Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
Add more filters

Publication year range
1.
Cell ; 182(3): 545-562.e23, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32621799

ABSTRACT

Scar tissue size following myocardial infarction is an independent predictor of cardiovascular outcomes, yet little is known about factors regulating scar size. We demonstrate that collagen V, a minor constituent of heart scars, regulates the size of heart scars after ischemic injury. Depletion of collagen V led to a paradoxical increase in post-infarction scar size with worsening of heart function. A systems genetics approach across 100 in-bred strains of mice demonstrated that collagen V is a critical driver of postinjury heart function. We show that collagen V deficiency alters the mechanical properties of scar tissue, and altered reciprocal feedback between matrix and cells induces expression of mechanosensitive integrins that drive fibroblast activation and increase scar size. Cilengitide, an inhibitor of specific integrins, rescues the phenotype of increased post-injury scarring in collagen-V-deficient mice. These observations demonstrate that collagen V regulates scar size in an integrin-dependent manner.


Subject(s)
Cicatrix/metabolism , Collagen Type V/deficiency , Collagen Type V/metabolism , Heart Injuries/metabolism , Myocardial Contraction/genetics , Myofibroblasts/metabolism , Animals , Cicatrix/genetics , Cicatrix/physiopathology , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Collagen Type III/genetics , Collagen Type III/metabolism , Collagen Type V/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Fibrosis/genetics , Fibrosis/metabolism , Gene Expression Regulation/genetics , Integrins/antagonists & inhibitors , Integrins/genetics , Integrins/metabolism , Isoproterenol/pharmacology , Male , Mechanotransduction, Cellular/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Atomic Force/instrumentation , Microscopy, Electron, Transmission , Myocardial Contraction/drug effects , Myofibroblasts/cytology , Myofibroblasts/pathology , Myofibroblasts/ultrastructure , Principal Component Analysis , Proteomics , RNA-Seq , Single-Cell Analysis
2.
Nature ; 620(7975): 855-862, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532930

ABSTRACT

Patients from historically under-represented racial and ethnic groups are enrolled in cancer clinical trials at disproportionately low rates in the USA1-3. As these patients often have limited English proficiency4-7, we hypothesized that one barrier to their inclusion is the cost to investigators of translating consent documents. To test this hypothesis, we evaluated more than 12,000 consent events at a large cancer centre and assessed whether patients requiring translated consent documents would sign consent documents less frequently in studies lacking industry sponsorship (for which the principal investigator pays the translation costs) than for industry-sponsored studies (for which the translation costs are covered by the sponsor). Here we show that the proportion of consent events for patients with limited English proficiency in studies not sponsored by industry was approximately half of that seen in industry-sponsored studies. We also show that among those signing consent documents, the proportion of consent documents translated into the patient's primary language in studies without industry sponsorship was approximately half of that seen in industry-sponsored studies. The results suggest that the cost of consent document translation in trials not sponsored by industry could be a potentially modifiable barrier to the inclusion of patients with limited English proficiency.


Subject(s)
Clinical Trials as Topic , Communication Barriers , Consent Forms , Drug Industry , Research Personnel , Translations , Humans , Consent Forms/economics , Translating , Clinical Trials as Topic/economics , Drug Industry/economics , Research Personnel/economics
3.
Cell ; 142(3): 456-67, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20691904

ABSTRACT

RNA import into mammalian mitochondria is considered essential for replication, transcription, and translation of the mitochondrial genome but the pathway(s) and factors that control this import are poorly understood. Previously, we localized polynucleotide phosphorylase (PNPASE), a 3' --> 5' exoribonuclease and poly-A polymerase, in the mitochondrial intermembrane space, a location lacking resident RNAs. Here, we show a new role for PNPASE in regulating the import of nuclear-encoded RNAs into the mitochondrial matrix. PNPASE reduction impaired mitochondrial RNA processing and polycistronic transcripts accumulated. Augmented import of RNase P, 5S rRNA, and MRP RNAs depended on PNPASE expression and PNPASE-imported RNA interactions were identified. PNPASE RNA processing and import activities were separable and a mitochondrial RNA targeting signal was isolated that enabled RNA import in a PNPASE-dependent manner. Combined, these data strongly support an unanticipated role for PNPASE in mediating the translocation of RNAs into mitochondria.


Subject(s)
Mitochondria/metabolism , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA/metabolism , Animals , Cell Line , Gene Knockout Techniques , Humans , Mice , Mice, Inbred C57BL , Polyribonucleotide Nucleotidyltransferase/genetics , RNA Processing, Post-Transcriptional , Ribonuclease P/metabolism , Saccharomyces cerevisiae/metabolism
4.
Nat Immunol ; 12(11): 1086-95, 2011 Oct 02.
Article in English | MEDLINE | ID: mdl-21964609

ABSTRACT

The presence of immune memory at pathogen-entry sites is a prerequisite for protection. Nevertheless, the mechanisms that warrant immunity at peripheral interfaces are not understood. Here we show that the nonclassical major histocompatibility complex (MHC) class I molecule thymus leukemia antigen (TL), induced on dendritic cells interacting with CD8αα on activated CD8αß(+) T cells, mediated affinity-based selection of memory precursor cells. Furthermore, constitutive expression of TL on epithelial cells led to continued selection of mature CD8αß(+) memory T cells. The memory process driven by TL and CD8αα was essential for the generation of CD8αß(+) memory T cells in the intestine and the accumulation of highly antigen-sensitive CD8αß(+) memory T cells that form the first line of defense at the largest entry port for pathogens.


Subject(s)
Dendritic Cells/metabolism , Listeriosis/immunology , Membrane Glycoproteins/metabolism , Precursor Cells, T-Lymphoid/metabolism , T-Lymphocytes/metabolism , Animals , Antigens/immunology , Antigens/metabolism , CD8 Antigens/metabolism , Cell Differentiation , Clonal Selection, Antigen-Mediated , Dendritic Cells/immunology , Dendritic Cells/pathology , Immunity, Mucosal/genetics , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transgenes/genetics
5.
J Biol Chem ; 294(14): 5420-5429, 2019 04 05.
Article in English | MEDLINE | ID: mdl-29463682

ABSTRACT

Pluripotent stem cells (PSCs) are highly proliferative cells characterized by robust metabolic demands to power rapid division. For many years considered a passive component or "passenger" of cell-fate determination, cell metabolism is now starting to take center stage as a driver of cell fate outcomes. This review provides an update and analysis of our current understanding of PSC metabolism and its role in self-renewal, differentiation, and somatic cell reprogramming to pluripotency. Moreover, we present evidence on the active roles metabolism plays in shaping the epigenome to influence patterns of gene expression that may model key features of early embryonic development.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Epigenesis, Genetic/physiology , Pluripotent Stem Cells/metabolism , Animals , Humans , Pluripotent Stem Cells/cytology
6.
Nature ; 510(7505): 397-401, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24828042

ABSTRACT

Metabolism and ageing are intimately linked. Compared with ad libitum feeding, dietary restriction consistently extends lifespan and delays age-related diseases in evolutionarily diverse organisms. Similar conditions of nutrient limitation and genetic or pharmacological perturbations of nutrient or energy metabolism also have longevity benefits. Recently, several metabolites have been identified that modulate ageing; however, the molecular mechanisms underlying this are largely undefined. Here we show that α-ketoglutarate (α-KG), a tricarboxylic acid cycle intermediate, extends the lifespan of adult Caenorhabditis elegans. ATP synthase subunit ß is identified as a novel binding protein of α-KG using a small-molecule target identification strategy termed drug affinity responsive target stability (DARTS). The ATP synthase, also known as complex V of the mitochondrial electron transport chain, is the main cellular energy-generating machinery and is highly conserved throughout evolution. Although complete loss of mitochondrial function is detrimental, partial suppression of the electron transport chain has been shown to extend C. elegans lifespan. We show that α-KG inhibits ATP synthase and, similar to ATP synthase knockdown, inhibition by α-KG leads to reduced ATP content, decreased oxygen consumption, and increased autophagy in both C. elegans and mammalian cells. We provide evidence that the lifespan increase by α-KG requires ATP synthase subunit ß and is dependent on target of rapamycin (TOR) downstream. Endogenous α-KG levels are increased on starvation and α-KG does not extend the lifespan of dietary-restricted animals, indicating that α-KG is a key metabolite that mediates longevity by dietary restriction. Our analyses uncover new molecular links between a common metabolite, a universal cellular energy generator and dietary restriction in the regulation of organismal lifespan, thus suggesting new strategies for the prevention and treatment of ageing and age-related diseases.


Subject(s)
Caenorhabditis elegans/drug effects , Ketoglutaric Acids/pharmacology , Longevity/physiology , Mitochondrial Proton-Translocating ATPases/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , HEK293 Cells , Humans , Jurkat Cells , Longevity/drug effects , Longevity/genetics , Mice , Mitochondrial Proton-Translocating ATPases/genetics , Protein Binding
7.
EMBO J ; 34(2): 138-53, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25476451

ABSTRACT

Recent studies link changes in energy metabolism with the fate of pluripotent stem cells (PSCs). Safe use of PSC derivatives in regenerative medicine requires an enhanced understanding and control of factors that optimize in vitro reprogramming and differentiation protocols. Relative shifts in metabolism from naïve through "primed" pluripotent states to lineage-directed differentiation place variable demands on mitochondrial biogenesis and function for cell types with distinct energetic and biosynthetic requirements. In this context, mitochondrial respiration, network dynamics, TCA cycle function, and turnover all have the potential to influence reprogramming and differentiation outcomes. Shifts in cellular metabolism affect enzymes that control epigenetic configuration, which impacts chromatin reorganization and gene expression changes during reprogramming and differentiation. Induced PSCs (iPSCs) may have utility for modeling metabolic diseases caused by mutations in mitochondrial DNA, for which few disease models exist. Here, we explore key features of PSC energy metabolism research in mice and man and the impact this work is starting to have on our understanding of early development, disease modeling, and potential therapeutic applications.


Subject(s)
Energy Metabolism/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Humans , Mice
8.
Analyst ; 145(1): 97-106, 2019 Dec 16.
Article in English | MEDLINE | ID: mdl-31746831

ABSTRACT

Cell cycle deregulation is a cancer hallmark that has stimulated the development of mitotic inhibitors with differing mechanisms of action. Quantitative phase imaging (QPI) is an emerging approach for determining cancer cell sensitivities to chemotherapies in vitro. Cancer cell fates in response to mitotic inhibitors are agent- and dose-dependent. Fates that lead to chromosomal instabilities may result in a survival advantage and drug resistance. Conventional techniques for quantifying cell fates are incompatible with growth inhibition assays that produce binary live/dead results. Therefore, we used QPI to quantify post-mitotic fates of G0/G1 synchronized HeLa cervical adenocarcinoma and M202 melanoma cells during 24 h of escalating-dose exposures to mitotic inhibitors, including microtubule inhibitors paclitaxel and colchicine, and an Aurora kinase A inhibitor, VX-680. QPI determined cell fates by measuring changes in cell biomass, morphology, and mean phase-shift. Cell fates fell into three groups: (1) bipolar division from drug failure; (2) cell death or sustained mitotic arrest; and (3) aberrant endocycling or multipolar division. In this proof-of-concept study, colchicine was most effective in producing desirable outcomes of sustained mitotic arrest or death throughout its dosing range, whereas both paclitaxel and VX-680 yielded dose-dependent multipolar divisions or endocycling, respectively. Furthermore, rapid completion of mitosis associated with bipolar divisions whereas prolonged mitosis associated with multipolar divisions or cell death. Overall, QPI measurement of drug-induced cancer cell fates provides a tool to inform the development of candidate agents by quantifying the dosing ranges over which suboptimal inhibitor choices lead to undesirable, aberrant cancer cell fates.


Subject(s)
Antineoplastic Agents/pharmacology , Colchicine/pharmacology , Mitosis/drug effects , Paclitaxel/pharmacology , Piperazines/pharmacology , Aurora Kinase A/antagonists & inhibitors , Cell Line, Tumor , Humans , Proof of Concept Study , Protein Kinase Inhibitors/pharmacology , Tubulin Modulators/pharmacology
9.
Semin Cell Dev Biol ; 52: 76-83, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26828436

ABSTRACT

Human pluripotent stem cells (hPSCs) have great potential in regenerative medicine because they can differentiate into any cell type in the body. Genome integrity is vital for human development and for high fidelity passage of genetic information across generations through the germ line. To ensure genome stability, hPSCs maintain a lower rate of mutation than somatic cells and undergo rapid apoptosis in response to DNA damage and additional cell stresses. Furthermore, cellular metabolism and the cell cycle are also differentially regulated between cells in pluripotent and differentiated states and can aid in protecting hPSCs against DNA damage and damaged cell propagation. Despite these safeguards, clinical use of hPSC derivatives could be compromised by tumorigenic potential and possible malignant transformation from failed to differentiate cells. Since hPSCs and mature cells differentially respond to cell stress, it may be possible to specifically target undifferentiated cells for rapid apoptosis in mixed cell populations to enable safer use of hPSC-differentiated cells in patients.


Subject(s)
Apoptosis/physiology , Induced Pluripotent Stem Cells/physiology , Mitochondria/physiology , Animals , Cellular Reprogramming/physiology , Humans , Induced Pluripotent Stem Cells/cytology
11.
EMBO J ; 33(13): 1454-73, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24825347

ABSTRACT

Much of the mechanism by which Wnt signaling drives proliferation during oncogenesis is attributed to its regulation of the cell cycle. Here, we show how Wnt/ß-catenin signaling directs another hallmark of tumorigenesis, namely Warburg metabolism. Using biochemical assays and fluorescence lifetime imaging microscopy (FLIM) to probe metabolism in vitro and in living tumors, we observe that interference with Wnt signaling in colon cancer cells reduces glycolytic metabolism and results in small, poorly perfused tumors. We identify pyruvate dehydrogenase kinase 1 (PDK1) as an important direct target within a larger gene program for metabolism. PDK1 inhibits pyruvate flux to mitochondrial respiration and a rescue of its expression in Wnt-inhibited cancer cells rescues glycolysis as well as vessel growth in the tumor microenvironment. Thus, we identify an important mechanism by which Wnt-driven Warburg metabolism directs the use of glucose for cancer cell proliferation and links it to vessel delivery of oxygen and nutrients.


Subject(s)
Colonic Neoplasms/metabolism , Glucose/metabolism , Glycolysis , Neovascularization, Pathologic/metabolism , Tumor Microenvironment , Wnt Signaling Pathway , Animals , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Glucose/genetics , Humans , Mice , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Oxygen Consumption/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
12.
Anal Chem ; 90(5): 3299-3306, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29381859

ABSTRACT

We report the development of high-speed live-cell interferometry (HSLCI), a new multisample, multidrug testing platform for directly measuring tumor therapy response via real-time optical cell biomass measurements. As a proof of concept, we show that HSLCI rapidly profiles changes in biomass in BRAF inhibitor (BRAFi)-sensitive parental melanoma cell lines and in their isogenic BRAFi-resistant sublines. We show reproducible results from two different HSLCI platforms at two institutions that generate biomass kinetic signatures capable of discriminating between BRAFi-sensitive and -resistant melanoma cells within 24 h. Like other quantitative phase imaging (QPI) modalities, HSLCI is well-suited to noninvasive measurements of single cells and cell clusters, requiring no fluorescence or dye labeling. HSLCI is substantially faster and more sensitive than field-standard growth inhibition assays, and in terms of the number of cells measured simultaneously, the number of drugs tested in parallel, and temporal measurement range, it exceeds the state of the art by more than 10-fold. The accuracy and speed of HSLCI in profiling tumor cell heterogeneity and therapy resistance are promising features of potential tools to guide patient therapeutic selections.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Interferometry/methods , Melanoma/classification , Protein Kinase Inhibitors/pharmacology , Biomass , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Humans , Kinetics , Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics
13.
Nat Methods ; 12(5): 439-44, 2015 May.
Article in English | MEDLINE | ID: mdl-25849636

ABSTRACT

We report a high-throughput platform for delivering large cargo elements into 100,000 cells in 1 min. Our biophotonic laser-assisted surgery tool (BLAST) generates an array of microcavitation bubbles that explode in response to laser pulsing, forming pores in adjacent cell membranes through which cargo is gently driven by pressurized flow. The platform delivers large items including bacteria, enzymes, antibodies and nanoparticles into diverse cell types with high efficiency and cell viability. We used this platform to explore the intracellular lifestyle of Francisella novicida and discovered that the iglC gene is unexpectedly required for intracellular replication even after phagosome escape into the cell cytosol.


Subject(s)
Francisella/physiology , Lasers , Microbubbles , Animals , Cell Line , Gene Expression Regulation, Bacterial/physiology , Humans
14.
Mol Cell ; 39(6): 873-85, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20864035

ABSTRACT

During an immune response, B cells undergo rapid proliferation and activation-induced cytidine deaminase (AID)-dependent remodeling of immunoglobulin (IG) genes within germinal centers (GCs) to generate memory B and plasma cells. Unfortunately, the genotoxic stress associated with the GC reaction also promotes most B cell malignancies. Here, we report that exogenous and intrinsic AID-induced DNA strand breaks activate ATM, which signals through an LKB1 intermediate to inactivate CRTC2, a transcriptional coactivator of CREB. Using genome-wide location analysis, we determined that CRTC2 inactivation unexpectedly represses a genetic program that controls GC B cell proliferation, self-renewal, and differentiation while opposing lymphomagenesis. Inhibition of this pathway results in increased GC B cell proliferation, reduced antibody secretion, and impaired terminal differentiation. Multiple distinct pathway disruptions were also identified in human GC B cell lymphoma patient samples. Combined, our data show that CRTC2 inactivation, via physiologic DNA damage response signaling, promotes B cell differentiation in response to genotoxic stress.


Subject(s)
B-Lymphocytes/cytology , Cell Cycle Proteins/metabolism , Cell Differentiation/immunology , Cytidine Deaminase/genetics , DNA Damage/immunology , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , AMP-Activated Protein Kinase Kinases , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/radiation effects , Animals , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/radiation effects , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Gene Expression/drug effects , Gene Expression/immunology , Gene Expression/radiation effects , Gene Expression Regulation/immunology , Germinal Center/cytology , Humans , Immunoglobulin Class Switching/physiology , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Metformin/pharmacology , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/radiation effects , Plasma Cells/cytology , Plasma Cells/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Signal Transduction/radiation effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
15.
Nat Methods ; 11(12): 1221-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25423019

ABSTRACT

Cell mass, volume and growth rate are tightly controlled biophysical parameters in cellular development and homeostasis, and pathological cell growth defines cancer in metazoans. The first measurements of cell mass were made in the 1950s, but only recently have advances in computer science and microfabrication spurred the rapid development of precision mass-quantifying approaches. Here we discuss available techniques for quantifying the mass of single live cells with an emphasis on relative features, capabilities and drawbacks for different applications.


Subject(s)
Biophysics/methods , Cell Proliferation , Cell Size , Interferometry/methods , Cell Differentiation , Humans
16.
EMBO Rep ; 16(6): 753-68, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25916856

ABSTRACT

T-cell-dependent antigenic stimulation drives the differentiation of B cells into antibody-secreting plasma cells and memory B cells, but how B cells regulate this process is unclear. We show that LKB1 expression in B cells maintains B-cell quiescence and prevents the premature formation of germinal centers (GCs). Lkb1-deficient B cells (BKO) undergo spontaneous B-cell activation and secretion of multiple inflammatory cytokines, which leads to splenomegaly caused by an unexpected expansion of T cells. Within this cytokine response, increased IL-6 production results from heightened activation of NF-κB, which is suppressed by active LKB1. Secreted IL-6 drives T-cell activation and IL-21 production, promoting T follicular helper (TFH ) cell differentiation and expansion to support a ~100-fold increase in steady-state GC B cells. Blockade of IL-6 secretion by BKO B cells inhibits IL-21 expression, a known inducer of TFH -cell differentiation and expansion. Together, these data reveal cell intrinsic and surprising cell extrinsic roles for LKB1 in B cells that control TFH -cell differentiation and GC formation, and place LKB1 as a central regulator of T-cell-dependent humoral immunity.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Germinal Center/physiology , Lymphocyte Activation , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , T-Lymphocytes, Helper-Inducer/immunology , AMP-Activated Protein Kinases , Animals , Cell Differentiation , Interleukin-6/immunology , Interleukin-6/metabolism , Interleukins/immunology , Mice , NF-kappa B/genetics , T-Lymphocytes, Helper-Inducer/physiology
18.
EMBO J ; 30(24): 4860-73, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22085932

ABSTRACT

It has been assumed, based largely on morphologic evidence, that human pluripotent stem cells (hPSCs) contain underdeveloped, bioenergetically inactive mitochondria. In contrast, differentiated cells harbour a branched mitochondrial network with oxidative phosphorylation as the main energy source. A role for mitochondria in hPSC bioenergetics and in cell differentiation therefore remains uncertain. Here, we show that hPSCs have functional respiratory complexes that are able to consume O(2) at maximal capacity. Despite this, ATP generation in hPSCs is mainly by glycolysis and ATP is consumed by the F(1)F(0) ATP synthase to partially maintain hPSC mitochondrial membrane potential and cell viability. Uncoupling protein 2 (UCP2) plays a regulating role in hPSC energy metabolism by preventing mitochondrial glucose oxidation and facilitating glycolysis via a substrate shunting mechanism. With early differentiation, hPSC proliferation slows, energy metabolism decreases, and UCP2 is repressed, resulting in decreased glycolysis and maintained or increased mitochondrial glucose oxidation. Ectopic UCP2 expression perturbs this metabolic transition and impairs hPSC differentiation. Overall, hPSCs contain active mitochondria and require UCP2 repression for full differentiation potential.


Subject(s)
Cell Differentiation , Energy Metabolism , Ion Channels/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Adenosine Triphosphate , Cell Line , Glycolysis , Humans , Hydrolysis , Ion Channels/genetics , Mitochondrial Proteins/genetics , Oxygen Consumption , Pluripotent Stem Cells/ultrastructure , Reactive Oxygen Species/metabolism , Uncoupling Protein 2
19.
Am J Hum Genet ; 91(5): 919-27, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23084290

ABSTRACT

A subset of nuclear-encoded RNAs has to be imported into mitochondria for the proper replication and transcription of the mitochondrial genome and, hence, for proper mitochondrial function. Polynucleotide phosphorylase (PNPase or PNPT1) is one of the very few components known to be involved in this poorly characterized process in mammals. At the organismal level, however, the effect of PNPase dysfunction and impaired mitochondrial RNA import are unknown. By positional cloning, we identified a homozygous PNPT1 missense mutation (c.1424A>G predicting the protein substitution p.Glu475Gly) of a highly conserved PNPase residue within the second RNase-PH domain in a family affected by autosomal-recessive nonsyndromic hearing impairment. In vitro analyses in bacteria, yeast, and mammalian cells showed that the identified mutation results in a hypofunctional protein leading to disturbed PNPase trimerization and impaired mitochondrial RNA import. Immunohistochemistry revealed strong PNPase staining in the murine cochlea, including the sensory hair cells and the auditory ganglion neurons. In summary, we show that a component of the mitochondrial RNA-import machinery is specifically required for auditory function.


Subject(s)
Exoribonucleases/genetics , Hearing Loss, Sensorineural/genetics , Mutation , RNA Transport/genetics , RNA/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Chromosome Mapping , Cochlea/metabolism , Cochlea/pathology , Consanguinity , Exons , Exoribonucleases/chemistry , Exoribonucleases/metabolism , Female , Gene Expression , Hearing Loss, Sensorineural/metabolism , Humans , Male , Mice , Models, Molecular , Molecular Sequence Data , Pedigree , Protein Conformation , RNA, Mitochondrial , Zebrafish/genetics , Zebrafish/metabolism
20.
Cancer Cell ; 12(6): 572-85, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18068633

ABSTRACT

Enhanced mesenchymal expression of FGF10 led to the formation of multifocal PIN or prostate cancer. Inhibition of epithelial FGFR1 signaling using DN FGFR1 led to reversal of the cancer phenotype. A subset of the FGF10-induced carcinoma was serially transplantable. Paracrine FGF10 led to an increase in epithelial androgen receptor and synergized with cell-autonomous activated AKT. Our observations indicate that stromal FGF10 expression may facilitate the multifocal histology observed in prostate adenocarcinoma and suggest the FGF10/FGFR1 axis as a potential therapeutic target in treating hormone-sensitive or refractory prostate cancer. We also show that transient exposure to a paracrine growth factor may be sufficient for the initiation of oncogenic transformation.


Subject(s)
Epithelial Cells/metabolism , Fibroblast Growth Factor 10/metabolism , Paracrine Communication , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Epithelium/pathology , Genes, Dominant , Male , Mesoderm/pathology , Mice , Neoplasm Transplantation , Organ Specificity , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Regeneration
SELECTION OF CITATIONS
SEARCH DETAIL