Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Nat Methods ; 17(5): 481-494, 2020 05.
Article in English | MEDLINE | ID: mdl-32251396

ABSTRACT

Diverse microbial ecosystems underpin life in the sea. Among these microbes are many unicellular eukaryotes that span the diversity of the eukaryotic tree of life. However, genetic tractability has been limited to a few species, which do not represent eukaryotic diversity or environmentally relevant taxa. Here, we report on the development of genetic tools in a range of protists primarily from marine environments. We present evidence for foreign DNA delivery and expression in 13 species never before transformed and for advancement of tools for eight other species, as well as potential reasons for why transformation of yet another 17 species tested was not achieved. Our resource in genetic manipulation will provide insights into the ancestral eukaryotic lifeforms, general eukaryote cell biology, protein diversification and the evolution of cellular pathways.


Subject(s)
DNA/administration & dosage , Eukaryota/physiology , Green Fluorescent Proteins/metabolism , Marine Biology , Models, Biological , Transformation, Genetic , Biodiversity , Ecosystem , Environment , Eukaryota/classification , Species Specificity
3.
Cell Calcium ; 86: 102127, 2020 03.
Article in English | MEDLINE | ID: mdl-31954928

ABSTRACT

Mitochondrial free calcium is critically linked to the regulation of cellular metabolism. Free ionic calcium concentration within these organelles is determined by the interplay between two processes: exchange across the mitochondrial inner membrane and calcium-buffering within the matrix. During stimulated calcium uptake, calcium is primarily buffered by orthophosphate, preventing calcium toxicity while allowing for well-regulated yet elevated calcium loads. However, if limited to orthophosphates only, this buffering system is expected to lead to the irreversible formation of insoluble precipitates, which are not observed in living cells, under physiological conditions. Here, we demonstrate that the regulation of free mitochondrial calcium requires the presence of free inorganic polyphosphate (polyP) within the organelle. We found that the overexpression of a mitochondrial-targeted enzyme hydrolyzing polyP leads to the loss of the cellular ability to maintain elevated calcium concentrations within the organelle, following stimulated cytoplasmic signal. We hypothesize that the presence of polyP prevents the formation of calcium-phosphate insoluble clusters, allowing for the maintenance of elevated free calcium levels, during stimulated calcium uptake.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Polyphosphates/pharmacology , Adenosine Triphosphate/pharmacology , Benzoates/metabolism , Bridged Bicyclo Compounds/metabolism , Calcium Signaling/drug effects , Cell Membrane Permeability/drug effects , Cycloheptanes/metabolism , HEK293 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Models, Biological , Ruthenium Red/metabolism , Sesquiterpenes/metabolism
4.
Anal Biochem ; 552: 38-44, 2018 07 01.
Article in English | MEDLINE | ID: mdl-28693989

ABSTRACT

Mitochondrial permeability transition (PT) is a phenomenon of an increase of the inner membrane permeability in response to an excessive matrix calcium accumulation. PTP is caused by the opening of the large weakly selective channel. Molecular composition and regulation of permeability transition pore (PTP) are not well understood. Here we used isolated mitochondria to investigate dependence of PTP activation on the osmotic pressure. We found that in low osmotic strength solution calcium-induced PTP is significantly inhibited. We propose that this effect is linked to the changes in the curvature of the mitochondrial inner membrane. This interpretation is consistent with the idea about the importance of ATP synthase dimerization in modulation of the PTP activity.


Subject(s)
Light , Microscopy, Electron/methods , Microscopy, Fluorescence/methods , Mitochondrial Membrane Transport Proteins/metabolism , Osmotic Pressure , Scattering, Radiation , Dimerization , Mitochondrial Membrane Transport Proteins/ultrastructure , Mitochondrial Permeability Transition Pore , Proton-Translocating ATPases/metabolism
5.
J Cereb Blood Flow Metab ; 37(8): 3027-3041, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27909264

ABSTRACT

The effects of global mitochondrial calcium (Ca2+) uniporter (MCU) deficiency on hypoxic-ischemic (HI) brain injury, neuronal Ca2+ handling, bioenergetics and hypoxic preconditioning (HPC) were examined. Forebrain mitochondria isolated from global MCU nulls displayed markedly reduced Ca2+ uptake and Ca2+-induced opening of the membrane permeability transition pore. Despite evidence that these effects should be neuroprotective, global MCU nulls and wild-type (WT) mice suffered comparable HI brain damage. Energetic stress enhanced glycolysis and depressed Complex I activity in global MCU null, relative to WT, cortical neurons. HI reduced forebrain NADH levels more in global MCU nulls than WT mice suggesting that increased glycolytic consumption of NADH suppressed Complex I activity. Compared to WT neurons, pyruvate dehydrogenase (PDH) was hyper-phosphorylated in MCU nulls at several sites that lower the supply of substrates for the tricarboxylic acid cycle. Elevation of cytosolic Ca2+ with glutamate or ionomycin decreased PDH phosphorylation in MCU null neurons suggesting the use of alternative mitochondrial Ca2+ transport. Under basal conditions, global MCU nulls showed similar increases of Ca2+ handling genes in the hippocampus as WT mice subjected to HPC. We propose that long-term adaptations, common to HPC, in global MCU nulls compromise resistance to HI brain injury and disrupt HPC.


Subject(s)
Brain/metabolism , Calcium Channels/metabolism , Energy Metabolism , Glycolysis , Hypoxia-Ischemia, Brain/metabolism , Neurons/metabolism , Animals , Brain/pathology , Calcium/metabolism , Calcium Channels/genetics , Cell Survival , Cells, Cultured , Female , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/prevention & control , Ischemic Preconditioning , Male , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Neurons/pathology , Primary Cell Culture
6.
J Bioenerg Biomembr ; 49(1): 113-119, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27665468

ABSTRACT

Calcium (Ca2+) plays diverse roles in all living organisms ranging from bacteria to humans. It is a structural element for bones, an essential mediator of excitation-contraction coupling, and a universal second messenger in the regulation of ion channel, enzyme and gene expression activities. In mitochondria, Ca2+ is crucial for the control of energy production and cellular responses to metabolic stress. Ca2+ uptake by the mitochondria occurs by the uniporter mechanism. The Mitochondrial Ca2+ Uniporter (MCU) protein has recently been identified as a core component responsible for mitochondrial Ca2+ uptake. MCU knockout (MCU KO) studies have identified a number of important roles played by this high capacity uptake pathway. Interestingly, this work has also shown that MCU-mediated Ca2+ uptake is not essential for vital cell functions such as muscle contraction, energy metabolism and neurotransmission. Although mitochondrial Ca2+ uptake was markedly reduced, MCU KO mitochondria still contained low but detectable levels of Ca2+. In view of the fundamental importance of Ca2+ for basic cell signalling, this finding suggests the existence of other currently unrecognized pathways for Ca2+ entry. We review the experimental evidence for the existence of alternative Ca2+ influx mechanisms and propose how these mechanisms may play an integral role in mitochondrial Ca2+ signalling.


Subject(s)
Calcium/metabolism , Ion Transport , Mitochondria/metabolism , Calcium/physiology , Metabolic Networks and Pathways , Mitochondrial Swelling
7.
Biochem Soc Trans ; 44(1): 7-12, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26862181

ABSTRACT

Mitochondrial permeability transition pore (mPTP) is a large channel located in the mitochondrial inner membrane. The opening of mPTP during pathological calcium overload leads to the membrane depolarization and disruption of ATP production. mPTP activation has been implicated as a central event during the process of stress-induced cell death. mPTP is a supramolecular complex composed of many proteins. Recent studies suggest that mitochondrial ATPase plays the central role in the formation of mPTP. However, the structure of the central conducting pore part of mPTP (mPTPore) remains elusive. Here we review current models proposed for the mPTPore and involvement of polyP in its formation and regulation. We discuss the underestimated role of polyP as an effector and a putative structural component of the mPTPore. We propose the hypothesis that inclusion of polyP can explain such properties of mPTP activity as calcium activation, selectivity and voltage-dependence.


Subject(s)
Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/metabolism , Polyphosphates/metabolism , Animals , Humans , Mitochondrial Permeability Transition Pore , Models, Biological
8.
Oncotarget ; 6(19): 17221-36, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-25980497

ABSTRACT

The cold and menthol receptor TRPM8 is highly expressed in prostate and prostate cancer (PC). Recently, we identified that TRPM8 is as an ionotropic testosterone receptor. The TRPM8 mRNA is expressed in early prostate tumors with high androgen levels, while anti-androgen therapy greatly reduces its expression. Here, from the chromatin-immunoprecipitation (ChIP) analysis, we found that an androgen response element (ARE) mediates androgen regulation of trpm8. Furthermore, using immunofluorescence, calcium-imaging and planar lipid bilayers, we identified that TRPM8 channel is functionally regulated by androgens in the prostate. Although TRPM8 mRNA is expressed at high levels, we found that the TRPM8 protein undergoes ubiquitination and degradation in PC cells. The mass-spectrometry analysis of TRPM8, immunoprecipitated from LNCaP cells identified ubiquitin-like modifier-activating enzyme 1 (UBA1). PYR-41, a potent inhibitor of initial enzyme in the ubiquitination cascade, UBA1, increased TRPM8 activity on the plasma membrane (PM) of LNCaP cells. Furthermore, PYR-41-mediated PMTRPM8 activity was accompanied by enhanced activation of p53 and Caspase-9. Interestingly, we found that the trpm8 promoter possesses putative binding sites for p53 and that the overexpression of p53 increased the TRPM8 mRNA levels. In addition to the genomic regulation of TRPM8 by AR and p53, our findings indicate that the testosterone-induced PMTRPM8 activity elicits Ca2+ uptake, subsequently causing apoptotic cell death. These findings support the strategy of rescuing PMTRPM8 expression as a new therapeutic application through the regulation of PC cell growth and proliferation.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , TRPM Cation Channels/metabolism , Androgens/metabolism , Calcium/metabolism , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mass Spectrometry , Prostatic Neoplasms/genetics , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Receptors, Androgen/genetics , Response Elements/genetics , TRPM Cation Channels/genetics , Tissue Array Analysis , Transfection
9.
Biochim Biophys Acta ; 1847(2): 231-240, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25448536

ABSTRACT

The permeability transition pore (PTP) is a large channel of the mitochondrial inner membrane, the opening of which is the central event in many types of stress-induced cell death. PTP opening is induced by elevated concentrations of mitochondrial calcium. It has been demonstrated that spermine and other polyamines can delay calcium-induced swelling of isolated mitochondria, suggesting their role as inhibitors of the mitochondrial PTP. Here we further investigated the mechanism by which spermine inhibits the calcium-induced, cyclosporine A (CSA) -sensitive PTP by using three indicators: 1) calcium release from the mitochondria detected with calcium green, 2) mitochondrial membrane depolarization using TMRM, and 3) mitochondrial swelling by measuring light absorbance. We found that despite calcium release and membrane depolarization, indicative of PTP activation, mitochondria underwent only partial swelling in the presence of spermine. This was in striking contrast to the high-amplitude swelling detected in control mitochondria and in mitochondria treated with the PTP inhibitor CSA. We conclude that spermine selectively prevents opening of the high-conductance state, while allowing activation of the lower conductance state of the PTP. We propose that the existence of lower conductance, stress-induced PTP might play an important physiological role, as it is expected to allow the release of toxic levels of calcium, while keeping important molecules (e.g., NAD) within the mitochondrial matrix.


Subject(s)
Calcium/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Spermine/pharmacology , Animals , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling/drug effects , Osmolar Concentration , Rats , Rats, Sprague-Dawley
10.
J Biol Chem ; 290(5): 2659-69, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25480783

ABSTRACT

The transient receptor potential ion channel of the melastatin subfamily, TRPM8, is a major cold receptor in the peripheral nervous system. Along with the sensory neurons, the TRPM8 protein is highly expressed in the prostate epithelial cells, and this expression is regulated by androgens. Here we investigated the expression and intracellular localization of the TRPM8 channel in relationship to androgens. We performed experiments using human prostate tissues obtained from healthy individuals and patients with prostate cancer at various stages of the disease as well as in cultured cells. Using an immunohistochemistry approach, we detected an intensive colocalization pattern of the TRPM8 protein with endogenous androgens in all tissues tested, suggesting possible interactions. Co-immunoprecipitation experiments performed using cultured prostate epithelial cells, prostate cancer cells, and HEK-293 cells stably expressing TRPM8 further confirmed direct binding of the steroid hormone, testosterone, to the TRPM8 protein. Applications of picomolar concentrations of testosterone to the primary human prostate cells, endogenously expressing TRPM8, elicited Ca(2+) responses and channel currents, and those were inhibited in the presence of TRPM8 antagonist, N-(2-aminoethyl)-N-(4-(benzyloxy)-3-methoxybenzyl)thiophene-2-carboxamide hydrochloride. These results indicate that the TRPM8 channel is physically associated with testosterone and suggest that, in addition to a genomic role, testosterone plays a role in direct regulation of the TRPM8 channel function.


Subject(s)
Receptors, Androgen/metabolism , TRPM Cation Channels/metabolism , Testosterone/metabolism , Cell Line , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Male , Protein Binding
11.
J Biol Chem ; 290(5): 2670-88, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25480785

ABSTRACT

Testosterone is a key steroid hormone in the development of male reproductive tissues and the regulation of the central nervous system. The rapid signaling mechanism induced by testosterone affects numerous behavioral traits, including sexual drive, aggressiveness, and fear conditioning. However, the currently identified testosterone receptor(s) is not believed to underlie the fast signaling, suggesting an orphan pathway. Here we report that an ion channel from the transient receptor potential family, TRPM8, commonly known as the cold and menthol receptor is the major component of testosterone-induced rapid actions. Using cultured and primary cell lines along with the purified TRPM8 protein, we demonstrate that testosterone directly activates TRPM8 channel at low picomolar range. Specifically, testosterone induced TRPM8 responses in primary human prostate cells, PC3 prostate cancer cells, dorsal root ganglion neurons, and hippocampal neurons. Picomolar concentrations of testosterone resulted in full openings of the purified TRPM8 channel in planar lipid bilayers. Furthermore, acute applications of testosterone on human skin elicited a cooling sensation. Our data conclusively demonstrate that testosterone is an endogenous and highly potent agonist of TRPM8, suggesting a role of TRPM8 channels well beyond their well established function in somatosensory neurons. This discovery may further imply TRPM8 channel function in testosterone-dependent behavioral traits.


Subject(s)
Receptors, Androgen/metabolism , TRPM Cation Channels/metabolism , Testosterone/metabolism , Calcium/metabolism , Gene Expression/drug effects , Gene Expression/genetics , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Lipid Bilayers/metabolism , Protein Binding/drug effects , Testosterone/pharmacology , Transient Receptor Potential Channels/metabolism
12.
ACS Chem Biol ; 9(9): 2101-10, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25007079

ABSTRACT

Inorganic polyphosphate (polyP) is a polymer composed of many orthophosphates linked together by phosphoanhydride bonds. Recent studies demonstrate that in addition to its important role in the function of microorganisms, polyP plays multiple important roles in the pathological and physiological function of higher eukaryotes, including mammalians. However, due to the dramatically lower abundance of polyP in mammalian cells when comparing to microorganisms, its investigation poses an experimental challenge. Here, we present the identification of novel fluorescent probes that allow for specific labeling of synthetic polyP in vitro as well as endogenous polyP in living cells. These probes demonstrate high selectivity for the labeling of polyP that was not sensitive to a number of ubiquitous organic polyphosphates, notably RNA. Use of these probes allowed us to demonstrate the real time detection of polyP release from lysosomes in live cells. Furthermore, we have been able to detect the increased levels of polyP in cells with Parkinson's disease related mutations.


Subject(s)
Benzimidazoles/metabolism , Brain/metabolism , Fluorescent Dyes/chemistry , Molecular Imaging/methods , Naphthalenes/metabolism , Polyphosphates/analysis , Animals , Astrocytes/metabolism , Benzimidazoles/analysis , Benzimidazoles/chemistry , Cells, Cultured , Dimethyl Sulfoxide/chemistry , Disease Models, Animal , Drosophila melanogaster , Fluorescent Dyes/analysis , Humans , Indoles/analysis , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Lysosomes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Naphthalenes/analysis , Naphthalenes/chemistry , Parkinson Disease/metabolism , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Rats , Reproducibility of Results
13.
PLoS One ; 8(9): e75812, 2013.
Article in English | MEDLINE | ID: mdl-24086638

ABSTRACT

Poly(3-hydroxybutyrate) (PHB) is a polyester of 3-hydroxybutyric acid (HB) that is ubiquitously present in all organisms. In higher eukaryotes PHB is found in the length of 10 to 100 HB units and can be present in free form as well as in association with proteins and inorganic polyphosphate. It has been proposed that PHB can mediate ion transport across lipid bilayer membranes. We investigated the ability of PHB to interact with living cells and isolated mitochondria and the effects of these interactions on membrane ion transport. We performed experiments using a fluorescein derivative of PHB (fluo-PHB). We found that fluo-PHB preferentially accumulated inside the mitochondria of HeLa cells. Accumulation of fluo-PHB induced mitochondrial membrane depolarization. This membrane depolarization was significantly delayed by the inhibitor of the mitochondrial permeability transition pore - Cyclosporin A. Further experiments using intact cells as well as isolated mitochondria confirmed that the effects of PHB directly linked to its ability to facilitate ion transport, including calcium, across the membranes. We conclude that PHB demonstrates ionophoretic properties in biological membranes and this effect is most profound in mitochondria due to the selective accumulation of the polymer in this organelle.


Subject(s)
Hydroxybutyrates/pharmacology , Mitochondria/drug effects , Mitochondrial Membranes/drug effects , Permeability/drug effects , Polyesters/pharmacology , Calcium/metabolism , Cell Line, Tumor , Cyclosporine/metabolism , HeLa Cells , Humans , Ion Transport/drug effects , Ion Transport/physiology , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/physiology , Mitochondrial Permeability Transition Pore , Prohibitins
14.
J Biol Chem ; 288(35): 25309-25317, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23873936

ABSTRACT

The intracellular lactate shuttle hypothesis posits that lactate generated in the cytosol is oxidized by mitochondrial lactate dehydrogenase (LDH) of the same cell. To examine whether skeletal muscle mitochondria oxidize lactate, mitochondrial respiratory oxygen flux (JO2) was measured during the sequential addition of various substrates and cofactors onto permeabilized rat gastrocnemius muscle fibers, as well as isolated mitochondrial subpopulations. Addition of lactate did not alter JO2. However, subsequent addition of NAD(+) significantly increased JO2, and was abolished by the inhibitor of mitochondrial pyruvate transport, α-cyano-4-hydroxycinnamate. In experiments with isolated subsarcolemmal and intermyofibrillar mitochondrial subpopulations, only subsarcolemmal exhibited NAD(+)-dependent lactate oxidation. To further investigate the details of the physical association of LDH with mitochondria in muscle, immunofluorescence/confocal microscopy and immunoblotting approaches were used. LDH clearly colocalized with mitochondria in intact, as well as permeabilized fibers. LDH is likely localized inside the outer mitochondrial membrane, but not in the mitochondrial matrix. Collectively, these results suggest that extra-matrix LDH is strategically positioned within skeletal muscle fibers to functionally interact with mitochondria.


Subject(s)
L-Lactate Dehydrogenase/metabolism , Mitochondria, Muscle/metabolism , Mitochondrial Membranes/enzymology , Muscle Proteins/metabolism , Muscle, Skeletal/enzymology , Animals , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Coumaric Acids/pharmacology , Lactic Acid/metabolism , Oxidation-Reduction/drug effects , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Pyruvic Acid/metabolism , Rats , Rats, Sprague-Dawley
15.
Cell Calcium ; 54(2): 86-94, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23702223

ABSTRACT

Polyhydroxybutyrate (PHB) is a biological polymer which belongs to the class of polyesters and is ubiquitously present in all living organisms. Mammalian mitochondrial membranes contain PHB consisting of up to 120 hydroxybutyrate residues. Roles played by PHB in mammalian mitochondria remain obscure. It was previously demonstrated that PHB of the size similar to one found in mitochondria mediates calcium transport in lipid bilayer membranes. We hypothesized that the presence of PHB in mitochondrial membrane might play a significant role in mitochondrial calcium transport. To test this, we investigated how the induction of PHB hydrolysis affects mitochondrial calcium transport. Mitochondrial PHB was altered enzymatically by targeted expression of bacterial PHB hydrolyzing enzyme (PhaZ7) in mitochondria of mammalian cultured cells. The expression of PhaZ7 induced changes in mitochondrial metabolism resulting in decreased mitochondrial membrane potential in HepG2 but not in U87 and HeLa cells. Furthermore, it significantly inhibited mitochondrial calcium uptake in intact HepG2, U87 and HeLa cells stimulated by the ATP or by the application of increased concentrations of calcium to the digitonin permeabilized cells. Calcium uptake in PhaZ7 expressing cells was restored by mimicking calcium uniporter properties with natural electrogenic calcium ionophore - ferutinin. We propose that PHB is a previously unrecognized important component of the mitochondrial calcium uptake system.


Subject(s)
Calcium/metabolism , Hydroxybutyrates/metabolism , Mitochondria/metabolism , Polyesters/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Cell Line, Tumor , Cell Survival , Female , HeLa Cells , Hep G2 Cells , Humans , Membrane Potential, Mitochondrial/physiology , Prohibitins , Transfection
16.
Reprod Biol Endocrinol ; 7: 51, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19463178

ABSTRACT

BACKGROUND: In the fusion pathway of trophoblast differentiation, stem villous cytotrophoblast cells proliferate and daughter cells differentiate and fuse with existing syncytiotrophoblast to maintain the multi-nucleated layer. Integrin-linked kinase (ILK) is highly expressed in 1st and 2nd trimester villous cytotrophoblast cells, yet barely detectable in syncytiotrophoblast, thus we examined the potential role of ILK in aiding trophoblast fusion. METHODS: The temporal/spatial expression and activity of ILK were determined in BeWo cells undergoing syncytialization by immunoblot and immunofluorescence analyses. BeWo cells were also transfected with pEGFP expression vectors containing wildtype or two mutant ILK cDNA constructs. The incidence of cell fusion in transfected cells grown under syncytialization conditions was then scored by the presence or absence of E-cadherin immunostaining. Beta-hCG expression in transfected cells, a marker of syncytiotrophoblast hormonal differentiation, was also similarly assessed. RESULTS: ILK catalytic activity increased and ILK began to increasingly localize to BeWo cell nuclei during syncytialization in correlation with increased pAkt and Snail protein expression. Syncytialization was also significantly elevated (p < 0.05) in BeWo cells expressing constitutively active (ca)-ILK vs cells containing empty vector or dn-ILK. Furthermore, cytoplasmic Beta-hCG expression markedly increased (p < 0.05) in cells expressing wt- and ca-ILK. CONCLUSION: ILK-facilitated syncytialization is dependent, at least in part, on ILK catalytic activity while hormonal differentiation appears dependent on both ILK-associated protein interactions and catalytic activity. This study demonstrates that ILK plays a novel role in BeWo syncytialization and differentiation, perhaps through an ILK-Akt-Snail pathway, and implicates ILK in the same process in villous cytotrophoblasts in vivo.


Subject(s)
Giant Cells/cytology , Giant Cells/enzymology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Trophoblasts/cytology , Trophoblasts/enzymology , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Cell Line, Tumor , Choriocarcinoma , Chorionic Gonadotropin, beta Subunit, Human/genetics , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Enzymologic/physiology , Green Fluorescent Proteins/genetics , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Transfection , Uterine Neoplasms
SELECTION OF CITATIONS
SEARCH DETAIL
...