Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
Add more filters

Publication year range
1.
Cell ; 173(6): 1481-1494.e13, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29706543

ABSTRACT

Global profiling of protein expression through the cell cycle has revealed subsets of periodically expressed proteins. However, expression levels alone only give a partial view of the biochemical processes determining cellular events. Using a proteome-wide implementation of the cellular thermal shift assay (CETSA) to study specific cell-cycle phases, we uncover changes of interaction states for more than 750 proteins during the cell cycle. Notably, many protein complexes are modulated in specific cell-cycle phases, reflecting their roles in processes such as DNA replication, chromatin remodeling, transcription, translation, and disintegration of the nuclear envelope. Surprisingly, only small differences in the interaction states were seen between the G1 and the G2 phase, suggesting similar hardwiring of biochemical processes in these two phases. The present work reveals novel molecular details of the cell cycle and establishes proteome-wide CETSA as a new strategy to study modulation of protein-interaction states in intact cells.


Subject(s)
Cell Cycle , Protein Interaction Mapping , Cell Division , Chromatin/chemistry , Cluster Analysis , DNA Replication , G1 Phase , G2 Phase , Humans , K562 Cells , Nuclear Envelope , Proteome , Proteomics/methods
2.
Cell ; 148(1-2): 259-72, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22225612

ABSTRACT

Identification of the factors critical to the tumor-initiating cell (TIC) state may open new avenues in cancer therapy. Here we show that the metabolic enzyme glycine decarboxylase (GLDC) is critical for TICs in non-small cell lung cancer (NSCLC). TICs from primary NSCLC tumors express high levels of the oncogenic stem cell factor LIN28B and GLDC, which are both required for TIC growth and tumorigenesis. Overexpression of GLDC and other glycine/serine enzymes, but not catalytically inactive GLDC, promotes cellular transformation and tumorigenesis. We found that GLDC induces dramatic changes in glycolysis and glycine/serine metabolism, leading to changes in pyrimidine metabolism to regulate cancer cell proliferation. In the clinic, aberrant activation of GLDC correlates with poorer survival in lung cancer patients, and aberrant GLDC expression is observed in multiple cancer types. This link between glycine metabolism and tumorigenesis may provide novel targets for advancing anticancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Cell Transformation, Neoplastic , Glycine Dehydrogenase (Decarboxylating)/metabolism , Lung Neoplasms/metabolism , Amino Acid Sequence , Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Fetal Proteins/metabolism , Glycine/metabolism , Humans , Molecular Sequence Data , Neoplasms/enzymology , Neoplasms/genetics , RNA-Binding Proteins , Sequence Alignment , Serine/metabolism , Thermus thermophilus/enzymology , Transplantation, Heterologous
3.
PLoS Biol ; 18(10): e3000903, 2020 10.
Article in English | MEDLINE | ID: mdl-33075054

ABSTRACT

Genetic diversity in offspring is induced by meiotic recombination, which is initiated between homologs at >200 sites originating from meiotic double-strand breaks (DSBs). Of this initial pool, only 1-2 DSBs per homolog pair will be designated to form meiotic crossovers (COs), where reciprocal genetic exchange occurs between parental chromosomes. Cyclin-dependent kinase 2 (CDK2) is known to localize to so-called "late recombination nodules" (LRNs) marking incipient CO sites. However, the role of CDK2 kinase activity in the process of CO formation remains uncertain. Here, we describe the phenotype of 2 Cdk2 point mutants with elevated or decreased activity, respectively. Elevated CDK2 activity was associated with increased numbers of LRN-associated proteins, including CDK2 itself and the MutL homolog 1 (MLH1) component of the MutLγ complex, but did not lead to increased numbers of COs. In contrast, reduced CDK2 activity leads to the complete absence of CO formation during meiotic prophase I. Our data suggest an important role for CDK2 in regulating MLH1 focus numbers and that the activity of this kinase is a key regulatory factor in the formation of meiotic COs.


Subject(s)
Crossing Over, Genetic , Cyclin-Dependent Kinase 2/metabolism , Meiosis , Animals , Apoptosis , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Chromosome Pairing , DNA Repair , Histones/metabolism , Ligases/metabolism , Male , Meiotic Prophase I , Metaphase , Mice, Inbred C57BL , Mice, Transgenic , MutL Protein Homolog 1/metabolism , Pachytene Stage , Sex Chromosomes/genetics , Spermatozoa/cytology , Spermatozoa/metabolism , Synaptonemal Complex/metabolism , Telomere/metabolism
4.
PLoS Comput Biol ; 18(5): e1010140, 2022 05.
Article in English | MEDLINE | ID: mdl-35613161

ABSTRACT

Methylenetetrahydrofolate dehydrogenase/cyclohydrolase (MTHFD2) is a new drug target that is expressed in cancer cells but not in normal adult cells, which provides an Achilles heel to selectively kill cancer cells. Despite the availability of crystal structures of MTHFD2 in the inhibitor- and cofactor-bound forms, key information is missing due to technical limitations, including (a) the location of absolutely required Mg2+ ion, and (b) the substrate-bound form of MTHFD2. Using computational modeling and simulations, we propose that two magnesium ions are present at the active site whereby (i) Arg233, Asp225, and two water molecules coordinate [Formula: see text], while [Formula: see text] together with Arg233 stabilize the inorganic phosphate (Pi); (ii) Asp168 and three water molecules coordinate [Formula: see text], and [Formula: see text] further stabilizes Pi by forming a hydrogen bond with two oxygens of Pi; (iii) Arg201 directly coordinates the Pi; and (iv) through three water-mediated interactions, Asp168 contributes to the positioning and stabilization of [Formula: see text], [Formula: see text] and Pi. Our computational study at the empirical valence bond level allowed us also to elucidate the detailed reaction mechanisms. We found that the dehydrogenase activity features a proton-coupled electron transfer with charge redistribution connected to the reorganization of the surrounding water molecules which further facilitates the subsequent cyclohydrolase activity. The cyclohydrolase activity then drives the hydration of the imidazoline ring and the ring opening in a concerted way. Furthermore, we have uncovered that two key residues, Ser197/Arg233, are important factors in determining the cofactor (NADP+/NAD+) preference of the dehydrogenase activity. Our work sheds new light on the structural and kinetic framework of MTHFD2, which will be helpful to design small molecule inhibitors that can be used for cancer treatment.


Subject(s)
Aminohydrolases , Methylenetetrahydrofolate Dehydrogenase (NADP) , Aminohydrolases/chemistry , Aminohydrolases/metabolism , Kinetics , Magnesium , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mitochondria/metabolism , Water
5.
Cell Mol Life Sci ; 80(1): 4, 2022 Dec 07.
Article in English | MEDLINE | ID: mdl-36477411

ABSTRACT

Excessive fat accumulation in the liver has become a major health threat worldwide. Unresolved fat deposition in the liver can go undetected until it develops into fatty liver disease, followed by steatohepatitis, fibrosis, cirrhosis, and eventually hepatocellular carcinoma. Lipid deposition in the liver is governed by complex communication, primarily between metabolic organs. This can be mediated by hormones, organokines, and also, as has been more recently discovered, metabolites. Although how metabolites from peripheral organs affect the liver is well documented, the effect of metabolic players released from the liver during the development of fatty liver disease or associated comorbidities needs further attention. Here we focus on interorgan crosstalk based on metabolites released from the liver and how these molecules act as signaling molecules in peripheral tissues. Due to the liver's specific role, we are covering lipid and bile mechanism-derived metabolites. We also discuss the high sucrose intake associated with uric acid release from the liver. Excessive fat deposition in the liver during fatty liver disease development reflects disrupted metabolic processes. As a response, the liver secretes a variety of signaling molecules as well as metabolites which act as a footprint of the metabolic disruption. In the coming years, the reciprocal exchange of metabolites between the liver and other metabolic organs will gain further importance and will help to better understand the development of fatty liver disease and associated diseases.


Subject(s)
Liver Diseases , Humans , Lipids
6.
PLoS Genet ; 16(11): e1009084, 2020 11.
Article in English | MEDLINE | ID: mdl-33147210

ABSTRACT

The liver possesses a remarkable regenerative capacity based partly on the ability of hepatocytes to re-enter the cell cycle and divide to replace damaged cells. This capability is substantially reduced upon chronic damage, but it is not clear if this is a cause or consequence of liver disease. Here, we investigate whether blocking hepatocyte division using two different mouse models affects physiology as well as clinical liver manifestations like fibrosis and inflammation. We find that in P14 Cdk1Liv-/- mice, where the division of hepatocytes is abolished, polyploidy, DNA damage, and increased p53 signaling are prevalent. Cdk1Liv-/- mice display classical markers of liver damage two weeks after birth, including elevated ALT, ALP, and bilirubin levels, despite the lack of exogenous liver injury. Inflammation was further studied using cytokine arrays, unveiling elevated levels of CCL2, TIMP1, CXCL10, and IL1-Rn in Cdk1Liv-/- liver, which resulted in increased numbers of monocytes. Ablation of CDK2-dependent DNA re-replication and polyploidy in Cdk1Liv-/- mice reversed most of these phenotypes. Overall, our data indicate that blocking hepatocyte division induces biological processes driving the onset of the disease phenotype. It suggests that the decrease in hepatocyte division observed in liver disease may not only be a consequence of fibrosis and inflammation, but also a pathological cue.


Subject(s)
Cell Division/physiology , Hepatocytes/physiology , Liver Cirrhosis/metabolism , Animals , Apoptosis/physiology , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Cycle , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Fibrosis/physiopathology , Hepatitis/metabolism , Hepatitis/physiopathology , Hepatocytes/metabolism , Inflammation/pathology , Liver/metabolism , Liver/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Male , Mice , Mice, Knockout , Signal Transduction
7.
Int J Mol Sci ; 24(2)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36674842

ABSTRACT

The PRDM family of methyltransferases has been implicated in cellular proliferation and differentiation and is deregulated in human diseases, most notably in cancer. PRDMs are related to the SET domain family of methyltransferases; however, from the 19 PRDMs only a few PRDMs with defined enzymatic activities are known. PRDM15 is an uncharacterized transcriptional regulator, with significant structural disorder and lack of defined small-molecule binding pockets. Many aspects of PRDM15 are yet unknown, including its structure, substrates, reaction mechanism, and its methylation profile. Here, we employ a series of computational approaches for an exploratory investigation of its potential substrates and reaction mechanism. Using the knowledge of PRDM9 and current knowledge of PRDM15 as basis, we tried to identify genuine substrates of PRDM15. We start from histone-based peptides and learn that the native substrates of PRDM15 may be non-histone proteins. In the future, a combination of sequence-based approaches and signature motif analysis may provide new leads. In summary, our results provide new information about the uncharacterized methyltransferase, PRDM15.


Subject(s)
Methyltransferases , Neoplasms , Humans , Methyltransferases/metabolism , Methylation , Histones/genetics , Histones/metabolism , DNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Histone-Lysine N-Methyltransferase/metabolism
8.
Int J Mol Sci ; 24(3)2023 Jan 31.
Article in English | MEDLINE | ID: mdl-36768986

ABSTRACT

The aim of this work was to investigate the effect of the whole-body deletion of p27 on the activity of brown adipose tissue and the susceptibility to develop obesity and glucose homeostasis disturbances in mice, especially when subjected to a high fat diet. p27 knockout (p27-/-) and wild type (WT) mice were fed a normal chow diet or a high fat diet (HFD) for 10-weeks. Body weight and composition were assessed. Insulin and glucose tolerance tests and indirect calorimetry assays were performed. Histological analysis of interscapular BAT (iBAT) was carried out, and expression of key genes/proteins involved in BAT function were characterized by qPCR and Western blot. iBAT activity was estimated by 18F-fluorodeoxyglucose (18FDG) uptake with microPET. p27-/- mice were more prone to develop obesity and insulin resistance, exhibiting increased size of all fat depots. p27-/- mice displayed a higher respiratory exchange ratio. iBAT presented larger adipocytes in p27-/- HFD mice, accompanied by downregulation of both Glut1 and uncoupling protein 1 (UCP1) in parallel with defective insulin signalling. Moreover, p27-/- HFD mice exhibited impaired response to cold exposure, characterized by a reduced iBAT 18FDG uptake and difficulty to maintain body temperature when exposed to cold compared to WT HFD mice, suggesting reduced thermogenic capacity. These data suggest that p27 could play a role in BAT activation and in the susceptibility to develop obesity and insulin resistance.


Subject(s)
Adipose Tissue, Brown , Insulin Resistance , Animals , Mice , Adipose Tissue, Brown/metabolism , Diet, High-Fat/adverse effects , Fluorodeoxyglucose F18/metabolism , Insulin/metabolism , Insulin Resistance/genetics , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Thermogenesis
9.
Semin Cell Dev Biol ; 107: 54-62, 2020 11.
Article in English | MEDLINE | ID: mdl-32386818

ABSTRACT

Cyclin-dependent kinases (CDKs) are activated by cyclins, which play important roles in dictating the actions of CDK/cyclin complexes. Cyclin binding influences the substrate specificity of these complexes in addition to their susceptibility to inhibition or degradation. CDK/cyclin complexes are best known to promote cell cycle progression in the mitotic cell cycle but are also crucial for important cellular processes not strictly associated with cellular division. This chapter primarily explores the understudied topic of CDK/cyclin complex functionality during the DNA damage response. We detail how CDK/cyclin complexes perform dual roles both as targets of DNA damage checkpoint signaling as well as effectors of DNA repair. Additionally, we discuss the potential CDK-independent roles of cyclins in these processes and the impact of such roles in human diseases such as cancer. Our goal is to place the spotlight on these important functions of cyclins either acting as independent entities or within CDK/cyclin complexes which have attracted less attention in the past. We consider that this will be important for a more complete understanding of the intricate functions of cell cycle proteins in the DNA damage response.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Animals , Cell Proliferation/genetics , DNA Damage/genetics , DNA Repair/genetics , Genomic Instability/genetics , Humans
10.
Proteins ; 90(1): 123-130, 2022 01.
Article in English | MEDLINE | ID: mdl-34318530

ABSTRACT

Lignin is one of the world's most abundant organic polymers, and 2-pyrone-4,6-dicarboxylate lactonase (LigI) catalyzes the hydrolysis of 2-pyrone-4,6-dicarboxylate (PDC) in the degradation of lignin. The pH has profound effects on enzyme catalysis and therefore we studied this in the context of LigI. We found that changes of the pH mostly affects surface residues, while the residues at the active site are more subject to changes of the surrounding microenvironment. In accordance with this, a high pH facilitates the deprotonation of the substrate. Detailed free energy calculations by the empirical valence bond (EVB) approach revealed that the overall hydrolysis reaction is more likely when the three active site histidines (His31, His33 and His180) are protonated at the ɛ site, however, protonation at the δ site may be favored during specific steps of the reaction. Our studies have uncovered the determinant role of the protonation state of the active site residues His31, His33 and His180 in the hydrolysis of PDC.


Subject(s)
Carboxylic Ester Hydrolases/chemistry , Catalytic Domain , Histidine/chemistry , Carboxylic Ester Hydrolases/metabolism , Catalysis , Histidine/metabolism , Hydrolysis , Lignin/chemistry , Lignin/metabolism , Protons
11.
Gastroenterology ; 160(6): 2103-2118, 2021 05.
Article in English | MEDLINE | ID: mdl-33465371

ABSTRACT

BACKGROUND & AIMS: Liver tight junctions (TJs) establish tissue barriers that isolate bile from the blood circulation. TJP2/ZO-2-inactivating mutations cause progressive cholestatic liver disease in humans. Because the underlying mechanisms remain elusive, we characterized mice with liver-specific inactivation of Tjp2. METHODS: Tjp2 was deleted in hepatocytes, cholangiocytes, or both. Effects on the liver were assessed by biochemical analyses of plasma, liver, and bile and by electron microscopy, histology, and immunostaining. TJ barrier permeability was evaluated using fluorescein isothiocyanate-dextran (4 kDa). Cholic acid (CA) diet was used to assess susceptibility to liver injury. RESULTS: Liver-specific deletion of Tjp2 resulted in lower Cldn1 protein levels, minor changes to the TJ, dilated canaliculi, lower microvilli density, and aberrant radixin and bile salt export pump (BSEP) distribution, without an overt increase in TJ permeability. Hepatic Tjp2-defcient mice presented with mild progressive cholestasis with lower expression levels of bile acid transporter Abcb11/Bsep and detoxification enzyme Cyp2b10. A CA diet tolerated by control mice caused severe cholestasis and liver necrosis in Tjp2-deficient animals. 1,4-Bis[2-(3,5-dichloropyridyloxy)]benzene ameliorated CA-induced injury by enhancing Cyp2b10 expression, and ursodeoxycholic acid provided partial improvement. Inactivating Tjp2 separately in hepatocytes or cholangiocytes showed only mild CA-induced liver injury. CONCLUSION: Tjp2 is required for normal cortical distribution of radixin, canalicular volume regulation, and microvilli density. Its inactivation deregulated expression of Cldn1 and key bile acid transporters and detoxification enzymes. The mice provide a novel animal model for cholestatic liver disease caused by TJP2-inactivating mutations in humans.


Subject(s)
Bile Canaliculi/metabolism , Chemical and Drug Induced Liver Injury/genetics , Cholestasis/genetics , Tight Junctions/metabolism , Zonula Occludens-2 Protein/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Bile Acids and Salts/metabolism , Bile Canaliculi/pathology , Chemical and Drug Induced Liver Injury/drug therapy , Cholagogues and Choleretics/therapeutic use , Cholic Acid , Claudin-1/metabolism , Cytochrome P450 Family 2/metabolism , Cytoskeletal Proteins/metabolism , Epithelial Cells , Female , Fibrosis , Genetic Predisposition to Disease , Hepatocytes , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mutation , Oxazoles/therapeutic use , Permeability , Protective Factors , RNA, Messenger/metabolism , Steroid Hydroxylases/metabolism , Tight Junctions/ultrastructure , Ursodeoxycholic Acid/therapeutic use , Zonula Occludens-2 Protein/deficiency
12.
Development ; 146(21)2019 11 06.
Article in English | MEDLINE | ID: mdl-31582414

ABSTRACT

The ability of men to remain fertile throughout their lives depends upon establishment of a spermatogonial stem cell (SSC) pool from gonocyte progenitors, and thereafter balancing SSC renewal versus terminal differentiation. Here, we report that precise regulation of the cell cycle is crucial for this balance. Whereas cyclin-dependent kinase 2 (Cdk2) is not necessary for mouse viability or gametogenesis stages prior to meiotic prophase I, mice bearing a deregulated allele (Cdk2Y15S ) are severely deficient in spermatogonial differentiation. This allele disrupts an inhibitory phosphorylation site (Tyr15) for the kinase WEE1. Remarkably, Cdk2Y15S/Y15S mice possess abnormal clusters of mitotically active SSC-like cells, but these are eventually removed by apoptosis after failing to differentiate properly. Analyses of lineage markers, germ cell proliferation over time, and single cell RNA-seq data revealed delayed and defective differentiation of gonocytes into SSCs. Biochemical and genetic data demonstrated that Cdk2Y15S is a gain-of-function allele causing elevated kinase activity, which underlies these differentiation defects. Our results demonstrate that precise regulation of CDK2 kinase activity in male germ cell development is crucial for the gonocyte-to-spermatogonia transition and long-term spermatogenic homeostasis.


Subject(s)
Cell Differentiation , Cell Lineage , Cyclin-Dependent Kinase 2/metabolism , Germ Cells/enzymology , Spermatogonia/cytology , Alleles , Animals , Apoptosis , CRISPR-Cas Systems , Cell Proliferation , Cluster Analysis , Crosses, Genetic , Germ Cells/cytology , Heterozygote , Homeostasis , Male , Mass Spectrometry , Meiosis , Mice , Mutagenesis, Site-Directed , Phenotype , Phosphorylation , RNA, Small Cytoplasmic/metabolism , Seminiferous Tubules/metabolism , Spermatogenesis , Spermatogonia/metabolism , Testis/metabolism , Transcriptome
13.
Cell Mol Life Sci ; 77(17): 3423-3439, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31722069

ABSTRACT

SUGCT (C7orf10) is a mitochondrial enzyme that synthesizes glutaryl-CoA from glutarate in tryptophan and lysine catabolism, but it has not been studied in vivo. Although mutations in Sugct lead to Glutaric Aciduria Type 3 disease in humans, patients remain largely asymptomatic despite high levels of glutarate in the urine. To study the disease mechanism, we generated SugctKO mice and uncovered imbalanced lipid and acylcarnitine metabolism in kidney in addition to changes in the gut microbiome. After SugctKO mice were treated with antibiotics, metabolites were comparable to WT, indicating that the microbiome affects metabolism in SugctKO mice. SUGCT loss of function contributes to gut microbiota dysbiosis, leading to age-dependent pathological changes in kidney, liver, and adipose tissue. This is associated with an obesity-related phenotype that is accompanied by lipid accumulation in kidney and liver, as well as "crown-like" structures in adipocytes. Furthermore, we show that the SugctKO kidney pathology is accelerated and exacerbated by a high-lysine diet. Our study highlights the importance of non-essential genes with no readily detectable early phenotype, but with substantial contributions to the development of age-related pathologies, which result from an interplay between genetic background, microbiome, and diet in the health of mammals.


Subject(s)
Aging , Coenzyme A-Transferases/genetics , Gastrointestinal Microbiome , Metabolic Syndrome/pathology , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Bacteria/isolation & purification , Carnitine/analogs & derivatives , Carnitine/metabolism , Coenzyme A-Transferases/deficiency , Dietary Supplements , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Humans , Kidney/metabolism , Kidney/pathology , Lipid Metabolism , Liver/metabolism , Liver/pathology , Lysine/administration & dosage , Metabolic Syndrome/metabolism , Metabolome/drug effects , Mice , Mice, Knockout , Obesity/metabolism , Obesity/pathology , Tryptophan/metabolism
14.
J Biol Chem ; 294(21): 8325-8335, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30940725

ABSTRACT

Interleukin (IL)-1 family cytokines potently regulate inflammation, with the majority of the IL-1 family proteins being secreted from immune cells via unconventional pathways. In many cases, secretion of IL-1 cytokines appears to be closely coupled to cell death, yet the secretory mechanisms involved remain poorly understood. Here, we studied the secretion of the three best-characterized members of the IL-1 superfamily, IL-1α, IL-1ß, and IL-18, in a range of conditions and cell types, including murine bone marrow-derived and peritoneal macrophages, human monocyte-derived macrophages, HeLa cells, and mouse embryonic fibroblasts. We discovered that IL-1ß and IL-18 share a common secretory pathway that depends upon membrane permeability and can operate in the absence of complete cell lysis and cell death. We also found that the pathway regulating the trafficking of IL-1α is distinct from the pathway regulating IL-1ß and IL-18. Although the release of IL-1α could also be dissociated from cell death, it was independent of the effects of the membrane-stabilizing agent punicalagin, which inhibited both IL-1ß and IL-18 release. These results reveal that in addition to their role as danger signals released from dead cells, IL-1 family cytokines can be secreted in the absence of cell death. We propose that models used in the study of IL-1 release should be considered context-dependently.


Subject(s)
Bone Marrow Cells/metabolism , Interleukin-18/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Macrophages, Peritoneal/metabolism , Animals , Bone Marrow Cells/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , HeLa Cells , Humans , Hydrolyzable Tannins/pharmacology , Macrophages, Peritoneal/cytology , Mice , Protein Transport/drug effects
15.
Lab Invest ; 100(5): 696-711, 2020 05.
Article in English | MEDLINE | ID: mdl-31915367

ABSTRACT

A hallmark of polycystic kidney diseases (PKDs) is aberrant proliferation, which leads to the formation and growth of renal cysts. Proliferation is mediated by cyclin-dependent kinases (Cdks), and the administration of roscovitine (a pan-Cdk inhibitor) attenuates renal cystic disease in juvenile cystic kidney (jck) mice. Cdk2 is a key regulator of cell proliferation, but its specific role in PKD remains unknown. The aim of this study was to test the hypothesis that Cdk2 deficiency reduces renal cyst growth in PKD. Three studies were undertaken: (i) a time course (days 28, 56, and 84) of cyclin and Cdk activity was examined in jck mice and compared with wild-type mice; (ii) the progression was compared in jck mice with or without Cdk2 ablation from birth; and (iii) the effect of sirolimus (an antiproliferative agent) on Cdk2 activity in jck mice was investigated. Renal disease in jck mice was characterized by diffuse tubular cyst growth, interstitial inflammation and fibrosis, and renal impairment, peaking on day 84. Renal cell proliferation peaked during earlier stages of disease (days 28-56), whereas the expression of Cdk2-cyclin partners (A and E) and Cdk1 and 2 activity, was maximal in the later stages of disease (days 56-84). Cdk2 ablation did not attenuate renal disease progression and was associated with persistent Cdk1 activity. In contrast, the postnatal treatment of jck mice with sirolimus reduced both Cdk2 and Cdk1 activity and reduced renal cyst growth. In conclusion, (i) the kinetics of Cdk2 and Cdk2-cyclin partners did not correlate with proliferation in jck mice; and (ii) the absence of Cdk2 did not alter renal cyst growth, most likely due to compensation by Cdk1. Taken together, these data suggest that Cdk2 is dispensable for the proliferation of cystic epithelial cells and progression of PKD.


Subject(s)
Cyclin-Dependent Kinase 2 , Polycystic Kidney Diseases , Animals , Cell Proliferation , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Disease Models, Animal , Disease Progression , Female , Kidney/cytology , Kidney/drug effects , Kidney/pathology , Male , Mice , Polycystic Kidney Diseases/metabolism , Polycystic Kidney Diseases/pathology , Sirolimus/pharmacology
16.
Am J Hum Genet ; 101(3): 391-403, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28886341

ABSTRACT

In five separate families, we identified nine individuals affected by a previously unidentified syndrome characterized by growth retardation, spine malformation, facial dysmorphisms, and developmental delays. Using homozygosity mapping, array CGH, and exome sequencing, we uncovered bi-allelic loss-of-function CDK10 mutations segregating with this disease. CDK10 is a protein kinase that partners with cyclin M to phosphorylate substrates such as ETS2 and PKN2 in order to modulate cellular growth. To validate and model the pathogenicity of these CDK10 germline mutations, we generated conditional-knockout mice. Homozygous Cdk10-knockout mice died postnatally with severe growth retardation, skeletal defects, and kidney and lung abnormalities, symptoms that partly resemble the disease's effect in humans. Fibroblasts derived from affected individuals and Cdk10-knockout mouse embryonic fibroblasts (MEFs) proliferated normally; however, Cdk10-knockout MEFs developed longer cilia. Comparative transcriptomic analysis of mutant and wild-type mouse organs revealed lipid metabolic changes consistent with growth impairment and altered ciliogenesis in the absence of CDK10. Our results document the CDK10 loss-of-function phenotype and point to a function for CDK10 in transducing signals received at the primary cilia to sustain embryonic and postnatal development.


Subject(s)
Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/physiology , Developmental Disabilities/genetics , Growth Disorders/genetics , Mutation , Spine/abnormalities , Spine/pathology , Animals , Cell Cycle , Cell Proliferation , Cells, Cultured , Child , Child, Preschool , Cilia/metabolism , Cilia/pathology , Developmental Disabilities/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Growth Disorders/pathology , Humans , Infant , Male , Mice , Mice, Knockout , Pedigree , Phosphorylation , Signal Transduction , Spine/metabolism
17.
Proc Natl Acad Sci U S A ; 114(3): 592-597, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28031483

ABSTRACT

Telomere attachment to the nuclear envelope (NE) is a prerequisite for chromosome movement during meiotic prophase I that is required for pairing of homologous chromosomes, synapsis, and homologous recombination. Here we show that Speedy A, a noncanonical activator of cyclin-dependent kinases (Cdks), is specifically localized to telomeres in prophase I male and female germ cells in mice, and plays an essential role in the telomere-NE attachment. Deletion of Spdya in mice disrupts telomere-NE attachment, and this impairs homologous pairing and synapsis and leads to zygotene arrest in male and female germ cells. In addition, we have identified a telomere localization domain on Speedy A covering the distal N terminus and the Cdk2-binding Ringo domain, and this domain is essential for the localization of Speedy A to telomeres. Furthermore, we found that the binding of Cdk2 to Speedy A is indispensable for Cdk2's localization on telomeres, suggesting that Speedy A and Cdk2 might be the initial components that are recruited to the NE for forming the meiotic telomere complex. However, Speedy A-Cdk2-mediated telomere-NE attachment is independent of Cdk2 activation. Our results thus indicate that Speedy A and Cdk2 might mediate the initial telomere-NE attachment for the efficient assembly of the telomere complex that is essential for meiotic prophase I progression.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase 2/metabolism , Animals , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cyclin-Dependent Kinase 2/chemistry , Enzyme Activation , Female , Male , Meiotic Prophase I/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Envelope/metabolism , Oocytes/cytology , Oocytes/metabolism , Protein Interaction Domains and Motifs , Spermatocytes/cytology , Spermatocytes/metabolism , Telomere/metabolism
18.
J Biol Chem ; 293(50): 19387-19399, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30366983

ABSTRACT

Bone mass is maintained by a balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption. Although recent genetic studies have uncovered various mechanisms that regulate osteoblast differentiation, the molecular basis of osteoblast proliferation remains unclear. Here, using an osteoblast-specific loss-of-function mouse model, we demonstrate that cyclin-dependent kinase 1 (Cdk1) regulates osteoblast proliferation and differentiation. Quantitative RT-PCR analyses revealed that Cdk1 is highly expressed in bone and is down-regulated upon osteoblast differentiation. We also noted that Cdk1 is dispensable for the bone-anabolic effects of parathyroid hormone (PTH). Cdk1 deletion in osteoblasts led to osteoporosis in adult mice due to low bone formation, but did not affect osteoclast formation in vivo Cdk1 overexpression in osteoblasts promoted proliferation, and conversely, Cdk1 knockdown inhibited osteoblast proliferation and promoted differentiation. Of note, we provide direct evidence that PTH's bone-anabolic effects occur without enhancing osteoblast proliferation in vivo Furthermore, we found that Cdk1 expression in osteoblasts is essential for bone fracture repair. These findings may help reduce the risk of nonunion after bone fracture and identify patients at higher risk for nonresponse to PTH treatment. Collectively, our results indicate that Cdk1 is essential for osteoblast proliferation and that it functions as a molecular switch that shifts osteoblast proliferation to maturation. We therefore conclude that Cdk1 plays an important role in bone formation.


Subject(s)
Bone and Bones/drug effects , Bone and Bones/physiology , CDC2 Protein Kinase/deficiency , CDC2 Protein Kinase/genetics , Gene Knockout Techniques , Osteogenesis/genetics , Parathyroid Hormone/pharmacology , 3T3 Cells , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Fractures, Bone/physiopathology , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Wound Healing/drug effects
19.
Bioessays ; 39(9)2017 09.
Article in English | MEDLINE | ID: mdl-28752618

ABSTRACT

The maintenance of cell size homeostasis has been studied for years in different cellular systems. With the focus on 'what regulates cell size', the question 'why cell size needs to be maintained' has been largely overlooked. Recent evidence indicates that animal cells exhibit nonlinear cell size dependent growth rates and mitochondrial metabolism, which are maximal in intermediate sized cells within each cell population. Increases in intracellular distances and changes in the relative cell surface area impose biophysical limitations on cells, which can explain why growth and metabolic rates are maximal in a specific cell size range. Consistently, aberrant increases in cell size, for example through polyploidy, are typically disadvantageous to cellular metabolism, fitness and functionality. Accordingly, cellular hypertrophy can potentially predispose to or worsen metabolic diseases. We propose that cell size control may have emerged as a guardian of cellular fitness and metabolic activity.


Subject(s)
Homeostasis/physiology , Animals , Cell Size , Humans , Hypertrophy/physiopathology , Mitochondria/metabolism , Mitochondria/physiology
20.
PLoS Genet ; 12(9): e1006310, 2016 09.
Article in English | MEDLINE | ID: mdl-27631493

ABSTRACT

The Greatwall kinase/Mastl is an essential gene that indirectly inhibits the phosphatase activity toward mitotic Cdk1 substrates. Here we show that although Mastl knockout (MastlNULL) MEFs enter mitosis, they progress through mitosis without completing cytokinesis despite the presence of misaligned chromosomes, which causes chromosome segregation defects. Furthermore, we uncover the requirement of Mastl for robust spindle assembly checkpoint (SAC) maintenance since the duration of mitotic arrest caused by microtubule poisons in MastlNULL MEFs is shortened, which correlates with premature disappearance of the essential SAC protein Mad1 at the kinetochores. Notably, MastlNULL MEFs display reduced phosphorylation of a number of proteins in mitosis, which include the essential SAC kinase MPS1. We further demonstrate that Mastl is required for multi-site phosphorylation of MPS1 as well as robust MPS1 kinase activity in mitosis. In contrast, treatment of MastlNULL cells with the phosphatase inhibitor okadaic acid (OKA) rescues the defects in MPS1 kinase activity, mislocalization of phospho-MPS1 as well as Mad1 at the kinetochore, and premature SAC silencing. Moreover, using in vitro dephosphorylation assays, we demonstrate that Mastl promotes persistent MPS1 phosphorylation by inhibiting PP2A/B55-mediated MPS1 dephosphorylation rather than affecting Cdk1 kinase activity. Our findings establish a key regulatory function of the Greatwall kinase/Mastl->PP2A/B55 pathway in preventing premature SAC silencing.


Subject(s)
CDC2 Protein Kinase/genetics , M Phase Cell Cycle Checkpoints/genetics , Microtubule-Associated Proteins/genetics , Protein Phosphatase 2/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Chromosome Segregation/genetics , Cytokinesis/genetics , Kinetochores/metabolism , Mice , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Microtubules/genetics , Microtubules/metabolism , Mitosis/genetics , Phosphorylation , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL