Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
1.
Clin Immunol ; 234: 108896, 2022 01.
Article in English | MEDLINE | ID: mdl-34848356

ABSTRACT

Little is known about the causes and mechanisms of ectopic immune responses, including different types of hypersensitivity, superantigens, and cytokine storms. Two of the most questionable phenomena observed in immunology are why the intensity and extent of immune responses to different antigens are different, and why some self-antigens are attacked as foreign. The secondary structure of the peptides involved in the immune system, such as the epitope-paratope interfaces plays a pivotal role in the resulting immune responses. Prolyl cis/trans isomerization plays a fundamental role in the form of the secondary structure and the folding of proteins. This review covers some of the emerging evidence indicating the impact of prolyl isomerization on protein conformation, aberration of immune responses, and the development of hypersensitivity reactions.


Subject(s)
Hypersensitivity/etiology , Peptidylprolyl Isomerase/physiology , Humans , Hypersensitivity/immunology , Immunity , Isomerism , Protein Folding , Protein Processing, Post-Translational , Protein Structure, Secondary
2.
Int J Biol Macromol ; 191: 40-50, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34534579

ABSTRACT

Peptidyl prolyl isomerases (PPIases) accelerate the rate limiting step of protein folding by catalyzing cis/trans isomerization of peptidyl prolyl bonds. The larger PPIases have been shown to be multi-domain proteins, with functions other than isomerization of the proline-containing peptide bond. Recently, a few smaller PPIases have also been described for their ability to stabilize folding intermediates. The yeast Fpr1 (FK506-sensitive proline rotamase) is a homologue of the mammalian prolyl isomerase FKBP12 (FK506-binding protein of 12 kDa). Its ability to stabilize stressed cellular proteins has not been reported yet. We had earlier reported upregulation of Fpr1 in yeast cells exposed to proteotoxic stress conditions. In this work, we show that yeast Fpr1 exhibits characteristics typical of a general chaperone of the proteostasis network. Aggregation of mutant huntingtin fragment was higher in Fpr1-deleted as compared to parental yeast cells. Overexpression of Fpr1 led to reduced protein aggregation by decreasing the amount of oligomers and diverting the aggregation pathway towards the formation of detergent-soluble species. This correlated well with higher survival of these cells. Purified and enzymatically active yeast Fpr1 was able to inhibit aggregation of mutant huntingtin fragment and luciferase in vitro in a concentration-dependent manner; suggesting a direct action for aggregation inhibitory action of Fpr1. Overexpression of yeast Fpr1 was able to protect E. coli cells against thermal shock. This work establishes the role of Fpr1 in the protein folding network and will be used for the identification of novel pharmacological leads in disease conditions.


Subject(s)
Molecular Chaperones/physiology , Peptidylprolyl Isomerase/physiology , Protein Aggregates , Proteostasis , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/enzymology , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Molecular Chaperones/genetics , Mutation , Peptidylprolyl Isomerase/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
3.
ACS Chem Biol ; 13(4): 1082-1089, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29543429

ABSTRACT

Skp and SurA are both periplasmic chaperones involved in the biogenesis of Escherichia coli ß-barrel outer membrane proteins (OMPs). It is commonly assumed that SurA plays a major role whereas Skp is a minor factor. However, there is no molecular evidence for whether their roles are redundant. Here, by using different dilution methods, we obtained monodisperse and aggregated forms of OmpC and studied their interactions with Skp and SurA by single-molecule fluorescence resonance energy transfer and fluorescence correlation spectroscopy. We found that Skp can dissolve aggregated OmpC while SurA cannot convert aggregated OmpC into the monodisperse form and the conformations of OmpC recognized by the two chaperones as well as their stoichiometries of binding are different. Our study demonstrates the functional distinctions between Skp and SurA. In particular, the role of Skp is not redundant and is probably more significant under stress conditions.


Subject(s)
Carrier Proteins/physiology , DNA-Binding Proteins/physiology , Escherichia coli Proteins/physiology , Molecular Chaperones/physiology , Peptidylprolyl Isomerase/physiology , Single Molecule Imaging , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli Proteins/metabolism , Molecular Chaperones/metabolism , Peptidylprolyl Isomerase/metabolism , Porins/metabolism , Protein Aggregates
4.
Nucleic Acids Res ; 46(5): 2459-2478, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29361176

ABSTRACT

FK506 binding proteins (FKBPs) catalyze the interconversion of cis-trans proline conformers in proteins. Importantly, FK506 drugs have anti-cancer and neuroprotective properties, but the effectors and mechanisms underpinning these properties are not well understood because the cellular function(s) of most FKBP proteins are unclear. FKBP25 is a nuclear prolyl isomerase that interacts directly with nucleic acids and is associated with several DNA/RNA binding proteins. Here, we show the catalytic FKBP domain binds microtubules (MTs) directly to promote their polymerization and stabilize the MT network. Furthermore, FKBP25 associates with the mitotic spindle and regulates entry into mitosis. This interaction is important for mitotic spindle dynamics, as we observe increased chromosome instability in FKBP25 knockdown cells. Finally, we provide evidence that FKBP25 association with chromatin is cell-cycle regulated by Protein Kinase C phosphorylation. This disrupts FKBP25-DNA contacts during mitosis while maintaining its interaction with the spindle apparatus. Collectively, these data support a model where FKBP25 association with chromatin and MTs is carefully choreographed to ensure faithful genome duplication. Additionally, they highlight that FKBP25 is a MT-associated FK506 receptor and potential therapeutic target in MT-associated diseases.


Subject(s)
Cell Cycle , Microtubules/metabolism , Peptidylprolyl Isomerase/metabolism , Tacrolimus Binding Proteins/metabolism , Cell Line , DNA/metabolism , Genomic Instability , Humans , Mitosis , Peptidylprolyl Isomerase/physiology , Phosphorylation , Polymerization , Protein Kinase C/metabolism , Tacrolimus Binding Proteins/physiology
5.
Infect Immun ; 84(10): 3034-46, 2016 10.
Article in English | MEDLINE | ID: mdl-27481256

ABSTRACT

The Gram-positive bacterium Listeria monocytogenes transitions from an environmental organism to an intracellular pathogen following its ingestion by susceptible mammalian hosts. Bacterial replication within the cytosol of infected cells requires activation of the central virulence regulator PrfA followed by a PrfA-dependent induction of secreted virulence factors. The PrfA-induced secreted chaperone PrsA2 and the chaperone/protease HtrA contribute to the folding and stability of select proteins translocated across the bacterial membrane. L. monocytogenes strains that lack both prsA2 and htrA exhibit near-normal patterns of growth in broth culture but are severely attenuated in vivo We hypothesized that, in the absence of PrsA2 and HtrA, the increase in PrfA-dependent protein secretion that occurs following bacterial entry into the cytosol results in misfolded proteins accumulating at the bacterial membrane with a subsequent reduction in intracellular bacterial viability. Consistent with this hypothesis, the introduction of a constitutively activated allele of prfA (prfA*) into ΔprsA2 ΔhtrA strains was found to essentially inhibit bacterial growth at 37°C in broth culture. ΔprsA2 ΔhtrA strains were additionally found to be defective for cell invasion and vacuole escape in selected cell types, steps that precede full PrfA activation. These data establish the essential requirement for PrsA2 and HtrA in maintaining bacterial growth under conditions of PrfA activation. In addition, chaperone function is required for efficient bacterial invasion and rapid vacuole lysis within select host cell types, indicating roles for PrsA2/HtrA prior to cytosolic PrfA activation and the subsequent induction of virulence factor secretion.


Subject(s)
Heat-Shock Proteins/physiology , Listeria monocytogenes/pathogenicity , Listeriosis/microbiology , Molecular Chaperones/physiology , Peptidylprolyl Isomerase/physiology , Serine Endopeptidases/physiology , Animals , Cytoplasm/microbiology , Epithelial Cells/microbiology , Glucuronidase/metabolism , Humans , Listeria monocytogenes/growth & development , Listeria monocytogenes/physiology , Macrophages/microbiology , Mice , Molecular Chaperones/metabolism , Protein Folding , Protein Stability , Virulence Factors/metabolism
6.
Proc Natl Acad Sci U S A ; 113(33): E4794-800, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27482090

ABSTRACT

Outer membrane protein (OMP) biogenesis is critical to bacterial physiology because the cellular envelope is vital to bacterial pathogenesis and antibiotic resistance. The process of OMP biogenesis has been studied in vivo, and each of its components has been studied in isolation in vitro. This work integrates parameters and observations from both in vivo and in vitro experiments into a holistic computational model termed "Outer Membrane Protein Biogenesis Model" (OMPBioM). We use OMPBioM to assess OMP biogenesis mathematically in a global manner. Using deterministic and stochastic methods, we are able to simulate OMP biogenesis under varying genetic conditions, each of which successfully replicates experimental observations. We observe that OMPs have a prolonged lifetime in the periplasm where an unfolded OMP makes, on average, hundreds of short-lived interactions with chaperones before folding into its native state. We find that some periplasmic chaperones function primarily as quality-control factors; this function complements the folding catalysis function of other chaperones. Additionally, the effective rate for the ß-barrel assembly machinery complex necessary for physiological folding was found to be higher than has currently been observed in vitro. Overall, we find a finely tuned balance between thermodynamic and kinetic parameters maximizes OMP folding flux and minimizes aggregation and unnecessary degradation. In sum, OMPBioM provides a global view of OMP biogenesis that yields unique insights into this essential pathway.


Subject(s)
Bacterial Outer Membrane Proteins/biosynthesis , Molecular Chaperones/physiology , Periplasm/metabolism , Bacterial Outer Membrane Proteins/chemistry , Carrier Proteins/physiology , Escherichia coli Proteins/physiology , Heat-Shock Proteins/physiology , Peptidylprolyl Isomerase/physiology , Periplasmic Proteins/physiology , Protein Aggregates , Protein Folding , Serine Endopeptidases/physiology , Stochastic Processes
7.
Adv Exp Med Biol ; 883: 271-94, 2015.
Article in English | MEDLINE | ID: mdl-26621473

ABSTRACT

In the dense cellular environment, protein misfolding and inter-molecular protein aggregation compete with protein folding. Chaperones associate with proteins to prevent misfolding and to assist in folding to the native state. In Escherichia coli, the chaperones trigger factor, DnaK/DnaJ/GrpE, and GroEL/ES are the major chaperones responsible for insuring proper de novo protein folding. With multitudes of proteins produced by the bacterium, the chaperones have to be selective for their substrates. Yet, chaperone selectivity cannot be too specific. Recent biochemical and high-throughput studies have provided important insights highlighting the strategies used by chaperones in maintaining proteostasis in the cell. Here, we discuss the substrate networks and cooperation among these protein folding chaperones.


Subject(s)
Chaperonin 60/physiology , Escherichia coli Proteins/physiology , HSP70 Heat-Shock Proteins/physiology , Molecular Chaperones/physiology , Peptidylprolyl Isomerase/physiology , Chaperonin 10/chemistry , Chaperonin 10/physiology , Escherichia coli Proteins/chemistry , HSP70 Heat-Shock Proteins/chemistry , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/physiology , Peptidylprolyl Isomerase/chemistry , Protein Folding
8.
Am J Chin Med ; 43(7): 1439-52, 2015.
Article in English | MEDLINE | ID: mdl-26477796

ABSTRACT

Cudraxanthone H (CH) is a natural compound isolated from a methanol extract of the root bark of Cudrania tricuspidata, a herbal plant also known as Moraceae. However, the effect of CH on human cancer cells has not been reported previously. The aim of this study was to investigate the anticancer effects and mechanism of action of CH on oral squamous cell carcinoma (OSCC) cells. CH exerted significant antiproliferative effects on OSCC cells in dose- and time-dependent manners. CH also induced apoptosis in OSCC cells, as evidenced by an increased percentage of cells in the sub-G1 phase of the cell cycle, annexin V-positive/propidium iodide-negative cells, and nuclear morphology. This antiproliferative effect of CH was associated with a marked reduction in the expression of cyclin D1 and cyclin E, with a concomitant induction of cyclin-dependent kinase inhibitor (CDKI) expression (p21 and p27). CH inhibited the phosphorylation and degradation of IκB-α and the nuclear translocation of NF-κB p65. Furthermore, CH treatment down-regulated PIN1 mRNA and protein expression in a dose-dependent manner. PIN1 overexpression by infection with adenovirus-PIN1 (Ad-PIN1) attenuated the CH-induced growth-inhibiting and apoptosis-inducing effects, blocked CH-enhanced CDKI expression and restored cyclin levels. In contrast, inhibiting PIN1 expression via juglone exerted the opposite effects. The present study is the first to demonstrate antiproliferative and apoptosis-inducing effects of CH, which exerts its effects by inhibiting NF-κB and PIN1. These data suggest that it might be a novel alternative chemotherapeutic agent for use in the treatment of oral cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , NF-kappa B/physiology , Peptidylprolyl Isomerase/physiology , Phytotherapy , Signal Transduction/drug effects , Signal Transduction/genetics , Xanthones/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Humans , Moraceae/chemistry , Mouth Neoplasms/drug therapy , NIMA-Interacting Peptidylprolyl Isomerase , Xanthones/isolation & purification
9.
Mol Cell ; 59(4): 677-84, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26236013

ABSTRACT

The cytosolic fraction of the tumor suppressor p53 activates the apoptotic effector protein BAX to trigger apoptosis. Here we report that p53 activates BAX through a mechanism different from that associated with activation by BH3 only proteins (BIM and BID). We observed that cis-trans isomerization of proline 47 (Pro47) within p53, an inherently rare molecular event, was required for BAX activation. The prolyl isomerase Pin1 enhanced p53-dependent BAX activation by catalyzing cis-trans interconversion of p53 Pro47. Our results reveal a signaling mechanism whereby proline cis-trans isomerization in one protein triggers conformational and functional changes in a downstream signaling partner. Activation of BAX through the concerted action of cytosolic p53 and Pin1 may integrate cell stress signals to induce a direct apoptotic response.


Subject(s)
Apoptosis , Peptidylprolyl Isomerase/physiology , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism , Cell Line, Tumor , Humans , Kinetics , NIMA-Interacting Peptidylprolyl Isomerase , Proline/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Stereoisomerism , Tumor Suppressor Protein p53/chemistry , bcl-2-Associated X Protein/chemistry
10.
Biochem Biophys Res Commun ; 464(2): 487-92, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26150353

ABSTRACT

Aging of tendon stem/progenitor cells (TSPCs) can lead to tissue degeneration and subsequent injury. However, the molecular mechanisms controlling TSPC aging are not completely understood. In the present study, we investigated the role of Pin1 in aging of human TSPCs. Pin1 mRNA and protein expression levels were significantly decreased during prolonged in vitro culture of human TSPCs. Furthermore, overexpression of Pin1 delayed the progression of cellular senescence, as confirmed by downregulation of senescence-associated ß-galactosidase, increased telomerase activity and decreased levels of the senescence marker, p16(INK4A). Conversely, Pin1 siRNA transfection promoted senescence in TSPCs. In addition, miR-140-5p regulated Pin1 expression at the translational level via directly targeting its 3'UTR. Our results collectively demonstrate that Pin1 acts as an important regulator of TSPC aging.


Subject(s)
Cellular Senescence/physiology , Peptidylprolyl Isomerase/physiology , Stem Cells/cytology , Tendons/cytology , Cells, Cultured , Gene Knockdown Techniques , Humans , MicroRNAs/physiology , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/genetics
11.
Zhongguo Dang Dai Er Ke Za Zhi ; 17(5): 496-501, 2015 May.
Article in Chinese | MEDLINE | ID: mdl-26014703

ABSTRACT

OBJECTIVE: To explore the effect of silence of Pin1 expression on hyperoxia-induced apoptosis in alveolar epithelial cells A549. METHODS: A549 cells were divided into four groups: control, hyperoxia, negative lentivirus and Pin1-shRNA hyperoxia. The hyperoxia group was exposed to a mixture of 95%O2 and 5%CO2 for 10 minutes. Then cells were cultured in a closed environment. After 24 hours, the changes of morphology were observed under an inverted microscope. Cell apoptosis was detected by flow cytometry (FCM). The expression of X-linked inhibitor of apoptosis protein (XIAP) and Caspase-9 were detected by immunohistochemistry. The production of reactive oxygen species (ROS) and cellular mitochondria membrane potential (△Ψm) were determined by fluorescence microscopy. RESULTS: Under the inverted microscope, the A549 cells grew slowly and the changes in morphology of the cells were most obvious in the hyperoxia and negative lentivirus groups. The changes in morphology of A549 cells were obviously improved in the Pin1-shRNA hyperoxia group. The FCM results showed that the apoptosis rate of A549 cells increased, Caspase-9 expression increased, XIAP expression decreased, mitochondrial ROS production increased and mitochondrial membrane potential decreased in the hyperoxia and negative lentivirus groups compared with the control group (P<0.05). Compared with the hyperoxia and negative lentivirus groups, the apoptosis rate of A549 cells decreased, Caspase-9 expression decreased, XIAP expression increased, mitochondrial ROS production decreased and mitochondrial membrane potential increased in the Pin1-shRNA hyperoxia group (P<0.05), although the levels of the indexes did not reach to those of the control group. CONCLUSIONS: Silencing of Pin1 could suppress hyperoxia-induced apoptosis of A549 cells.


Subject(s)
Apoptosis , Hyperoxia/pathology , Peptidylprolyl Isomerase/physiology , Caspase 9/genetics , Humans , Membrane Potential, Mitochondrial , NIMA-Interacting Peptidylprolyl Isomerase , Reactive Oxygen Species/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics
12.
Exp Biol Med (Maywood) ; 240(3): 403-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25662955

ABSTRACT

Pin1 is a peptidyl-prolyl isomerase which plays a critical role in many diseases including cancer and Alzheimer's disease. The essential role of Pin1 is to affect stability, localization or function of phosphoproteins by catalyzing structural changes. Among the collection of Pin1 substrates, many have been shown to be involved in regulating cell cycle progression. The cell cycle disorder caused by dysregulation of these substrates is believed to be a common phenomenon in cancer. A number of recent studies have revealed possible functions of several important Pin1-binding cell cycle regulators. Investigating the involvement of Pin1 in the cell cycle may assist in the development of future cancer therapeutics. In this review, we summarize current knowledge regarding the network of Pin1 substrates and Pin1 regulators in cell cycle progression. In G1/S progression, cyclin D1, RB, p53, p27, and cyclin E are all well-known cell cycle regulators that are modulated by Pin1. During G2/M transition, our lab has shown that Aurora A suppresses Pin1 activity through phosphorylation at Ser16 and cooperates with hBora to modulate G2/M transition. We conclude that Pin1 may be thought of as a molecular timer which modulates cell cycle progression networks.


Subject(s)
Cell Cycle/physiology , Peptidylprolyl Isomerase/physiology , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Phosphoproteins/physiology , Signal Transduction/physiology
13.
Apoptosis ; 20(2): 224-34, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25563855

ABSTRACT

Eosinophils (Eos) are potent inflammatory cells and abundantly present in the sputum and lung of patients with allergic asthma. During both transit to and residence in the lung, Eos contact prosurvival cytokines, particularly IL-3, IL-5 and GM-CSF, that attenuate cell death. Cytokine signaling modulates the expression and function of a number of intracellular pro- and anti-apoptotic molecules. Both intrinsic mitochondrial and extrinsic receptor-mediated pathways are affected. This article discusses the fundamental role of the extracellular and intracellular molecules that initiate and control survival decisions by human Eos and highlights the role of the cis-trans isomerase, Pin1 in controlling these processes.


Subject(s)
Apoptosis , Eosinophils/physiology , Animals , Apoptosis Regulatory Proteins/physiology , Cell Survival , Cytokines/physiology , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/physiology , Signal Transduction
14.
Eur Heart J ; 36(13): 817-28, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-24801072

ABSTRACT

AIM: Diabetes is a major driver of cardiovascular disease, but the underlying mechanisms remain elusive. Prolyl-isomerase Pin1 recognizes specific peptide bonds and modulates function of proteins altering cellular homoeostasis. The present study investigates Pin1 role in diabetes-induced vascular disease. METHODS AND RESULTS: In human aortic endothelial cells (HAECs) exposed to high glucose, up-regulation of Pin1-induced mitochondrial translocation of pro-oxidant adaptor p66(Shc) and subsequent organelle disruption. In this setting, Pin1 recognizes Ser-116 inhibitory phosphorylation of endothelial nitric oxide synthase (eNOS) leading to eNOS-caveolin-1 interaction and reduced NO availability. Pin1 also mediates hyperglycaemia-induced nuclear translocation of NF-κB p65, triggering VCAM-1, ICAM-1, and MCP-1 expression. Indeed, gene silencing of Pin1 in HAECs suppressed p66(Shc)-dependent ROS production, restored NO release and blunted NF-kB p65 nuclear translocation. Consistently, diabetic Pin1(-/-) mice were protected against mitochondrial oxidative stress, endothelial dysfunction, and vascular inflammation. Increased expression and activity of Pin1 were also found in peripheral blood monocytes isolated from diabetic patients when compared with age-matched healthy controls. Interestingly, enough, Pin1 up-regulation was associated with impaired flow-mediated dilation, increased urinary 8-iso-prostaglandin F2α and plasma levels of adhesion molecules. CONCLUSIONS: Pin1 drives diabetic vascular disease by causing mitochondrial oxidative stress, eNOS dysregulation as well as NF-kB-induced inflammation. These findings provide molecular insights for novel mechanism-based therapeutic strategies in patients with diabetes.


Subject(s)
Diabetic Angiopathies/prevention & control , Mitochondrial Diseases/prevention & control , Oxidative Stress/physiology , Peptidylprolyl Isomerase/physiology , Analysis of Variance , Animals , Aorta/metabolism , Case-Control Studies , Cells, Cultured , Chemokine CCL2/metabolism , Cytochromes c/biosynthesis , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Gene Knockdown Techniques , Glucose/pharmacology , Humans , Hyperglycemia/physiopathology , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , NIMA-Interacting Peptidylprolyl Isomerase , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism , Shc Signaling Adaptor Proteins/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1 , Up-Regulation/physiology , Vascular Cell Adhesion Molecule-1/metabolism , Vasculitis/physiopathology
15.
J Prosthodont Res ; 58(4): 193-207, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25311991

ABSTRACT

Periodontal ligament (PDL) plays critical roles in the development and maintenance of periodontium such as tooth eruption and dissipation of masticatory force. The mechanical properties of PDL are mainly derived from fibrillar type I collagen, the most abundant extracellular component. The biosynthesis of type I collagen is a long, complex process including a number of intra- and extracellular post-translational modifications. The final modification step is the formation of covalent intra- and intermolecular cross-links that provide collagen fibrils with stability and connectivity. It is now clear that collagen post-translational modifications are regulated by groups of specific enzymes and associated molecules in a tissue-specific manner; and these modifications appear to change in response to mechanical force. This review focuses on the effect of mechanical loading on collagen biosynthesis and fibrillogenesis in PDL with emphasis on the post-translational modifications of collagens, which is an important molecular aspect to understand in the field of prosthetic dentistry.


Subject(s)
Collagen Type I/biosynthesis , Collagen Type I/genetics , Periodontal Ligament/metabolism , Animals , Epigenesis, Genetic , Gene Expression Regulation, Developmental/genetics , Humans , Molecular Chaperones/physiology , Peptidylprolyl Isomerase/physiology , Periodontal Ligament/physiology , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/physiology , Prolyl Hydroxylases/physiology , Prosthodontics , Protein Processing, Post-Translational
16.
Cell Mol Life Sci ; 71(17): 3311-25, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24760129

ABSTRACT

By virtue of their general ability to bind (hold) translocating or unfolding polypeptides otherwise doomed to aggregate, molecular chaperones are commonly dubbed "holdases". Yet, chaperones also carry physiological functions that do not necessitate prevention of aggregation, such as altering the native states of proteins, as in the disassembly of SNARE complexes and clathrin coats. To carry such physiological functions, major members of the Hsp70, Hsp110, Hsp100, and Hsp60/CCT chaperone families act as catalytic unfolding enzymes or unfoldases that drive iterative cycles of protein binding, unfolding/pulling, and release. One unfoldase chaperone may thus successively convert many misfolded or alternatively folded polypeptide substrates into transiently unfolded intermediates, which, once released, can spontaneously refold into low-affinity native products. Whereas during stress, a large excess of non-catalytic chaperones in holding mode may optimally prevent protein aggregation, after the stress, catalytic disaggregases and unfoldases may act as nanomachines that use the energy of ATP hydrolysis to repair proteins with compromised conformations. Thus, holding and catalytic unfolding chaperones can act as primary cellular defenses against the formation of early misfolded and aggregated proteotoxic conformers in order to avert or retard the onset of degenerative protein conformational diseases.


Subject(s)
Molecular Chaperones/physiology , Protein Folding , Adenosine Triphosphate/physiology , Animals , Catalysis , Escherichia coli Proteins/physiology , Heat-Shock Proteins/physiology , Humans , Models, Biological , Molecular Chaperones/chemistry , Neurodegenerative Diseases/metabolism , Peptidylprolyl Isomerase/physiology , Protein Binding , Protein Conformation , Protein Transport , Proteostasis Deficiencies/metabolism , Stress, Physiological
17.
Life Sci ; 102(2): 98-104, 2014 May 02.
Article in English | MEDLINE | ID: mdl-24657892

ABSTRACT

AIMS: Cardiac hypertrophy is elicited by endothelin (ET)-1 as well as other neurohumoral factors, hemodynamic overload, and oxidative stress; HMG-CoA reductase inhibitors (statins) were shown to inhibit cardiac hypertrophy partly via the anti-oxidative stress. One of their common intracellular pathways is the phosphorylation cascade of MEK signaling. Pin1 specifically isomerizes the phosphorylated protein with Ser/Thr-Pro bonds and regulates their activity through conformational changes. There is no report whether the Pin1 activation contributes to ET-1-induced cardiomyocyte hypertrophy and whether the Pin1 inactivation contributes to the inhibitory effect of statins. The aim of this study was to reveal these questions. MAIN METHODS: We assessed neonatal rat cardiomyocyte hypertrophy using ET-1 and fluvastatin by the cell surface area, ANP mRNA expression, JNK and c-Jun phosphorylation, and [(3)H]-leucine incorporation. KEY FINDINGS: Fluvastatin inhibited ET-1-induced increase in the cell surface area, ANP expression, and [(3)H]-leucine incorporation; and it suppressed the signaling cascade from JNK to c-Jun. The phosphorylated Pin1 level, an inactive form, was decreased by ET-1; however, it reached basal level by fluvastatin. Furthermore, Pin1 overexpression clearly elicited cardiomyocyte hypertrophy, which was inhibited by fluvastatin. SIGNIFICANCE: This is the first report that ET-1-induced cardiomyocyte hypertrophy is mediated through the Pin1 activation and that the inhibitory effect of fluvastatin on cardiomyocyte hypertrophy would partly be attributed to the suppression of the Pin1 function. This study firstly suggests that Pin1 determines the size of hypertrophied cardiomyocyte by regulating the activity of phosphorylated molecules and that statins exert their pleiotropic effects partly via Pin1 inactivation.


Subject(s)
Cardiomegaly/prevention & control , Endothelin-1/toxicity , Fatty Acids, Monounsaturated/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Indoles/pharmacology , Myocytes, Cardiac/metabolism , Peptidylprolyl Isomerase/physiology , Animals , Animals, Newborn , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Cells, Cultured , Endothelin-1/antagonists & inhibitors , Fatty Acids, Monounsaturated/therapeutic use , Fluvastatin , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Indoles/therapeutic use , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/antagonists & inhibitors , Peptidylprolyl Isomerase/biosynthesis , Rats , Rats, Sprague-Dawley , Treatment Outcome
18.
Oncogene ; 33(11): 1438-47, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-23542176

ABSTRACT

Estrogen receptor-alpha (ERα) is an important biomarker used to classify and direct therapy decisions in breast cancer (BC). Both ERα protein and its transcript, ESR1, are used to predict response to tamoxifen therapy, yet certain tumors have discordant levels of ERα protein and ESR1, which is currently unexplained. Cellular ERα protein levels can be controlled post-translationally by the ubiquitin-proteasome pathway through a mechanism that depends on phosphorylation at residue S118. Phospho-S118 (pS118-ERα) is a substrate for the peptidyl prolyl isomerase, Pin1, which mediates cis-trans isomerization of the pS118-P119 bond to enhance ERα transcriptional function. Here, we demonstrate that Pin1 can increase ERα protein without affecting ESR1 transcript levels by inhibiting proteasome-dependent receptor degradation. Pin1 disrupts ERα ubiquitination by interfering with receptor interactions with the E3 ligase, E6AP, which also is shown to bind pS118-ERα. Quantitative in situ assessments of ERα protein, ESR1, and Pin1 in human tumors from a retrospective cohort show that Pin1 levels correlate with ERα protein but not to ESR1 levels. These data show that ERα protein is post-translationally regulated by Pin1 in a proportion of breast carcinomas. As Pin1 impacts both ERα protein levels and transactivation function, these data implicate Pin1 as a potential surrogate marker for predicting outcome of ERα-positive BC.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Peptidylprolyl Isomerase/physiology , Cell Line, Tumor , Female , Humans , NIMA-Interacting Peptidylprolyl Isomerase , Phosphorylation , Proteolysis , Ubiquitination
19.
Intern Emerg Med ; 9(3): 303-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23054412

ABSTRACT

Helicobacter pylori infection is characterized by an inflammatory infiltrate, consisting mainly of neutrophils and T cells. This study was undertaken to evaluate the type of gastric T cell response elicited by the secreted peptidyl prolyl cis, trans-isomerase of H. pylori (HP0175) in patients with distal gastric adenocarcinoma. The cytokine profile and the effector functions of gastric tumor-infiltrating lymphocytes (TILs) specific for HP0175 was investigated in 20 patients with distal gastric adenocarcinoma and H. pylori infection. The helper function of HP0175-specific TILs for monocyte MMP-2, MMP-9, and VEGF production was also investigated. TILs cells from H. pylori infected patients with distal gastric adenocarcinoma produced Interleukin (IL)-17 and IL-21 in response to HP0175. HP0175-specific TILs showed poor cytolytic activity while expressing helper activity for monocyte MMP-2, MMP-9 and VEGF production. These findings indicate that HP0175 is able to drive gastric Th17 response. Thus, HP0175, by promoting pro-inflammatory low cytotoxic TIL response, matrix degradation and pro-angiogenic pathways, may provide a link between H. pylori and gastric cancer.


Subject(s)
Adenocarcinoma/immunology , Helicobacter pylori/metabolism , Peptidylprolyl Isomerase/biosynthesis , Peptidylprolyl Isomerase/physiology , Stomach Neoplasms/immunology , Th17 Cells/immunology , Female , Humans , Inflammation/immunology , Male , Middle Aged , Tumor Cells, Cultured
20.
FEBS Lett ; 587(22): 3640-7, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24113655

ABSTRACT

Peptidyl-prolyl isomerase 1 (Pin1) is the only enzyme known to catalyze isomerization of the pSer/Thr-Pro peptide bond. Pin1 induces conformational change of substrates and subsequently regulates diverse cellular processes. However, its role in osteoblast differentiation is not well understood. Here we show that Pin1 enhances osteoblast differentiation. Pin1 interacts and affects the protein stability and transcriptional activity of an important osteogenic transcriptional factor Runx2. Our results indicate that this regulation is likely due to suppression of poly-ubiquitination-mediated proteasomal degradation of Runx2. Our current finding suggests that Pin1 is a novel regulator of osteoblast differentiation that acts through the regulation of Runx2 function.


Subject(s)
Core Binding Factor Alpha 1 Subunit/metabolism , Osteoblasts/enzymology , Peptidylprolyl Isomerase/physiology , Animals , Cell Differentiation , Core Binding Factor Alpha 1 Subunit/chemistry , Gene Expression Regulation , HEK293 Cells , Humans , MAP Kinase Kinase Kinases/metabolism , Mice , NIMA-Interacting Peptidylprolyl Isomerase , Osteoblasts/physiology , Peptidylprolyl Isomerase/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Stability , Proteolysis , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...