity demonstrate that mild hypomorphic mutations in the sodium pump significantly improve longevity, which was verified using the Na(+), K(+) ATPase antagonist ouabain. The isolation and characterization of a series of new missense alleles of ATPalpha in Drosophila provides the foundation for further studies of these neurological diseases and the role of sodium pump impairment in animal longevity.


Subject(s)
Mutation , Nervous System Diseases/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Alleles , Animals , Behavior, Animal , Drosophila melanogaster , Heterozygote , Longevity , Models, Genetic , Mutation, Missense , Neurodegenerative Diseases/genetics , Sequence Analysis, DNA , Trachea/metabolism
11.
Development ; 134(5): 999-1009, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17267446

ABSTRACT

Epithelial tubes are the functional units of many organs, but little is known about how tube sizes are established. Using the Drosophila tracheal system as a model, we previously showed that mutations in varicose (vari) cause tubes to become elongated without increasing cell number. Here we show vari is required for accumulation of the tracheal size-control proteins Vermiform and Serpentine in the tracheal lumen. We also show that vari is an essential septate junction (SJ) gene encoding a membrane associated guanylate kinase (MAGUK). In vivo analyses of domains important for MAGUK scaffolding functions demonstrate that while the Vari HOOK domain is essential, the L27 domain is dispensable. Phylogenetic analyses reveal that Vari helps define a new MAGUK subgroup that includes mammalian PALS2. Importantly, both Vari and PALS2 are basolateral, and the interaction of Vari with the cell-adhesion protein Neurexin IV parallels the interaction of PALS2 and another cell-adhesion protein, Necl-2. Vari therefore bolsters the similarity between Drosophila and vertebrate epithelial basolateral regions, which had previously been limited to the common basolateral localization of Scrib, Dlg and Lgl, proteins required for epithelial polarization at the beginning of embryogenesis. However, by contrast to Scrib, Dlg and Lgl, Vari is not required for cell polarity but rather is part of a cell-adhesion complex. Thus, Vari fundamentally extends the similarity of Drosophila and vertebrate basolateral regions from sharing only polarity complexes to sharing both polarity and cell-adhesion complexes.


Subject(s)
Drosophila Proteins/physiology , Drosophila/physiology , Guanylate Cyclase/physiology , Intercellular Junctions/metabolism , Membrane Proteins/physiology , Amidohydrolases/metabolism , Amino Acid Sequence , Animals , Cell Adhesion , Cell Adhesion Molecules, Neuronal/metabolism , Cell Polarity , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Guanylate Cyclase/genetics , Guanylate Kinases/genetics , Guanylate Kinases/physiology , Membrane Proteins/genetics , Molecular Sequence Data , Phylogeny , Trachea/growth & development , Trachea/physiology
12.
Development ; 134(1): 147-55, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164420

ABSTRACT

The heterodimeric Na,K-ATPase has been implicated in vertebrate and invertebrate epithelial cell junctions, morphogenesis and oncogenesis, but the mechanisms involved are unclear. We previously showed that the Drosophila Na,K-ATPase is required for septate junction (SJ) formation and that of the three beta-subunit loci, only Nrv2 isoforms support epithelial SJ barrier function and tracheal tube-size control. Here we show that Nrv1 is endogenously co-expressed with Nrv2 in the epidermis and tracheal system, but Nrv1 has a basolateral localization and appears to be excluded from the Nrv2-containing SJs. When the normally neuronal Nrv3 is expressed in epithelial cells, it does not associate with SJs. Thus, the beta-subunit is a key determinant of Na,K-ATPase subcellular localization as well as function. However, localization of the Na,K-ATPase to SJs is not sufficient for junctional activity because although several Nrv2/Nrv3 chimeric beta-subunits localize to SJs, only those containing the extracellular domain of Nrv2 have junctional activity. Junctional activity is also specific to different alpha-subunit isoforms, with only some isoforms from the major alpha-subunit locus being able to provide full barrier function and produce normal tracheal tubes. Importantly, mutations predicted to inactivate ATPalpha catalytic function do not compromise junctional activity, demonstrating that the Drosophila Na,K-ATPase has an ion-pump-independent role in junction formation and tracheal morphogenesis. These results define new functions for the intensively studied Na,K-ATPase. Strikingly, the rat alpha1 isoform has full junctional activity and can rescue Atpalpha-null mutants to viability, suggesting that the Na,K-ATPase has an evolutionarily conserved role in junction formation and function.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/growth & development , Epithelium/metabolism , Intercellular Junctions/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Trachea/embryology , Alternative Splicing , Animals , Animals, Genetically Modified , Drosophila/embryology , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Embryo, Nonmammalian , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Structure, Tertiary , Sodium-Potassium-Exchanging ATPase/genetics
13.
Genetics ; 174(4): 2255-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17057238

ABSTRACT

We report new w- fluorescent balancers scorable from stage 13 through adulthood that bear a nuclear-localized yellow fluorescent protein marker directly driven by dfd and GMR enhancer elements. The utility of this marker is enhanced by identification of an anti-GFP/yellow fluorescent protein (YFP) serum that is compatible with heat fixation.


Subject(s)
Bacterial Proteins/genetics , Chromosomes/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Genetic Markers , Homeodomain Proteins/genetics , Luminescent Proteins/genetics , Animals , Bacterial Proteins/metabolism , Cell Nucleus/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Luminescent Proteins/metabolism , Molecular Sequence Data , Protein Transport
14.
Curr Biol ; 15(2): R70-2, 2005 Jan 26.
Article in English | MEDLINE | ID: mdl-15668162

ABSTRACT

In animals, rearrangements of the junctions between cells are fundamental to morphogenesis. Recent work has revealed the acrobatics that Drosophila tracheal cells perform as they intercalate to form unicellular tubes.


Subject(s)
Adherens Junctions/physiology , Drosophila/embryology , Gene Expression Regulation, Developmental , Morphogenesis , Respiratory System/cytology , Animals , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Epithelium/embryology , Extracellular Matrix Proteins/metabolism , Homeodomain Proteins/metabolism , Respiratory System/embryology , Signal Transduction/physiology , Transcription Factors/metabolism
16.
Development ; 130(20): 4963-74, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12930776

ABSTRACT

Although the correct architecture of epithelial tubes is crucial for the function of organs such as the lung, kidney and vascular system, little is known about the molecular mechanisms that control tube size. We show that mutations in the ATPalpha alpha and nrv2 beta subunits of the Na+/K+ ATPase cause Drosophila tracheal tubes to have increased lengths and expanded diameters. ATPalpha and nrv2 mutations also disrupt stable formation of septate junctions, structures with some functional and molecular similarities to vertebrate tight junctions. The Nrv2 beta subunit isoforms have unique tube size and junctional functions because Nrv2, but not other Drosophila Na+/K+ ATPase beta subunits, can rescue nrv2 mutant phenotypes. Mutations in known septate junctions genes cause the same tracheal tube-size defects as ATPalpha and nrv2 mutations, indicating that septate junctions have a previously unidentified role in epithelial tube-size control. Double mutant analyses suggest that tube-size control by septate junctions is mediated by at least two discernable pathways, although the paracellular diffusion barrier function does not appear to involved because tube-size control and diffusion barrier function are genetically separable. Together, our results demonstrate that specific isoforms of the Na+/K+ ATPase play a crucial role in septate junction function and that septate junctions have multiple distinct functions that regulate paracellular transport and epithelial tube size.


Subject(s)
Drosophila/growth & development , Epithelium/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Sodium-Potassium-Exchanging ATPase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...