Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 7(15): 19575-88, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26988909

ABSTRACT

It has long been known that the resting potential of tumor cells is depolarized relative to their normal counterparts. More recent work has provided evidence that resting potential is not just a readout of cell state: it regulates cell behavior as well. Thus, the ability to control resting potential in vivo would provide a powerful new tool for the study and treatment of tumors, a tool capable of revealing living-state physiological information impossible to obtain using molecular tools applied to isolated cell components. Here we describe the first use of optogenetics to manipulate ion-flux mediated regulation of membrane potential specifically to prevent and cause regression of oncogene-induced tumors. Injection of mutant-KRAS mRNA induces tumor-like structures with many documented similarities to tumors, in Xenopus tadpoles. We show that expression and activation of either ChR2D156A, a blue-light activated cation channel, or Arch, a green-light activated proton pump, both of which hyperpolarize cells, significantly lowers the incidence of KRAS tumor formation. Excitingly, we also demonstrate that activation of co-expressed light-activated ion translocators after tumor formation significantly increases the frequency with which the tumors regress in a process called normalization. These data demonstrate an optogenetic approach to dissect the biophysics of cancer. Moreover, they provide proof-of-principle for a novel class of interventions, directed at regulating cell state by targeting physiological regulators that can over-ride the presence of mutations.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/radiation effects , Light , Optogenetics/methods , Animals , Antineoplastic Agents/pharmacology , Archaeal Proteins/genetics , Cell Transformation, Neoplastic/drug effects , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/radiation effects , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Membrane Potentials/radiation effects , Mutation , Optogenetics/instrumentation , Proto-Oncogene Proteins p21(ras)/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rhodopsin/genetics , Xenopus laevis
2.
Oncotarget ; 5(10): 3287-306, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24830454

ABSTRACT

The microenvironment is increasingly recognized as a crucial aspect of cancer. In contrast and complement to the field's focus on biochemical factors and extracellular matrix, we characterize a novel aspect of host:tumor interaction - endogenous bioelectric signals among non-excitable somatic cells. Extending prior work focused on the bioelectric state of cancer cells themselves, we show for the first time that the resting potentials of distant cells are critical for oncogene-dependent tumorigenesis. In the Xenopus laevis tadpole model, we used human oncogenes such as mutant KRAS to drive formation of tumor-like structures that exhibited overproliferation, increased nuclear size, hypoxia, acidity, and leukocyte attraction. Remarkably, misexpression of hyperpolarizing ion channels at distant sites within the tadpole significantly reduced the incidence of these tumors. The suppression of tumorigenesis could also be achieved by hyperpolarization using native CLIC1 chloride channels, suggesting a treatment modality not requiring gene therapy. Using a dominant negative approach, we implicate HDAC1 as the mechanism by which resting potential changes affect downstream cell behaviors. Based on published data on the voltage-mediated changes of butyrate flux through the SLC5A8 transporter, we present a model linking resting potentials of host cells to the ability of oncogenes to initiate tumorigenesis. Antibiotic data suggest that the relevant butyrate is generated by a native bacterial species, identifying a novel link between the microbiome and cancer that is mediated by alterations in bioelectric signaling.


Subject(s)
Carcinogenesis , Membrane Potentials/physiology , Oncogenes/physiology , Tumor Microenvironment/physiology , Animals , Embryo, Nonmammalian , Humans , Immunohistochemistry , Xenopus laevis
3.
Front Physiol ; 5: 519, 2014.
Article in English | MEDLINE | ID: mdl-25646081

ABSTRACT

In addition to the immediate microenvironment, long-range signaling may be an important component of cancer. Molecular-genetic analyses have implicated gap junctions-key mediators of cell-cell communication-in carcinogenesis. We recently showed that the resting voltage potential of distant cell groups is a key determinant of metastatic transformation and tumor induction. Here, we show in the Xenopus laevis model that gap junctional communication (GJC) is a modulator of the long-range bioelectric signaling that regulates tumor formation. Genetic disruption of GJC taking place within tumors, within remote host tissues, or between the host and tumors significantly lowers the incidence of tumors induced by KRAS mutations. The most pronounced suppression of tumor incidence was observed upon GJC disruption taking place farther away from oncogene-expressing cells, revealing a role for GJC in distant cells in the control of tumor growth. In contrast, enhanced GJC communication through the overexpression of wild-type connexin Cx26 increased tumor incidence. Our data confirm a role for GJC in tumorigenesis, and reveal that this effect is non-local. Based on these results and on published data on movement of ions through GJs, we present a quantitative model linking the GJC coupling and bioelectrical state of cells to the ability of oncogenes to initiate tumorigenesis. When integrated with data on endogenous bioelectric signaling during left-right patterning, the model predicts differential tumor incidence outcomes depending on the spatial configurations of gap junction paths relative to tumor location and major anatomical body axes. Testing these predictions, we found that the strongest influence of GJ modulation on tumor suppression by hyperpolarization occurred along the embryonic left-right axis. Together, these data reveal new, long-range aspects of cancer control by the host's physiological parameters.

4.
Dis Model Mech ; 6(3): 595-607, 2013 May.
Article in English | MEDLINE | ID: mdl-23471912

ABSTRACT

Understanding mechanisms that orchestrate cell behavior into appropriately patterned tissues and organs within the organism is an essential element of preventing, detecting and treating cancer. Bioelectric signals (resting transmembrane voltage potential gradients in all cells) underlie an important and broadly conserved set of control mechanisms that regulate pattern formation. We tested the role of transmembrane potential in tumorigenesis mediated by canonical oncogenes in Xenopus laevis. Depolarized membrane potential (Vmem) was a characteristic of induced tumor-like structures (ITLSs) generated by overexpression of Gli1, Kras(G12D), Xrel3 or p53(Trp248). This bioelectric signature was also present in precursor ITLS sites. Vmem is a bioelectric marker that reveals ITLSs before they become histologically and morphologically apparent. Moreover, voltage was functionally important: overexpression of hyperpolarizing ion transporters caused a return to normal Vmem and significantly reduced ITLS formation in vivo. To characterize the molecular mechanism by which Vmem change regulates ITLS phenotypes, we performed a suppression screen. Vmem hyperpolarization was transduced into downstream events via Vmem-regulated activity of SLC5A8, a sodium-butyrate exchanger previously implicated in human cancer. These data indicate that butyrate, a histone deacetylase (HDAC) inhibitor, might be responsible for transcriptional events that mediate suppression of ITLSs by hyperpolarization. Vmem is a convenient cellular parameter by which tumors induced by human oncogenes can be detected in vivo and represents a new diagnostic modality. Moreover, control of resting membrane potential is functionally involved in the process by which oncogene-bearing cells depart from normal morphogenesis programs to form tumors. Modulation of Vmem levels is a novel and promising strategy for tumor normalization.


Subject(s)
Cell Transformation, Neoplastic/pathology , Disease Models, Animal , Membrane Potentials , Xenopus laevis/physiology , Animals , Butyrates/metabolism , Embryo, Nonmammalian/pathology , Genetic Testing , Humans , Models, Biological , Mutant Proteins/metabolism , Oncogenes , Suppression, Genetic , Tumor Suppressor Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics
5.
Cold Spring Harb Protoc ; 2012(6): 683-90, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22661444

ABSTRACT

Xenopus laevis is an ideal model system for investigating dynamic morphogenetic processes during embryogenesis, regeneration, and homeostasis. Our understanding of these events has been greatly facilitated by lineage labeling, that is, marking a cell or a group of cells and all their descendants using vital dyes, fluorescent molecules, or transplantation techniques. Unfortunately, these strategies are limited in their spatiotemporal resolution: They do not allow long-term dynamic in vivo imaging, are generally invasive, and labeling is restricted to cells on the surface. Genetically encoded fluorescent proteins (FPs), on the other hand, provide excellent alternative methods to traditional lineage labeling, enabling labeling with high spatiotemporal resolution and tracking of cellular and subcellular structures to study patterning events. Over the past decade, FPs have evolved to allow fine control of their spectral properties (in a defined region of interest) for greater labeling specificity. One example is EosFP, which is a protein cloned from the scleractinian coral Lobophyllia hemprichii that can be photoconverted from green to red fluorescence state with near-ultraviolet (UV) light irradiation. Here, we describe EosFP-photoconversion of Xenopus embryos to track cells during developmental and regenerative processes using a metal-halide- or xenon-arc-based fluorescent microscope system, which provides a simpler, less expensive alternative to photoconversion using laser microscopy.


Subject(s)
Cell Movement , Embryo, Nonmammalian/cytology , Fluorescence , Luminescent Proteins/metabolism , Staining and Labeling/methods , Xenopus laevis/embryology , Animals , Luminescent Proteins/genetics , Ultraviolet Rays
6.
Cold Spring Harb Protoc ; 2012(4): 447-52, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22474651

ABSTRACT

Xenopus laevis is an ideal model system for investigating the mechanisms of pattern formation. The ability to express exogenous mRNA or introduce morpholinos into cleavage-stage Xenopus embryos has allowed gain- and loss-of-function experiments that reveal molecular-genetic control of development and regeneration. However, injection of mRNAs into cleavage-stage embryos provides limited spatio-temporal control: It is difficult to limit targeting to small regions (e.g., inducing foci of expression) and the fate map does not facilitate targeting some tissues, such as those of the tail. Likewise, early injection can result in unwanted developmental defects because mRNA can be translated long before the desired time point. These are especially important limitations when studying developmental and regenerative processes during the gastrula to tailbud stages. Although transgenic techniques allow precise control over spatio-temporal expression of genes when the appropriate promoter is available, the process of creating stable transgenic animals is time-consuming. Electroporation provides an alternative method for delivering mRNA and other nucleic acids, enabling the targeting of single cells or groups of cells at any stage of development. This protocol describes detailed electroporation parameters for the transfection of mRNA into a wide range of tissues in embryos at gastrula to tailbud stages, with high efficiency and expression as early as 4 h post electroporation.


Subject(s)
Electroporation/methods , RNA, Messenger/genetics , RNA, Messenger/metabolism , Xenopus laevis/embryology , Animals , Embryo, Nonmammalian/embryology , Xenopus laevis/genetics , Xenopus laevis/growth & development
7.
Mol Cell ; 35(1): 82-92, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19595718

ABSTRACT

Large-scale expansions of DNA repeats are implicated in numerous hereditary disorders in humans. We describe a yeast experimental system to analyze large-scale expansions of triplet GAA repeats responsible for the human disease Friedreich's ataxia. When GAA repeats were placed into an intron of the chimeric URA3 gene, their expansions caused gene inactivation, which was detected on the selective media. We found that the rates of expansions of GAA repeats increased exponentially with their lengths. These rates were only mildly dependent on the repeat's orientation within the replicon, whereas the repeat-mediated replication fork stalling was exquisitely orientation dependent. Expansion rates were significantly elevated upon inactivation of the replication fork stabilizers, Tof1 and Csm3, but decreased in the knockouts of postreplication DNA repair proteins, Rad6 and Rad5, and the DNA helicase Sgs1. We propose a model for large-scale repeat expansions based on template switching during replication fork progression through repetitive DNA.


Subject(s)
Friedreich Ataxia/genetics , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/genetics , Yeasts/genetics , DNA Replication/genetics , Gene Expression Regulation, Fungal , Humans , Introns/genetics , Iron-Binding Proteins/genetics , Plasmids/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Frataxin
SELECTION OF CITATIONS
SEARCH DETAIL
...