Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(24): 3464-3476, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28114277

ABSTRACT

Megakaryoblastic Leukemia 1 and 2 (MKL1/2) are transcriptional coactivators of Serum Response Factor (SRF) with an essential role for hepatocellular carcinoma (HCC) growth and oncogene-induced senescence. In this report, we identified myoferlin as a novel MKL/SRF target gene by gene expression profiling and verification in vivo in HCC xenografts. Myoferlin was overexpressed in human and murine HCCs triggered by conditional expression of constitutively active SRF-VP16 protein in hepatocytes. Furthermore, myoferlin was required for HCC cell invasion, proliferation and anchorage-independent cell growth. We provide evidence that myoferlin is a crucial gene target of MKL1/2 mediating its effect on oncogene-induced senescence by modulating the activation state of the EGFR and downstream MAPK and p16-/Rb pathways. Depletion of myoferlin in tumour cells from SRF-VP16-derived murine HCCs induced a senescence phenotype. These findings identify MKL1/2 and myoferlin as novel therapeutic targets to treat human HCC by a senescence-inducing strategy.


Subject(s)
Calcium-Binding Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Gene Expression Profiling/methods , Liver Neoplasms/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Serum Response Factor/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Calcium-Binding Proteins/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/genetics , Membrane Proteins/genetics , Mice , Muscle Proteins/genetics , NIH 3T3 Cells , Neoplasm Invasiveness , Neoplasm Transplantation
2.
Int Immunopharmacol ; 29(1): 158-65, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26033492

ABSTRACT

Specialized epithelial cells in the respiratory tract such as solitary chemosensory cells and brush cells sense the luminal content and initiate protective reflexes in response to the detection of potentially harmful substances. The majority of these cells are cholinergic and utilize the canonical taste signal transduction cascade to detect "bitter" substances such as bacterial quorum sensing molecules. Utilizing two different mouse strains reporting expression of choline acetyltransferase (ChAT), the synthesizing enzyme of acetylcholine (ACh), we detected cholinergic cells in the submucosal glands of the murine larynx and trachea. These cells were localized in the ciliated glandular ducts and were neither found in the collecting ducts nor in alveolar or tubular segments of the glands. ChAT expression in tracheal gland ducts was confirmed by in situ hybridization. The cholinergic duct cells expressed the brush cell marker proteins, villin and cytokeratin-18, and were immunoreactive for components of the taste signal transduction cascade (Gα-gustducin, transient receptor potential melastatin-like subtype 5 channel = TRPM5, phospholipase C(ß2)), but not for carbonic anhydrase IV. Furthermore, these cells expressed the bitter taste receptor Tas2r131, as demonstrated utilizing an appropriate reporter mouse strain. Our study identified a previously unrecognized presumptive chemosensory cell type in the duct of the airway submucosal glands that likely utilizes ACh for paracrine signaling. We propose that these cells participate in infection-sensing mechanisms and initiate responses assisting bacterial clearance from the lower airways.


Subject(s)
Acetylcholine/metabolism , Chemoreceptor Cells/metabolism , Epithelial Cells/metabolism , Larynx/cytology , Trachea/cytology , Animals , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Gene Expression Regulation, Enzymologic , Green Fluorescent Proteins , Mice , Mice, Inbred C57BL , Mice, Transgenic
3.
J Dent Res ; 94(7): 945-54, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25838461

ABSTRACT

In organized tissues, the precise geometry and the overall shape are critical for the specialized functions that the cells carry out. Odontoblasts are major matrix-producing cells of the tooth and have also been suggested to participate in sensory transmission. However, refined morphologic data on these important cells are limited, which hampers the analysis and understanding of their cellular functions. We took advantage of fluorescent color-coding genetic tracing to visualize and reconstruct in 3 dimensions single odontoblasts, pulp cells, and their assemblages. Our results show distinct structural features and compartments of odontoblasts at different stages of maturation, with regard to overall cellular shape, formation of the main process, orientation, and matrix deposition. We demonstrate previously unanticipated contacts between the processes of pulp cells and odontoblasts. All reported data are related to mouse incisor tooth. We also show that odontoblasts express TRPM5 and Piezo2 ion channels. Piezo2 is expressed ubiquitously, while TRPM5 is asymmetrically distributed with distinct localization to regions proximal to and within odontoblast processes.


Subject(s)
Imaging, Three-Dimensional/methods , Odontoblasts/cytology , Ameloblasts/cytology , Ameloblasts/ultrastructure , Animals , Cell Compartmentation , Cell Nucleus/ultrastructure , Cell Shape , Cell Surface Extensions/ultrastructure , Dental Pulp/cytology , Dental Pulp/ultrastructure , Dentin/ultrastructure , Extracellular Matrix/ultrastructure , Fluorescent Antibody Technique , Incisor/cytology , Incisor/ultrastructure , Ion Channels/ultrastructure , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/ultrastructure , Mice , Mice, Transgenic , Microscopy, Electron, Scanning/methods , Odontoblasts/ultrastructure , TRPM Cation Channels/ultrastructure
4.
Pflugers Arch ; 466(12): 2177-89, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24633576

ABSTRACT

Transient receptor potential cation channel, subfamily M, member 7 (TRPM7) is a cation channel covalently linked to a protein kinase domain. TRPM7 is ubiquitously expressed and regulates key cellular processes such as Mg(2+) homeostasis, motility, and proliferation. TRPM7 is involved in anoxic neuronal death, cardiac fibrosis, and tumor growth. The goal of this work was to identify small molecule activators of the TRPM7 channel and investigate their mechanism of action. We used an aequorin bioluminescence-based assay to screen for activators of the TRPM7 channel. Valid candidates were further characterized using patch clamp electrophysiology. We identified 20 drug-like compounds with various structural backbones that can activate the TRPM7 channel. Among them, the δ opioid antagonist naltriben was studied in greater detail. Naltriben's action was selective among the TRP channels tested. Naltriben activates TRPM7 currents without prior depletion of intracellular Mg(2+) even under conditions of low PIP2. Moreover, naltriben interfered with the effect of the TRPM7 inhibitor NS8593. Finally, our experiments with TRPM7 variants carrying mutations in the pore, TRP, and kinase domains indicate that the site of TRPM7 activation by this small-molecule ligand is most likely located in or near the TRP domain. In conclusion, we identified the first organic small-molecule activators of TRPM7 channels, thus providing new experimental tools to study TRPM7 function in native cellular environments.


Subject(s)
Small Molecule Libraries/pharmacology , TRPM Cation Channels/agonists , Action Potentials , Animals , HEK293 Cells , Humans , Magnesium/metabolism , Mice , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Small Molecule Libraries/chemistry , TRPM Cation Channels/metabolism
6.
Histochem Cell Biol ; 137(4): 483-97, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22261922

ABSTRACT

The luminal composition of the auditory tube influences its function. The mechanisms involved in the monitoring are currently not known. For the lower respiratory epithelium, such a sentinel role is carried out by cholinergic brush cells. Here, using two different mouse strains expressing eGFP under the control of the promoter of choline acetyltransferase (ChAT), we show the presence of solitary cholinergic villin-positive brush cells also in the mouse auditory tube epithelium. They express the vesicular acetylcholine (ACh) transporter and proteins of the taste transduction pathway such as α-gustducin, phospholipase C beta 2 (PLC(ß2)) and transient receptor potential cation channel subfamily M member 5 (TRPM5). Immunoreactivity for TRPM5 and PLCß2 was found regularly, whereas α-gustducin was absent in approximately 15% of the brush cells. Messenger RNA for the umami taste receptors (TasR), Tas1R1 and 3, and for the bitter receptors, Tas2R105 and Tas2R108, involved in perception of cycloheximide and denatonium were detected in the auditory tube. Using a transgenic mouse that expresses eGFP under the promotor of the nicotinic ACh receptor α3-subunit, we identified cholinoceptive nerve fibers that establish direct contacts to brush cells in the auditory tube. A subpopulation of these fibers displayed also CGRP immunoreactivity. Collectively, we show for the first time the presence of brush cells in the auditory tube. These cells are equipped with all proteins essential for sensing the composition of the luminal microenvironment and for communication of the changes to the CNS via attached sensory nerve fibers.


Subject(s)
Chemoreceptor Cells/cytology , Cholinergic Neurons/cytology , Eustachian Tube/cytology , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tongue/cytology
7.
Br J Pharmacol ; 166(4): 1357-76, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22242975

ABSTRACT

BACKGROUND AND PURPOSE: Transient receptor potential cation channel subfamily M member 7 (TRPM7) is a bifunctional protein comprising a TRP ion channel segment linked to an α-type protein kinase domain. TRPM7 is essential for proliferation and cell growth. Up-regulation of TRPM7 function is involved in anoxic neuronal death, cardiac fibrosis and tumour cell proliferation. The goal of this work was to identify non-toxic inhibitors of the TRPM7 channel and to assess the effect of blocking endogenous TRPM7 currents on the phenotype of living cells. EXPERIMENTAL APPROACH: We developed an aequorin bioluminescence-based assay of TRPM7 channel activity and performed a hypothesis-driven screen for inhibitors of the channel. The candidates identified were further assessed electrophysiologically and in cell biological experiments. KEY RESULTS: TRPM7 currents were inhibited by modulators of small conductance Ca²âº -activated K⁺ channels (K(Ca)2.1-2.3; SK) channels, including the antimalarial plant alkaloid quinine, CyPPA, dequalinium, NS8593, SKA31 and UCL 1684. The most potent compound NS8593 (IC50 1.6 µM) specifically targeted TRPM7 as compared with other TRP channels, interfered with Mg²âº -dependent regulation of TRPM7 channel and inhibited the motility of cultured cells. NS8593 exhibited full and reversible block of native TRPM7-like currents in HEK 293 cells, freshly isolated smooth muscle cells, primary podocytes and ventricular myocytes. CONCLUSIONS AND IMPLICATIONS: This study reveals a tight overlap in the pharmacological profiles of TRPM7 and K(Ca)2.1-2.3 channels. NS8593 acts as a negative gating modulator of TRPM7 and is well-suited to study functional features and cellular roles of endogenous TRPM7.


Subject(s)
Drug Discovery , Magnesium/metabolism , Membrane Transport Modulators/pharmacology , Potassium Channel Blockers/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , TRPM Cation Channels/antagonists & inhibitors , 1-Naphthylamine/adverse effects , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , Animals , Calcium Signaling/drug effects , Cell Movement/drug effects , Cells, Cultured , HEK293 Cells , Humans , Membrane Potentials/drug effects , Membrane Transport Modulators/adverse effects , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Podocytes/cytology , Podocytes/drug effects , Podocytes/metabolism , Potassium Channel Blockers/adverse effects , Protein Isoforms/antagonists & inhibitors , Protein Serine-Threonine Kinases , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
8.
Oncogene ; 31(35): 3913-23, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22139079

ABSTRACT

Deleted in Liver Cancer 1 (DLC1) is a tumor suppressor whose allele is lost in 50% of liver, breast, lung and 70% of colon cancers. Here, we show that the transcriptional coactivators Megakaryoblastic Leukemia 1 and 2 (MKL1/2) are constitutively localized to the nucleus in hepatocellular and mammary carcinoma cells that lack DLC1. Moreover, DLC1 loss and MKL1 nuclear localization correlate in primary human hepatocellular carcinoma. Nuclear accumulation of MKL1 in DLC1-deficient cancer cells is accomplished by activation of the RhoA/actin signaling pathway and concomitant impairment of MKL1 phosphorylation, which results in constitutive activation of MKL1/2 target genes. We provide evidence that MKL1/2 mediates cancerous transformation in DLC1-deficient hepatocellular and mammary carcinoma cells. Depletion of MKL1/2 suppresses cell migration, cell proliferation and anchorage-independent cell growth induced by DLC1 loss.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Hepatocellular/metabolism , DNA-Binding Proteins/metabolism , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , Liver Neoplasms/metabolism , Oncogene Proteins, Fusion/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Breast Neoplasms/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Cell Transformation, Neoplastic , GTPase-Activating Proteins/metabolism , Humans , Liver Neoplasms/genetics , Signal Transduction , Trans-Activators , Tumor Suppressor Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
9.
Adv Exp Med Biol ; 661: 187-200, 2010.
Article in English | MEDLINE | ID: mdl-20204731

ABSTRACT

Hypoxic pulmonary vasoconstriction (HPV) is an essential mechanism of the lung matching blood perfusion to ventilation during local alveolar hypoxia. HPV thus optimizes pulmonary gas exchange. In contrast chronic and generalized hypoxia leads to pulmonary vascular remodeling with subsequent pulmonary hypertension and right heart hypertrophy. Among other non-selective cation channels, the family of classical transient receptor potential channels (TRPC) has been shown to be expressed in pulmonary arterial smooth muscle cells. Among this family, TRPC6 is essential for the regulation of acute HPV in mice. Against this background, in this chapter we give an overview about the TRPC family and their role in HPV.


Subject(s)
Hypoxia/metabolism , Lung , Protein Isoforms/metabolism , Pulmonary Artery/physiology , Transient Receptor Potential Channels/metabolism , Vasoconstriction/physiology , Animals , Calcium/metabolism , Humans , Lung/blood supply , Lung/metabolism , Phylogeny , Protein Isoforms/chemistry , Protein Isoforms/classification , Protein Isoforms/genetics , Protein Structure, Secondary , Signal Transduction/physiology , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/classification , Transient Receptor Potential Channels/genetics
10.
Thromb Haemost ; 103(2): 262-70, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20126834

ABSTRACT

The mammalian transient receptor potential (TRP) superfamily of non-selective cation channels can be divided into six major families. Among them, the "classical" or "canonical" TRPC family is most closely related to Drosophila TRP, the founding member of the superfamily. All seven channels of this family designated TRPC1-7 share the common property of receptor-operated activation through phospholipase C (PLC)-coupled receptors, but their regulation by store-dependent mechanisms involving the proteins STIM and ORAi is still discussed controversially. This review will focus on the proposed functions of TRPC proteins in cells of the vascular system (e.g. platelets, smooth muscle cells and endothelial cells) and will present data concerning their physiological functions analysed in isolated tissues with down-regulated channel activity and in gene-deficient mouse models.


Subject(s)
Transient Receptor Potential Channels/physiology , Blood Platelets/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Type C Phospholipases/metabolism
11.
Eur Respir J ; 32(6): 1639-51, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19043010

ABSTRACT

Hypoxic pulmonary vasoconstriction (HPV), also known as the von Euler-Liljestrand mechanism, is a physiological response to alveolar hypoxia which distributes pulmonary capillary blood flow to alveolar areas of high oxygen partial pressure. Impairment of this mechanism may result in hypoxaemia. Under conditions of chronic hypoxia generalised vasoconstriction of the pulmonary vasculature in concert with hypoxia-induced vascular remodelling leads to pulmonary hypertension. Although the principle of HPV was recognised decades ago, its exact pathway still remains elusive. Neither the oxygen sensing process nor the exact pathway underlying HPV is fully deciphered yet. The effector pathway is suggested to include L-type calcium channels, nonspecific cation channels and voltage-dependent potassium channels, whereas mitochondria and nicotinamide adenine dinucleotide phosphate oxidases are discussed as oxygen sensors. Reactive oxygen species, redox couples and adenosine monophosphate-activated kinases are under investigation as mediators of hypoxic pulmonary vasoconstriction. Moreover, the role of calcium sensitisation, intracellular calcium stores and direction of change of reactive oxygen species is still under debate. In this context the present article focuses on the basic mechanisms of hypoxic pulmonary vasoconstriction and also outlines differences in current concepts that have been suggested for the regulation of hypoxic pulmonary vasoconstriction.


Subject(s)
Hypoxia , Vasoconstriction , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Capillaries/metabolism , Humans , Models, Biological , NADPH Oxidases/metabolism , Oxidation-Reduction , Oxygen/metabolism , Pressure , Pulmonary Artery/pathology , Pulmonary Circulation/physiology , Reactive Oxygen Species/metabolism , Signal Transduction
12.
Clin Exp Allergy ; 38(9): 1548-58, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18631347

ABSTRACT

BACKGROUND: Non-selective cation influx through canonical transient receptor potential channels (TRPCs) is thought to be an important event leading to airway inflammation. TRPC6 is highly expressed in the lung, but its role in allergic processes is still poorly understood. OBJECTIVE: The purpose of this study was to evaluate the role of TRPC6 in airway hyperresponsiveness (AHR) and allergic inflammation of the lung. METHODS: Methacholine-induced AHR was assessed by head-out body plethysmography of wild type (WT) and TRPC6(-/-) mice. Experimental airway inflammation was induced by intraperitoneal ovalbumin (OVA) sensitization, followed by OVA aerosol challenges. Allergic inflammation and mucus production were analysed 24 h after the last allergen challenge. RESULTS: Methacholine-induced AHR and agonist-induced contractility of tracheal rings were increased in TRPC6(-/-) mice compared with WT mice, most probably due to compensatory up-regulation of TRPC3 in airway smooth muscle cells. Most interestingly, when compared with WT mice, TRPC6(-/-) mice exhibited reduced allergic responses after allergen challenge as evidenced by a decrease in airway eosinophilia and blood IgE levels, as well as decreased levels of T-helper type 2 (Th2) cytokines (IL-5, IL-13) in the bronchoalveolar lavage. However, lung mucus production after allergen challenge was not altered by TRPC6 deficiency. CONCLUSIONS: TRPC6 deficiency inhibits specific allergic immune responses, pointing to an important immunological function of this cation channel in Th2 cells, eosinophils, mast cells and B cells.


Subject(s)
Bronchial Hyperreactivity/metabolism , Hypersensitivity/metabolism , TRPC Cation Channels/physiology , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Cells, Cultured , Epithelial Cells/metabolism , Guinea Pigs , Hypersensitivity/immunology , Hypersensitivity/physiopathology , Immunoglobulin E/blood , In Vitro Techniques , Interleukin-13/metabolism , Interleukin-5/metabolism , Leukocytes/pathology , Lung/immunology , Lung/metabolism , Mice , Mice, Knockout , Mucus/metabolism , Muscle Contraction , Muscle, Smooth/metabolism , Muscle, Smooth/physiopathology , Ovalbumin/immunology , Pulmonary Eosinophilia/immunology , Pulmonary Eosinophilia/pathology , TRPC Cation Channels/biosynthesis , TRPC Cation Channels/genetics , TRPC6 Cation Channel , Trachea/metabolism , Trachea/physiopathology
13.
Oncogene ; 27(12): 1737-48, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-17906699

ABSTRACT

Neuropeptide hormones like bombesin/gastrin-releasing peptide, galanin or bradykinin, acting via auto and paracrine growth loops, represent the principal mitogens of small cell lung cancer (SCLC). These mitogenic neuropeptides activate G(q/11)-coupled receptors which stimulate phospholipase Cbeta activity, followed by rises of the intracellular calcium concentration ([Ca2+](i)) and activation of protein kinase C (PKC). We report here that proline-rich tyrosine kinase Pyk2 is highly expressed in SCLC cells and provides a functional link between neuropeptide-induced increases in [Ca2+](i) and tumor cell proliferation. Activation of Pyk2 and its association with Src kinases critically depends on the elevation of [Ca2+](i), but is independent of PKC. Src kinase activities are crucial for neuropeptide-mediated GTP-loading of Ras and activation of extracellular signal-regulated kinases in SCLC cells. Pyk2 and Src kinases essentially contribute to anchorage-independent proliferation of SCLC cells. Inhibition of either Pyk2 or Src kinases by lentiviral RNAi or pharmacological inhibition with PP2, respectively, attenuated basal and neuropeptide-elicited survival and proliferation of SCLC cells in liquid culture and in soft agar. Thus, neuropeptides stimulate anchorage-independent survival and proliferation of SCLC cells via pathways involving Pyk2 and Src kinases. Therefore, Ca2+-induced Pyk2/Src complex formation may be a rewarding molecular target for novel therapeutic strategies in SCLC cells.


Subject(s)
Carcinoma, Small Cell/enzymology , Cell Proliferation , Focal Adhesion Kinase 2/physiology , Galanin/physiology , Lung Neoplasms/enzymology , src-Family Kinases/physiology , Calcium/physiology , Carcinoma, Small Cell/pathology , Cell Line , Cell Line, Tumor , Enzyme Activation/genetics , Focal Adhesion Kinase 2/biosynthesis , Focal Adhesion Kinase 2/genetics , Focal Adhesion Kinase 2/metabolism , Humans , Lung Neoplasms/pathology , Proline/metabolism , src-Family Kinases/metabolism
14.
Handb Exp Pharmacol ; (179): 125-41, 2007.
Article in English | MEDLINE | ID: mdl-17217054

ABSTRACT

TRPC6 is a Ca(2+)-permeable non-selective cation channel expressed in brain, smooth muscle containing tissues and kidney, as well as in immune and blood cells. Channel homomers heterologously expressed have a characteristic doubly rectifying current-voltage relationship and are six times more permeable for Ca2+ than for Na+. In smooth muscle tissues, however, Na+ influx and activation of voltage-gated calcium channels by membrane depolarization rather than Ca2+ elevation by TRPC6 channels is the driving force for contraction. TRPC6 channels are directly activated by the second messenger diacylglycerol (DAG) and regulated by specific tyrosine or serine phosphorylation. Extracellular Ca2+ has inhibitory effects, while Ca2+/calmodulin acting from the intracellular side has potentiator effects on channel activity. Given its specific expression, TRPC6 is likely to play a number of physiological roles. Studies with TRPC6(-/-) mice suggest a role for the channel in the regulation of vascular and pulmonary smooth muscle contraction. TRPC6 was identified as an essential component of the slit diaphragm architecture of kidney podocytes. Other functions in immune and blood cells, as well as in brain and in smooth muscle-containing tissues such as stomach, colon and myometrium, remain elusive.


Subject(s)
TRPC Cation Channels/genetics , TRPC Cation Channels/physiology , Animals , Gene Expression Regulation/physiology , Humans , Ion Channels/metabolism , TRPC Cation Channels/biosynthesis , TRPC6 Cation Channel
15.
Exp Clin Endocrinol Diabetes ; 113(5): 245-7, 2005 May.
Article in English | MEDLINE | ID: mdl-15926107

ABSTRACT

The receptors for the trophic hormones luteinizing hormone (LH), follicle-stimulating hormone (FSH), and thyrotropin (TSH) play a central role in endocrinology. These receptors face the challenge to accommodate large heterodimeric glycoprotein ligands within their extracellular hormone-binding domain. Until recently, the mechanism of hormone binding and consequently the mode of receptor activation remained enigmatic. By solving the crystal structure of human follicle-stimulating hormone bound to the receptor's hormone binding domain, it has become clear that the follicle-stimulating hormone receptor grabs the glycoprotein hormone in a hand-clasp mode resulting in a hormone orientation perpendicular to the long axis of the ligand-binding domain. These findings have important ramifications for our understanding of the molecular mechanism of receptor activation and may provide a rational basis for the development of small, non-peptidic FSH receptor ligands.


Subject(s)
Follicle Stimulating Hormone/chemistry , Follicle Stimulating Hormone/metabolism , Receptors, FSH/metabolism , Animals , Binding Sites , Crystallization , Dimerization , Humans , Molecular Structure , Protein Structure, Secondary , Receptors, FSH/chemistry
16.
Naunyn Schmiedebergs Arch Pharmacol ; 371(4): 334-41, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15902429

ABSTRACT

Investigations into Drosophila mutants with impaired vision due to mutations in the transient receptor potential gene (trp) initiated a systematic search for TRP homologs in other species, finally leading to the discovery of a whole new family of plasma membrane cation channels involved in multiple physiological processes. Among the recently discovered TRP cation channels two homologous proteins, TRPM6 and TRPM7, display unique domain compositions and biophysical properties. These remarkable genes are vital for Mg(2+) homeostasis in vertebrates and, if disrupted, lead to cell death or human disease.


Subject(s)
Magnesium/metabolism , Phosphotransferases/physiology , Signal Transduction/physiology , TRPM Cation Channels/physiology , Animals , Base Sequence , Humans , Mice , Molecular Sequence Data , Phosphotransferases/genetics , Phosphotransferases/metabolism , Protein Serine-Threonine Kinases , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
19.
J Clin Endocrinol Metab ; 86(9): 4429-33, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549687

ABSTRACT

Sporadic and familial nonautoimmune hyperthyroidism are very rarely occurring diseases. Within the last years constitutively activating TSH receptor mutations were identified as one possible pathomechanism. Except for S281N in the extracellular N-terminal domain, all other germline mutations are located in the transmembrane domains 2, 3, 5, 6, and 7 of the TSH receptor, whereas no mutation was reported in transmembrane domains 1 and 4 to date. Here we report the first family with a constitutively active TSHR mutation in transmembrane domain 1 resulting in a substitution of the conserved Gly(431) for Ser. This mutation was found in the investigated patient, his father, and the paternal grandmother. As known from other familial cases of nonautoimmune hyperthyroidism, the age of onset of the disease was variable, ranging from early childhood in the patient and his father to adolescence in the grandmother. Functional characterization of this mutation showed a constitutive activation of the G(s)/adenylyl cyclase system. Moreover, this germline mutation also activates the G(q/11)/phospholipase C pathway. The importance of Gly(431) for receptor quiescence is supported further by introduction of other mutations at this position, all leading to constitutive receptor activity. Our data show now that constitutively activating mutations can be found in the entire transmembrane domain region of the TSH receptor, indicating the important role of all parts of the transmembrane domain region for maintaining the inactive receptor conformation.


Subject(s)
DNA/genetics , Hyperthyroidism/genetics , Mutation, Missense/genetics , Receptors, Thyrotropin/genetics , Adenylyl Cyclases/genetics , Animals , COS Cells , Child, Preschool , Cyclic AMP/metabolism , Female , GTP-Binding Protein alpha Subunits, Gs/metabolism , Genome , Humans , Hyperthyroidism/blood , Male , Pedigree , Plasmids/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Hormones/blood , Thyrotropin/metabolism , Type C Phospholipases/metabolism
20.
Novartis Found Symp ; 239: 68-79; discussion 80-4, 150-9, 2001.
Article in English | MEDLINE | ID: mdl-11529317

ABSTRACT

It has only recently been fully realized that G protein-coupled receptors and G proteins play crucial roles in the regulation of cell growth, differentiation and even tumour formation. Naturally occurring mutations in G protein-coupled receptors and in G protein alpha subunits result in uncontrolled cellular proliferation resulting in distinct human diseases. One important mechanism to transduce mitogenic signals from the cell membrane to the cell nucleus is the engagement of the extracellular signal-regulated kinase (ERK)-mitogen-activated protein kinase (MAPK) cascade. A multitude of distinct signal transduction pathways have been deciphered which connect G proteins with the ERK cascade. Both receptor and non-receptor tyrosine kinases play pivotal roles in these signalling pathways. Mitogenic signalling by G protein-coupled receptors can be regarded as a complex interplay between signals emanating from different classes of cell surface receptors which ultimately converge upon a small subset of central signalling proteins in the cell. The characterization of receptor-, G protein- and tyrosine kinase-specific contributions to mitogenic signalling in a particular cell and the identification of proteins serving as a point of convergence in the mitogenic signalling cascade may ultimately allow the design of novel pharmacological approaches to treat diseases involving unrestricted cell proliferation.


Subject(s)
GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , GTP-Binding Proteins/metabolism , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Receptors, Cell Surface/metabolism , Calcium/metabolism , Cyclic AMP/metabolism , Cytoskeleton , Enzyme Activation , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Ligands , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...