Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biomolecules ; 13(5)2023 04 22.
Article in English | MEDLINE | ID: mdl-37238590

ABSTRACT

Cells in the body are exposed to dynamic external and internal environments, many of which cause cell damage. The cell's response to this damage, broadly called the stress response, is meant to promote survival and repair or remove damage. However, not all damage can be repaired, and sometimes, even worse, the stress response can overtax the system itself, further aggravating homeostasis and leading to its loss. Aging phenotypes are considered a manifestation of accumulated cellular damage and defective repair. This is particularly apparent in the primary cell type of the articular joint, the articular chondrocytes. Articular chondrocytes are constantly facing the challenge of stressors, including mechanical overloading, oxidation, DNA damage, proteostatic stress, and metabolic imbalance. The consequence of the accumulation of stress on articular chondrocytes is aberrant mitogenesis and differentiation, defective extracellular matrix production and turnover, cellular senescence, and cell death. The most severe form of stress-induced chondrocyte dysfunction in the joints is osteoarthritis (OA). Here, we summarize studies on the cellular effects of stressors on articular chondrocytes and demonstrate that the molecular effectors of the stress pathways connect to amplify articular joint dysfunction and OA development.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Oxidative Stress/physiology , Cartilage, Articular/pathology , Osteoarthritis/pathology , Cellular Senescence , Chondrocytes
2.
Sci Adv ; 9(14): eadg0731, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37018401

ABSTRACT

Women experience osteoporosis at higher rates than men. Aside from hormones, the mechanisms driving sex-dependent bone mass regulation are not well understood. Here, we demonstrate that the X-linked H3K4me2/3 demethylase KDM5C regulates sex-specific bone mass. Loss of KDM5C in hematopoietic stem cells or bone marrow monocytes increases bone mass in female but not male mice. Mechanistically, loss of KDM5C impairs the bioenergetic metabolism, resulting in impaired osteoclastogenesis. Treatment with the KDM5 inhibitor reduces osteoclastogenesis and energy metabolism of both female mice and human monocytes. Our report details a sex-dependent mechanism for bone homeostasis, connecting epigenetic regulation to osteoclast metabolism and positions KDM5C as a potential target for future treatment of osteoporosis in women.


Subject(s)
Osteoclasts , Osteoporosis , Animals , Female , Humans , Male , Mice , Energy Metabolism , Epigenesis, Genetic , Histone Demethylases/metabolism , Osteoclasts/metabolism
3.
bioRxiv ; 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36865269

ABSTRACT

Women experience osteoporosis at higher rates than men. Aside from hormones, the mechanisms driving sex-dependent bone mass regulation are not well-understood. Here, we demonstrate that the X-linked H3K4me2/3 demethylase KDM5C regulates sex-specific bone mass. Loss of KDM5C in hematopoietic stem cells or bone marrow monocytes (BMM) increases bone mass in female but not male mice. Mechanistically, loss of KDM5C impairs the bioenergetic metabolism resulting in impaired osteoclastogenesis. Treatment with the KDM5 inhibitor reduces osteoclastogenesis and energy metabolism of both female mice and human monocytes. Our report details a novel sex-dependent mechanism for bone homeostasis, connecting epigenetic regulation to osteoclast metabolism, and positions KDM5C as a target for future treatment of osteoporosis in women. One-Sentence Summary: KDM5C, an X-linked epigenetic regulator, controls female bone homeostasis by promoting energy metabolism in osteoclasts.

4.
Cell Rep ; 42(1): 112012, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36680774

ABSTRACT

Long bones are generated by mesoderm-derived skeletal progenitor/stem cells (SSCs) through endochondral ossification, a process of sequential chondrogenic and osteogenic differentiation tightly controlled by the synergy between intrinsic and microenvironment cues. Here, we report that loss of TRIM28, a transcriptional corepressor, in mesoderm-derived cells expands the SSC pool, weakens SSC osteochondrogenic potential, and endows SSCs with properties of ectoderm-derived neural crest cells (NCCs), leading to severe defects of skeletogenesis. TRIM28 preferentially enhances H3K9 trimethylation and DNA methylation on chromatin regions more accessible in NCCs; loss of this silencing upregulates neural gene expression and enhances neurogenic potential. Moreover, TRIM28 loss causes hyperexpression of GREM1, which is an extracellular signaling factor promoting SSC self-renewal and SSC neurogenic potential by activating AKT/mTORC1 signaling. Our results suggest that TRIM28-mediated chromatin silencing establishes a barrier for maintaining the SSC lineage trajectory and preventing a transition to ectodermal fate by regulating both intrinsic and microenvironment cues.


Subject(s)
Osteogenesis , Tripartite Motif-Containing Protein 28 , Cell Differentiation/genetics , Chromatin , Gene Expression , Proto-Oncogene Proteins c-akt/genetics , Stem Cells , TOR Serine-Threonine Kinases/genetics , Animals , Mice , Tripartite Motif-Containing Protein 28/metabolism , Signal Transduction
5.
Cells ; 11(17)2022 08 31.
Article in English | MEDLINE | ID: mdl-36078118

ABSTRACT

The modification of proteins by small ubiquitin-related modifier (SUMO) molecules, SUMOylation, is a key post-translational modification involved in a variety of biological processes, such as chromosome organization, DNA replication and repair, transcription, nuclear transport, and cell signaling transduction. In recent years, emerging evidence has shown that SUMOylation regulates the development and homeostasis of the skeletal system, with its dysregulation causing skeletal diseases, suggesting that SUMOylation pathways may serve as a promising therapeutic target. In this review, we summarize the current understanding of the molecular mechanisms by which SUMOylation pathways regulate skeletal cells in physiological and disease contexts.


Subject(s)
Small Ubiquitin-Related Modifier Proteins , Sumoylation , Homeostasis , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin/metabolism
6.
Article in English | MEDLINE | ID: mdl-30733708

ABSTRACT

RNF216, encoding an E3 ubiquitin ligase, has been identified as a causative gene for Gordon Holmes syndrome, characterized by ataxia, dementia, and hypogonadotropic hypogonadism. However, it is still elusive how deficiency in RNF216 leads to hypogonadotropic hypogonadism. In this study, by using GN11 immature GnRH neuronal cell line, we demonstrated an important role of RNF216 in the GnRH neuron migration. RNA interference of RNF216 inhibited GN11 cell migration, but had no effect on the proliferation of GN11 cells or GnRH expression. Knockdown of RNF216 increased the protein levels of its targets, Arc and Beclin1. RNAi of Beclin1, but not Arc, normalized the suppressive effect caused by RNF216 knockdown. As Beclin1 plays a critical role in the autophagy regulation, we further demonstrated that RNAi of RNF216 led to increase in autophagy, and autophagy inhibitor CQ and 3-MA rescued the GN11 cell migration deficit caused by RNF216 knockdown. We further demonstrated that pharmacological increase autophagy by rapamycin could suppress the GN11 cell migration. We thus have identified that RNF216 regulates the migration of GnRH neuron by suppressing Beclin1 mediated autophagy, and indicated a potential contribution of autophagy to the hypogonadotropic hypogonadism.

7.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 43(5): 461-468, 2018 May 28.
Article in Chinese | MEDLINE | ID: mdl-29886459

ABSTRACT

OBJECTIVE: To investigate the mechanism for the synergistic effect of interferon regulatory factor 4 (IRF4) and microphthalmia-associated transcription factor (MITF) on tyrosinase (TYR) promoter.
 Methods: The synergistic transcriptional effect, subcellular localization, and protein-protein interaction for IRF4 and MITF were observed by luciferase assay, immunofluorescence, GST-pull down, and co-immunoprecipitation, respectively.
 Results: IRF4 and MITF proteins were co-expressed in the cell nucleus. IRF4 augmented the transcriptional function of MITF (but not the mutant MITF) to activate the expression of the TYR promoter, but with no effect on other MITF-specific target promoters. IRF4 alone did not affect TYR promoter significantly. No direct interaction between the two proteins was noted.
 Conclusion: IRF4 and MITF exert a specifically synergistic effect on activation of TYR promoter through IRF4-mediated upregulation of transcriptional function of MITF. This synergistic effect is mainly regulated by MITF; DNA might be involved in the interaction between the two proteins.


Subject(s)
Cell Nucleus/metabolism , Interferon Regulatory Factors/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Monophenol Monooxygenase/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Luciferases
8.
J Bone Miner Res ; 33(10): 1773-1784, 2018 10.
Article in English | MEDLINE | ID: mdl-29750835

ABSTRACT

Single-nucleotide polymorphisms in the LRP1 gene coding sequence are associated with low bone mass, and cell culture studies suggest that LRP1 plays a role in osteoblast proliferation and osteoblast-mediated osteoclastogenesis. However, the in vivo function of LRP1 in bone homeostasis has not been explored. In this work, we studied the osteoclast-specific role of LRP1 in bone homeostasis using a Ctsk-Cre;Lrp1f/f mouse model on the C57BL/6J background. These mice had a dramatically decreased trabecular bone mass with markedly more osteoclasts, while the osteoblast activity was unaffected or slightly increased. The cortical bone parameters were largely unaltered. Upon RANKL treatment, Lrp1-deficient bone marrow monocytes more efficiently differentiated into osteoclasts and showed elevated p65 NFκB and p38 signaling. Consistently, Lrp1-overexpressing Raw264.7 cells were desensitized to RANKL-induced p38 and p65 activation and osteoclastogenesis. Moreover, RANKL treatment led to a sharp decrease of LRP1 protein and RNA in BMMs. Overall, our data suggest that osteoclast-expressed LRP1 is a crucial regulator of bone mass. It inhibits the NFκB and p38 pathways and lessens the efficiency of RANKL-induced osteoclastogenesis. © 2018 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption/metabolism , NF-kappa B/metabolism , Osteoclasts/metabolism , Osteogenesis , RANK Ligand/pharmacology , Receptors, LDL/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Resorption/pathology , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Bone and Bones/pathology , Cathepsin K/genetics , Cell Differentiation/drug effects , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Organ Size/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Phenotype , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/genetics , Signal Transduction/drug effects , Tumor Suppressor Proteins/genetics , X-Ray Microtomography
9.
Nat Commun ; 9(1): 646, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422621

ABSTRACT

In the originally published version of this Article, the positions of the final two authors in the author list were inadvertently inverted during the production process. This error has now been corrected in both the PDF and HTML versions of the Article.

10.
Sci Rep ; 8(1): 2545, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416046

ABSTRACT

Sumoylation is a post-translational modification process having an important influence in mesenchymal stem cell (MSC) differentiation. Thus, sumoylation-modulating chemicals might be used to control MSC differentiation for skeletal tissue engineering. In this work, we studied how the differentiation of mouse bone marrow stromal cells (mBMSCs) is affected by ginkgolic acid (GA), a potent sumoylation inhibitor also reported to inhibit histone acetylation transferase (HAT). Our results show that GA promoted the differentiation of mBMSCs into adipocytes when cultured in osteogenic medium. Moreover, mBMSCs pre-treated with GA showed enhanced pre-adipogenic gene expression and were more efficiently differentiated into adipocytes when subsequently cultured in the adipogenic medium. However, when GA was added at a later stage of adipogenesis, adipocyte maturation was markedly inhibited, with a dramatic down-regulation of multiple lipogenesis genes. Moreover, we found that the effects of garcinol, a HAT inhibitor, differed from those of GA in regulating adipocyte commitment and adipocyte maturation of mBMSCs, implying that the GA function in adipogenesis is likely through its activity as a sumoylation inhibitor, not as a HAT inhibitor. Overall, our studies revealed an unprecedented role of GA in MSC differentiation and provide new mechanistic insights into the use of GA in clinical applications.


Subject(s)
Adipogenesis/drug effects , Bone Marrow Cells , Mesenchymal Stem Cells , Salicylates/pharmacology , Sumoylation/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Histone Acetyltransferases/antagonists & inhibitors , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Terpenes/pharmacology
11.
Nat Commun ; 9(1): 143, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321472

ABSTRACT

The development, growth, and renewal of skeletal tissues rely on the function of osteochondroprogenitors (OCPs). Protein sumoylation/desumoylation has emerged as a pivotal mechanism for stem cell/progenitor homeostasis, and excessive sumoylation has been associated with cell senescence and tissue aging, but its role in regulating OCP function is unclear. Here we show that postnatal loss of the desumoylase SUMO1/sentrin-specific peptidase 6 (SENP6) causes premature aging. OCP-specific SENP6 knockout mice exhibit smaller skeletons, with elevated apoptosis and cell senescence in OCPs and chondrocytes. In Senp6 ‒/‒ cells, the two most significantly elevated pathways are p53 signaling and senescence-associated secreted phenotypes (SASP), and Trp53 loss partially rescues the skeletal and cellular phenotypes caused by Senp6 loss. Furthermore, SENP6 interacts with, desumoylates, and stabilizes TRIM28, suppressing p53 activity. Our data reveals a crucial role of the SENP6-p53 axis in maintaining OCP homeostasis during skeletal development.


Subject(s)
Apoptosis/genetics , Bone Development/genetics , Cellular Senescence/genetics , Chondrocytes/cytology , Homeostasis/genetics , Osteoblasts/cytology , Peptide Hydrolases/metabolism , Stem Cells/cytology , Tumor Suppressor Protein p53/metabolism , Aging, Premature/genetics , Animals , Bone and Bones/cytology , Bone and Bones/diagnostic imaging , Bone and Bones/metabolism , Chondrocytes/metabolism , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Mice , Mice, Knockout , Osteoblasts/metabolism , Peptide Hydrolases/genetics , Stem Cells/metabolism , Sumoylation , Tripartite Motif-Containing Protein 28/metabolism , X-Ray Microtomography
12.
Hum Mol Genet ; 26(15): 2949-2960, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28486600

ABSTRACT

Ciliopathies form a group of inherited disorders sharing several clinical manifestations because of abnormal cilia formation or function, and few treatments have been successful against these disorders. Here, we report a mouse model with mutated Sclt1 gene, which encodes a centriole distal appendage protein important for ciliogenesis. Sodium channel and clathrin linker 1 (SCLT1) mutations were associated with the oral-facial-digital syndrome (OFD), an autosomal recessive ciliopathy. The Sclt1-/- mice exhibit typical ciliopathy phenotypes, including cystic kidney, cleft palate and polydactyly. Sclt1-loss decreases the number of cilia in kidney; increases proliferation and apoptosis of renal tubule epithelial cells; elevates protein kinase A, extracellular signal-regulated kinases, SMAD and signal transducer and activator of transcription 3 (STAT3) pathways; and enhances pro-inflammation and pro-fibrosis pathways with disease progression. Embryonic kidney cyst formation of Sclt1-/- mice was effectively reduced by an anti-STAT3 treatment using pyrimethamine. Overall, we reported a new mouse model for the OFD; and our data suggest that STAT3 inhibition may be a promising treatment for SCLT1-associated cystic kidney.


Subject(s)
STAT3 Transcription Factor/metabolism , Sodium Channels/metabolism , Animals , Cilia/metabolism , Ciliopathies/genetics , Ciliopathies/metabolism , Cysts/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Kidney/metabolism , Kidney Diseases, Cystic/etiology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , MAP Kinase Signaling System , Mice , Mice, Transgenic , Models, Animal , Mutation , Phenotype , STAT3 Transcription Factor/genetics , Signal Transduction , Sodium Channels/genetics
13.
Oncotarget ; 7(40): 66087-66099, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27602774

ABSTRACT

The anticancer small molecule MLN4924, a Nedd8-activating enzyme (NAE) inhibitor, triggers cell-cycle arrest, apoptosis, and senescence in cancer cells. In this study, we demonstrate that MLN4924 suppresses osteosarcoma cell proliferation by inducing G2/M cell cycle arrest and apoptosis. Our results indicate that MLN4924 stabilizes the retinoid orphan nuclear receptor alpha (RORα) by decreasing its ubiquitination. RNA interference of RORα attenuates the anti-proliferative effect of MLN4924 in U2OS osteosarcoma cells. MLN4924 up-regulates the expression of p21 and Bmal1, two transcriptional targets of RORα. However, p21 plays a minimal role in the anti-proliferative effect of MLN4924 in U2OS osteosarcoma cells. In contrast, Bmal1 suppression by siRNA attenuates the anti-proliferative effect of MLN4924 in U2OS osteosarcoma cells, indicating that the MLN4924-mediated cell growth inhibition is mediated by Bmal1. These results show MLN4924 to be a promising therapeutic agent for the treatment of osteosarcoma and suggest that MLN4924-induced tumor growth inhibition is mediated by the circadian clock components RORα and Bmal1.


Subject(s)
ARNTL Transcription Factors/metabolism , Bone Neoplasms/drug therapy , Cell Proliferation/drug effects , Circadian Clocks , Cyclopentanes/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Osteosarcoma/drug therapy , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Humans , Mice , Mice, Nude , Osteosarcoma/metabolism , Osteosarcoma/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Biochem Biophys Res Commun ; 469(3): 501-6, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26687946

ABSTRACT

Mutations in Prokineticin receptor 2 (PKR2), a G-protein-coupled receptor, have been identified in patients with Kallmann syndrome and/or idiopathic hypogonadotropic hypogonadism, characterized by delayed puberty and infertility. In this study, we performed yeast two-hybrid screening by using PKR2 C-terminus (amino acids 333-384) as a bait, and identified Snapin as a novel interaction partner for PKR2. The interaction of Snapin and PKR2 was confirmed in GST pull-down and co-immunoprecipitation studies. We further demonstrated that two α-helix domains in Snapin are required for the interaction. And the interactive motifs of PKR2 were mapped to YFK (343-345) and HWR (351-353), which shared a similar sequence of two aromatic amino acids followed by a basic amino acid. Disruption of Snapin-PKR2 interaction did not affect PKR2 signaling, but increased the ligand-induced degradation, implying a role of Snapin in the trafficking of PKR2.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/chemistry , Receptors, Peptide/metabolism , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/metabolism , Amino Acid Sequence , Binding Sites , HEK293 Cells , Humans , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping
15.
J Biol Chem ; 289(22): 15518-26, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24753254

ABSTRACT

Mutations in the G protein-coupled prokineticin receptor 2 (PKR2) are known to cause Kallmann syndrome and idiopathic hypogonadotropic hypogonadism manifesting with delayed puberty and infertility. Some of the mutant receptors are not routed to the cell surface; instead, they are trapped in the cellular secretory pathway. The cell-permeant agonists/antagonists have been used to rescue some membrane receptors that are not targeted onto the cell membrane. Here, we chose three disease-associated mutations (W178S, G234D, and P290S), which all resulted in retention of PKR2 intracellularly. We show that a small molecule PKR2 antagonist (A457) dramatically increased cell surface expression and rescued the function of P290S PKR2, but had no effect on W178S and G234D PKR2. Furthermore, we also tested chemical chaperone glycerol on the cell surface expression and function of PKR2 mutants. Treatment with 10% glycerol significantly increased the cell surface expression and signaling of P290S and W178S PKR2. These data demonstrate that some Kallmann syndrome-associated, intracellularly retained mutant PKR2 receptors can be functionally rescued, suggesting a potential treatment strategy for patients bearing such mutations.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Kallmann Syndrome/genetics , Kallmann Syndrome/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , Animals , CHO Cells , Cricetulus , Cryoprotective Agents/pharmacology , Glycerol/pharmacology , HEK293 Cells , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Humans , Kallmann Syndrome/drug therapy , Membrane Proteins/genetics , Membrane Proteins/metabolism , Point Mutation , Protein Transport/genetics , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Peptide/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics
16.
Cell Signal ; 26(5): 1118-24, 2014 May.
Article in English | MEDLINE | ID: mdl-24509228

ABSTRACT

Prokineticins (PKs) are a pair of signal factors involved in many physiological processes by binding to two closely related G-protein-coupled receptors (GPCRs), PKR1 and PKR2. We recently demonstrated that PKR2 undergoes rapid ligand-induced endocytosis, and PKR2 recycles back to the plasma membrane after the removal of ligand. However, little is known about the molecular mechanisms underlying the PKR2 endocytosis. Here, we studied the involvement of GPCR kinase 2 (GRK2), ß-arrestins, clathrin and protein kinase C (PKC) in the PKR2 endocytosis. Our results indicated that PK2-induced PKR2 endocytosis is GRK2- and clathrin-dependent, but ß-arrestin-independent. PKC activation also induced PKR2 endocytosis; however, PKC activation is not necessary for the PK2-induced PKR2 endocytosis. PK2 stimulation induced a transient activation of extracellular signal regulated kinase 1/2 (ERK1/2) on PKR2 expressing cells. The internalization and PKC activation are not required for the PK2-induced ERK1/2 activation. Our results indicated that PK2-induced ERK1/2 activation may involve the released ßγ subunits of G-protein, phospholipase C ß and MEK activation.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Peptide/metabolism , Arrestins/metabolism , Endocytosis/drug effects , Enzyme Activation/drug effects , G-Protein-Coupled Receptor Kinase 2/metabolism , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , HEK293 Cells , Humans , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Kinase C/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Sphingosine/pharmacology , beta-Arrestins
17.
Biochem Biophys Res Commun ; 439(1): 12-7, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23969157

ABSTRACT

Mutations in the G-protein-coupled receptor PROKR2 have been identified in patients with idiopathic hypogonadotropic hypogonadism (IHH) and Kallmann syndrome (KS) manifesting with delayed puberty and infertility. Recently, the homozygous mutation V274D was identified in a man displaying KS with an apparent reversal of hypogonadism. The affected amino acid, valine 274, is located at the junction region of the third intracellular loop (IL3) and the sixth transmembrane domain (TM6). In this study, we first studied the effect of V274D and related mutations (V274A, V274T, and V274R) on the signaling activity and cell surface expression of PROKR2. Our data indicate that a charged amino acid substitution at residue 274 of PROKR2 results in low cell surface expression and loss-of-function. Furthermore, we studied the effects of two clusters of basic amino acids located at the proximal region of Val274 on the cell surface expression and function of PROKR2. The deletion of RRK (270-272) resulted in undetectable cell surface expression, whereas RKR (264-266)-deleted PROKR2 was expressed normally on the cell surface but showed loss-of-function due to a deficiency in G-protein coupling. Our data indicate that the distal region of the IL3 of PROKR2 may differentially influence receptor trafficking and G-protein coupling.


Subject(s)
Receptors, G-Protein-Coupled/chemistry , Receptors, Peptide/chemistry , Amino Acid Motifs , Amino Acid Substitution , Cell Membrane/metabolism , Gene Deletion , HEK293 Cells , Humans , Hypogonadism/genetics , Kallmann Syndrome/genetics , Models, Molecular , Mutation , Protein Sorting Signals , Protein Structure, Tertiary , Protein Transport , Receptors, G-Protein-Coupled/physiology , Receptors, Peptide/physiology , Signal Transduction , Valine/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...