Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Biophys Res Commun ; 715: 150001, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38676996

ABSTRACT

The skeletal muscle is a pivotal organ involved in the regulation of both energy metabolism and exercise capacity. There is no doubt that exercise contributes to a healthy life through the consumption of excessive energy or the release of myokines. Skeletal muscles exhibit insulin sensitivity and can rapidly uptake blood glucose. In addition, they can undergo non-shivering thermogenesis through actions of both the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) and small peptide, sarcolipin, resulting in systemic energy metabolism. Accordingly, the maintenance of skeletal muscles is important for both metabolism and exercise. Prolyl isomerase Pin1 is an enzyme that converts the cis-trans form of proline residues and controls substrate function. We have previously reported that Pin1 plays important roles in insulin release, thermogenesis, and lipolysis. However, the roles of Pin1 in skeletal muscles remains unknown. To clarify this issue, we generated skeletal muscle-specific Pin1 knockout mice. Pin1 deficiency had no effects on muscle weights, morphology and ratio of fiber types. However, they showed exacerbated obesity or insulin resistance when fed with a high-fat diet. They also showed a lower ability to exercise than wild type mice did. We also found that Pin1 interacted with SERCA and elevated its activity, resulting in the upregulation of oxygen consumption. Overall, our study reveals that Pin1 in skeletal muscles contributes to both systemic energy metabolism and exercise capacity.


Subject(s)
Energy Metabolism , Muscle, Skeletal , NIMA-Interacting Peptidylprolyl Isomerase , Physical Conditioning, Animal , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Animals , Male , Mice , Diet, High-Fat , Energy Metabolism/genetics , Insulin Resistance , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
2.
J Dermatol ; 50(4): 462-471, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37006202

ABSTRACT

Atopic dermatitis (AD) is attributable to both a genetic predisposition and environmental factors. Among numerous cytokines involved in the pathogenesis of AD, interleukin-33 (IL-33), reportedly escaping exocytotically in response to a scratch, is abundantly expressed in the skin tissues of patients with AD and is postulated to induce inflammatory and autoimmune responses. In this study, we first demonstrated that peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (Pin1), a unique enzyme which isomerizes the proline residues of target proteins, is abundantly expressed in keratinocytes, and that the areas where it is present in the skin tissues of AD patients became expanded due to hyperkeratosis. Thus, we investigated the effects of Pin1 on the regulation of IL-33 expression using the human keratinocyte cell line HaCaT. Interestingly, silencing of the Pin1 gene or treatment with Pin1 inhibitors dramatically reduced IL-33 expressions in HaCaT cells, although Pin1 overexpression did not elevate it. Subsequently, we showed that Pin1 binds to STAT1 and the nuclear factor-kappaB (NF-κB) subunit p65. Silencing the Pin1 gene with small interfering RNAs significantly reduced the phosphorylation of p65, while no marked effects of Pin1 on the STAT1 pathway were detected. Thus, it is likely that Pin1 contributes to increased expression of IL-33 via the NF-κB subunit p65 in HaCaT cells, at least modestly. Nevertheless, further study is necessary to demonstrate the pathogenic roles of Pin1 and IL-33 in AD development.


Subject(s)
Dermatitis, Atopic , Peptidylprolyl Isomerase , Humans , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Interleukin-33/genetics , Interleukin-33/metabolism , HaCaT Cells/metabolism , Phosphorylation , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism
3.
Exp Cell Res ; 425(2): 113544, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36906101

ABSTRACT

Hepatic stellate cells (HSCs) produce extracellular matrixes (ECMs), such as collagen and fibronectin, in response to stimulation with transforming growth factor ß (TGFß). The massive ECM accumulation in the liver due to HSCs causes fibrosis which eventually leads to hepatic cirrhosis and hepatoma development. However, details of the mechanisms underlying continuous HSC activation are as yet poorly understood. We thus attempted to elucidate the role of Pin1, one of the prolyl isomerases, in the underlying mechanism(s), using the human HSC line LX-2. Treatment with Pin1 siRNAs markedly alleviated the TGFß-induced expressions of ECM components such as collagen 1a1/2, smooth muscle actin and fibronectin at both the mRNA and the protein level. Pin1 inhibitors also decreased the expressions of fibrotic markers. In addition, it was revealed that Pin1 associates with Smad2/3/4, and that four Ser/Thr-Pro motifs in the linker domain of Smad3 are essential for binding with Pin1. Pin1 significantly regulated Smad-binding element transcriptional activity without affecting Smad3 phosphorylations or translocation. Importantly, both Yes-associated protein (YAP) and WW domain-containing transcription regulator (TAZ) also participate in ECM induction, and upregulate Smad3 activity rather than TEA domain transcriptional factor transcriptional activity. Although Smad3 interacts with both TAZ and YAP, Pin1 facilitates the Smad3 association with TAZ, but not that with YAP. In conclusion, Pin1 plays pivotal roles in ECM component productions in HSCs through regulation of the interaction between TAZ and Smad3, and Pin1 inhibitors may have the potential to ameliorate fibrotic diseases.


Subject(s)
Fibronectins , Peptidylprolyl Isomerase , Humans , Peptidylprolyl Isomerase/genetics , Peptidylprolyl Isomerase/metabolism , Fibronectins/genetics , Fibronectins/metabolism , Hepatic Stellate Cells/metabolism , Transforming Growth Factor beta/metabolism , Liver Cirrhosis/pathology , Fibrosis , Extracellular Matrix/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism
4.
Cancer Med ; 12(7): 8464-8475, 2023 04.
Article in English | MEDLINE | ID: mdl-36583514

ABSTRACT

BACKGROUND: Prostate cancer (PCa) is a major cause of cancer morbidity and mortality for men globally, and androgen signaling clearly drives its onset and progression. Androgen receptor (AR) regulation is complex and remains elusive, despite several studies tackling these issues. Therefore, elucidating the mechanism(s) underlying AR regulation is a potentially promising approach to suppressing PCa. METHODS: We report that Par14, one isoform of the prolyl isomerases homologous to Pin1, is a critical regulator of AR transcriptional activity and is essential for PCa cell growth. RESULTS: Par14 was shown to be overexpressed in PCa, based on analyses of deposited data. Importantly, overexpression of Par14 significantly enhanced androgen-sensitive LNCap cell growth. In contrast, silencing of Par14 dramatically decreased cell growth in LNCap cells by causing cell cycle arrest. Mechanistically, silencing of the Par14 gene dramatically induced cyclin-dependent kinase inhibitor p21 at both the mRNA and the protein level through modulating the localization of p53. In addition, suppression of Par14 in LNCap cells was shown to downregulate the expressions of androgen response genes, at both the mRNA and the protein level, induced by dihydrotestosterone. Par14 was shown to directly associate with AR in nuclei via its DNA-binding domain and augment AR transcriptional activity. CONCLUSION: Thus, Par14 plays a critical role in PCa progression, and its enhancing effects on AR signaling are likely to be involved in the underlying molecular mechanisms. These findings suggest Par14 to be a promising therapeutic target for PCa.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Male , Humans , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Androgens/pharmacology , Androgens/metabolism , Cell Line, Tumor , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Cell Proliferation , RNA, Messenger/metabolism , Gene Expression Regulation, Neoplastic , NIMA-Interacting Peptidylprolyl Isomerase/genetics
5.
Metabolism ; 115: 154459, 2021 02.
Article in English | MEDLINE | ID: mdl-33279499

ABSTRACT

BACKGROUND: Lipolysis is essential for the supply of nutrients during fasting, the control of body weight, and remodeling of white adipose tissues and thermogenesis. In the obese state, lipolysis activity and the expression of adipose triglyceride lipase (ATGL), a rate-limiting enzyme, is suppressed. However, the mechanism underlying the regulation of ATGL remains largely unknown. We previously reported that a high-fat diet obviously increases protein levels of the prolyl isomerase, Pin1, in epididymal white adipose tissue (epiWAT) of mice and that Pin1 KO mice are resistant to developing obesity. RESULTS: The present study found that deletion of the Pin1 gene in epiWAT upregulated lipolysis and increased ATGL protein expression by ~2-fold. In addition, it was demonstrated that Pin1 directly associated with ATGL and enhanced its degradation through the ubiquitin proteasome system. Indeed, Pin1 overexpression decreased ATGL expression levels, whereas Pin1 knockdown by siRNA treatment upregulated ATGL protein levels without altering mRNA levels. Moreover, under a high fat diet (HFD)-fed condition, adipocyte-specific Pin1 KO (adipoPin1 KO) mice had 2-fold increase lipolytic activity and upregulated ß-oxidation-related gene expressions. These mice also gained less body weight, and had better glucose metabolism according to the results of glucose and insulin tolerance tests. CONCLUSION: Taken together, these results showed that Pin1 directly interacted with and degraded ATGL via a ubiquitin-proteasome system, consequently causing the downregulation of lipolysis. Therefore, Pin1 could be considered a target for the treatment of dyslipidemia and related disorders.


Subject(s)
Adipose Tissue/metabolism , Gene Expression Regulation , Lipase/metabolism , Lipolysis/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Obesity/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Diet, High-Fat , Glucose Tolerance Test , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase/genetics
6.
Cell Rep ; 26(12): 3221-3230.e3, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30893596

ABSTRACT

Non-shivering thermogenesis in adipocytes provides defense against low temperatures and obesity development, but the underlying regulatory mechanism remains to be fully clarified. Based on both markedly increased Pin1 expression in states of excess nutrition and resistance to obesity development in Pin1 null mice, we speculated that adipocyte Pin1 may play a role in thermogenic programs. Adipose-specific Pin1 knockout (adPin1 KO) mice showed enhanced transcription of thermogenic genes and tolerance to hypothermia when exposed to cold. In addition, adPin1 KO mice were resistant to high-fat diet-induced obesity and glucose intolerance. A series of experiments revealed that Pin1 binds to PRDM16 and thereby promotes its degradation through the ubiquitin-proteasome system. Consistent with these results, Pin1 deletion in differentiated adipocytes showed enhancement of thermogenic programs in response to the ß3 agonist CL316243 through the upregulation of PRDM16 proteins. These observations indicate that Pin1 is a negative regulator of non-shivering thermogenesis.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Proteolysis , Thermogenesis/physiology , Transcription Factors/metabolism , Adipocytes/cytology , Animals , DNA-Binding Proteins/genetics , Mice , Mice, Knockout , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Protein Binding , Transcription Factors/genetics , Transcription, Genetic/physiology , Ubiquitination/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...