179 results on '"PPM1D"'
Search Results
2. The role of PPM1D in cancer and advances in studies of its inhibitors
- Author
-
Deng, Wenhong, Li, Jieqing, Dorrah, Kimberly, Jimenez-Tapia, Denise, Arriaga, Brando, Hao, Qiongyu, Cao, Wei, Gao, Zhaoxia, Vadgama, Jay, and Wu, Yong
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Breast Cancer ,Cancer ,Women's Health ,2.1 Biological and endogenous factors ,5.1 Pharmaceuticals ,Antineoplastic Agents ,Gene Expression Regulation ,Neoplastic ,Molecular Structure ,Neoplasms ,Protein Phosphatase 2C ,PPM1D ,Phosphatase ,Inhibitor ,p53 ,Pharmacology and Pharmaceutical Sciences ,Oncology & Carcinogenesis ,Pharmacology and pharmaceutical sciences - Abstract
A greater understanding of factors causing cancer initiation, progression and evolution is of paramount importance. Among them, the serine/threonine phosphatase PPM1D, also referred to as wild-type p53-induced phosphatase 1 (Wip1) or protein phosphatase 2C delta (PP2Cδ), is emerging as an important oncoprotein due to its negative regulation on a number of crucial cancer suppressor pathways. Initially identified as a p53-regulated gene, PPM1D has been afterwards found amplified and more recently mutated in many human cancers such as breast cancer. The latest progress in this field further reveals that selective inhibition of PPM1D to delay tumor onset or reduce tumor burden represents a promising anti-cancer strategy. Here, we review the advances in the studies of the PPM1D activity and its relevance to various cancers, and recent progress in development of PPM1D inhibitors and discuss their potential application in cancer therapy. Consecutive research on PPM1D and its relationship with cancer is essential, as it ultimately contributes to the etiology and treatment of cancer.
- Published
- 2020
3. Jansen‐de Vries syndrome: Expansion of the PPM1D clinical and phenotypic spectrum in 34 families.
- Author
-
Wojcik, Monica H., Srivastava, Siddharth, Agrawal, Pankaj B., Balci, Tugce B., Callewaert, Bert, Calvo, Pier Luigi, Carli, Diana, Caudle, Michelle, Colaiacovo, Samantha, Cross, Laura, Demetriou, Kalliope, Drazba, Katy, Dutra‐Clarke, Marina, Edwards, Matthew, Genetti, Casie A., Grange, Dorothy K., Hickey, Scott E., Isidor, Bertrand, Küry, Sébastien, and Lachman, Herbert M.
- Abstract
Jansen‐de Vries syndrome (JdVS) is a neurodevelopmental condition attributed to pathogenic variants in Exons 5 and 6 of PPM1D. As the full phenotypic spectrum and natural history remain to be defined, we describe a large cohort of children and adults with JdVS. This is a retrospective cohort study of 37 individuals from 34 families with disease‐causing variants in PPM1D leading to JdVS. Clinical data were provided by treating physicians and/or families. Of the 37 individuals, 27 were male and 10 female, with median age 8.75 years (range 8 months to 62 years). Four families document autosomal dominant transmission, and 32/34 probands were diagnosed via exome sequencing. The facial gestalt, including a broad forehead and broad mouth with a thin and tented upper lip, was most recognizable between 18 and 48 months of age. Common manifestations included global developmental delay (35/36, 97%), hypotonia (25/34, 74%), short stature (14/33, 42%), constipation (22/31, 71%), and cyclic vomiting (6/35, 17%). Distinctive personality traits include a hypersocial affect (21/31, 68%) and moderate‐to‐severe anxiety (18/28, 64%). In conclusion, JdVS is a clinically recognizable neurodevelopmental syndrome with a characteristic personality and distinctive facial features. The association of pathogenic variants in PPM1D with cyclic vomiting bears not only medical attention but also further pathogenic and mechanistic evaluation. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
4. Characteristic analysis and identification of novel molecular biomarkers in elderly glioblastoma patients using the 2021 WHO Classification of Central Nervous System Tumors.
- Author
-
Yaning Wang, Junlin Li, Yaning Cao, Wenlin Chen, Hao Xing, Xiaopeng Guo, Yixin Shi, Yuekun Wang, Tingyu Liang, Liguo Ye, Delin Liu, Tianrui Yang, Yu Wang, and Wenbin Ma
- Subjects
OLDER patients ,BRAIN tumors ,CENTRAL nervous system tumors ,INTRACRANIAL hypertension - Abstract
Introduction: Elderly glioblastoma (GBM) patients is characterized by high incidence and poor prognosis. Currently, however, there is still a lack of adequate molecular characterization of elderly GBM patients. The fifth edition of the WHO Classification of Central Nervous System Tumors (WHO5) gives a new classification approach for GBM, and the molecular characteristics of elderly GBM patients need to be investigated under this new framework. Methods: The clinical and radiological features of patients with different classifications and different ages were compared. Potential prognostic molecular markers in elderly GBM patients under the WHO5 classification were found using Univariate Cox regression and Kaplan--Meier survival analysis. Results: A total of 226 patients were included in the study. The prognostic differences between younger and elderly GBM patients were more pronounced under the WHO5 classification. Neurological impairment was more common in elderly patients (p = 0.001), while intracranial hypertension (p = 0.034) and epilepsy (p = 0.038) were more common in younger patients. Elderly patients were more likely to have higher Ki-67(p = 0.013), and in elderly WHO5 GBM patients, KMT5B (p = 0.082), KRAS (p = 0.1) and PPM1D (p = 0.055) were each associated with overall survival (OS). Among them, KRAS and PPM1D were found to be prognostic features unique to WHO5 elderly GBM patients. Conclusion: Our study demonstrates that WHO5 classification can better distinguish the prognosis of elderly and younger GBM. Furthermore, KRAS and PPM1D may be potential prognostic predictors in WHO5 elderly GBM patients. The specific mechanism of these two genes in elderly GBM remains to be further studied. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Short stature leads to a diagnosis of Jansen–de Vries syndrome in two unrelated Taiwanese girls: A case report and literature review
- Author
-
Meng-Ju Melody Tsai, Ni-Chung Lee, Yin-Hsiu Chien, Wuh-Liang Hwu, and Yi-Ching Tung
- Subjects
PPM1D ,Short stature ,Intellectual disability ,Jansen–de Vries syndrome (JDVS) ,Medicine (General) ,R5-920 - Abstract
Short stature and intellectual disability are two of the major components of many dysmorphic syndromes. Jansen–de Vries syndrome (JDVS) is a rare syndromic disorder that was discovered recently using next-generation sequencing. It is characterized by hypotonia, developmental delay, a dysmorphic face, short stature, and high pain threshold and is caused by the variants of the protein phosphatase magnesium-dependent 1D (PPM1D) gene. Here, we report the first two cases of PPM1D mutations in Taiwan; both had de novo variants in exon 6. Both presented with short stature, developmental delay, and dysmorphic faces. In addition to the characteristics listed above, syndactyly was noted in one. Genetic studies should be considered when approaching a patient with growth retardation, intellectual disability, and other major or minor dysmorphisms.
- Published
- 2022
- Full Text
- View/download PDF
6. TC-hunter: identification of the insertion site of a transgenic gene within the host genome
- Author
-
Vanja Börjesson, Angela Martinez-Monleon, Susanne Fransson, Per Kogner, John Inge Johnsen, Jelena Milosevic, and Marcela Dávila López
- Subjects
Next-generation sequencing ,Transgenic insertion site ,PPM1D ,Chimeric reads ,Discordant read pairs ,Transgenic organisms ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background Transgenic animal models are crucial for the study of gene function and disease, and are widely utilized in basic biological research, agriculture and pharma industries. Since the current methods for generating transgenic animals result in the random integration of the transgene under study, the phenotype may be compromised due to disruption of known genes or regulatory regions. Unfortunately, most of the tools that predict transgene insertion sites from high-throughput data are not publicly available or not properly maintained. Results We implemented TC-hunter, Transgene-Construct hunter, an open tool that identifies transgene insertion sites and provides simple reports and visualization aids. It relies on common tools used in the analysis of high-throughput data and makes use of chimeric reads and discordant read pairs to identify and support the transgenic insertion site. To demonstrate its applicability, we applied TC-hunter to four transgenic mice samples harboring the human PPM1D gene, a model used in the study of malignant tumor development. We identified the transgenic insertion site in each sample and experimentally validated them with Touchdown-polymerase chain reaction followed by Sanger sequencing. Conclusions TC-hunter is an accessible bioinformatics tool that can automatically identify transgene insertion sites from DNA sequencing data with high sensitivity (98%) and precision (92.45%). TC-hunter is a valuable tool that can aid in evaluating any potential phenotypic complications due to the random integration of the transgene and can be accessed at https://github.com/bcfgothenburg/SSF .
- Published
- 2022
- Full Text
- View/download PDF
7. Development of Antibody-like Proteins Targeting the Oncogenic Ser/Thr Protein Phosphatase PPM1D.
- Author
-
Ikeura, Megumi, Tashiro, Hiroto, Yamagata, Yuka, Saito, Hikaru, Kobayashi, Tamaki, Mizunuma, Masataka, Yamazaki, Kazuki, Baba, Keisuke, Furukawa, Kazuhiro, and Chuman, Yoshiro
- Subjects
ONCOGENIC proteins ,PHOSPHOPROTEIN phosphatases ,RECOMBINANT proteins ,SMALL molecules ,DOSAGE forms of drugs - Abstract
PPM1D, a protein Ser/Thr phosphatase, is overexpressed in various cancers and functions as an oncogenic protein by inactivating the p53 pathway. Therefore, molecules that bind PPM1D are expected to be useful anti-cancer agents. In this study, we constructed a phage display library based on the antibody-like small molecule protein adnectin and screened for PPM1D-specific binding molecules. We identified two adnectins, PMDB-1 and PMD-24, that bind PPM1D specific B-loop and PPM1D430 as targets, respectively. Specificity analyses of these recombinant proteins using other Ser/Thr protein phosphatases showed that these molecules bind to only PPM1D. Expression of PMDB-1 in breast cancer-derived MCF-7 cells overexpressing endogenous PPM1D stabilized p53, indicating that PMDB-1 functions as an inhibitor of PPM1D. Furthermore, MTT assay exhibited that MCF-7 cells expressing PMDB-1 showed inhibition of cell proliferation. These data suggest that the adnectin PMDB-1 identified in this study can be used as a lead compound for anti-cancer drugs targeting intracellular PPM1D. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
8. Clonal hematopoiesis and cardiovascular disease in cancer patients and survivors.
- Author
-
Fuster, José J.
- Abstract
Cancer genomes have long been known to carry a high number of somatic mutations distributed across many genes. However, recent sequencing studies have unveiled that non-cancerous cells also carry a considerable number of somatic mutations, which are acquired continuously through the lifespan. Accordingly, the pathophysiological relevance of somatic mutagenesis beyond cancer has become a topic of intensive research. Human genetic studies and experiments in mice have shown that some somatic mutations in the hematopoietic system provide a competitive advantage to the mutant cell and allow its clonal expansion. This phenomenon, termed clonal hematopoiesis, is typically driven by mutations in known oncogenes and tumor suppressor genes, and it is associated with a higher risk of hematological malignancies. Unexpectedly, accumulating genetic and experimental evidence strongly suggest that clonal hematopoiesis, at least when driven by certain mutations, also contributes causally to the development of cardiovascular disease and, therefore, represents a new cardiovascular risk factor. While clonal hematopoiesis is relatively common in healthy individuals, especially among the elderly, it is particularly frequent in cancer patients and survivors. Hence, it has emerged as a candidate contributor to the increased risk of cardiovascular complications in cancer patients. This review summarizes our current understanding of the connection between clonal hematopoiesis and cardiovascular disease, with a special focus on the available evidence linking clonal hematopoiesis to cardiovascular disorders that are frequent in cancer patients and survivors. • Somatic mutations that drive clonal hematopoiesis represent a new cardiovascular risk factor. • Clonal hematopoiesis is particularly common in elderly individuals and cancer patients. • Some clonal hematopoiesis-related mutations contribute directly to cardiovascular disease. • Cytotoxic therapies facilitate clonal hematopoiesis driven by mutations in DNA damage response genes. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
9. TRPM2-AS promotes the malignancy of osteosarcoma cells by targeting miR-15b-5p/PPM1D axis.
- Author
-
Cai, Yingchun, Yang, Yudan, Zhang, Xudong, Ma, Qingqing, and Li, Mengyi
- Subjects
LINCRNA ,MICRORNA ,OSTEOSARCOMA - Abstract
Osteosarcoma (OS) is a malignant tumor with a low survival rate and a high incidence rate worldwide. Although research has reported the involvement of long non-coding RNAs (lncRNAs) in the pathogenesis of OS cells, the role of TRPM2-AS, miR-15b-5p, and PPM1D in OS progression remains unclear. This study aimed to examine the interaction of the TRPM2-AS/miR-15b-5p/PPM1D axis in OS cells to gain new insights into the molecular mechanism and pathogenesis of OS. After performing in vitro functional assays, we discovered that TRPM2-AS was overexpressed in OS cells. TRPM2-AS silencing impaired OS cell viability, proliferation, and migration, while it induced apoptosis in OS cells in vitro. Our experimental analysis also revealed that PPM1D is a direct target of miR-15b-5p. TRPM2-AS silencing was found to reverse the tumorigenic effect of the miR-15b-5p inhibitor, while the miR-15b-5p inhibitor restored the inhibition of OS caused by silencing PPM1D. Moreover, our findings revealed that miR-15b-5p exerted its tumor-suppressive role by directly targeting PPM1D. In conclusion, this study suggests that TRPM2-AS could promote OS cell malignancy by sponging miR-15b-5p/PPM1D axis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
10. Short stature leads to a diagnosis of Jansen-de Vries syndrome in two unrelated Taiwanese girls: A case report and literature review.
- Author
-
Tsai, Meng-Ju Melody, Lee, Ni-Chung, Chien, Yin-Hsiu, Hwu, Wuh-Liang, and Tung, Yi-Ching
- Subjects
SHORT stature ,BODY dysmorphic disorder ,PAIN threshold ,PHOSPHOPROTEIN phosphatases ,INTELLECTUAL disabilities - Abstract
Short stature and intellectual disability are two of the major components of many dysmorphic syndromes. Jansen-de Vries syndrome (JDVS) is a rare syndromic disorder that was discovered recently using next-generation sequencing. It is characterized by hypotonia, developmental delay, a dysmorphic face, short stature, and high pain threshold and is caused by the variants of the protein phosphatase magnesium-dependent 1D (PPM1D) gene. Here, we report the first two cases of PPM1D mutations in Taiwan; both had de novo variants in exon 6. Both presented with short stature, developmental delay, and dysmorphic faces. In addition to the characteristics listed above, syndactyly was noted in one. Genetic studies should be considered when approaching a patient with growth retardation, intellectual disability, and other major or minor dysmorphisms. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
11. TC-hunter: identification of the insertion site of a transgenic gene within the host genome.
- Author
-
Börjesson, Vanja, Martinez-Monleon, Angela, Fransson, Susanne, Kogner, Per, Johnsen, John Inge, Milosevic, Jelena, and López, Marcela Dávila
- Subjects
REGULATOR genes ,TRANSGENIC animals ,TRANSGENIC mice ,DNA sequencing ,GENES - Abstract
Background: Transgenic animal models are crucial for the study of gene function and disease, and are widely utilized in basic biological research, agriculture and pharma industries. Since the current methods for generating transgenic animals result in the random integration of the transgene under study, the phenotype may be compromised due to disruption of known genes or regulatory regions. Unfortunately, most of the tools that predict transgene insertion sites from high-throughput data are not publicly available or not properly maintained. Results: We implemented TC-hunter, Transgene-Construct hunter, an open tool that identifies transgene insertion sites and provides simple reports and visualization aids. It relies on common tools used in the analysis of high-throughput data and makes use of chimeric reads and discordant read pairs to identify and support the transgenic insertion site. To demonstrate its applicability, we applied TC-hunter to four transgenic mice samples harboring the human PPM1D gene, a model used in the study of malignant tumor development. We identified the transgenic insertion site in each sample and experimentally validated them with Touchdown-polymerase chain reaction followed by Sanger sequencing. Conclusions: TC-hunter is an accessible bioinformatics tool that can automatically identify transgene insertion sites from DNA sequencing data with high sensitivity (98%) and precision (92.45%). TC-hunter is a valuable tool that can aid in evaluating any potential phenotypic complications due to the random integration of the transgene and can be accessed at https://github.com/bcfgothenburg/SSF. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. PPM1D regulates p21 expression via dephoshporylation at serine 123
- Author
-
Cao, Ruibing, Zhang, Jin, Zhang, Min, and Chen, Xinbin
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,1.1 Normal biological development and functioning ,Animals ,Blotting ,Western ,Cell Cycle ,Cyclin-Dependent Kinase Inhibitor p21 ,DNA Primers ,Dogs ,Dose-Response Relationship ,Drug ,Mutagenesis ,Phosphoprotein Phosphatases ,Phosphorylation ,Plasmids ,Protein Isoforms ,Protein Phosphatase 2C ,Reverse Transcriptase Polymerase Chain Reaction ,Serine ,p21 ,p53 ,phosphatase ,PPM1D ,Wip1 ,ATM ,ataxia telangiectasia mutated ,CDKs ,cyclin-dependent kinases ,Cip1 ,Cdk interacting protein 1 ,PCNA ,proliferating cell nuclear antigen ,PP2C ,type 2C protein phosphatases ,PPM1D ,Protein phosphatase Mg2+/Mn2+ dependent 1D ,WAF1 ,wild-type p53 activated factor 1 ,Wip1 ,wild-type p53 induced phosphatase 1 ,Developmental Biology ,Biochemistry and cell biology - Abstract
The cyclin-dependent kinase inhibitor p21 plays a critical role in regulating cell cycle and cell proliferation. We previously cloned the dog p21 gene and found that unlike human p21, dog p21 is expressed as 2 isoforms due to the proline-directed phosphorylation at serine 123 (S123). Here, we identified that PPM1D, also called Wip1 and a Mg(2+)-dependent phosphatase, dephosphorylates dog p21 protein at serine 123. Specifically, we showed that the level of S123-phosphorylated dog p21 is increased by a PPM1D inhibitor in a dose-dependent manner. We also showed that over-expression of PPM1D decreases, whereas knockdown of PPM1D increases, the level of S123-phosphorylated dog p21 regardless of p53. Additionally, in vitro phosphatase assay was performed and showed that phosphorylated S123 in dog p21 is dephosphorylated by recombinant rPPM1D, which contains the catalytic domain of human PPM1D (residue 1-420), but not by the phosphatase dead rPPM1D (D314A). Furthermore, dephosphorylation of S123 by rPPM1D can be abrogated by PPM1D inhibitor or by withdrawal of Mg(2+). Finally, we showed that upon PPM1D inhibition, the level of S123-phosphorylated dog p21 was increased, concomitantly with decreased expression of cyclin A, cyclin B, Rb, and PCNA. Together, our results indicate that PPM1D functions as a phosphatase of dog p21 at serine 123 and plays a role in cell cycle control via p21.
- Published
- 2015
13. Crystal structure and mechanistic studies of the PPM1D serine/threonine phosphatase catalytic domain.
- Author
-
Kumar JP, Kosek D, Durell SR, Miller Jenkins LM, Debnath S, Coussens NP, Hall MD, Appella DH, Dyda F, Mazur SJ, and Appella E
- Subjects
- Humans, Crystallography, X-Ray, Magnesium metabolism, Magnesium chemistry, Molecular Dynamics Simulation, Kinetics, Phosphoprotein Phosphatases chemistry, Phosphoprotein Phosphatases metabolism, Phosphoprotein Phosphatases genetics, Protein Phosphatase 2C metabolism, Protein Phosphatase 2C chemistry, Protein Phosphatase 2C genetics, Catalytic Domain
- Abstract
Protein phosphatase 1D (PPM1D, Wip1) is induced by the tumor suppressor p53 during DNA damage response signaling and acts as an oncoprotein in several human cancers. Although PPM1D is a potential therapeutic target, insights into its atomic structure were challenging due to flexible regions unique to this family member. Here, we report the first crystal structure of the PPM1D catalytic domain to 1.8 Å resolution. The structure reveals the active site with two Mg
2+ ions bound, similar to other structures. The flap subdomain and B-loop, which are crucial for substrate recognition and catalysis, were also resolved, with the flap forming two short helices and three short β-strands that are followed by an irregular loop. Unexpectedly, a nitrogen-oxygen-sulfur bridge was identified in the catalytic domain. Molecular dynamics simulations and kinetic studies provided further mechanistic insights into the regulation of PPM1D catalytic activity. In particular, the kinetic experiments demonstrated a magnesium concentration-dependent lag in PPM1D attaining steady-state velocity, a feature of hysteretic enzymes that show slow transitions compared with catalytic turnover. All combined, these results advance the understanding of PPM1D function and will support the development of PPM1D-targeted therapeutics., Competing Interests: Conflicts of interests The authors declare that they have no conflicts of interest with the contents of this article., (Published by Elsevier Inc.)- Published
- 2024
- Full Text
- View/download PDF
14. Novel truncating variant of PPM1D penultimate exon in a Chinese patient with Jansen‐de Vries syndrome
- Author
-
Zhuoguang Li, Caiqi Du, Cai Zhang, Mini Zhang, Yanqin Ying, Yan Liang, and Xiaoping Luo
- Subjects
exome sequencing ,intellectual disability ,Jansen‐de Vries syndrome ,PPM1D ,variant ,Genetics ,QH426-470 - Abstract
Abstract Background Jansen‐de Vries syndrome is a rare autosomal dominant neurodevelopmental disorder caused by pathogenic variants in the last and penultimate exons of the PPM1D gene. It is characterized by delayed psychomotor development, intellectual disability with speech delay, behavioral abnormalities, and dysmorphic features. Up to date, only 17 affected patients have been reported worldwide (no report in Chinese). Methods Here, we analyzed the clinical data and genetic test results of a Chinese patient with Jansen‐de Vries syndrome admitted in our hospital in May 2019. Results We report a 9‐month‐old boy carrying a pathogenic variant (c.1254_1255del, p.(V419Qfs*14)) in PPM1D exon 5, which can account for his phenotype. Most of his clinical features overlap with the reported phenotype, such as growth retardation, feeding difficulties, constipation, congenital abnormalities (such as atrial septal defect, ventricular septal defect, and patent ductus arteriosus), small hands and feet with broad forehead, low‐set posteriorly rotated ears, wide mouth with thin upper lip and pointed chin; however, he also presented with additional features like hepatomegaly and left inguinal hernia. Conclusion This is the first published case of Jansen‐de Vries syndrome in Chinese population, which will help us to enrich the clinical spectrum of this syndrome.
- Published
- 2020
- Full Text
- View/download PDF
15. Epileptic encephalopathy caused by biallelic mutation in PPM1D: a case report
- Author
-
Hind AlMaghthawi, Marwan Nashabat, and Majid Alfadhel
- Subjects
ppm1d ,phosphatase ,epileptic encephalopathy ,intellectual disability ,Genetics ,QH426-470 - Abstract
Background: PPM1D gene encodes for metal-dependent protein phosphatase. Its function includes the inhibition of some tumor suppressor genes, DNA damage response, and cell cycle control. Germline heterozygous de novo mutations in this gene were reported to cause intellectual disability and hypotonia. Case Presentation: We report a 40-month-old girl with an intractable seizure disorder, microcephaly, and global developmental delay. She had frequent epileptiform discharges on electroencephalography. Molecular investigations showed a homozygous truncating mutation in the PPM1D gene. Both parents were healthy heterozygous carriers. Conclusion: This is the first time in the literature to describe a homozygous biallelic mutation in the PPM1D gene, which resulted in epileptic encephalopathy, microcephaly, and global developmental delay. PPM1D mutations could be inherited as autosomal recessive with asymptomatic heterozygote carriers. [JBCGenetics 2018; 1(1.000): 47-50]
- Published
- 2018
- Full Text
- View/download PDF
16. Protein Phosphatase Magnesium-Dependent 1δ (PPM1D) Expression as a Prognostic Marker in Adult Supratentorial Diffuse Astrocytic and Oligodendroglial Tumors
- Author
-
Hui Jeong Jeong, Chang Gok Woo, Bora Lee, Shin Kwang Khang, Soo Jeong Nam, and Jene Choi
- Subjects
PPM1D ,IDH1 ,Mutation ,Diffuse astrocytic and oligodendroglial tumors ,Supratentorial gliomas ,Molecular marker ,Pathology ,RB1-214 - Abstract
Background Protein phosphatase magnesium-dependent 1δ (PPM1D) is a p53-induced serine/threonine phosphatase, which is overexpressed in various human cancers. A recent study reported that a mutation in the PPM1D gene is associated with poor prognosis in brainstem gliomas. In this study, we evaluated the utility of PPM1D as a prognostic biomarker of adult supratentorial diffuse astrocytic and oligodendroglial tumors. Methods To investigate PPM1D protein expression, mRNA expression, and copy number changes, immunohistochemistry, RNAscope in situ hybridization, and fluorescence in situ hybridization were performed in 84 adult supratentorial diffuse gliomas. We further analyzed clinical characteristics and overall survival (OS) according to PPM1D protein expression, and examined its correlation with other glioma biomarkers such as isocitrate dehydrogenase (IDH) mutation, and p53 expression. Results Forty-six cases (54.8%) were PPM1D-positive. PPM1D expression levels were significantly correlated with PPM1D transcript levels (p= .035), but marginally with PPM1D gene amplification (p=.079). Patients with high-grade gliomas showed a higher frequency of PPM1D expression than those with low-grade gliomas (p
- Published
- 2018
- Full Text
- View/download PDF
17. PPM1D accelerates proliferation and metastasis of osteosarcoma by activating PKP2.
- Author
-
HE, X.-L., XIAO, Q., ZHOU, Z.-P., and HUI, C.-Y.
- Abstract
OBJECTIVE: This project aims to elucidate the diagnostic and prognostic values of PPM1D in osteosarcoma and the molecular mechanism. PATIENTS AND METHODS: PPM1D levels in osteosarcoma and adjacent tissues were detected. Pathological information of included osteosarcoma patients was collected for analyzing the relationship between PPM1D and prognosis of osteosarcoma. Regulatory effects of PPM1D on in vivo and in vitro progressions of osteosarcoma were assessed by generating xenograft model in nude mice and PPM1D knockdown models in MG63 and U2OS cells, respectively. The involvement of PKP2, the target gene of PPM1D in osteosarcoma progression was finally evaluated. RESULTS: PPM1D was upregulated in osteosarcoma tissues than adjacent ones. High level of PPM1D indicated higher risks of distant metastasis and worse prognosis in osteosarcoma. In vivo knockdown of PPM1D contributed to a delay in tumor growth of osteosarcoma in nude mice. PKP2, as the downstream gene targeting PPM1D, was highly expressed in osteosarcoma tissues and positively correlated to PPM1D level. The overexpression of PKP2 was able to abolish the inhibited proliferative and migratory abilities in osteosarcoma cells with PPM1D knockdown. CONCLUSIONS: PPM1D triggers proliferative and migratory abilities of osteosarcoma by positively regulating PKP2, which can be served as an effective diagnostic marker for osteosarcoma in the early phase. [ABSTRACT FROM AUTHOR]
- Published
- 2021
18. Novel truncating variant of PPM1D penultimate exon in a Chinese patient with Jansen‐de Vries syndrome.
- Author
-
Li, Zhuoguang, Du, Caiqi, Zhang, Cai, Zhang, Mini, Ying, Yanqin, Liang, Yan, and Luo, Xiaoping
- Subjects
CHINESE people ,ATRIAL septal defects ,PATENT ductus arteriosus ,VENTRICULAR septal defects ,SYNDROMES ,RECESSIVE genes ,DUCTUS arteriosus - Abstract
Background: Jansen‐de Vries syndrome is a rare autosomal dominant neurodevelopmental disorder caused by pathogenic variants in the last and penultimate exons of the PPM1D gene. It is characterized by delayed psychomotor development, intellectual disability with speech delay, behavioral abnormalities, and dysmorphic features. Up to date, only 17 affected patients have been reported worldwide (no report in Chinese). Methods: Here, we analyzed the clinical data and genetic test results of a Chinese patient with Jansen‐de Vries syndrome admitted in our hospital in May 2019. Results: We report a 9‐month‐old boy carrying a pathogenic variant (c.1254_1255del, p.(V419Qfs*14)) in PPM1D exon 5, which can account for his phenotype. Most of his clinical features overlap with the reported phenotype, such as growth retardation, feeding difficulties, constipation, congenital abnormalities (such as atrial septal defect, ventricular septal defect, and patent ductus arteriosus), small hands and feet with broad forehead, low‐set posteriorly rotated ears, wide mouth with thin upper lip and pointed chin; however, he also presented with additional features like hepatomegaly and left inguinal hernia. Conclusion: This is the first published case of Jansen‐de Vries syndrome in Chinese population, which will help us to enrich the clinical spectrum of this syndrome. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
19. Jansen de Vries syndrome:report of four new patients and review of the literature
- Author
-
Tuiskula, A. (Anna), Rahikkala, E. (Elisa), Kero, A. (Andreina), Haanpää, M. K. (Maria K.), Avela, K. (Kristiina), Tuiskula, A. (Anna), Rahikkala, E. (Elisa), Kero, A. (Andreina), Haanpää, M. K. (Maria K.), and Avela, K. (Kristiina)
- Abstract
Jansen de Vries syndrome (JDVS, OMIM: 617450) is a rare neurodevelopmental disorder associated with hypotonia, behavioral features, high threshold to pain, short stature, ophthalmological abnormalities, dysmorphism and occasionally a structural cardiac condition. It is caused by truncating variants of the last and penultimate exons of PPM1D. So far, 21 patients with JVDS have been reported in the literature. Here, we describe four novel cases of JVDS and review the current literature. Notably, our patients 1, 3 and 4 do not have intellectual disability albeit they have significant developmental difficulties. Thus, the phenotype may span from a classic intellectual disability syndrome to a milder neurodevelopmental disorder. Interestingly, two of our patients have received successful growth hormone treatment. Considering the phenotype of all the known JDVS patients, a cardiological consultation is recommended, as at least 7/25 patients showed a structural cardiac defect. Episodic fever and vomiting may associate with hypoglycemia and may even mimic a metabolic disorder. We also report the first JDVS patient with a mosaic gene defect and a mild neurodevelopmental phenotype.
- Published
- 2023
20. WIP1 dephosphorylation of p27Kip1 Serine 140 destabilizes p27Kip1 and reverses anti-proliferative effects of ATM phosphorylation.
- Author
-
Choi, Byung-Kwon, Fujiwara, Kenichiro, Dayaram, Tajhal, Darlington, Yolanda, Dickerson, Joshua, Goodell, Margaret A., and Donehower, Lawrence A.
- Subjects
DNA repair ,SERINE/THREONINE kinases ,CYCLIN-dependent kinase inhibitors ,SERINE ,DEPHOSPHORYLATION ,PROTEIN stability ,PHOSPHORYLATION ,CYCLIN-dependent kinase inhibitor-2A ,CELL cycle - Abstract
The phosphoinositide-3-kinase like kinases (PIKK) such as ATM and ATR play a key role in initiating the cellular DNA damage response (DDR). One key ATM target is the cyclin-dependent kinase inhibitor p27
Kip1 that promotes G1 arrest. ATM activates p27Kip1 -induced arrest in part through phosphorylation of p27Kip1 at Serine 140. Here we show that this site is dephosphorylated by the type 2C serine/threonine phosphatase, WIP1 (Wildtype p53-Induced Phosphatase-1), encoded by the PPM1D gene. WIP1 has been shown to dephosphorylate numerous ATM target sites in DDR proteins, and its overexpression and/or mutation has often been associated with oncogenesis. We demonstrate that wildtype, but not phosphatase-dead WIP1, efficiently dephosphorylates p27Kip1 Ser140 both in vitro and in cells and that this dephosphorylation is sensitive to the WIP1-specific inhibitor GSK 2830371. Increased expression of wildtype WIP1 reduces stability of p27Kip1 while increased expression of similar amounts of phosphatase-dead WIP1 has no effect on p27Kip1 protein stability. Overexpression of wildtype p27Kip1 reduces cell proliferation and colony forming capability relative to the S140A (constitutively non-phosphorylated) form of p27. Thus, WIP1 plays a significant role in homeostatic modulation of p27Kip1 activity following activation by ATM. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
21. Sensitivity of Cells with Various Levels of Ppm1d Expression to Classical Chemotherapeutic Drugs for Colorectal Cancer Treatment.
- Author
-
Kochetkova, E. Yu., Grigorash, B. B., and Demidov, O. N.
- Abstract
Mutations and amplifications of the Ppm1d gene encoding Wip1 phosphatase have been found in various tumors. Recent studies have shown that the presence of Wip1 stable mutant after chemotherapy suggests that Wip1 is involved in the resistance of malignant cells to chemotherapeutic drugs. In the present work, we investigated the role of Wip1 in the response of colon cancer cells to the antitumor drugs 5-fluorouracil and oxaliplatin. Cell lines with an increased level of Ppm1d expression were obtained with acquired using lentiviral transduction. It has been shown that Wip1 overexpression maintains the cell viability of cells exposed to oxaliplatinum, while deletion of the Ppm1d gene decreases the viability and clonogenicity both under the combined and single exposure to these drugs. These findings suggest that an increased Wip1 level in cancer cells during chemotherapy may contribute to the development of cancer cell resistance to antitumor therapy. Methods aimed at reducing the Wip1 level will increase the efficiency of colon cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
22. Two unrelated girls with intellectual disability associated with a truncating mutation in the PPM1D penultimate exon.
- Author
-
Kuroda, Yukiko, Murakami, Hiroaki, Yokoi, Takayuki, Kumaki, Tatsuro, Enomoto, Yumi, Tsurusaki, Yoshinori, and Kurosawa, Kenji
- Subjects
- *
INTELLECTUAL disabilities , *FRAMESHIFT mutation , *DEVELOPMENTAL delay , *EXONS (Genetics) , *GIRLS , *GENETICS - Abstract
PPM1D truncating mutations in the last and penultimate exons of the gene have been associated with intellectual disability (ID) syndrome. Only 15 affected patients to-date have been reported with mild-to-severe ID, autistic behavior, anxiety and dysmorphic features. Here, we describe the clinical characteristics and underlying genetics of two unrelated girls with moderate developmental delay and dysmorphic features associated with novel mutations in PPM1D exon 5. The dysmorphic features demonstrated by these two patients are consistent with previously reported patients, including broad forehead, thin upper lip, brachydactyly, and hypoplastic nails. We identified a de novo PPM1D mutation in exon 5 of each patient (c.1250_1251insACCA p.V419Tfs*16 and c.1256_1257insCAAG p.S421Qfs*14) by panel sequencing for 4,813 disease-related genes. Both patients also had frameshift mutations (at different positions) that resulted in the same estimated termination codon at 434. These additional reports add to the growing literature on PPM1D -associated ID syndrome and help delineate the clinical phenotype and genetic basis. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
23. Inhibition of protein phosphatase PPM1D enhances retinoic acid-induced differentiation in human embryonic carcinoma cell line.
- Author
-
Ogasawara, Sari, Chuman, Yoshiro, Michiba, Takahiro, Kamada, Rui, Imagawa, Toshiaki, and Sakaguchi, Kazuyasu
- Subjects
- *
PHOSPHOPROTEIN phosphatases , *EXTRACELLULAR signal-regulated kinases , *CELL lines , *CELL differentiation , *ALKALINE phosphatase , *CELLULAR control mechanisms - Abstract
The protein phosphatase PPM1D (Wip1) was originally identified as a p53 target product. Activation of PPM1D through various mechanism promotes the tumorigenic potential of various cancers by suppressing p53 and other DNA damage response proteins. New functions of PPM1D have recently been revealed in physiological processes such as cell differentiation. However, the regulatory mechanisms of signalling pathway to maintain stemness and induce cell differentiation are still unclear. Here we report that PPM1D modulates retinoic acid (RA) signalling. PPM1D knockdown resulted in decreased alkaline phosphatase activity of the human teratocarcinoma cell line NT2/D1. Inhibition of PPM1D-induced cell differentiation and decreased gene expression of the stem cell marker Oct-4 (POU5F1). RA-induced cell differentiation was promoted by reducing PPM1D activity. RA treatment elicited activation of the MEK-ERK pathway and induced rapid and transient activation of the extracellular signal-regulated kinase 1/2 (ERK-1/2). PPM1D dephosphorylated a phosphopeptide with the TEY motif in ERK-1/2 in vitro. Moreover, phosphorylation of ERK-1/2 was facilitated by PPM1D inhibition. Our study shows that PPM1D plays an important role in maintaining the undifferentiation state and a new function in RA-induced ERK regulation and cell differentiation. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
24. The crossroads of cancer therapies and clonal hematopoiesis.
- Author
-
Singh A and Balasubramanian S
- Subjects
- Humans, Clonal Hematopoiesis, Hematopoiesis genetics, Mutation, Receptors, Chimeric Antigen, Neoplasms genetics, Neoplasms therapy
- Abstract
The intricate interplay between Clonal Hematopoiesis (CH) and the repercussions of cancer therapies has garnered significant research focus in recent years. Previously perceived as an age-related phenomenon, CH is now closely linked to inflammation ("Inflammaging") and cancer, impacting leukemogenesis, cancer progression, and treatment responses. This review explores the complex interplay between CH and diverse cancer therapies, including chemotherapy, targeted treatments, radiation, stem cell transplants, CAR-T cell therapy, and immunotherapy, like immune checkpoint inhibitors. Notably, knowledge about post-chemotherapy CH mutation/acquisition has evolved from a de novo incident to more of a clonal selection process. Chemotherapy and radiation exposure, whether therapeutic or environmental, increases CH risk, particularly in genes like TP53 and PPM1D. Environmental toxins, especially in high-risk environments like post-disaster sites or space exploration, are associated with CH. CH affects clinical outcomes in stem cell transplant scenarios, including engraftment, survival, and t-MN development. The presence of CH also alters CAR-T cell therapy responses and impacts the efficacy and toxicity of immunotherapies. Furthermore, specific mutations like DNMT3A and TET2 thrive under inflammatory stress, influencing therapy outcomes and justifying the ongoing tailored interventions in clinical trials. This review underscores the critical need to integrate CH analysis into personalized medicine, enhancing risk assessments and refining treatment strategies. As we progress, multidisciplinary collaboration and comprehensive studies are imperative. Understanding CH's impact, especially concerning genotoxic stressors, will inform screening, surveillance, and early detection strategies, decreasing the risk of therapy-related myeloid neoplasms and revolutionizing cancer treatment paradigms., Competing Interests: Declaration of competing interest The following represents disclosure information provided by authors of this manuscript. Abhay Singh: Advisory Board: Rigel Pharmaceuticals. Suresh Balasubramanian: No potential conflicts of interest., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
25. Diagnosis of Li‐Fraumeni Syndrome: Differentiating TP53 germline mutations from clonal hematopoiesis.
- Author
-
Weber‐Lassalle, Konstantin, Harter, Philipp, Hauke, Jan, Ernst, Corinna, Kommoss, Stefan, Marmé, Frederik, Weber‐Lassalle, Nana, Prieske, Katharina, Dietrich, Dimo, Borde, Julika, Pohl‐Rescigno, Esther, Reuss, Alexander, Ataseven, Beyhan, Engel, Christoph, Stingl, Julia C., Schmutzler, Rita K., and Hahnen, Eric
- Abstract
The Li‐Fraumeni cancer predisposition syndrome (LFS1) presents with a variety of tumor types and the TP53 gene is covered by most diagnostic cancer gene panels. We demonstrate that deleterious TP53 variants identified in blood‐derived DNA of 523 patients with ovarian cancer (AGO‐TR1 trial) were not causal for the patients' ovarian cancer in three out of six TP53‐positive cases. In three out of six patients, deleterious TP53 mutations were identified with low variant fractions in blood‐derived DNA but not in the tumor of the patient seeking advice. The analysis of the TP53 and PPM1D genes, both intimately involved in chemotherapy‐induced and/or age‐related clonal hematopoiesis (CH), in 523 patients and 1,053 age‐matched female control individuals revealed that CH represents a frequent event following chemotherapy, affecting 26 of the 523 patients enrolled (5.0%). Considering that TP53 mutations may arise from chemotherapy‐induced CH, our findings help to avoid false‐positive genetic diagnoses of LFS1. We demonstrate that deleterious TP53 variants identified in blood‐derived DNA of 523 patients with ovarian cancer (AGO‐TR1 trial; NCT02222883) were not causal for the patients' ovarian cancer in 3 of 6 TP53‐positive patients. The analysis of the TP53 and PPM1D genes, both intimately involved in chemotherapy‐induced and/or age‐related clonal hematopoiesis, in all patients and 1,053 age‐matched female control individuals revealed that clonal hematopoiesis represents a frequent event following chemotherapy, affecting 26 of the 523 patients enrolled (5.0%). Considering that TP53 mutations may arise from chemotherapy‐induced clonal hematopoiesis, our findings help to avoid false‐positive genetic diagnoses of Li‐Fraumeni Syndrome. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
26. ATR kinase regulates its attenuation via PPM1D phosphatase recruitment to chromatin during recovery from DNA replication stress signalling.
- Author
-
BHATTACHARYA, DEBADRITA, HIREGANGE, DISHA, and RAO, BASUTHKAR J.
- Subjects
- *
PROTEIN kinases , *EUKARYOTES , *DNA damage , *DNA replication , *KINASE genetics , *CHROMATIN , *PHOSPHATASES - Abstract
In eukaryotes, in response to replication stress, DNA damage response kinase, ATR is activated, whose signalling abrogation leads to cell lethality due to aberrant fork remodelling and excessive origin firing. Here we report that inhibition of ATR kinase activity specifically during replication stress recovery results in persistent ATR signalling, evidenced by the presence of ATR-dependent phosphorylation marks (γH2AX, pChk1 and pRad17) and delayed cell cycle re-entry. Further, such disruption of ATR signalling attenuation leads to double-strand breaks, fork collapse and thereby 'replication catastrophe'. PPM1D phosphatase, a nucleolar localized protein, relocates to chromatin during replication stress and reverts back to nucleolus following stress recovery, under the control of ATR kinase action. Inhibition of ATR kinase activity, specifically during post replication stress, triggers dislodging of the chromatin-bound PPM1D from nucleus to cytoplasm followed by its degradation, thereby leading to persistence of activated ATR marks in the nuclei. Chemical inhibition of PPM1D activity or SiRNA mediated depletion of the protein during post replication stress recovery 'phenocopies' ATR kinase inhibition by failing to attenuate ATR signalling. Collectively, our observations suggest a novel role of ATR kinase in mediating its own signal attenuation via PPM1D recruitment to chromatin as an essential mechanism for restarting the stalled forks, cell-cycle re-entry and cellular recovery from replication stress. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
27. PPM1D is a potential prognostic biomarker and correlates with immune cell infiltration in hepatocellular carcinoma
- Author
-
Xianhai Mao, Sulai Liu, Yu Jiang, Chuang Peng, Zhangtao Yu, Nanhui Yu, Le Wang, Bo Jiang, Mengting Cai, Zhihua Zhang, Yinghui Song, Xiehong Liu, and Lianhong Zou
- Subjects
Aging ,Carcinoma, Hepatocellular ,Phosphatase ,Wip1 ,T-Lymphocytes, Regulatory ,Monocytes ,Immune system ,Biomarkers, Tumor ,medicine ,Humans ,Macrophage ,RNA, Messenger ,prognostic biomarker ,Neoplasm Staging ,business.industry ,Macrophages ,Liver Neoplasms ,Cancer ,hepatocellular carcinoma ,Dendritic Cells ,T-Lymphocytes, Helper-Inducer ,Cell Biology ,PPM1D ,medicine.disease ,M2 Macrophage ,Gene Expression Regulation, Neoplastic ,Protein Phosphatase 2C ,Gene expression profiling ,immune infiltrates ,Hepatocellular carcinoma ,Cancer research ,Immunohistochemistry ,business ,Research Paper - Abstract
Background Protein phosphatase magnesium-dependent 1 delta (PPM1D), also referred to as wild-type p53-induced phosphatase 1 (Wip1) or protein phosphatase 2C delta (PP2Cδ), is an oncogenic nuclear serine/threonine phosphatase belonging to the PP2C family. However, the knowledge regarding PPM1D mRNA expression, tumor immunity, and the prognosis in hepatocellular carcinoma (HCC) is scanty. Methods We analyzed PPM1D, including its expression in both the normal and tumor tissue using the Sangerbox database and Tumor Immune Estimation Resource (TIMER). We evaluated its correlation with prognosis in different tumor types by the Kaplan-Meier plotter and Gene Expression Profiling Interactive Analysis (GEPIA). The correlations between PPM1D and the cancer immune infiltrates were determined using TIMER. The correlations between PPM1D expression and gene marker sets of the immune infiltrates were established by both the TIMER and GEPIA. Immunohistochemistry was performed to detect the expression of Wip1 protein encoded by PPM1D in HCC, and the relationship between Wip1 expression and the prognosis of HCC were analyzed. Results We found out that PPM1D mRNA expression was significantly higher in several human cancers, including HCC, than in the corresponding normal human tissues. The PPM1D mRNA high expression in HCC was significantly correlated with poor prognosis. The expression was associated with progression-free survival (PFS) in multiple HCC patients' cohorts (PFS HR = 1.5, P = 0.0066). This was especially in early stage (stage 1) and AJCC_T 1 of HCC. Besides, PPM1D mRNA expression indicated a positive correlation with tumor-infiltrating Monocytes, tumor-associated macrophages (TAMs), M1 Macrophage, M2 Macrophage, dendritic cells (DCs), T-helper (Th) and Treg. Wip1 was higher in HCC than paracancerous tissue. High expression of Wip1 was associated with poor prognosis of HCC. Conclusion Our findings suggested that PPM1D mRNA is critical in activating tumor immunity. Besides, they implied that PPM1D could be a potential prognostic biomarker for cancer progression. Moreover, it correlated with tumor immune cell infiltration in HCC.
- Published
- 2021
- Full Text
- View/download PDF
28. Development of Antibody-like Proteins Targeting the Oncogenic Ser/Thr Protein Phosphatase PPM1D
- Author
-
Megumi Ikeura, Hiroto Tashiro, Yuka Yamagata, Hikaru Saito, Tamaki Kobayashi, Masataka Mizunuma, Kazuki Yamazaki, Keisuke Baba, Kazuhiro Furukawa, and Yoshiro Chuman
- Subjects
Process Chemistry and Technology ,Chemical Engineering (miscellaneous) ,Bioengineering ,protein phosphatase ,adnectin ,phage display library ,PPM1D ,anti-cancer drug - Abstract
PPM1D, a protein Ser/Thr phosphatase, is overexpressed in various cancers and functions as an oncogenic protein by inactivating the p53 pathway. Therefore, molecules that bind PPM1D are expected to be useful anti-cancer agents. In this study, we constructed a phage display library based on the antibody-like small molecule protein adnectin and screened for PPM1D-specific binding molecules. We identified two adnectins, PMDB-1 and PMD-24, that bind PPM1D specific B-loop and PPM1D430 as targets, respectively. Specificity analyses of these recombinant proteins using other Ser/Thr protein phosphatases showed that these molecules bind to only PPM1D. Expression of PMDB-1 in breast cancer-derived MCF-7 cells overexpressing endogenous PPM1D stabilized p53, indicating that PMDB-1 functions as an inhibitor of PPM1D. Furthermore, MTT assay exhibited that MCF-7 cells expressing PMDB-1 showed inhibition of cell proliferation. These data suggest that the adnectin PMDB-1 identified in this study can be used as a lead compound for anti-cancer drugs targeting intracellular PPM1D.
- Published
- 2022
- Full Text
- View/download PDF
29. Mg2+依赖性蛋白磷酸酶1δ的生物信息学分析.
- Author
-
刘畅 and 刘安
- Abstract
Protein phosphatase magnesium-dependent 1δ(PPM1D) is a potential prognostic marker and therapeutic target for hepatocellular carcinoma. But its carcinogenic mechanism and prognostic value have not been fully elucidated. To gain insightful information of PPM1D protein, bioinformatics methods are applied to analyze the hereditary conservation, tissue expression, subcellular localization, chemical properties, space structure and protein interaction networks of PPM1D protein. The PPM1D protein is comprised of 605 amino acid residues which belong to the PP2C super family. It is a hydrophilic unstable protein without signal peptide and trans-membrane region. It is mainly located in the nucleus and the main secondary structure elements are random coil. It contains several phosphorylation, acetylation, methylation and ubiquitination sites. Interactive proteins with PPM1D are mainly cell cycle checkpoint proteins and cell damage and repair-related proteins.We analyze the relationship between PPM1D protein and cancer and the theoretical basis of PPM1D as the cancer marker, which provide some references for further study of the protein and its participation in signal pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
30. Wip1: A candidate phosphatase for cancer diagnosis and treatment.
- Author
-
Oghabi Bakhshaiesh, Tayebeh, Majidzadeh-A, Keivan, and Esmaeili, Rezvan
- Subjects
- *
CANCER diagnosis , *CANCER treatment , *PHOSPHATASES , *DNA damage , *TUMOR suppressor proteins - Abstract
The critical regulatory mechanisms in numerous cellular pathways including cell survival and DNA damage response mostly depend on phosphorylation and dephosphorylation of proteins. The serine/threonine phosphatase wild-type p53-induced phosphatase 1 (Wip1) is a growth-promoting phosphatase and its numerous downstream targets are important tumor suppressors. Here, we review the Wip1 activity and its relevance to cancer as an oncoprotein. Consecutive investigations about Wip1 and its relation to cancer is critical, as these studies ultimately contribute to the etiology of cancer. A number of innovative studies have recently investigated the importance of Wip1 as a new candidate for cancer diagnosis and prognosis. Accordingly, we discuss the present challenges of using Wip1 as a target for cancer treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
31. De Novo Truncating Mutations in the Last and Penultimate Exons of PPM1D Cause an Intellectual Disability Syndrome.
- Author
-
Jansen, Sandra, Geuer, Sinje, Pfundt, Rolph, Brough, Rachel, Ghongane, Priyanka, Herkert, Johanna C., Marco, Elysa J., Willemsen, Marjolein H., Kleefstra, Tjitske, Hannibal, Mark, Shieh, Joseph T., Lynch, Sally Ann, Flinter, Frances, FitzPatrick, David R., Gardham, Alice, Bernhard, Birgitta, Ragge, Nicola, Newbury-Ecob, Ruth, Bernier, Raphael, and Kvarnung, Malin
- Subjects
- *
INTELLECTUAL disabilities , *EXONS (Genetics) , *GENETIC mutation , *PHOSPHATASES , *CELLULAR signal transduction - Abstract
Intellectual disability (ID) is a highly heterogeneous disorder involving at least 600 genes, yet a genetic diagnosis remains elusive in ∼35%–40% of individuals with moderate to severe ID. Recent meta-analyses statistically analyzing de novo mutations in >7,000 individuals with neurodevelopmental disorders highlighted mutations in PPM1D as a possible cause of ID. PPM1D is a type 2C phosphatase that functions as a negative regulator of cellular stress-response pathways by mediating a feedback loop of p38-p53 signaling, thereby contributing to growth inhibition and suppression of stress-induced apoptosis. We identified 14 individuals with mild to severe ID and/or developmental delay and de novo truncating PPM1D mutations. Additionally, deep phenotyping revealed overlapping behavioral problems (ASD, ADHD, and anxiety disorders), hypotonia, broad-based gait, facial dysmorphisms, and periods of fever and vomiting. PPM1D is expressed during fetal brain development and in the adult brain. All mutations were located in the last or penultimate exon, suggesting escape from nonsense-mediated mRNA decay. Both PPM1D expression analysis and cDNA sequencing in EBV LCLs of individuals support the presence of a stable truncated transcript, consistent with this hypothesis. Exposure of cells derived from individuals with PPM1D truncating mutations to ionizing radiation resulted in normal p53 activation, suggesting that p53 signaling is unaffected. However, a cell-growth disadvantage was observed, suggesting a possible effect on the stress-response pathway. Thus, we show that de novo truncating PPM1D mutations in the last and penultimate exons cause syndromic ID, which provides additional insight into the role of cell-cycle checkpoint genes in neurodevelopmental disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
32. Jansen de Vries syndrome: Report of four new patients and review of the literature.
- Author
-
Tuiskula, Anna, Rahikkala, Elisa, Kero, Andreina, Haanpää, Maria K., and Avela, Kristiina
- Subjects
- *
SHORT stature , *MILD cognitive impairment , *PAIN threshold , *DISABILITIES , *STATURE - Abstract
Jansen de Vries syndrome (JDVS, OMIM: 617450) is a rare neurodevelopmental disorder associated with hypotonia, behavioral features, high threshold to pain, short stature, ophthalmological abnormalities, dysmorphism and occasionally a structural cardiac condition. It is caused by truncating variants of the last and penultimate exons of PPM1D. So far, 21 patients with JVDS have been reported in the literature. Here, we describe four novel cases of JVDS and review the current literature. Notably, our patients 1, 3 and 4 do not have intellectual disability albeit they have significant developmental difficulties. Thus, the phenotype may span from a classic intellectual disability syndrome to a milder neurodevelopmental disorder. Interestingly, two of our patients have received successful growth hormone treatment. Considering the phenotype of all the known JDVS patients, a cardiological consultation is recommended, as at least 7/25 patients showed a structural cardiac defect. Episodic fever and vomiting may associate with hypoglycemia and may even mimic a metabolic disorder. We also report the first JDVS patient with a mosaic gene defect and a mild neurodevelopmental phenotype. • Truncating variants of the penultimate and last exons of PPM1D cause Jansen de Vries syndrome. • The phenotype varies from intellectual disability to milder cognitive impairment. • Some patients with Jansen de Vries syndrome may benefit from growth hormone therapy. • A cardiology consultation would be beneficial for children with this condition. • Jansen de Vries syndrome may mimic a metabolic disorder. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
33. Proteomics and phosphoproteomics profiling in glutamatergic neurons and microglia in an iPSC model of Jansen de Vries Syndrome.
- Author
-
Aguilan JT, Pedrosa E, Dolstra H, Baykara RN, Barnes J, Zhang J, Sidoli S, and Lachman HM
- Abstract
Background: Jansen de Vries Syndrome (JdVS) is a rare neurodevelopmental disorder (NDD) caused by gain-of-function (GOF) truncating mutations in PPM1D exons 5 or 6. PPM1D is a serine/threonine phosphatase that plays an important role in the DNA damage response (DDR) by negatively regulating TP53 (P53). JdVS-associated mutations lead to the formation of a truncated PPM1D protein that retains catalytic activity and has a GOF effect because of reduced degradation. Somatic PPM1D exons 5 and 6 truncating mutations are well-established factors in a number of cancers, due to excessive dephosphorylation and reduced function of P53 and other substrates involved in DDR. Children with JdVS have a variety of neurodevelopmental, psychiatric, and physical problems. In addition, a small fraction has acute neuropsychiatric decompensation apparently triggered by infection or severe non-infectious environmental stress factors., Methods: To understand the molecular basis of JdVS, we developed an induced pluripotent stem cell (iPSC) model system. iPSCs heterozygous for the truncating variant ( PPM1D
+/tr ), were made from a patient, and control lines engineered using CRISPR-Cas9 gene editing. Proteomics and phosphoprotemics analyses were carried out on iPSC-derived glutamatergic neurons and microglia from three control and three PPM1D+/tr iPSC lines. We also analyzed the effect of the TLR4 agonist, lipopolysaccharide, to understand how activation of the innate immune system in microglia could account for acute behavioral decompensation., Results: One of the major findings was the downregulation of POGZ in unstimulated microglia. Since loss-of-function variants in the POGZ gene are well-known causes of autism spectrum disorder, the decrease in PPM1D+/tr microglia suggests this plays a role in the neurodevelopmental aspects of JdVS. In addition, neurons, baseline, and LPS-stimulated microglia show marked alterations in the expression of several E3 ubiquitin ligases, most notably UBR4, and regulators of innate immunity, chromatin structure, ErbB signaling, and splicing. In addition, pathway analysis points to overlap with neurodegenerative disorders., Limitations: Owing to the cost and labor-intensive nature of iPSC research, the sample size was small., Conclusions: Our findings provide insight into the molecular basis of JdVS and can be extrapolated to understand neuropsychiatric decompensation that occurs in subgroups of patients with ASD and other NDDs.- Published
- 2023
- Full Text
- View/download PDF
34. LZAP is a novel Wip1 binding partner and positive regulator of its phosphatase activity in vitro.
- Author
-
Wamsley, J. Jacob, Issaeva, Natalia, An, Hanbing, Lu, Xinyuan, Donehower, Lawrence A., and Yarbrough, Wendell G.
- Published
- 2017
- Full Text
- View/download PDF
35. Identification of PPM1D as an essential Ulk1 phosphatase for genotoxic stress-induced autophagy.
- Author
-
Torii, Satoru, Yoshida, Tatsushi, Arakawa, Satoko, Honda, Shinya, Nakanishi, Akira, and Shimizu, Shigeomi
- Abstract
Autophagy is an evolutionary conserved process that degrades subcellular constituents. Unlike starvation-induced autophagy, the molecular mechanism of genotoxic stress-induced autophagy has not yet been fully elucidated. In this study, we analyze the molecular mechanism of genotoxic stress-induced autophagy and identify an essential role of dephosphorylation of the Unc51-like kinase 1 (Ulk1) at Ser
637 , which is catalyzed by the protein phosphatase 1D magnesium-dependent delta isoform (PPM1D). We show that after exposure to genotoxic stress, PPM1D interacts with and dephosphorylates Ulk1 at Ser637 in a p53-dependent manner. The PPM1D-dependent Ulk1 dephosphorylation triggers Ulk1 puncta formation and induces autophagy. This happens not only in mouse embryonic fibroblasts but also in primary thymocytes, where the genetic ablation of PPM1D reduces the dephosphorylation of Ulk1 at Ser637 , inhibits autophagy, and accelerates apoptosis induced by X-ray irradiation. This acceleration of apoptosis is caused mainly by the inability of the autophagic machinery to degrade the proapoptotic molecule Noxa. These findings indicate that the PPM1D-Ulk1 axis plays a pivotal role in genotoxic stress-induced autophagy. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
36. PPM1D exerts its oncogenic properties in human pancreatic cancer through multiple mechanisms.
- Author
-
Wu, Bo, Guo, Bo-Min, Kang, Jie, Deng, Xian-Zhao, Fan, You-Ben, Zhang, Xiao-Ping, and Ai, Kai-Xing
- Abstract
Protein phosphatase, Mg/Mn dependent, 1D (PPM1D) is emerging as an oncogene by virtue of its negative control on several tumor suppressor pathways. However, the clinical significance of PPM1D in pancreatic cancer (PC) has not been defined. In this study, we determined PPM1D expression in human PC tissues and cell lines and their irrespective noncancerous controls. We subsequently investigated the functional role of PPM1D in the migration, invasion, and apoptosis of MIA PaCa-2 and PANC-1 PC cells in vitro and explored the signaling pathways involved. Furthermore, we examined the role of PPM1D in PC tumorigenesis in vivo. Our results showed that PPM1D is overexpressed in human PC tissues and cell lines and significantly correlated with tumor growth and metastasis. PPM1D promotes PC cell migration and invasion via potentiation of the Wnt/β-catenin pathway through downregulation of apoptosis-stimulating of p53 protein 2 (ASPP2). In contrast to PPM1D, our results showed that ASPP2 is downregulated in PC tissues. Additionally, PPM1D suppresses PC cell apoptosis via inhibition of the p38 MAPK/p53 pathway through both dephosphorylation of p38 MAPK and downregulation of ASPP2. Furthermore, PPM1D promotes PC tumor growth in vivo. Our results demonstrated that PPM1D is an oncogene in PC. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
37. Protein phosphatase magnesium-dependent 1δ is a novel tumor marker and target in hepatocellular carcinoma.
- Author
-
Xu, Zhi, Cao, Chunxiang, Xia, Haiyan, Shi, Shujing, Hong, Lingzhi, Wei, Xiaowei, Gu, Dongying, Bian, Jianmin, Liu, Zijun, Huang, Wenbin, Zhang, Yixin, He, Song, Lee, Nikki, and Chen, Jinfei
- Abstract
Hepatocellular carcinoma (HCC) is a lethal liver malignancy worldwide. In this study, we reported that protein phosphatase magnesium-dependent 1δ (PPM1D) was highly expressed in the majority of HCC cases (approximately 59%) and significantly associated with high serum α-fetoprotein (AFP) level ( P = 0.044). Kaplan- Meier and Cox regression data indicated that PPM1D overexpression was an independent predictor of HCCspecific overall survival (HR, 2.799; 95% CI, 1.346-5.818, P = 0.006). Overexpressing PPM1D promoted cell viability and invasion, whereas RNA interference-mediated knockdown of PPM1D inhibited proliferation, invasion, and migration of cultured HCC cells. In addition, PPM1D suppression by small interfering RNA decreased the tumorigenicity of HCC cells in vivo. Overall, results suggest that PPM1D is a potential prognostic marker and therapeutic target for HCC. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
38. PPM1D Is a Therapeutic Target in Childhood Neural Tumors
- Author
-
Johnsen, Jelena Milosevic, Diana Treis, Susanne Fransson, Gabriel Gallo-Oller, Baldur Sveinbjörnsson, Nina Eissler, Keiji Tanino, Kazuyasu Sakaguchi, Tommy Martinsson, Malin Wickström, Per Kogner, and John Inge
- Subjects
neuroblastoma ,medulloblastoma ,chromosome 17q gain ,p53 ,WIP1 ,PPM1D ,neoplasms - Abstract
Childhood medulloblastoma and high-risk neuroblastoma frequently present with segmental gain of chromosome 17q corresponding to aggressive tumors and poor patient prognosis. Located within the 17q-gained chromosomal segments is PPM1D at chromosome 17q23.2. PPM1D encodes a serine/threonine phosphatase, WIP1, that is a negative regulator of p53 activity as well as key proteins involved in cell cycle control, DNA repair and apoptosis. Here, we show that the level of PPM1D expression correlates with chromosome 17q gain in medulloblastoma and neuroblastoma cells, and both medulloblastoma and neuroblastoma cells are highly dependent on PPM1D expression for survival. Comparison of different inhibitors of WIP1 showed that SL-176 was the most potent compound inhibiting medulloblastoma and neuroblastoma growth and had similar or more potent effects on cell survival than the MDM2 inhibitor Nutlin-3 or the p53 activator RITA. SL-176 monotherapy significantly suppressed the growth of established medulloblastoma and neuroblastoma xenografts in nude mice. These results suggest that the development of clinically applicable compounds inhibiting the activity of WIP1 is of importance since PPM1D activating mutations, genetic gain or amplifications and/or overexpression of WIP1 are frequently detected in several different cancers.
- Published
- 2021
- Full Text
- View/download PDF
39. PPM1D Is a Therapeutic Target in Childhood Neural Tumors
- Author
-
Jelena Milosevic, Diana Treis, Susanne Fransson, Gabriel Gallo-Oller, Baldur Sveinbjörnsson, Nina Eissler, Keiji Tanino, Kazuyasu Sakaguchi, Tommy Martinsson, Malin Wickström, Per Kogner, and John Inge Johnsen
- Subjects
chromosome 17q gain ,p53 ,neuroblastoma ,PPM1D ,WIP1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medulloblastoma ,neoplasms ,RC254-282 ,Article - Abstract
Simple Summary Medulloblastoma and neuroblastoma are childhood tumors of the central nervous system or the peripheral nervous system, respectively. These are the most common and deadly tumors of childhood. A common genetic feature of medulloblastoma and neuroblastoma is frequent segmental gain or amplification of chromosome 17q. Located on chromosome 17q23.2 is PPM1D which encodes WIP1, a phosphatase that acts as a regulator of p53 and DNA repair. Overexpression of WIP1 correlates with poor patient prognosis. We investigated the effects of genetic or pharmacologic inhibition of WIP1 activity and found that medulloblastoma and neuroblastoma cells were strongly dependent on WIP1 expression for survival. We also tested a number of small molecule inhibitors of WIP1 and show that SL-176 was the most effective compound suppressing the growth of medulloblastoma and neuroblastoma in vitro and in vivo. Abstract Childhood medulloblastoma and high-risk neuroblastoma frequently present with segmental gain of chromosome 17q corresponding to aggressive tumors and poor patient prognosis. Located within the 17q-gained chromosomal segments is PPM1D at chromosome 17q23.2. PPM1D encodes a serine/threonine phosphatase, WIP1, that is a negative regulator of p53 activity as well as key proteins involved in cell cycle control, DNA repair and apoptosis. Here, we show that the level of PPM1D expression correlates with chromosome 17q gain in medulloblastoma and neuroblastoma cells, and both medulloblastoma and neuroblastoma cells are highly dependent on PPM1D expression for survival. Comparison of different inhibitors of WIP1 showed that SL-176 was the most potent compound inhibiting medulloblastoma and neuroblastoma growth and had similar or more potent effects on cell survival than the MDM2 inhibitor Nutlin-3 or the p53 activator RITA. SL-176 monotherapy significantly suppressed the growth of established medulloblastoma and neuroblastoma xenografts in nude mice. These results suggest that the development of clinically applicable compounds inhibiting the activity of WIP1 is of importance since PPM1D activating mutations, genetic gain or amplifications and/or overexpression of WIP1 are frequently detected in several different cancers.
- Published
- 2021
40. High Expression of PPM1D Induces Tumors Phenotypically Similar to TP53 Loss-of-Function Mutations in Mice
- Author
-
Milosevic, Jelena, Fransson, Susanne, Gulyas, Miklos, Olsen, Thale K., Gallo-Oller, Gabriel, Treis, Diana, Elfman, Lotta H. M., Wilhelm, Margareta, Martinsson, Tommy, Baryawno, Ninib, Kogner, Per, and Johnsen, John Inge
- Subjects
p53 ,Cancer och onkologi ,adenocarcinoma ,Clinical Laboratory Medicine ,PPM1D ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lymphoma ,Article ,genetically engineered mouse model ,Klinisk laboratoriemedicin ,neuroblastoma ,Cancer and Oncology ,hemic and lymphatic diseases ,RC254-282 - Abstract
Simple Summary Aberrant expression of the PPM1D gene which encodes a phosphatase called WIP1 is frequently observed in cancers of different origins. WIP1 is a negative regulator of the tumor suppressor p53. Improper inactivation of p53 results in genomic instability and can induce neoplastic transformation. We show that overexpression of PPM1D induces tumors in mice similar to cancers harboring p53 mutations. Our results suggest that PPM1D can act as an oncogenic driver by inducing genomic instability, impaired growth arrest, and apoptotic escape that can result in neoplastic transformation and malignant tumor development. Abstract PPM1D is a negative regulator of p53 and genomic aberrations resulting in increased activity of PPM1D have been observed in cancers of different origins, indicating that PPM1D has oncogenic properties. We established a transgenic mouse model overexpressing PPM1D and showed that these mice developed a wide variety of cancers. PPM1D-expressing mice developed tumors phenotypically and genetically similar to tumors in mice with dysfunctional p53. T-cell lymphoblastic lymphoma was the most frequent cancer observed in these mice (55%) followed by adenocarcinomas (24%), leukemia (12%) and other solid tumors including neuroblastoma. Characterization of T-cell lymphomas in mice overexpressing PPM1D demonstrates Pten-deletion and p53-accumulation similar to mice with p53 loss-of-function. Also, Notch1 mutations which are recurrently observed in T-cell acute lymphoblastic lymphoma (T-ALL) were frequently detected in PPM1D-transgenic mice. Hence, PPM1D acts as an oncogenic driver in connection with cellular stress, suggesting that the PPM1D gene status and expression levels should be investigated in TP53 wild-type tumors.
- Published
- 2021
- Full Text
- View/download PDF
41. Novel truncating PPM1D mutation in a patient with intellectual disability.
- Author
-
Porrmann, Joseph, Rump, Andreas, Hackmann, Karl, Di Donato, Nataliya, Kahlert, Anne-Karin, Wagner, Johannes, Jahn, Arne, Eger, Ines, Flury, Monika, Schrock, Evelin, Tzschach, Andreas, and Gieldon, Laura
- Subjects
- *
INTELLECTUAL disabilities , *GENETIC mutation , *SYMPTOMS , *GASTROINTESTINAL diseases , *PHENOTYPES - Abstract
Abstract Truncating mutations in the last and penultimate exons of the PPM1D gene were recently described as a cause for mild to severe intellectual disability in fourteen patients. Feeding difficulties, periods of fever and vomiting as well as a high pain threshold were described as additional characteristic features and the disorder was subsequently termed "intellectual developmental disorder with gastrointestinal difficulties and high pain threshold (IDDGIP)" in the OMIM database (MIM # 617450). Here we report on an additional patient carrying a novel de novo truncating mutation NM_003620.3: c.1535del, p.(Asn512Ilefs*2) in the last exon of PPM1D. While the patient showed features overlapping with the reported phenotype, such as a short stature and small hands and feet, he also presented with additional features like cleft lip and palate and an aberrant right subclavian artery. Notably, the patient did not have any gastrointestinal difficulties or periods of fever, indicating variability of the phenotype of patients with PPM1D mutations. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
42. Wip1 contributes to the adaptation of HepG2 human liver cancer cells to stress hormone-induced DNA damage.
- Author
-
Li, Gaoxiang, Qian, Yazhi, Chen, Yuzhu, Cao, Mingyue, Yang, Xiaozhou, Kong, Dexin, Wang, Guiping, An, Haiyan, Yang, Nan, Huang, Wei, and Liu, Yanyong
- Subjects
- *
DNA repair , *DNA damage , *LIVER cells , *LIVER cancer , *CANCER cells , *CELL death , *DEXAMETHASONE , *PROTEIN kinases - Abstract
Numerous studies have shown that the release of stress hormones resulting from repeated exposure to chronic psychological stress increases DNA damage and promotes tumorigenesis. However, the mechanisms that enable cancerous cells adapt to stress hormone-induced DNA damage and survive remain unclear. The present study aimed to investigate the impact of stress hormones on the survival of liver cancer cells and the underlying mechanism. HepG2 human liver cancer cells were treated with dexamethasone (DEX), epinephrine (EPI) and norepinephrine (NE) and subjected to the testing of DNA damage, cell survival and cell apoptosis by alkaline comet assay, CCK-8 viability assay and flow cytometry, respectively. The protein expression levels of DNA damage response factors were determined by western blotting analysis. The results revealed that treatment of HepG2 cells with DEX, EPI and NE induced DNA damage without affecting cell survival or inducing apoptosis. The protein levels of wild-type p53-induced phosphatase 1 (Wip1), a type 2C family serine/threonine phosphatase, were increased, and the dephosphorylation of DNA damage response factors, including phosphorylated (p-)ataxia-telangiectasia mutated and p-checkpoint kinase 2, occurred following treatment with DEX, EPI and NE. In addition, a cycloheximide chase assay was performed to explore the protein stability under treatment with stress hormones. Compared with vehicle-treated cells, Wip1 exhibited increased protein stability in stress hormone-treated HepG2 cells. Eventually, the depletion of Wip1 using small interfering RNA verified the role of Wip1 in the modulation of stress hormone-induced DNA damage. These findings suggest that cancerous cells likely adapt to stress hormone-induced DNA damage via Wip1 upregulation. The present study provides an insight into the underlying mechanism that links chronic psychological stress with tumor growth and progression. [ABSTRACT FROM AUTHOR]
- Published
- 2023
43. miR-16-5p enhances sensitivity to RG7388 through targeting PPM1D expression (WIP1) in Childhood Acute Lymphoblastic Leukemia.
- Author
-
Zanjirband M, Rahgozar S, and Aberuyi N
- Abstract
Aim: Given the encouraging results of the p53-Mdm2 inhibitor RG7388 in clinical trials and the vital function of miR-16-5p in suppressing cell proliferation, the aim of the present study was to investigate the combined impact of RG7388 and miR-16-5p overexpression on the childhood acute lymphoblastic leukemia (chALL). Methods: miRTarBase and miRDB, along with KEGG and STRING databases, were used to predict miR-16-5p target genes and explore protein-protein interaction networks, respectively. B- and T-lymphoblastic cell lines, in addition to patient primary cells, were treated with RG7388. Ectopic overexpression of miR-16-5p in Nalm6 cell line was induced through cell electroporation and transfection of microRNA mimics was confirmed by qRT-PCR. Cell viability was evaluated using the MTT assay. Western blot analyses were performed to evaluate the effects of RG7388 and miR-16-5p upregulation on the protein levels of p53 and its downstream target genes in chALL cells. Paired sample t-test was employed for statistical analyses. Results: MTT assay showed RG7388-induced cytotoxicity in wild-type p53 Nalm6 cell line and p53 functional patient primary cells. However, CCRF-CEM and p53 non-functional leukemic cells indicated drug resistance. Western blot analyses validated the bioinformatics results, confirming the downregulation of WIP1, p53 stabilization, as well as overexpression of p21
WAF1 and Mdm2 proteins in Nalm6 cells transfected with miR-16-5p. Moreover, enhanced sensitivity to RG7388 was observed in the transfected cells. Conclusion: This is the first study indicating the mechanistic importance of miR-16-5p overexpression in chALL and its inhibitory role in leukemia treatment when combined with the p53-Mdm2 antagonist, RG7388. These findings might be useful for researchers and clinicians to pave the way for better management of chALL., Competing Interests: All authors declared that there are no conflicts of interest., (© The Author(s) 2023.)- Published
- 2023
- Full Text
- View/download PDF
44. PPM1D overexpression predicts poor prognosis in non-small cell lung cancer.
- Author
-
Yang, Hua, Gao, Xiao-Yu, Li, Ping, and Jiang, Ting-Shu
- Abstract
It has been reported that protein phosphatase, Mg/Mn dependent, 1D (PPM1D) plays an important role in cancer tumorigenesis. However, the clinical and functional significance of PPM1D expression has not been characterized previously in non-small cell lung cancer (NSCLC). The purpose of this study was to assess PPM1D expression and to explore its contribution to NSCLC. We examined PPM1D messenger RNA (mRNA) expression in 53 NSCLC tissues and matched adjacent noncancerous tissues by quantitative reverse transcription PCR (qRT-PCR). Furthermore, the PPM1D protein expression was analyzed by immunohistochemistry in 157 NSCLC samples. The relationship between PPM1D expression and clinicopathological features was analyzed by appropriate statistics. Kaplan-Meier analysis and Cox proportional hazards regression models were used to investigate the correlation between PPM1D expression and prognosis of NSCLC patients. The relative mRNA expression of PPM1D was significantly elevated in NSCLC tissues as compared with adjacent noncancerous tissues ( P < 0.001). The high expression of PPM1D in NSCLC tissues was significantly correlated with tumor grade ( P = 0.006), tumor size ( P = 0.017), clinical stage ( P = 0.001), and lymph node metastases ( P = 0.002). Kaplan-Meier survival analysis revealed that high PPM1D expression correlated with poor prognosis of NSCLC patients ( P < 0.001). Multivariate analysis showed that PPM1D expression was an independent prognostic marker for overall survival of NSCLC patients. In conclusion, PPM1D plays an important role in the progression of NSCLC. PPM1D may potentially be used as an independent biomarker for the prognostic evaluation of NSCLC. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
45. Gynecologic Cancer InterGroup (GCIG) Consensus Review for Clear Cell Carcinoma of the Ovary.
- Author
-
Okamoto, Aikou, Glasspool, Rosalind M., Mabuchi, Seiji, Matsumura, Noriomi, Nomura, Hiroyuki, Itamochi, Hiroaki, Takano, Masashi, Takano, Tadao, Susumu, Nobuyuki, Aoki, Daisuke, Konishi, Ikuo, Covens, Alan, Ledermann, Jonathan, Mezzazanica, Delia, Steer, Christopher, Millan, David, Mcneish, Iain A., Pfisterer, Jacobus, Kang, Sokbom, and Gladieff, Laurence
- Abstract
Clear cell carcinoma of the ovary (CCC) is a histologic subtype of epithelial ovarian cancer with a distinct clinical behavior. There are marked geographic differences in the prevalence of CCC. The CCC is more likely to be detected at an early stage than high-grade serous cancers, and when confined within the ovary, the prognosis is good. However, advanced disease is associated with a very poor prognosis and resistance to standard treatment. Cytoreductive surgery should be performed for patients with stage II, III, or IV disease. An international phase III study to compare irinotecan/cisplatin and paclitaxel/carboplatin as adjuvant chemotherapy for stage IIV CCC has completed enrollment (GCIG/JGOG3017). Considering the frequent PIK3CA mutation in CCC, dual inhibitors targeting PI3K, AKT in the mTOR pathway, are promising. Performing these trials and generating the evidence will require considerable international collaboration. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
46. Targeting the PPM1D phenotype; 2,4-bisarylthiazoles cause highly selective apoptosis in PPM1D amplified cell-lines.
- Author
-
Cheeseman, Matthew D., Faisal, Amir, Rayter, Sydonia, Barbeau, Olivier R., Kalusa, Andrew, Westlake, Maura, Burke, Rosemary, Swan, Michael, van Montfort, Rob, Linardopoulos, Spiros, and Jones, Keith
- Subjects
- *
PHOSPHOPROTEIN phosphatases , *THIAZOLE derivatives , *APOPTOSIS , *GENE amplification , *CELL lines , *ONCOGENES , *HEALTH outcome assessment - Abstract
The metal-dependent phosphatase PPM1D (WIP1) is an important oncogene in cancer, with over-expression of the protein being associated with significantly worse clinical outcomes. In this communication we describe the discovery and optimization of novel 2,4-bisarylthiazoles that phenocopy the knockdown of PPM1D, without inhibiting its phosphatase activity. These compounds cause growth inhibition at nanomolar concentrations, induce apoptosis, activate p53 and display impressive cell-line selectivity. The results demonstrate the potential for targeting phenotypes in drug discovery when tackling challenging targets or unknown mechanisms. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
47. PPM1D is a prognostic marker and therapeutic target in colorectal cancer.
- Author
-
TIAN-SHU PENG, YONG-HENG HE, TIAN NIE, XIANG-DANG HU, HAI-YAN LU, JIAN YI, YUN-FEI SHUAI, and MIN LUO
- Subjects
- *
PHOSPHOPROTEIN phosphatases , *BIOMARKERS , *COLON cancer treatment , *PROTEIN expression , *IMMUNOHISTOCHEMISTRY , *SMALL interfering RNA , *CELL proliferation - Abstract
Protein phosphatase, Mg2+/Mn2+ dependent, 1D (PPM1D) has been associated with carcinogenesis. The present study investigated PPM1D expression as a potential biomarker in colorectal cancer (CRC). PPM1D expression was assessed using immunohistochemistry in 368 patients with CRC. The correlation between PPM1D expression, clinicopathological features and prognosis was analyzed. PPM1D small interfering (si)RNA-induced PPM1D silencing was performed in CRC cell lines to assess the effect of PPM1D on tumor cell proliferation and invasion in vitro. A total of 68.48% (252/368) of the CRC samples displayed high PPM1D expression. By contrast, only 9.24% (34/368) of the matched non-cancerous tissue samples exhibited high PPM1D expression. High PPM1D expression was correlated with node metastasis (P=0.0024), distant metastasis (P<0.001) and TNM stage (P=0.0016). Kaplan-Meier survival analysis revealed that patients with low PPM1D expression had significantly longer survival than those with high PPM1D expression (P=0.012). Moreover, multivariate analyses demonstrated that high PPM1D expression was an independent prognostic factor for overall survival (hazard ratio = 0.24; 95% confidence interval, 0.13-0.86; P=0.004). Furthermore, PPM1D gene silencing was found to significantly reduce the proliferation and invasion of CRC cells in vitro. These findings suggest a role for PPM1D as a prognostic marker and potential therapeutic target in CRC. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
48. Prognostic value of PPM1D in 800 gastric cancer patients.
- Author
-
DAN MA, CHAO-JUN ZHANG, ZU-LIN CHEN, and HUA YANG
- Subjects
- *
PHOSPHOPROTEIN phosphatases , *STOMACH cancer patients , *MAGNESIUM in the body , *GENE expression , *MULTIVARIATE analysis , *IMMUNOHISTOCHEMISTRY - Abstract
Protein phosphatase magnesium-dependent 1 delta (PPM1D) has recently been associated with tumor biology. However, the expression pattern and clinical significance of PPM1D in gastric cancer (GC) have yet to be elucidated. The present study aimed to investigate the clinical and prognostic significance of PPM1D in GC. PPM1D expression was assessed in 800 patients with GC using immunohistochemistry and tissue samples were divided into a PPM1D-positive and -negative group. The correlation between PPM1D expression and clinicopathological parameters or prognosis was investigated. PPM1D expression was significantly higher in GC tissue than in adjacent normal tissue (48 versus 9.5%; P<0.001). PPM1D positivity was significantly correlated with nodal status, distant metastasis and vascular invasion. Survival analysis indicated that the five-year survival rate in the PPM1D-positive group was significantly lower than that in the PPM1D-negative group (41 versus 72%; p=0.0012). Furthermore, the association between PPM1D positivity and survival rate was still significant following regulation of other prognostic markers in a multivariate analysis [hazard ratio (HR), 6.572; 95% confidence interval (CI), 3.108-13.471; P=0.0018]. In conclusion, the present study suggested that PPM1D positivity is associated with GC invasion and metastasis, and proposed PPM1D positivity as an indicator of unfavorable prognosis in patients with GC. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
49. The role of PPM1D in cancer and advances in studies of its inhibitors
- Author
-
Yong Wu, Jieqing Li, Wenhong Deng, Jay Vadgama, Denise Jimenez-Tapia, Wei Cao, Kimberly Dorrah, Brando Arriaga, Zhaoxia Gao, and Qiongyu Hao
- Subjects
0301 basic medicine ,p53 ,Inhibitor ,Phosphatase ,Antineoplastic Agents ,RM1-950 ,Pyruvate dehydrogenase phosphatase ,Selective inhibition ,Article ,law.invention ,Serine ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,law ,Neoplasms ,Breast Cancer ,Genetics ,Medicine ,2.1 Biological and endogenous factors ,Oncology & Carcinogenesis ,Aetiology ,Gene ,Cancer ,Pharmacology ,Neoplastic ,Molecular Structure ,business.industry ,General Medicine ,Pharmacology and Pharmaceutical Sciences ,medicine.disease ,PPM1D ,Gene Expression Regulation, Neoplastic ,Protein Phosphatase 2C ,030104 developmental biology ,Gene Expression Regulation ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Cancer research ,Suppressor ,Therapeutics. Pharmacology ,Development of treatments and therapeutic interventions ,business - Abstract
A greater understanding of factors causing cancer initiation, progression and evolution is of paramount importance. Among them, the serine/threonine phosphatase PPM1D, also referred to as wild-type p53-induced phosphatase 1 (Wip1) or protein phosphatase 2C delta (PP2Cδ), is emerging as an important oncoprotein due to its negative regulation on a number of crucial cancer suppressor pathways. Initially identified as a p53-regulated gene, PPM1D has been afterwards found amplified and more recently mutated in many human cancers such as breast cancer. The latest progress in this field further reveals that selective inhibition of PPM1D to delay tumor onset or reduce tumor burden represents a promising anti-cancer strategy. Here, we review the advances in the studies of the PPM1D activity and its relevance to various cancers, and recent progress in development of PPM1D inhibitors and discuss their potential application in cancer therapy. Consecutive research on PPM1D and its relationship with cancer is essential, as it ultimately contributes to the etiology and treatment of cancer.
- Published
- 2020
50. Genetic variants and mutations of PPM1D control the response to DNA damage.
- Author
-
Dudgeon, Crissy, Shreeram, Sathyavageeswaran, Kan Tanoue, Mazur, Sharlyn J., Sayadi, Ahmed, Robinson, Robert C., Appella, Ettore, and Bulavin, Dmitry V.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.