Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cell Rep ; 30(11): 3851-3863.e6, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32187554

ABSTRACT

Cancer therapy is limited, in part, by lack of specificity. Thus, identifying molecules that are selectively expressed by, and relevant for, cancer cells is of paramount medical importance. Here, we show that peptidyl-prolyl-cis-trans-isomerase (PPIase) FK506-binding protein 10 (FKBP10)-positive cells are present in cancer lesions but absent in the healthy parenchyma of human lung. FKBP10 expression negatively correlates with survival of lung cancer patients, and its downregulation causes a dramatic diminution of lung tumor burden in mice. Mechanistically, our results from gain- and loss-of-function assays show that FKBP10 boosts cancer growth and stemness via its PPIase activity. Also, FKBP10 interacts with ribosomes, and its downregulation leads to reduction of translation elongation at the beginning of open reading frames (ORFs), particularly upon insertion of proline residues. Thus, our data unveil FKBP10 as a cancer-selective molecule with a key role in translational reprogramming, stem-like traits, and growth of lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Biosynthesis , Tacrolimus Binding Proteins/metabolism , Animals , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peptidylprolyl Isomerase/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Ribosomes/metabolism
2.
Cell Rep ; 18(8): 1858-1868, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28228253

ABSTRACT

Cancer stem cells (CSCs) have high tumorigenic capacity. Here, we show that stem-like traits of specific human cancer cells are reduced by overexpression of the histone deacetylase sirtuin 6 (SIRT6). SIRT6-sensitive cancer cells bear mutations that activate phosphatidylinositol-3-kinase (PI3K) signaling, and overexpression of SIRT6 reduces growth, progression, and grade of breast cancer in a mouse model with PI3K activation. Tumor metabolomic and transcriptomic analyses reveal that SIRT6 overexpression dampens PI3K signaling and stem-like characteristics and causes metabolic rearrangements in this cancer model. Ablation of a PI3K activating mutation in otherwise isogenic cancer cells is sufficient to convert SIRT6-sensitive into SIRT6-insensitive cells. SIRT6 overexpression suppresses PI3K signaling at the transcriptional level and antagonizes tumor sphere formation independent of its histone deacetylase activity. Our data identify SIRT6 as a putative molecular target that hinders stemness of tumors with PI3K activation.


Subject(s)
Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Sirtuins/metabolism , Acetylation , Animals , Carcinogenesis/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mutation/physiology , Signal Transduction/physiology , Transcription, Genetic/physiology
3.
Front Pharmacol ; 4: 61, 2013.
Article in English | MEDLINE | ID: mdl-23675350

ABSTRACT

Gene targeting strategies have become a powerful technology for elucidating mammalian gene function. The recently generated knockout (KO)-first strategy produces a KO at the RNA processing level and also allows for the generation of conditional KO alleles by combining FLP/FRT and Cre/loxP systems, thereby providing high flexibility in gene manipulation. However, this multipurpose KO-first cassette might produce hypomorphic rather than complete KOs if the RNA processing module is bypassed. Moreover, the generation of a conditional phenotype is also dependent on specific activity of Cre recombinase. Here, we report the use of an efficient molecular biological approach to test pannexin1 (Panx1) mRNA expression in global and conditional Panx1 KO mice derived from the KO-first mouse line, Panx1(tm1a(KOMP)Wtsi). Using qRT-PCR, we demonstrate that tissues from wild-type (WT) mice show a range of Panx1 mRNA expression levels, with highest expression in trigeminal ganglia, bladder and spleen. Unexpectedly, we found that in mice homozygous for the KO-first allele, Panx1 mRNA expression is not abolished but reduced by 70% compared to that of WT tissues. Thus, Panx1 KO-first mice present a hypomorphic phenotype. Crosses of Panx1 KO-first with FLP deleter mice generated Panx1(f/f) mice. Further crosses of the latter mice with mGFAP-Cre or NFH-Cre mice were used to generate astrocyte- and neuron-specific Panx1 deletions, respectively. A high incidence of ectopic Cre expression was found in offspring of both types of conditional Panx1 KO mice. Our study demonstrates that Panx1 expression levels in the global and conditional Panx1 KO mice derived from KO-first mouse lines must be carefully characterized to ensure modulation of Panx1 gene expression. The precise quantitation of Panx1 expression and its relation to function is expected to provide a foundation for future efforts aimed at deciphering the role of Panx1 under physiological and pathological conditions.

4.
J Mol Endocrinol ; 51(1): 49-58, 2013.
Article in English | MEDLINE | ID: mdl-23613279

ABSTRACT

Channels formed by the gap junction protein Connexin36 (CX36) contribute to the proper control of insulin secretion. We previously demonstrated that chronic exposure to glucose decreases Cx36 levels in insulin-secreting cells in vitro. Here, we investigated whether hyperglycemia also regulates Cx36 in vivo. Using a model of continuous glucose infusion in adult rats, we showed that prolonged (24-48 h) hyperglycemia reduced the Cx36 gene Gjd2 mRNA levels in pancreatic islets. Accordingly, prolonged exposure to high glucose concentrations also reduced the expression and function of Cx36 in the rat insulin-producing INS-1E cell line. The glucose effect was blocked after inhibition of the cAMP/PKA pathway and was associated with an overexpression of the inducible cAMP early repressor ICER-1/ICER-1γ, which binds to a functional cAMP-response element in the promoter of the Cx36 gene Gjd2. The involvement of this repressor was further demonstrated using an antisense strategy of ICER-1 inhibition, which prevented glucose-induced downregulation of Cx36. The data indicate that chronic exposure to glucose alters the in vivo expression of Cx36 by the insulin-producing ß-cells through ICER-1/ICER-1γ overexpression. This mechanism may contribute to the reduced glucose sensitivity and altered insulin secretion, which contribute to the pathophysiology of diabetes.


Subject(s)
Connexins/genetics , Cyclic AMP Response Element Modulator/genetics , Gene Expression Regulation , Hyperglycemia/genetics , Hyperglycemia/metabolism , Islets of Langerhans/metabolism , Animals , Blood Glucose , Cell Line, Tumor , Connexins/metabolism , Cyclic AMP Response Element Modulator/metabolism , Glucose/metabolism , Male , Rats , Gap Junction delta-2 Protein
5.
Autophagy ; 9(4): 550-67, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23380587

ABSTRACT

Osmotic homeostasis is fundamental for most cells, which face recurrent alterations of environmental osmolality that challenge cell viability. Protein damage is a consequence of hypertonic stress, but whether autophagy contributes to the osmoprotective response is unknown. Here, we investigated the possible implications of autophagy and microtubule organization on the response to hypertonic stress. We show that hypertonicity rapidly induced long-lived protein degradation, LC3-II generation and Ptdlns3K-dependent formation of LC3- and ATG12-positive puncta. Lysosomotropic agents chloroquine and bafilomycin A 1, but not nutrient deprivation or rapamycin treatment, further increased LC3-II generation, as well as ATG12-positive puncta, indicating that hypertonic stress increases autophagic flux. Autophagy induction upon hypertonic stress enhanced cell survival since cell death was increased by ATG12 siRNA-mediated knockdown and reduced by rapamycin. We additionally showed that hypertonicity induces fast reorganization of microtubule networks, which is associated with strong reorganization of microtubules at centrosomes and fragmentation of Golgi ribbons. Microtubule remodeling was associated with pericentrosomal clustering of ATG12-positive autolysosomes that colocalized with SQSTM1/p62 and ubiquitin, indicating that autophagy induced by hypertonic stress is at least partly selective. Efficient autophagy by hypertonic stress required microtubule remodeling and was DYNC/dynein-dependent as autophagosome clustering was enhanced by paclitaxel-induced microtubule stabilization and was reduced by nocodazole-induced tubulin depolymerization as well as chemical (EHNA) or genetic [DCTN2/dynactin 2 (p50) overexpression] interference of DYNC activity. The data document a general and hitherto overlooked mechanism, where autophagy and microtubule remodeling play prominent roles in the osmoprotective response.


Subject(s)
Autophagy/drug effects , Hypertonic Solutions/pharmacology , Microtubules/metabolism , Phagosomes/metabolism , Stress, Physiological/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Dynactin Complex , Dyneins/metabolism , Humans , LLC-PK1 Cells , Lysosomes/drug effects , Lysosomes/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/drug effects , Phagosomes/drug effects , Swine
6.
PLoS One ; 7(7): e41535, 2012.
Article in English | MEDLINE | ID: mdl-22848521

ABSTRACT

Connexin36 (Cx36) plays an important role in insulin secretion by controlling the intercellular synchronization of Ca(2+) transients induced during stimulation. The lack of drugs acting on Cx36 channels is a major limitation in further unraveling the molecular mechanism underlying this effect. To screen for such drugs, we have developed an assay allowing for a semi-automatic, fluorimetric quantification of Ca(2+) transients in large populations of MIN6 cells. Here, we show that (1) compared to control cells, MIN6 cells with reduced Cx36 expression or function showed decreased synchrony of glucose-induced Ca(2+) oscillations; (2) glibenclamide, a sulphonylurea which promotes Cx36 junctions and coupling, increased the number of synchronous MIN6 cells, whereas quinine, an antimalarial drug which inhibits Cx36-dependent coupling, decreased this proportion; (3) several drugs were identified that altered the intercellular Ca(2+) synchronization, cell coupling and distribution of Cx36; (4) some of them also affected insulin content. The data indicate that the intercellular synchronization of Ca(2+) oscillations provides a reliable and non-invasive measurement of Cx36-dependent coupling, which is useful to identify novel drugs affecting the function of ß-cells, neurons, and neuron-related cells that express Cx36.


Subject(s)
Biological Clocks/physiology , Calcium/metabolism , Connexins/metabolism , Animals , Antimalarials/pharmacology , Biological Clocks/drug effects , Cell Line , Connexins/genetics , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Mice , Quinine/pharmacology , Gap Junction delta-2 Protein
7.
J Membr Biol ; 245(5-6): 263-73, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22729650

ABSTRACT

The insulin-producing ß cells of pancreatic islets are coupled by connexin36 (Cx36) channels. To investigate what controls the expression of this connexin, we have investigated its pattern during mouse pancreas development, and the influence of three transcription factors that are critical for ß-cell development and differentiation. We show that (1) the Cx36 gene (Gjd2) is activated early in pancreas development and is markedly induced at the time of the surge of the transcription factors that determine ß-cell differentiation; (2) the cognate protein is detected about a week later and is selectively expressed by ß cells throughout the prenatal development of mouse pancreas; (3) a 2-kbp fragment of the Gjd2 promoter, which contains three E boxes for the binding of the bHLH factor Beta2/NeuroD1, ensures the expression of Cx36 by ß cells; and (4) Beta2/NeuroD1 binds to these E boxes and, in the presence of the E47 ubiquitous cofactor, transactivates the Gjd2 promoter. The data identify Cx36 as a novel early marker of ß cells and as a target of Beta2/NeuroD1, which is essential for ß-cell development and differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Connexins/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Chromatin Immunoprecipitation , Computational Biology , Gap Junctions/metabolism , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Protein Binding/genetics , Reverse Transcriptase Polymerase Chain Reaction , Gap Junction delta-2 Protein
8.
J Clin Invest ; 121(12): 4870-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22056383

ABSTRACT

Type 1 diabetes develops when most insulin-producing ß cells of the pancreas are killed by an autoimmune attack. The in vivo conditions modulating the sensitivity and resistance of ß cells to this attack remain largely obscure. Here, we show that connexin 36 (Cx36), a trans-membrane protein that forms gap junctions between ß cells in the pancreatic islets, protects mouse ß cells against both cytotoxic drugs and cytokines that prevail in the islet environment at the onset of type 1 diabetes. We documented that this protection was at least partially dependent on intercellular communication, which Cx36 and other types of connexin channels establish within pancreatic islets. We further found that proinflammatory cytokines decreased expression of Cx36 and that experimental reduction or augmentation of Cx36 levels increased or decreased ß cell apoptosis, respectively. Thus, we conclude that Cx36 is central to ß cell protection from toxic insults.


Subject(s)
Connexins/physiology , Diabetes Mellitus, Experimental/prevention & control , Islets of Langerhans/pathology , Alloxan/pharmacology , Alloxan/toxicity , Animals , Apoptosis/drug effects , Cell Communication , Cellular Microenvironment , Connexins/antagonists & inhibitors , Connexins/deficiency , Connexins/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Gap Junctions/physiology , Gene Dosage , Insulin/genetics , Interferon-gamma/toxicity , Interleukin-1beta/toxicity , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide/biosynthesis , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Recombinant Fusion Proteins/physiology , Streptozocin/pharmacology , Streptozocin/toxicity , Tumor Necrosis Factor-alpha/toxicity , Gap Junction delta-2 Protein
9.
PLoS One ; 6(9): e25178, 2011.
Article in English | MEDLINE | ID: mdl-21949881

ABSTRACT

Imbalance of the excitatory neurotransmitter glutamate and of the inhibitory neurotransmitter GABA is one of several causes of seizures. ATP has also been implicated in epilepsy. However, little is known about the mechanisms involved in the release of ATP from cells and the consequences of the altered ATP signaling during seizures. Pannexin1 (Panx1) is found in astrocytes and in neurons at high levels in the embryonic and young postnatal brain, declining in adulthood. Panx1 forms large-conductance voltage sensitive plasma membrane channels permeable to ATP that are also activated by elevated extracellular K(+) and following P2 receptor stimulation. Based on these properties, we hypothesized that Panx1 channels may contribute to seizures by increasing the levels of extracellular ATP. Using pharmacological tools and two transgenic mice deficient for Panx1 we show here that interference with Panx1 ameliorates the outcome and shortens the duration of kainic acid-induced status epilepticus. These data thus indicate that the activation of Panx1 in juvenile mouse hippocampi contributes to neuronal hyperactivity in seizures.


Subject(s)
Behavior, Animal/drug effects , Connexins/physiology , Epilepsy/prevention & control , Nerve Tissue Proteins/physiology , Seizures/prevention & control , Adenosine Triphosphate/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Blotting, Western , Brain/cytology , Brain/metabolism , Cells, Cultured , Epilepsy/chemically induced , Epilepsy/metabolism , Fluorescent Antibody Technique , Hippocampus/cytology , Hippocampus/metabolism , Kainic Acid/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Potassium/metabolism , Seizures/chemically induced , Seizures/metabolism , Status Epilepticus/metabolism
10.
Pediatr Res ; 70(2): 142-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21527868

ABSTRACT

Diabetes develops when the insulin needs of peripheral cells exceed the availability or action of the hormone. This situation results from the death of most beta-cells in type 1 diabetes, and from an inability of the beta-cell mass to adapt to increasing insulin needs in type 2 and gestational diabetes. We analyzed several lines of transgenic mice and showed that connexins (Cxs), the transmembrane proteins that form gap junctions, are implicated in the modulation of the beta-cell mass. Specifically, we found that the native Cx36 does not alter islet size or insulin content, whereas the Cx43 isoform increases both parameters, and Cx32 has a similar effect only when combined with GH. These findings open interesting perspectives for the in vitro and in vivo regulation of the beta-cell mass.


Subject(s)
Cell Size , Connexin 43/metabolism , Connexins/metabolism , Diabetes Mellitus/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Connexin 43/genetics , Connexins/genetics , Crosses, Genetic , Fluorescent Antibody Technique , Growth Hormone/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Radioimmunoassay , Statistics, Nonparametric , Gap Junction beta-1 Protein , Gap Junction delta-2 Protein
11.
Hum Mol Genet ; 19(2): 262-75, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19864490

ABSTRACT

Targeting of numerous transmembrane proteins to the cell surface is thought to depend on their recognition by cargo receptors that interact with the adaptor machinery for anterograde traffic at the distal end of the Golgi complex. We report here on consortin, a novel integral membrane protein that is predicted to be intrinsically disordered, i.e. that contains large segments whose native state is unstructured. We identified consortin as a binding partner of connexins, the building blocks of gap junctions. Consortin is located at the trans-Golgi network (TGN), in tubulovesicular transport organelles, and at the plasma membrane. It directly interacts with the TGN clathrin adaptors GGA1 and GGA2, and disruption of this interaction by expression of a consortin mutant lacking the acidic cluster-dileucine (DXXLL) GGA interaction motif causes an intracellular accumulation of several connexins. RNA interference-mediated silencing of consortin expression in HeLa cells blocks the cell surface targeting of these connexins, which accumulate intracellularly, whereas partial depletion and redistribution of the consortin pool slows down the intracellular degradation of gap junction plaques. Altogether, our results show that, by studying connexin trafficking, we have identified the first TGN cargo receptor for the targeting of transmembrane proteins to the plasma membrane. The identification of consortin provides in addition a potential target for therapies aimed at diseases in which connexin traffic is altered, including cardiac ischemia, peripheral neuropathies, cataracts and hearing impairment. Sequence accession numbers. GenBank: Human CNST cDNA, NM_152609; mouse Cnst cDNA, NM_146105.


Subject(s)
Carrier Proteins/metabolism , Cell Membrane/metabolism , Connexins/metabolism , Membrane Proteins/metabolism , trans-Golgi Network/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Carrier Proteins/genetics , Cell Membrane/genetics , Connexins/genetics , HeLa Cells , Humans , Membrane Proteins/genetics , Mice , Protein Binding , Protein Transport , trans-Golgi Network/genetics
12.
Hum Mol Genet ; 18(3): 428-39, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19000992

ABSTRACT

Previous studies have documented that the insulin-producing beta-cells of laboratory rodents are coupled by gap junction channels made solely of the connexin36 (Cx36) protein, and have shown that loss of this protein desynchronizes beta-cells, leading to secretory defects reminiscent of those observed in type 2 diabetes. Since human islets differ in several respects from those of laboratory rodents, we have now screened human pancreas, and islets isolated thereof, for expression of a variety of connexin genes, tested whether the cognate proteins form functional channels for islet cell exchanges, and assessed whether this expression changes with beta-cell function in islets of control and type 2 diabetics. Here, we show that (i) different connexin isoforms are differentially distributed in the exocrine and endocrine parts of the human pancreas; (ii) human islets express at the transcript level different connexin isoforms; (iii) the membrane of beta-cells harbors detectable levels of gap junctions made of Cx36; (iv) this protein is concentrated in lipid raft domains of the beta-cell membrane where it forms gap junctions; (v) Cx36 channels allow for the preferential exchange of cationic molecules between human beta-cells; (vi) the levels of Cx36 mRNA correlated with the expression of the insulin gene in the islets of both control and type 2 diabetics. The data show that Cx36 is a native protein of human pancreatic islets, which mediates the coupling of the insulin-producing beta-cells, and contributes to control beta-cell function by modulating gene expression.


Subject(s)
Connexins/metabolism , Gap Junctions/metabolism , Gene Expression , Insulin-Secreting Cells/metabolism , Insulin/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Cells, Cultured , Connexins/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Gap Junctions/genetics , Humans , Insulin/metabolism , Islets of Langerhans/metabolism , Pancreas/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Gap Junction delta-2 Protein
13.
Cell Commun Adhes ; 15(1): 143-54, 2008 May.
Article in English | MEDLINE | ID: mdl-18649186

ABSTRACT

Connexins and pannexins have been implicated in the formation of "hemichannels," which may account for the uptake and release of membrane-impermeant molecules in single cells. The in vivo existence of "hemichannels" and their protein composition is still debated. Investigations on these matters are complicated by the lack of adequate negative controls. In search for such essential controls, the authors have investigated transformed (MIN6 line) and primary insulin-producing cells. Here, the authors report that these cells, which express Cx36 and pannexin 1, cannot be shown to display functional "hemichannels," as evaluated by (1) uptake of the membrane-impermeant tracer ethidium bromide, whether in the presence or absence of extracellular Ca(2+), following stimulation of P2X(7) receptors, and after exposure to hypotonic medium; and (2) lack of exocytosis-independent release of endogenous ATP. Moreover, electrophysiological recordings indicated the absence of carbenoxolone-sensitive pannexin 1 currents evoked by membrane potentials above +30 mV. Thus, insulin-producing cells are expected to provide a useful tool in the further characterization of hemichannel composition, properties, and physiological relevance.


Subject(s)
Connexins/metabolism , Insulin-Secreting Cells/metabolism , Animals , Cell Line, Transformed , Cell Line, Tumor , Cells, Cultured , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Microscopy, Confocal
14.
Exp Cell Res ; 314(5): 997-1012, 2008 Mar 10.
Article in English | MEDLINE | ID: mdl-18258229

ABSTRACT

Connexin36 (Cx36) is the main connexin isoform expressed in neurons of the central nervous system (CNS) and in pancreatic beta-cells, i.e. two types of excitable cells that share - in spite of their different origins - a number of common features. Previous studies on Cx36 deficient mice have documented that loss of Cx36 resulted in phenotypic abnormalities in both the CNS and the pancreas which, however, could not be attributed to specific cell types due to the general deletion nature of the animal model used. Attempts to address this limitation using cell type specific deletions generated by the Cre/loxP strategy have so far been complicated by the lack of Cx36 expression from the floxed allele. We have now generated a conditional Cx36 deficient mouse mutant in which the coding region of Cx36 is flanked by loxP sites, followed by a cyan fluorescent protein (CFP) reporter gene. Here we show that Cx36 was still expressed from the floxed allele in neurons and pancreatic beta-cells. In these cells, a 30-60% decrease of this protein, relative to the expression level of the wildtype allele, did not significantly perturb cell coupling. The deletion of Cx36 by ubiquitously and cell type specifically expressed Cre recombinases revealed that CFP functions as a reliable reporter for Cx36 expression in brain neurons and to some extent in retina neurons, but not in pancreas. Loss of Cx36 by Cre-mediated recombination was documented at transcript and protein levels. Cell type specific deletion of Cx36 in the endocrine pancreas revealed major alterations in the basal as well as the glucose-induced insulin secretion, hence specifically attributing to pancreatic Cx36 an important regulatory role in the control of beta-cell function. Cell type specific deletion of Cx36 in the CNS by suitable Cre recombinases should also help to elucidate the functional role of Cx36 in different neuronal subtypes.


Subject(s)
Connexins/genetics , Connexins/physiology , Insulin-Secreting Cells/chemistry , Neurons/chemistry , Animals , Brain/cytology , Connexins/deficiency , Gene Expression Regulation , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Integrases , Mice , Retina/cytology , Tissue Distribution , Gap Junction delta-2 Protein
15.
Diabetes ; 56(4): 1078-86, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17395748

ABSTRACT

We studied the effect of gap junctional coupling on the excitability of beta-cells in slices of pancreas, which provide a normal environment for islet cells. The electrophysiological properties of beta-cells from mice (C57Bl/6 background) lacking the gap junction protein connexin36 (Cx36(-/-)) were compared with heterozygous (Cx36(+/-)) and wild-type littermates (Cx36(+/+)) and with frequently used wild-type NMRI mice. Most electrophysiological characteristics of beta-cells were found to be unchanged after the knockout of Cx36, except the density of Ca(2+) channels, which was increased in uncoupled cells. With closed ATP-sensitive K(+) (K(ATP)) channels, the electrically coupled beta-cells of Cx36(+/+) and Cx36(+/-) mice were hyperpolarized by the membrane potential of adjacent, inactive cells. Additionally, the hyperpolarization of one beta-cell could attenuate or even stop the electrical activity of nearby coupled cells. In contrast, beta-cells of Cx36(-/-) littermates with blocked K(ATP) channels rapidly depolarized and exhibited a continuous electrical activity. Absence of electrical coupling modified the electrophysiological properties of beta-cells consistent with the reported increase in basal insulin release and altered the switch on/off response of beta-cells during an acute drop of the glucose concentration. Our data indicate an important role for Cx36-gap junctions in modulating stimulation threshold and kinetics of insulin release.


Subject(s)
Connexins/physiology , Glucose/physiology , Insulin-Secreting Cells/physiology , Insulin/metabolism , Animals , Connexins/deficiency , Connexins/genetics , Electrophysiology , Insulin Secretion , Insulin-Secreting Cells/metabolism , Kinetics , Membrane Potentials , Mice , Mice, Inbred Strains , Mice, Knockout , Patch-Clamp Techniques , Gap Junction delta-2 Protein
16.
Diabetes ; 56(4): 950-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17229940

ABSTRACT

The transcription factor regulatory factor X (RFX)-3 regulates the expression of genes required for the growth and function of cilia. We show here that mouse RFX3 is expressed in developing and mature pancreatic endocrine cells during embryogenesis and in adults. RFX3 expression already is evident in early Ngn3-positive progenitors and is maintained in all major pancreatic endocrine cell lineages throughout their development. Primary cilia of hitherto unknown function present on these cells consequently are reduced in number and severely stunted in Rfx3(-/-) mice. This ciliary abnormality is associated with a developmental defect leading to a uniquely altered cellular composition of the islets of Langerhans. Just before birth, Rfx3(-/-) islets contain considerably less insulin-, glucagon-, and ghrelin-producing cells, whereas pancreatic polypeptide-positive cells are markedly increased in number. In adult mice, the defect leads to small and disorganized islets, reduced insulin production, and impaired glucose tolerance. These findings suggest that RFX3 participates in the mechanisms that govern pancreatic endocrine cell differentiation and that the presence of primary cilia on islet cells may play a key role in this process.


Subject(s)
DNA-Binding Proteins/physiology , Islets of Langerhans/physiology , Transcription Factors/physiology , Animals , Cilia/physiology , Cilia/ultrastructure , Crosses, Genetic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Female , Ghrelin , Glucose Tolerance Test , Islets of Langerhans/cytology , Mice , Mice, Knockout , Peptide Hormones/analysis , Pregnancy , RNA, Messenger/genetics , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Stem Cells/physiology , TATA-Box Binding Protein/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
17.
Arch Physiol Biochem ; 112(2): 74-81, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16931449

ABSTRACT

Most cell types are functionally coupled by connexin (Cx) channels, i.e. exchange cytoplasmic ions and small metabolites through gap junction domains of their membrane. This form of direct cell-to-cell communication occurs in all existing animals, whatever their position in the phylogenetic scale, and up to humans. Pancreatic beta-cells are no exception, and normally cross-talk with their neighbors via channels made of Cx36. These exchanges importantly contribute to coordinate and synchronize the function of individual cells within pancreatic islets, particularly in the context of glucose-induced insulin secretion. Compelling evidence now indicates that Cx36-mediated coupling, and/or the Cx36 protein per se, play significant regulatory roles in various beta-cell functions, ranging from the biosynthesis, storage and release of insulin. Recent preliminary data further suggest that the protein may also be implicated in the balance of beta-cell growth versus necrosis and apoptosis, and in the regulated expression of specific genes. Here, we review this evidence, discuss the possible involvement of Cx36 in the pathophysiology of diabetes, and evaluate the relevance of this connexin in the therapeutic approaches to the disease.


Subject(s)
Connexins/physiology , Insulin-Secreting Cells/metabolism , Animals , Connexins/genetics , Humans , Gap Junction delta-2 Protein
18.
J Clin Invest ; 116(2): 405-13, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16440062

ABSTRACT

To investigate the function of Cx43 during hypertension, we studied the mouse line Cx43KI32 (KI32), in which the coding region of Cx32 replaces that of Cx43. Within the kidneys of homozygous KI32 mice, Cx32 was expressed in cortical and medullary tubules, as well as in some extra- and intraglomerular vessels, i.e., at sites where Cx32 and Cx43 are found in WT mice. Under such conditions, renin expression was much reduced compared with that observed in the kidneys of WT and heterozygous KI32 littermates. After exposure to a high-salt diet, all mice retained a normal blood pressure. However, whereas the levels of renin were significantly reduced in the kidneys of WT and heterozygous KI32 mice, reaching levels comparable to those observed in homozygous littermates, they were not further affected in the latter animals. Four weeks after the clipping of a renal artery (the 2-kidney, 1-clip [2K1C] model), 2K1C WT and heterozygous mice showed an increase in blood pressure and in the circulating levels of renin, whereas 2K1C homozygous littermates remained normotensive and showed unchanged plasma renin activity. Hypertensive, but not normotensive, mice also developed cardiac hypertrophy. The data indicate that replacement of Cx43 by Cx32 is associated with decreased expression and secretion of renin, thus preventing the renin-dependent hypertension that is normally induced in the 2K1C model.


Subject(s)
Connexin 43/metabolism , Hypertension/metabolism , Renin/metabolism , Animals , Blood Pressure/physiology , Connexin 43/genetics , Connexins/genetics , Connexins/metabolism , Kidney Tubules/cytology , Kidney Tubules/metabolism , Mice , Mice, Inbred Strains , Mice, Transgenic , RNA, Messenger/metabolism , Renal Artery Obstruction , Renin/genetics , Sodium, Dietary , Gap Junction beta-1 Protein
19.
Biochim Biophys Acta ; 1719(1-2): 82-101, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16359942

ABSTRACT

Glands were the first type of tissues in which the permissive role of gap junctions in the cell-to-cell transfer of membrane-impermeant molecules was shown. During the 40 years that have followed this seminal finding, gap junctions have been documented in all types of multicellular secretory systems, whether of the exocrine, endocrine or pheromonal nature. Also, compelling evidence now indicates that gap junction-mediated coupling, and/or the connexin proteins per se, play significant regulatory roles in various aspects of gland functions, ranging from the biosynthesis, storage and release of a variety of secretory products, to the control of the growth and differentiation of secretory cells, and to the regulation of gland morphogenesis. This review summarizes this evidence in the light of recent reports.


Subject(s)
Cell Communication , Connexins/physiology , Endocrine Glands/metabolism , Exocrine Glands/metabolism , Gap Junctions/physiology , Animals , Calcium/metabolism , Cell Membrane/metabolism , Connexins/metabolism , Humans , Models, Biological , Protein Isoforms , Signal Transduction , Tissue Distribution , Transgenes
20.
Hum Mutat ; 25(6): 543-9, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15880785

ABSTRACT

Mutations in claudin 14 (CLDN14) cause nonsyndromic DFNB29 deafness in humans. The analysis of a murine model indicated that this phenotype is associated with degeneration of hair cells, possibly due to cation overload. However, the mechanism linking these alterations to CLDN14 mutations is unknown. To investigate this mechanism, we compared the ability of wild-type and missense mutant CLDN14 to form tight junctions. Ectopic expression in L mouse fibroblasts (LM cells) of wild-type CLDN14 protein induced the formation of tight junctions, while both the c.254T>A (p.V85D) mutant, previously identified in a Pakistani family, and the c.301 G>A (p.G101R) mutant, identified in this study through the screen of 183 Spanish and Greek patients affected with sporadic nonsyndromic deafness, failed to form such junctions. However, the two mutant proteins differed in their ability to localize at the plasma membrane. We further identified hitherto undescribed exons of CLDN14 that are utilized in alternative spliced transcripts. We demonstrated that different mutations of CLDN14 impaired by different mechanisms the ability of the protein to form tight junctions. Our results indicate that the ability of CLDN14 to be recruited to these junctions is crucial for the hearing process.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation/genetics , Tight Junctions/metabolism , Alternative Splicing/genetics , Amino Acid Sequence , Animals , Claudins , Dogs , Exons/genetics , Fibroblasts/cytology , Humans , Introns/genetics , Membrane Proteins/chemistry , Mice , Molecular Sequence Data , Protein Transport , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Tight Junctions/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...