Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Thorac Oncol ; 10(10): 1421-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25719266

ABSTRACT

BACKGROUND: Treatment guidelines provide recommendations but cannot account for the wide variability in patient-tumor characteristics in individual patients. We developed an on-line interactive decision tool to provide expert recommendations for specific patient scenarios in the first-line and maintenance settings for advanced non-small-cell lung cancer. We sought to determine how providing expert feedback would influence clinical decision-making. METHOD: Five lung cancer experts selected treatment for 96 different patient cases based on patient and/or tumor-specific features. These data were used to develop an on-line decision tool. Participant physicians entered variables for their patient scenario with treatment choices, and then received expert treatment recommendations for that scenario. To determine the impact on decision-making, users were asked whether the expert feedback impacted their original plan. RESULTS: A total of 442 individual physicians, of which 88% were from outside the United States, entered 653 cases, with report on impact in 389 cases. Expert feedback affected treatment choice in 73% of cases (23% changed and 50% confirmed decisions). For cases with epidermal growth factor receptor (EGFR) mutation or anaplastic lymphoma kinase (ALK) fusion, all experts selected targeted therapy whereas 51% and 58% of participants did not. Greater variability was seen between experts and participants for cases involving EGFR or ALK wild-type tumors. Participants were 2.5-fold more likely to change to expert recommended therapy for ALK fusions than for EGFR mutations (p = 0.017). CONCLUSION: This online tool for treatment decision-making resulted in a positive influence on clinician's decisions. This approach offers opportunities for improving quality of care and meets an educational need in application of new therapeutic paradigms.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Decision Making , Lung Neoplasms/therapy , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Humans , Internet , Lung Neoplasms/pathology , Telemedicine
2.
PLoS One ; 4(8): e6844, 2009 Aug 31.
Article in English | MEDLINE | ID: mdl-19718436

ABSTRACT

The four sperm-specific CatSper ion channel proteins are required for hyperactivated motility and male fertility, and for Ca(2+) entry evoked by alkaline depolarization. In the absence of external Ca(2+), Na(+) carries current through CatSper channels in voltage-clamped sperm. Here we show that CatSper channel activity can be monitored optically with the [Na(+)](i)-reporting probe SBFI in populations of intact sperm. Removal of external Ca(2+) increases SBFI signals in wild-type but not CatSper2-null sperm. The rate of the indicated rise of [Na(+)](i) is greater for sperm alkalinized with NH(4)Cl than for sperm acidified with propionic acid, reflecting the alkaline-promoted signature property of CatSper currents. In contrast, the [Na(+)](i) rise is slowed by candidate CatSper blocker HC-056456 (IC(50) approximately 3 microM). HC-056456 similarly slows the rise of [Ca(2+)](i) that is evoked by alkaline depolarization and reported by fura-2. HC-056456 also selectively and reversibly decreased CatSper currents recorded from patch-clamped sperm. HC-056456 does not prevent activation of motility by HCO(3) (-) but does prevent the development of hyperactivated motility by capacitating incubations, thus producing a phenocopy of the CatSper-null sperm. When applied to hyperactivated sperm, HC-056456 causes a rapid, reversible loss of flagellar waveform asymmetry, similar to the loss that occurs when Ca(2+) entry through the CatSper channel is terminated by removal of external Ca(2+). Thus, open CatSper channels and entry of external Ca(2+) through them sustains hyperactivated motility. These results indicate that pharmacological targeting of the CatSper channel may impose a selective late-stage block to fertility, and that high-throughput screening with an optical reporter of CatSper channel activity may identify additional selective blockers with potential for male-directed contraception.


Subject(s)
Calcium Channels/drug effects , Spermatozoa/drug effects , Animals , Calcium/metabolism , Ion Transport , Male , Mice , Sodium/metabolism
3.
Biol Chem ; 390(1): 67-73, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19007311

ABSTRACT

Apolipoprotein E receptor 2 (Apoer2) is a multifunctional transport and signaling receptor that regulates the uptake of selenium into the mouse brain and testis through endocytosis of selenoprotein P (Sepp1). Mice deficient in Apoer2 or Sepp1 are infertile, with kinked and hypomotile spermatozoa. They also develop severe neurological defects on a low selenium diet, due to a profound impairment of selenium uptake. Little is known about the function of Apoer2 in the testis beyond its role as a Sepp1 receptor. By contrast, in the brain, Apoer2 is an essential component of the Reelin signaling pathway, which is required for proper neuronal organization and synapse function. Using knock-in mice, we have functionally dissociated the signaling motifs in the Apoer2 cytoplasmic domain from Sepp1 uptake. Selenium concentration of brain and testis was normal in the knock-in mutants, in contrast to Apoer2 knock-outs. Thus, the neurological defects in the signaling impaired knock-in mice are not caused by a selenium uptake defect, but instead are a direct consequence of a disruption of the Reelin signal. Reduced sperm motility was observed in some of the knock-in mice, indicating a novel signaling role for Apoer2 in sperm development and function that is independent of selenium uptake.


Subject(s)
Endocytosis , Intracellular Space/metabolism , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/metabolism , Selenium/metabolism , Signal Transduction , Animals , Biological Transport , Brain/metabolism , Cytoplasm/metabolism , Gene Knock-In Techniques , LDL-Receptor Related Proteins , Male , Mice , Mutation , Protein Structure, Tertiary , Receptors, Lipoprotein/genetics , Reelin Protein , Sperm Motility , Spermatozoa/classification , Spermatozoa/cytology , Spermatozoa/metabolism , Testis/metabolism
4.
Mol Immunol ; 45(14): 3756-66, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18614237

ABSTRACT

T cell receptor signaling processes are controlled by the integrated actions of families of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPases). Several distinct cytosolic protein tyrosine phosphatases have been described that are able to negatively regulate TCR signaling pathways, including SHP-1, SHP-2, PTPH1, and PEP. Using PTPase substrate-trapping mutants and wild type enzymes, we determined that PTPN4/PTP-MEG1, a PTPH1-family member, could complex and dephosphorylate the ITAMs of the TCR zeta subunit. In addition, the substrate-trapping derivative augmented basal and TCR-induced activation of NF-kappaB in T cells. To characterize the contribution of this PTPase in T cells, we developed PTPN4-deficient mice. T cell development and TCR signaling events were comparable between wild type and PTPN4-deficient animals. The magnitude and duration of TCR-regulated ITAM phosphorylation, as well as overall protein phosphorylation, was unaltered in the absence of PTPN4. Finally, Th1- and Th2-derived cytokines and in vivo immune responses to Listeria monocytogenes were equivalent between wild type and PTPN4-deficient mice. These findings suggest that additional PTPases are involved in controlling ITAM phosphorylations.


Subject(s)
NF-kappa B/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 4/physiology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/physiology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Jurkat Cells , Kidney/cytology , Mice , NF-kappa B/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 4/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 4/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 4/metabolism , Receptors, Antigen, T-Cell/immunology , Transfection
6.
Proc Natl Acad Sci U S A ; 104(22): 9325-30, 2007 May 29.
Article in English | MEDLINE | ID: mdl-17517652

ABSTRACT

We previously identified a sperm-specific Na(+)/H(+) exchanger (sNHE) principally localized to the flagellum. Disruption of the sNHE gene in mice resulted in absolute male infertility associated with a complete loss of sperm motility. Here, we show that the sNHE-null spermatozoa fail to develop the cAMP-dependent protein tyrosine phosphorylation that coincides with the functional maturation occurring upon incubation in capacitating conditions in vitro. Both the sperm motility defect and the lack of induced protein tyrosine phosphorylation are rescued by the addition of cell-permeable cAMP analogs, suggesting that cAMP metabolism is impaired in spermatozoa lacking sNHE. Our analyses of the bicarbonate-dependent soluble adenylyl cyclase (sAC) signaling pathway in sNHE-null sperm cells reveal that sNHE is required for the expression of full-length sAC, and that it is important for the bicarbonate stimulation of sAC activity in spermatozoa. Furthermore, both codependent expression and coimmunoprecipitation experiments indicate that sNHE and sAC associate with each other. Thus, these two proteins appear to be components of a signaling complex at the sperm flagellar plasma membrane. We propose that the formation of this complex efficiently modulates intracellular pH and bicarbonate levels through the rapid and effective control of sAC and sNHE activities to facilitate sperm motility regulation.


Subject(s)
Adenylyl Cyclases/metabolism , Bicarbonates/metabolism , Gene Expression Regulation , Sodium-Hydrogen Exchangers/metabolism , Spermatozoa/metabolism , Adenylyl Cyclases/genetics , Animals , Cell Extracts , Cell Line , Cyclic AMP/metabolism , Enzyme Activation , Humans , Male , Mice , Phosphotyrosine/metabolism , Protein Binding , Sensitivity and Specificity , Sodium-Hydrogen Exchangers/genetics , Sperm Motility , Spermatozoa/cytology
7.
Proc Natl Acad Sci U S A ; 104(4): 1219-23, 2007 Jan 23.
Article in English | MEDLINE | ID: mdl-17227845

ABSTRACT

Mammalian spermatozoa become motile at ejaculation, but before they can fertilize the egg, they must acquire more thrust to penetrate the cumulus and zona pellucida. The forceful asymmetric motion of hyperactivated spermatozoa requires Ca2+ entry into the sperm tail by an alkalinization-activated voltage-sensitive Ca2+-selective current (ICatSper). Hyperactivation requires CatSper1 and CatSper2 putative ion channel genes, but the function of two other related genes (CatSper3 and CatSper4) is not known. Here we show that targeted disruption of murine CatSper3 or CatSper4 also abrogated ICatSper, sperm cell hyperactivated motility and male fertility but did not affect spermatogenesis or initial motility. Direct protein interactions among CatSpers, the sperm specificity of these proteins, and loss of ICatSper in each of the four CatSper-/- mice indicate that CatSpers are highly specialized flagellar proteins.


Subject(s)
Calcium Channels/physiology , Fertility/physiology , Protein Isoforms/physiology , Sperm Motility/physiology , Animals , Male , Mice , Molecular Sequence Data
8.
Mol Cell Endocrinol ; 250(1-2): 84-92, 2006 May 16.
Article in English | MEDLINE | ID: mdl-16413670

ABSTRACT

Successful natural reproduction normally requires vigorously motile spermatozoa. Using a signal peptide trapping strategy, we identified two new genes, a putative sperm Na+/H+ exchanger (sNHE) and the putative cation channel CatSper2, with unique and essential roles in sperm motility. Disruption of the sNHE or CatSper2 genes in mice caused male infertility due to immotile spermatozoa or failed motility hyperactivation, respectively, without other apparent abnormalities. The immotility phenotype of the sNHE null spermatozoa appears to result from an intimate association of sNHE and the atypical adenylyl cyclase (sAC), while a failure of calcium entry requiring an apparent CatSper1 and -2 heteromeric ion channel correlates with a hyperactivation defect in these null animals. The specific expression of sNHE and the CatSpers in spermatozoa and their required function in cell motility make them excellent potential targets for the development of novel male contraceptives.


Subject(s)
Calcium Channels/metabolism , Infertility, Male/genetics , Seminal Plasma Proteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Sperm Motility/genetics , Spermatozoa/physiology , Adenylyl Cyclases/metabolism , Animals , Calcium Channels/analysis , Calcium Channels/genetics , Cell Membrane/chemistry , Cell Membrane/metabolism , Contraceptive Agents, Male/pharmacology , Fertility/drug effects , Fertility/genetics , Male , Mice , Mutation , Seminal Plasma Proteins/analysis , Seminal Plasma Proteins/genetics , Signal Transduction , Sodium-Hydrogen Exchangers/analysis , Sodium-Hydrogen Exchangers/genetics , Spermatozoa/metabolism
9.
J Biol Chem ; 280(37): 32238-44, 2005 Sep 16.
Article in English | MEDLINE | ID: mdl-16036917

ABSTRACT

Among several candidate Ca(2+) entry channels in sperm, only CatSper1 and CatSper2 are known to have required roles in male fertility. Past work with CatSper1 null sperm indicates that a critical lesion in hyperactivated motility underlies the infertility phenotype and is associated with an absence of depolarization-evoked Ca(2+)entry. Here we show that failure of hyperactivation of CatSper2 null sperm similarly correlates with an absence of depolarization evoked Ca(2+) entry. Additional shared aspects of the phenotypes of CatSper1 and -2 null sperm include unperturbed regional distributions of conventional voltage-gated Ca(2+) channel proteins and robust acceleration of the flagellar beat by bicarbonate. Further study reveals that treatment of both wild-type and CatSper2 null sperm with procaine increases beat asymmetry, a characteristic of the flagellar waveform of hyperactivation. This partial rescue of the loss-of-hyperactivation phenotype suggests that an absence of CatSper2 precludes hyperactivation by preventing delivery of needed Ca(2+) messenger rather than by preventing flagellar responses to Ca(2+). CatSper2 null sperm also have an increased basal cAMP content and beat frequency. Protein kinase A inhibitor H89 lowers beat frequency to that of wild-type sperm, suggesting that CatSper2 is required for protein kinase A-mediated, tonic control of resting cAMP content. Relative to wild-type testis, CatSper1 and -2 null testes contain normal amounts of CatSper2 and -1 transcripts, respectively. However, CatSper1 null sperm lack CatSper2 protein and CatSper2 null sperm lack CatSper1 protein. Hence, stable expression of CatSper1 protein requires CatSper2 and vice versa. This co-dependent expression dictates identical loss-of-function sperm phenotypes for CatSper1 and -2 null mutants.


Subject(s)
Calcium Channels/genetics , Calcium Channels/physiology , Seminal Plasma Proteins/genetics , Seminal Plasma Proteins/physiology , Animals , Bicarbonates/pharmacology , Calcium/metabolism , Coloring Agents/pharmacology , Cyclic AMP/metabolism , Egtazic Acid/chemistry , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/pharmacology , Immunoblotting , Immunohistochemistry , Isoquinolines/pharmacology , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Phenotype , Procaine/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Seminal Plasma Proteins/chemistry , Sperm Capacitation , Spermatozoa/metabolism , Sulfonamides/pharmacology , Testis/metabolism
10.
Proc Natl Acad Sci U S A ; 100(25): 14869-74, 2003 Dec 09.
Article in English | MEDLINE | ID: mdl-14657366

ABSTRACT

Elevations of sperm Ca2+ seem to be responsible for an asymmetric form of motility called hyperactivation, which is first seen near the time of fertilization. The mechanism by which intracellular Ca2+ concentrations increase remains unknown despite considerable investigation. Although several prototypical voltage-gated calcium channels are present in spermatozoa, they are not essential for motility. Furthermore, the forward velocity and percentage of motility of spermatozoa are associated with infertility, but their importance relative to hyperactivation also remains unknown. We show here that disruption of the gene for a recently described sperm-specific voltage-gated cation channel, CatSper2, fails to significantly alter sperm production, protein tyrosine phosphorylation that is associated with capacitation, induction of the acrosome reaction, forward velocity, or percentage of motility, yet CatSper2-/- males are completely infertile. The defect that we identify in the null sperm cells is a failure to acquire hyperactivated motility, which seems to render spermatozoa incapable of generating the "power" needed for penetration of the extracellular matrix of the egg. A loss of power is suggested also by experiments in which the viscosity of the medium was increased after incubation of spermatozoa in normal capacitating conditions. In high-viscosity medium, CatSper2-null spermatozoa lost the ability to swim forward, whereas wild-type cells continued to move forward. Thus, CatSper2 is responsible for driving hyperactivated motility, and, even with typical sperm forward velocities, fertilization is not possible in the absence of this highly active form of motility.


Subject(s)
Calcium Channels/physiology , Fertilization , Seminal Plasma Proteins/physiology , Sperm Motility , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cations , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Embryo, Mammalian/cytology , Exons , Female , Flagella/physiology , Genetic Vectors , Genotype , Immunoblotting , Infertility , Male , Mice , Mice, Transgenic , Models, Genetic , Phenotype , Phosphorylation , Phosphotyrosine/chemistry , Seminal Plasma Proteins/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism , Stem Cells/metabolism , Time Factors
11.
Nat Cell Biol ; 5(12): 1117-22, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14634667

ABSTRACT

It has long been speculated that intracellular pH is a critical regulator of both invertebrate and vertebrate sperm motility, and sodium-hydrogen exchange has been suggested as a mediator of such pH(i) regulation in various instances. Two sodium-hydrogen exchangers (NHE1 and NHE5) are expressed in spermatozoa. However, elimination of the NHE1 gene fails to cause infertility, suggesting that normal sperm function is maintained in NHE1-null animals. Here, we used a functionally unbiased signal peptide trap screen to identify a novel sperm-specific NHE. The NHE contains 14 predicted transmembrane segments, including a potential voltage sensor and a consensus cyclic nucleotide-binding motif. Testis histology, sperm numbers and morphology were normal, but NHE-null males were completely infertile with severely diminished sperm motility. The addition of ammonium chloride, which elevates intracellular pH, partially rescued the motility and fertility defects. Surprisingly, cyclic AMP analogues almost completely rescued the motility and infertility phenotypes. The existence of this new sperm NHE provides an attractive contraceptive target, given its cell-specific expression and absolute requirement for fertility.


Subject(s)
Fertility/physiology , Intracellular Fluid/metabolism , Sodium-Hydrogen Exchangers/isolation & purification , Sperm Motility/physiology , Spermatozoa/metabolism , Ammonium Chloride/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/enzymology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , DNA, Complementary/analysis , DNA, Complementary/genetics , Female , Hydrogen-Ion Concentration/drug effects , Intracellular Fluid/drug effects , Male , Mice , Molecular Sequence Data , Mutation/genetics , Protein Structure, Tertiary/physiology , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Sodium-Hydrogen Exchangers/genetics
12.
Dev Growth Differ ; 36(6): 615-620, 1994 Dec.
Article in English | MEDLINE | ID: mdl-37281849

ABSTRACT

The fertilization layer of Xenopus laevis is formed upon egg activation by the binding of the cortical granule lectin (CGL) to its ligand in the egg extracellular matrix. Using Western blotting methods with biotinylated CGL as a probe, oviductal tissue extracts were examined to determine the site of origin of the CGL ligand. Three glycoprotein ligands of Mr s= > 250,000, 160,000, and 90,000 (reduced samples) were localized to the pars convoluta oviduct immediately posterior to the pars recta oviduct. The binding of CGL to these glycoproteins was inhibited in the presence of 200 mM galactose, but not with 200 mM mannose indicating a specific lectin interaction. The Mr s= > 250,000 and 90,000 glycoproteins were linked by disulfide bonds. In addition, these ligands were secreted from a more anterior region of the pars convoluta oviduct than the Mr =160,000 ligand. No binding of CGL was detected to pars recta secretory granule lysate components. The highest molecular weight CGL ligand seen in the pars convoluta corresponded to the CGL ligand in isolated fertilization envelopes. Thus, the CGL ligand involved in the formation of the fertilization layer is a product of the pars convoluta oviduct.

SELECTION OF CITATIONS
SEARCH DETAIL
...