259 results on '"Ari Elson"'
Search Results
2. Sorting Nexin 10 as a Key Regulator of Membrane Trafficking in Bone-Resorbing Osteoclasts: Lessons Learned From Osteopetrosis
- Author
-
Ari Elson, Merle Stein, Grace Rabie, Maayan Barnea-Zohar, Sabina Winograd-Katz, Nina Reuven, Moran Shalev, Juraj Sekeres, Moien Kanaan, Jan Tuckermann, and Benjamin Geiger
- Subjects
osteoclast ,osteopetrosis ,ARO ,sorting nexin ,SNX10 ,bone resorption ,Biology (General) ,QH301-705.5 - Abstract
Bone homeostasis is a complex, multi-step process, which is based primarily on a tightly orchestrated interplay between bone formation and bone resorption that is executed by osteoblasts and osteoclasts (OCLs), respectively. The essential physiological balance between these cells is maintained and controlled at multiple levels, ranging from regulated gene expression to endocrine signals, yet the underlying cellular and molecular mechanisms are still poorly understood. One approach for deciphering the mechanisms that regulate bone homeostasis is the characterization of relevant pathological states in which this balance is disturbed. In this article we describe one such “error of nature,” namely the development of acute recessive osteopetrosis (ARO) in humans that is caused by mutations in sorting nexin 10 (SNX10) that affect OCL functioning. We hypothesize here that, by virtue of its specific roles in vesicular trafficking, SNX10 serves as a key selective regulator of the composition of diverse membrane compartments in OCLs, thereby affecting critical processes in the sequence of events that link the plasma membrane with formation of the ruffled border and with extracellular acidification. As a result, SNX10 determines multiple features of these cells either directly or, as in regulation of cell-cell fusion, indirectly. This hypothesis is further supported by the similarities between the cellular defects observed in OCLs form various models of ARO, induced by mutations in SNX10 and in other genes, which suggest that mutations in the known ARO-associated genes act by disrupting the same plasma membrane-to-ruffled border axis, albeit to different degrees. In this article, we describe the population genetics and spread of the original arginine-to-glutamine mutation at position 51 (R51Q) in SNX10 in the Palestinian community. We further review recent studies, conducted in animal and cellular model systems, that highlight the essential roles of SNX10 in critical membrane functions in OCLs, and discuss possible future research directions that are needed for challenging or substantiating our hypothesis.
- Published
- 2021
- Full Text
- View/download PDF
3. Role of OSCAR Signaling in Osteoclastogenesis and Bone Disease
- Author
-
Iva R. Nedeva, Mattia Vitale, Ari Elson, Judith A. Hoyland, and Jordi Bella
- Subjects
osteoclastogenesis ,osteoclast-associated receptor ,OSCAR ,bone remodeling ,bone disease ,collagen ,Biology (General) ,QH301-705.5 - Abstract
Formation of mature bone-resorbing cells through osteoclastogenesis is required for the continuous remodeling and repair of bone tissue. In aging and disease this process may become aberrant, resulting in excessive bone degradation and fragility fractures. Interaction of receptor-activator of nuclear factor-κB (RANK) with its ligand RANKL activates the main signaling pathway for osteoclastogenesis. However, compelling evidence indicates that this pathway may not be sufficient for the production of mature osteoclast cells and that co-stimulatory signals may be required for both the expression of osteoclast-specific genes and the activation of osteoclasts. Osteoclast-associated receptor (OSCAR), a regulator of osteoclast differentiation, provides one such co-stimulatory pathway. This review summarizes our present knowledge of osteoclastogenesis signaling and the role of OSCAR in the normal production of bone-resorbing cells and in bone disease. Understanding the signaling mechanism through this receptor and how it contributes to the production of mature osteoclasts may offer a more specific and targeted approach for pharmacological intervention against pathological bone resorption.
- Published
- 2021
- Full Text
- View/download PDF
4. PTPRJ promotes osteoclast maturation and activity by inhibiting Cbl‐mediated ubiquitination of NFATc1 in late osteoclastogenesis
- Author
-
Moran Shalev, Sergey Kapishnikov, Esther Arman, Merle Stein, Isabelle Royal, Vlad Brumfeld, Ari Elson, Jan Tuckermann, and Yael Cohen-Sharir
- Subjects
Male ,0301 basic medicine ,Regulator ,Osteoclasts ,Receptor, Macrophage Colony-Stimulating Factor ,Protein tyrosine phosphatase ,Biochemistry ,Monocytes ,Bone resorption ,Osteoclast maturation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Multinucleate ,Ubiquitin ,Osteogenesis ,Osteoclast ,medicine ,Animals ,Proto-Oncogene Proteins c-cbl ,Molecular Biology ,Transcription factor ,Cells, Cultured ,NFATC Transcription Factors ,biology ,Chemistry ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,Ubiquitination ,Cell Biology ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Female - Abstract
Bone-resorbing osteoclasts (OCLs) are multinucleated phagocytes, whose central roles in regulating bone formation and homeostasis are critical for normal health and development. OCLs are produced from precursor monocytes in a multistage process that includes initial differentiation, cell-cell fusion, and subsequent functional and morphological maturation; the molecular regulation of osteoclastogenesis is not fully understood. Here, we identify the receptor-type protein tyrosine phosphatase PTPRJ as an essential regulator specifically of OCL maturation. Monocytes from PTPRJ-deficient (JKO) mice differentiate and fuse normally, but their maturation into functional OCLs and their ability to degrade bone are severely inhibited. In agreement, mice lacking PTPRJ throughout their bodies or only in OCLs exhibit increased bone mass due to reduced OCL-mediated bone resorption. We further show that PTPRJ promotes OCL maturation by dephosphorylating the M-CSF receptor (M-CSFR) and Cbl, thus reducing the ubiquitination and degradation of the key osteoclastogenic transcription factor NFATc1. Loss of PTPRJ increases ubiquitination of NFATc1 and reduces its amounts at later stages of osteoclastogenesis, thereby inhibiting OCL maturation. PTPRJ thus fulfills an essential and cell-autonomous role in promoting OCL maturation by balancing between the pro- and anti-osteoclastogenic activities of the M-CSFR and maintaining NFATc1 expression during late osteoclastogenesis.
- Published
- 2021
5. The roles of protein tyrosine phosphatases in bone-resorbing osteoclasts
- Author
-
Ari Elson and Moran Shalev
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Proteases ,Bone disease ,Osteoclasts ,Receptor-Like Protein Tyrosine Phosphatases ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,Cell Growth Processes ,Protein tyrosine phosphatase ,Bone resorption ,03 medical and health sciences ,0302 clinical medicine ,Osteoclast ,medicine ,Animals ,Humans ,PTEN ,Bone Resorption ,Phosphorylation ,Tyrosine ,Molecular Biology ,Cell Proliferation ,biology ,Chemistry ,Protein Tyrosine Phosphatase, Non-Receptor Type 6 ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,PTEN Phosphohydrolase ,Cell Differentiation ,Cell Biology ,Protein-Tyrosine Kinases ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Protein Tyrosine Phosphatases ,Signal Transduction - Abstract
Maintaining the proper balance between osteoblast-mediated production of bone and its degradation by osteoclasts is essential for health. Osteoclasts are giant phagocytic cells that are formed by fusion of monocyte-macrophage precursor cells; mature osteoclasts adhere to bone tightly and secrete protons and proteases that degrade its matrix. Phosphorylation of tyrosine residues in proteins, which is regulated by the biochemically-antagonistic activities of protein tyrosine kinases and protein tyrosine phosphatases (PTPs), is central in regulating the production of osteoclasts and their bone-resorbing activity. Here we review the roles of individual PTPs of the classical and dual-specificity sub-families that are known to support these processes (SHP2, cyt-PTPe, PTPRO, PTP-PEST, CD45) or to inhibit them (SHP1, PTEN, MKP1). Characterizing the functions of PTPs in osteoclasts is essential for complete molecular level understanding of bone resorption and for designing novel therapeutic approaches for treating bone disease.
- Published
- 2019
6. An SNX10-dependent mechanism downregulates fusion between mature osteoclasts
- Author
-
Fadi Thalji, Jan Tuckermann, Merle Stein, Sabina Winograd-Katz, Benjamin Geiger, Esther Arman, Moien Kanaan, Moran Shalev, Ari Elson, Lee Roth, Nina Reuven, Ofra Golani, and Maayan Barnea-Zohar
- Subjects
Mutant ,Osteoclasts ,Biology ,medicine.disease_cause ,Bone resorption ,03 medical and health sciences ,Myoblast fusion ,Mice ,0302 clinical medicine ,Osteoclast ,medicine ,Animals ,Bone Resorption ,Bone ,Cell fusion ,Sorting Nexins ,030304 developmental biology ,0303 health sciences ,Mutation ,Invadopodia and Podosomes ,Osteopetrosis ,Cell Biology ,medicine.disease ,Cell biology ,Sorting nexin ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,SNX10 ,Research Article - Abstract
Homozygosity for the R51Q mutation in sorting nexin 10 (SNX10) inactivates osteoclasts (OCLs) and induces autosomal recessive osteopetrosis in humans and in mice. We show here that the fusion of wild-type murine monocytes to form OCLs is highly regulated, and that its extent is limited by blocking fusion between mature OCLs. In contrast, monocytes from homozygous R51Q SNX10 mice fuse uncontrollably, forming giant dysfunctional OCLs that can become 10- to 100-fold larger than their wild-type counterparts. Furthermore, mutant OCLs display reduced endocytotic activity, suggesting that their deregulated fusion is due to alterations in membrane homeostasis caused by loss of SNX10 function. This is supported by the finding that the R51Q SNX10 protein is unstable and exhibits altered lipid-binding properties, and is consistent with a key role for SNX10 in vesicular trafficking. We propose that OCL size and functionality are regulated by a cell-autonomous SNX10-dependent mechanism that downregulates fusion between mature OCLs. The R51Q mutation abolishes this regulatory activity, leading to excessive fusion, loss of bone resorption capacity and, consequently, to an osteopetrotic phenotype in vivo. This article has an associated First Person interview with the joint first authors of the paper., Summary: Fusion of monocytes to become bone-resorbing osteoclasts is limited by an SNX10-dependent cell-autonomous mechanism. Loss of SNX10 function deregulates fusion and generates giant inactive osteoclasts.
- Published
- 2021
7. Role of OSCAR Signaling in Osteoclastogenesis and Bone Disease
- Author
-
Jordi Bella, Judith A. Hoyland, Ari Elson, Mattia Vitale, and Iva R. Nedeva
- Subjects
collagen ,musculoskeletal diseases ,0301 basic medicine ,Cell signaling ,Bone disease ,OSCAR ,030209 endocrinology & metabolism ,Review ,Biology ,Bone tissue ,Bone resorption ,Bone remodeling ,Cell and Developmental Biology ,03 medical and health sciences ,0302 clinical medicine ,Osteoclast ,medicine ,cell signaling ,lcsh:QH301-705.5 ,osteoclastogenesis ,bone remodeling ,Cell Biology ,medicine.disease ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,RANKL ,biology.protein ,bone disease ,osteoclast-associated receptor ,Signal transduction ,Developmental Biology - Abstract
Formation of mature bone-resorbing cells through osteoclastogenesis is required for the continuous remodeling and repair of bone tissue. In aging and disease this process may become aberrant, resulting in excessive bone degradation and fragility fractures. Interaction of receptor-activator of nuclear factor-κB (RANK) with its ligand RANKL activates the main signaling pathway for osteoclastogenesis. However, compelling evidence indicates that this pathway may not be sufficient for the production of mature osteoclast cells and that co-stimulatory signals may be required for both the expression of osteoclast-specific genes and the activation of osteoclasts. Osteoclast-associated receptor (OSCAR), a regulator of osteoclast differentiation, provides one such co-stimulatory pathway. This review summarizes our present knowledge of osteoclastogenesis signaling and the role of OSCAR in the normal production of bone-resorbing cells and in bone disease. Understanding the signaling mechanism through this receptor and how it contributes to the production of mature osteoclasts may offer a more specific and targeted approach for pharmacological intervention against pathological bone resorption.
- Published
- 2021
8. Unravelling the effect of an osteopetrotic Snx10 mutation on bone, muscle and other tissues
- Author
-
Benjamin Geiger, Polina Stepensky, F Thalji, Maayan Barnea, Merle Stein, Moien Kanaan, Ari Elson, Jan Tuckermann, Hila Elinav, Sabina Winograd-Katz, Esther Arman, Moran Shalev, and Ori Brenner
- Subjects
Mutation (genetic algorithm) ,Biology ,Molecular biology - Published
- 2019
9. Kinetic modeling of DUSP regulation in Herceptin-resistant HER2-positive breast cancer
- Author
-
Petronela Buiga, Lydia Tabernero, Jean-Marc Schwartz, and Ari Elson
- Subjects
Drug ,lcsh:QH426-470 ,MAP Kinase Signaling System ,Receptor, ErbB-2 ,media_common.quotation_subject ,Phosphatase ,Breast Neoplasms ,Biology ,Article ,Antineoplastic Agents, Immunological ,breast cancer ,Breast cancer ,Herceptin ,Trastuzumab ,Cell Line, Tumor ,Dual-specificity phosphatase ,Genetics ,medicine ,Humans ,skin and connective tissue diseases ,neoplasms ,Gene ,Genetics (clinical) ,media_common ,kinetic model ,Models, Theoretical ,medicine.disease ,lcsh:Genetics ,HEK293 Cells ,Drug Resistance, Neoplasm ,Cancer research ,biology.protein ,Female ,Signal transduction ,dual-specificity phosphatases ,Tyrosine kinase ,medicine.drug - Abstract
Background: HER2 (human epidermal growth factor 2)-positive breast cancer is an aggressive type of breast cancer characterized by the overexpression of the receptor-type protein tyrosine kinase HER2 or amplification of the HER2 gene. It is commonly treated by the drug trastuzumab (Herceptin), but resistance to its action frequently develops and limits its therapeutic benefit. Dual-specificity phosphatases (DUSPs) were previously highlighted as central regulators of HER2 signaling, therefore, understanding their role is crucial to designing new strategies to improve the efficacy of Herceptin treatment. We investigated whether inhibiting certain DUSPs re-sensitized Herceptin-resistant breast cancer cells to the drug. We built a series of kinetic models incorporating the key players of HER2 signaling pathways and simulating a range of inhibition intensities. The simulation results were compared to live tumor cells in culture, and showed good agreement with the experimental analyses. In particular, we observed that Herceptin-resistant DUSP16-silenced breast cancer cells became more responsive to the drug when treated for 72 h with Herceptin, showing a decrease in resistance, in agreement with the model predictions. Overall, we showed that the kinetic modeling of signaling pathways is able to generate predictions that assist experimental research in the identification of potential targets for cancer treatment.
- Published
- 2019
10. Modelling the role of dual specificity phosphatases in Herceptin resistant breast cancer cell lines
- Author
-
Jean-Marc Schwartz, Lydia Tabernero, Petronela Buiga, and Ari Elson
- Subjects
0301 basic medicine ,Systems biology ,Antineoplastic Agents ,Breast Neoplasms ,Drug resistance ,Models, Biological ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Structural Biology ,Trastuzumab ,Cell Line, Tumor ,Dual-specificity phosphatase ,medicine ,Humans ,skin and connective tissue diseases ,Gene ,Cell Proliferation ,biology ,Mechanism (biology) ,Cell growth ,Systems Biology ,Organic Chemistry ,Cancer ,medicine.disease ,Up-Regulation ,Gene Expression Regulation, Neoplastic ,Computational Mathematics ,030104 developmental biology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Dual-Specificity Phosphatases ,medicine.drug - Abstract
BackgroundBreast cancer remains the most lethal type of cancer for women. A significant proportion of breast cancer cases are characterised by overexpression of the human epidermal growth factor receptor 2 protein (HER2). These cancers are commonly treated by Herceptin (Trastuzumab), but resistance to drug treatment frequently develops in tumour cells. Dual-specificity phosphatases (DUSPs) are thought to play a role in the mechanism of resistance, since some of them were reported to be overexpressed in tumours resistant to Herceptin.ResultsWe used a systems biology approach to investigate how DUSP overexpression could favour cell proliferation and to predict how this mechanism could be reversed by targeted inhibition of selected DUSPs. We measured the expression of 20 DUSP genes in two breast cancer cell lines following long-term (6 months) exposure to Herceptin, after confirming that these cells had become resistant to the drug. We constructed several Boolean models including specific substrates of each DUSP, and showed that our models correctly account for resistance when overexpressed DUSPs were kept activated. We then simulated inhibition of both individual and combinations of DUSPs, and determined conditions under which the resistance could be reversed.ConclusionsThese results show how a combination of experimental analysis and modelling help to understand cell survival mechanisms in breast cancer tumours, and crucially enable us to generate testable predictions potentially leading to new treatments of resistant tumours.
- Published
- 2019
11. Phosphorylation of the phosphatase PTPROt at Tyr
- Author
-
Lee, Roth, Jean, Wakim, Elad, Wasserman, Moran, Shalev, Esther, Arman, Merle, Stein, Vlad, Brumfeld, Cari A, Sagum, Mark T, Bedford, Jan, Tuckermann, and Ari, Elson
- Subjects
Mice, Knockout ,Mice, 129 Strain ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,Osteoclasts ,Bone and Bones ,Mice, Inbred C57BL ,HEK293 Cells ,src-Family Kinases ,Animals ,Humans ,Tyrosine ,Bone Resorption ,Phosphorylation ,Cells, Cultured ,Signal Transduction - Abstract
Bone resorption by osteoclasts is essential for bone homeostasis. The kinase Src promotes osteoclast activity and is activated in osteoclasts by the receptor-type tyrosine phosphatase PTPROt. In other contexts, however, PTPROt can inhibit Src activity. Through in vivo and in vitro experiments, we show that PTPROt is bifunctional and can dephosphorylate Src both at its inhibitory residue Tyr
- Published
- 2019
12. Phosphorylation of the phosphatase PTPROt at Tyr 399 is a molecular switch that controls osteoclast activity and bone mass in vivo
- Author
-
Mark T. Bedford, Ari Elson, Jan Tuckermann, Esther Arman, Moran Shalev, Jean Wakim, Merle Stein, Vlad Brumfeld, Cari A. Sagum, Elad Wasserman, and Lee Roth
- Subjects
0303 health sciences ,biology ,Chemistry ,Kinase ,Phosphatase ,Cell Biology ,Protein tyrosine phosphatase ,Biochemistry ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Osteoclast ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Phosphorylation ,GRB2 ,Tyrosine ,Molecular Biology ,030304 developmental biology ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Bone resorption by osteoclasts is essential for bone homeostasis. The kinase Src promotes osteoclast activity and is activated in osteoclasts by the receptor-type tyrosine phosphatase PTPROt. In other contexts, however, PTPROt can inhibit Src activity. Through in vivo and in vitro experiments, we show that PTPROt is bifunctional and can dephosphorylate Src both at its inhibitory residue Tyr 527 and its activating residue Tyr 416 . Whereas wild-type and PTPROt knockout mice exhibited similar bone masses, mice in which a putative C-terminal phosphorylation site, Tyr 399 , in endogenous PTPROt was replaced with phenylalanine had increased bone mass and reduced osteoclast activity. Osteoclasts from the knock-in mice also showed reduced Src activity. Experiments in cultured cells and in osteoclasts derived from both mouse strains demonstrated that the absence of phosphorylation at Tyr 399 caused PTPROt to dephosphorylate Src at the activating site pTyr 416 . In contrast, phosphorylation of PTPROt at Tyr 399 enabled PTPROt to recruit Src through Grb2 and to dephosphorylate Src at the inhibitory site Tyr 527 , thus stimulating Src activity. We conclude that reversible phosphorylation of PTPROt at Tyr 399 is a molecular switch that selects between its opposing activities toward Src and maintains a coherent signaling output, and that blocking this phosphorylation event can induce physiological effects in vivo. Because most receptor-type tyrosine phosphatases contain potential phosphorylation sites at their C termini, we propose that preventing phosphorylation at these sites or its consequences may offer an alternative to inhibiting their catalytic activity to achieve therapeutic benefit.
- Published
- 2019
13. Protein tyrosine phosphatase alpha inhibits hypothalamic leptin receptor signaling and regulates body weight in vivo
- Author
-
Hilla Knobler, Ari Elson, Ivo Spiegel, Yael Kuperman, Yael Cohen-Sharir, Daniella Apelblat, Jeroen den Hertog, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
0301 basic medicine ,Leptin ,Male ,Receptor-Like Protein Tyrosine Phosphatases ,Protein tyrosine phosphatase ,Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics ,Biochemistry ,Mice ,0302 clinical medicine ,Physical Conditioning, Animal/physiology ,Obesity/metabolism ,Receptors ,Phosphorylation ,Non-U.S. Gov't ,Adiposity ,Mice, Knockout ,Chemistry ,Kinase ,Research Support, Non-U.S. Gov't ,Phosphorylation/physiology ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,digestive, oral, and skin physiology ,Physical Conditioning ,Hypothalamus ,Receptors, Leptin ,Receptors, Leptin/metabolism ,Female ,hormones, hormone substitutes, and hormone antagonists ,Biotechnology ,Signal Transduction ,medicine.medical_specialty ,Knockout ,Phosphatase ,Class 4/genetics ,Janus Kinase 2/metabolism ,Research Support ,Adiposity/physiology ,03 medical and health sciences ,Animal/physiology ,In vivo ,Internal medicine ,Physical Conditioning, Animal ,Genetics ,medicine ,Journal Article ,Animals ,Obesity ,Molecular Biology ,Leptin receptor ,Hypothalamus/metabolism ,Signal Transduction/physiology ,Body Weight ,Leptin/metabolism ,Janus Kinase 2 ,030104 developmental biology ,Endocrinology ,Body Weight/physiology ,030217 neurology & neurosurgery - Abstract
Understanding how body weight is regulated at the molecular level is essential for treating obesity. We show that female mice genetically lacking protein tyrosine phosphatase (PTP) receptor type α (PTPRA) exhibit reduced weight and adiposity and increased energy expenditure, and are more resistant to diet-induced obesity than matched wild-type control mice. These mice also exhibit reduced levels of circulating leptin and are leptin hypersensitive, suggesting that PTPRA inhibits leptin signaling in the hypothalamus. Male and female PTPRA-deficient mice fed a high-fat diet were leaner and displayed increased metabolic rates and lower circulating leptin levels, indicating that the effects of loss of PTPRA persist in the obese state. Molecularly, PTPRA down-regulates leptin receptor signaling by dephosphorylating the receptor-associated kinase JAK2, with which the phosphatase associates constitutively. In contrast to the closely related tyrosine phosphatase ε, leptin induces only weak phosphorylation of PTPRA at its C-terminal regulatory site Y789, and this does not affect the activity of PTPRA toward JAK2. PTPRA is therefore an inhibitor of hypothalamic leptin signaling in vivo and may prevent premature activation of leptin signaling, as well as return signaling to baseline after exposure to leptin.-Cohen-Sharir, Y., Kuperman, Y., Apelblat, D., den Hertog, J., Spiegel, I., Knobler, H., Elson, A. Protein tyrosine phosphatase alpha inhibits hypothalamic leptin receptor signaling and regulates body weight in vivo.
- Published
- 2019
14. Massive osteopetrosis caused by non-functional osteoclasts in R51Q SNX10 mutant mice
- Author
-
Maayan Barnea-Zohar, Sabina Winograd-Katz, Polina Stepensky, Jennifer Gerstung, Fadi Thalji, Merle Stein, Hila Elinav, Jan Tuckermann, Moien Kanaan, Benjamin Geiger, Esther Arman, Ari Elson, Ori Brenner, and Moran Shalev
- Subjects
0301 basic medicine ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Mutant ,Osteoclasts ,030209 endocrinology & metabolism ,Biology ,medicine.disease_cause ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Osteoclast ,medicine ,Animals ,Secretion ,Sorting Nexins ,Mutation ,Osteopetrosis ,medicine.disease ,Molecular biology ,Sorting nexin ,030104 developmental biology ,medicine.anatomical_structure ,Failure to thrive ,medicine.symptom ,Rare disease - Abstract
The R51Q mutation in sorting nexin 10 (SNX10) was shown to cause a lethal genetic disease in humans, namely autosomal recessive osteopetrosis (ARO). We describe here the first R51Q SNX10 knock-in mouse model and show that mice homozygous for this mutation exhibit massive, early-onset, and widespread osteopetrosis. The mutant mice exhibit multiple additional characteristics of the corresponding human disease, including stunted growth, failure to thrive, missing or impacted teeth, occasional osteomyelitis, and a significantly-reduced lifespan. Osteopetrosis in this model is the result of osteoclast inactivity that, in turn, is caused by absence of ruffled borders in the mutant osteoclasts and by their inability to secrete protons. These results confirm that the R51Q mutation in SNX10 is a causative factor in ARO and provide a model system for studying this rare disease.
- Published
- 2020
15. R51Q SNX10 induces osteopetrosis by promoting uncontrolled fusion of monocytes to form giant, non-functional osteoclasts
- Author
-
Moran Shalev, Moien Kanaan, Sabina Winograd-Katz, Polina Stepensky, Ori Brenner, Fadi Thalji, Esther Arman, Merle Stein, Hila Elinav, Jan Tuckermann, Benjamin Geiger, Ari Elson, and Maayan Barnea
- Subjects
Sorting nexin ,Mutation ,Precursor cell ,Mutant ,medicine ,Osteopetrosis ,Biology ,medicine.disease ,medicine.disease_cause ,Phenotype ,Bone resorption ,In vitro ,Cell biology - Abstract
SummaryThe molecular mechanisms that regulate fusion of monocytes into functional osteoclasts are virtually unknown. We describe a knock-in mouse model for the R51Q mutation in sorting nexin 10 (SNX10) that exhibits osteopetrosis and related symptoms of patients of autosomal recessive osteopetrosis linked to this mutation. Osteopetrosis arises in homozygous R51Q SNX10 mice due to a unique combination of reduced numbers of osteoclasts that are non-functional. Fusion of mutant monocytes is deregulated and occurs rapidly and continuously to form giant, non-functional osteoclasts. Mutant osteoclasts mature quickly and survive poorly in vitro, possibly accounting for their scarcity in vivo. These cells also exhibit impaired ruffled borders, which are required for bone resorption, providing an additional basis for the osteopetrotic phenotype. More broadly, we propose that the maximal size of osteoclasts is actively determined by a genetically-regulated, cell-autonomous mechanism that limits precursor cell fusion, and for which SNX10 is required.
- Published
- 2018
- Full Text
- View/download PDF
16. Regulation of dual specificity phosphatases in breast cancer during initial treatment with Herceptin: a Boolean model analysis
- Author
-
Jean-Marc Schwartz, Lydia Tabernero, Ari Elson, and Petronela Buiga
- Subjects
0301 basic medicine ,Cell signaling ,Systems biology ,DUSPs ,Antineoplastic Agents ,Breast Neoplasms ,Models, Biological ,03 medical and health sciences ,Breast cancer ,Herceptin ,Structural Biology ,Cell Line, Tumor ,Dual-specificity phosphatase ,medicine ,Cluster Analysis ,Humans ,Initial treatment ,Kinase activity ,skin and connective tissue diseases ,lcsh:QH301-705.5 ,Molecular Biology ,biology ,Research ,Applied Mathematics ,Boolean model ,Trastuzumab ,medicine.disease ,Computer Science Applications ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,lcsh:Biology (General) ,Modeling and Simulation ,Cancer cell ,Cancer research ,biology.protein ,Dual-Specificity Phosphatases ,Function (biology) - Abstract
Background 25% of breast cancer patients suffer from aggressive HER2-positive tumours that are characterised by overexpression of the HER2 protein or by its increased tyrosine kinase activity. Herceptin is a major drug used to treat HER2 positive breast cancer. Understanding the molecular events that occur when breast cancer cells are exposed to Herceptin is therefore of significant importance. Dual specificity phosphatases (DUSPs) are central regulators of cell signalling that function downstream of HER2, but their role in the cellular response to Herceptin is mostly unknown. This study aims to model the initial effects of Herceptin exposure on DUSPs in HER2-positive breast cancer cells using Boolean modelling. Results We experimentally measured expression time courses of 21 different DUSPs between 0 and 24 h following Herceptin treatment of human MDA-MB-453 HER2-positive breast cancer cells. We clustered these time courses into patterns of similar dynamics over time. In parallel, we built a series of Boolean models representing the known regulatory mechanisms of DUSPs and then demonstrated that the dynamics predicted by the models is in agreement with the experimental data. Furthermore, we used the models to predict regulatory mechanisms of DUSPs, where these mechanisms were partially known. Conclusions Boolean modelling is a powerful technique to investigate and understand signalling pathways. We obtained an understanding of different regulatory pathways in breast cancer and new insights on how these signalling pathways are activated. This method can be generalized to other drugs and longer time courses to better understand how resistance to drugs develops in cancer cells over time. Electronic supplementary material The online version of this article (10.1186/s12918-018-0534-5) contains supplementary material, which is available to authorized users.
- Published
- 2018
17. PTPe (RPTPe and Cyt-PTPe)
- Author
-
Ari Elson and Liat Rousso-Noori
- Published
- 2018
18. Adaptor Protein GRB2 Promotes Src Tyrosine Kinase Activation and Podosomal Organization by Protein-tyrosine Phosphatase ϵ in Osteoclasts
- Author
-
Ari Elson, Shawn S.-C. Li, Eynat Finkelshtein, Vidyasiri Vemulapalli, Einat Levy-Apter, and Mark T. Bedford
- Subjects
musculoskeletal diseases ,Mice, 129 Strain ,Integrin ,Osteoclasts ,Protein tyrosine phosphatase ,SH2 domain ,environment and public health ,Biochemistry ,Cell Adhesion ,Animals ,Humans ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Phosphorylation ,Cell adhesion ,Molecular Biology ,GRB2 Adaptor Protein ,Mice, Knockout ,biology ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Cell Differentiation ,Cell Biology ,Cell biology ,enzymes and coenzymes (carbohydrates) ,HEK293 Cells ,src-Family Kinases ,embryonic structures ,cardiovascular system ,biology.protein ,Cancer research ,GRB2 ,biological phenomena, cell phenomena, and immunity ,Cell Adhesion Molecules ,Protein Processing, Post-Translational ,Tyrosine kinase ,Signal Transduction ,Protein Binding ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The non-receptor isoform of protein-tyrosine phosphatase ϵ (cyt-PTPe) supports adhesion of bone-resorbing osteoclasts by activating Src downstream of integrins. Loss of cyt-PTPe reduces Src activity in osteoclasts, reduces resorption of mineralized matrix both in vivo and in cell culture, and induces mild osteopetrosis in young female PTPe KO mice. Activation of Src by cyt-PTPe is dependent upon this phosphatase undergoing phosphorylation at its C-terminal Tyr-638 by partially active Src. To understand how cyt-PTPe activates Src, we screened 73 Src homology 2 (SH2) domains for binding to Tyr(P)-638 of cyt-PTPe. The SH2 domain of GRB2 bound Tyr(P)-638 of cyt-PTPe most prominently, whereas the Src SH2 domain did not bind at all, suggesting that GRB2 may link PTPe with downstream molecules. Further studies indicated that GRB2 is required for activation of Src by cyt-PTPe in osteoclast-like cells (OCLs) in culture. Overexpression of GRB2 in OCLs increased activating phosphorylation of Src at Tyr-416 and of cyt-PTPe at Tyr-638; opposite results were obtained when GRB2 expression was reduced by shRNA or by gene inactivation. Phosphorylation of cyt-PTPe at Tyr-683 and its association with GRB2 are integrin-driven processes in OCLs, and cyt-PTPe undergoes autodephosphorylation at Tyr-683, thus limiting Src activation by integrins. Reduced GRB2 expression also reduced the ability of bone marrow precursors to differentiate into OCLs and reduced the fraction of OCLs in which podosomal adhesion structures assume organization typical of active, resorbing cells. We conclude that GRB2 physically links cyt-PTPe with Src and enables cyt-PTPe to activate Src downstream of activated integrins in OCLs.
- Published
- 2014
19. Stepping out of the shadows: Oncogenic and tumor-promoting protein tyrosine phosphatases
- Author
-
Ari Elson
- Subjects
0301 basic medicine ,Cell ,Phosphatase ,Protein tyrosine phosphatase ,Biology ,medicine.disease_cause ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Neoplasms ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Tyrosine ,Phosphorylation ,Tumor Suppressor Proteins ,Tyrosine phosphorylation ,Cell Biology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cell Transformation, Neoplastic ,chemistry ,Protein Tyrosine Phosphatases ,Carcinogenesis ,Tyrosine kinase - Abstract
Protein tyrosine phosphorylation is critical for proper function of cells and organisms. Phosphorylation is regulated by the concerted but generically opposing activities of tyrosine kinases (PTKs) and tyrosine phosphatases (PTPs), which ensure its proper regulation, reversibility, and ability to respond to changing physiological situations. Historically, PTKs have been associated mainly with oncogenic and pro-tumorigenic activities, leading to the generalization that protein dephosphorylation is anti-oncogenic and hence that PTPs are tumor-suppressors. In many cases PTPs do suppress tumorigenesis. However, a growing body of evidence indicates that PTPs act as dominant oncogenes and drive cell transformation in a number of contexts, while in others PTPs support transformation that is driven by other oncogenes. This review summarizes the known transforming and tumor-promoting activities of the classical, tyrosine specific PTPs and highlights their potential as drug targets for cancer therapy.
- Published
- 2017
20. Production of Osteoclasts for Studying Protein Tyrosine Phosphatase Signaling
- Author
-
Eynat, Finkelshtein, Einat, Levy-Apter, and Ari, Elson
- Subjects
Mice ,HEK293 Cells ,Bone Marrow ,Gene Knockdown Techniques ,Cell Culture Techniques ,Animals ,Humans ,Osteoclasts ,Bone Marrow Cells ,Protein Tyrosine Phosphatases ,Cells, Cultured ,Cell Proliferation ,Signal Transduction - Abstract
Osteoclasts, specialized cells that degrade bone, are key components of the cellular system that regulates and maintains bone homeostasis. Aberrant function of osteoclasts can lead to pathological loss or gain of bone mass, such as in osteopetrosis, osteoporosis, and several types of cancer that metastasize to bone. Phosphorylation of osteoclast proteins on tyrosine residues is critical for formation of osteoclasts and for their proper function and responses to physiological signals. Here we describe preparation and growth of osteoclasts from bone marrow of mice, use of viral vectors to downregulate expression of endogenous proteins and to express exogenous proteins in osteoclasts, and analysis of signaling processes triggered by M-CSF, estrogen, and physical contact with matrix in these cells.
- Published
- 2016
21. The PTPROt tyrosine phosphatase functions as an obligate haploinsufficient tumor suppressor in vivo in B-cell chronic lymphocytic leukemia
- Author
-
Eszter Bakos, Matthias P. Kramer, Idit Shachar, Esther Arman, Shirly Becker-Herman, Ari Elson, and Jean Wakim
- Subjects
0301 basic medicine ,Male ,Cancer Research ,Programmed cell death ,Chronic lymphocytic leukemia ,Protein tyrosine phosphatase ,Haploinsufficiency ,Biology ,Molecular oncology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Growth factor receptor ,immune system diseases ,hemic and lymphatic diseases ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Phosphorylation ,Molecular Biology ,Mice, Knockout ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,breakpoint cluster region ,Cell cycle ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Female ,Signal transduction ,Signal Transduction - Abstract
The tyrosine phosphatase PTPROt is a suggested tumor suppressor (TS) in B-cell chronic lymphocytic leukemia (CLL), and its expression is reduced in this disease. In order to examine how reduced PTPROt expression affects CLL in vivo we induced CLL in PTPROt-targeted mice. Unexpectedly, loss of both Ptprot alleles delayed disease detection and progression and lengthened survival relative to mice carrying two intact alleles, indicating that PTPROt fulfills a novel tumor-promoting role in CLL. Tumor cells from mice lacking PTPROt exhibited reduced B-cell receptor (BCR)-induced signaling, as well as increased apoptosis and autophagy. Inhibition of BCR/Src signaling in CLL cells induced their apoptosis, indicating that these findings are linked causally. These results suggest a cell-autonomous mechanism for the weakened CLL phenotype of PTPROt-deficient mice and uncover non-redundant roles for PTPROt in support of BCR signaling and survival of CLL cells. In contrast, loss of only one Ptprot allele induced earlier detection and progression of CLL and reduced survival, consistent with a tumor-suppressing role for PTPROt. Tumor cells from mice lacking one or both Ptprot allele exhibited increased interleukin-10 (IL-10) expression and signaling, factors known to support CLL; cells lacking one Ptprot alleles exhibited normal BCR signaling and cell death rates. We conclude that loss of one Ptprot allele promotes CLL, most likely by activating IL-10 signaling. Loss of both Ptprot alleles also reduces BCR signaling and increases cell death rates, offsetting the IL-10 effects and reducing the severity of the disease. PTPROt thus functions as an obligate haploinsufficient TS in CLL, where its expression levels determine its role as a promoter or inhibitor of the tumorigenic process in mice. Partial loss of PTPROt generates the strongest disease phenotype, suggesting that its intermediate expression levels in CLL are selected for.
- Published
- 2016
22. Regulation of receptor-type protein tyrosine phosphatases by their C-terminal tail domains
- Author
-
Tsviya Olender, Maayan Barnea, Ari Elson, and Mark T. Bedford
- Subjects
0301 basic medicine ,animal structures ,Binding Sites ,Amino Acid Motifs ,Receptor-Like Protein Tyrosine Phosphatases ,Protein tyrosine phosphatase ,Receptor type ,Biology ,SH2 domain ,Biochemistry ,Models, Biological ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Animals ,Humans ,Tyrosine ,Amino Acid Sequence ,Phosphorylation ,Sequence motif ,Function (biology) ,Protein Binding - Abstract
Protein tyrosine phosphatases (PTPs) perform specific functions in vivo, despite being vastly outnumbered by their substrates. Because of this and due to the central roles PTPs play in regulating cellular function, PTP activity is regulated by a large variety of molecular mechanisms. We review evidence that indicates that the divergent C-terminal tail sequences (C-terminal domains, CTDs) of receptor-type PTPs (RPTPs) help regulate RPTP function by controlling intermolecular associations in a way that is itself subject to physiological regulation. We propose that the CTD of each RPTP defines an ‘interaction code’ that helps determine molecules it will interact with under various physiological conditions, thus helping to regulate and diversify PTP function.
- Published
- 2016
23. Protein Tyrosine Kinase-6 (PTK6)
- Author
-
Dominique M. Donato, Steven K. Hanks, Kenneth A. Jacobson, M. P. Suresh Jayasekara, Zhan-Guo Gao, Francesca Deflorian, John Papaconstantinou, Ching-Chyuan Hsieh, James H. DeFord, Krassimira Alexieva-Botcheva, Carl W. Anderson, Sheng-Wei Yang, Yuan-Hao Hsu, Linya You, Xiang-Jiao Yang, Miles D. Houslay, Joanna E. Gawecka, Joe W. Ramos, Maria Aparecida Nagai, Jonathan H. Clarke, Robin F. Irvine, Su Jun Lim, Willis X. Li, Sujeet Kumar, Ponniah Selvakumar, Rajendra K. Sharma, Takashi Sasaki, Jun Kotera, Kenji Omori, Lomon So, David A. Fruman, Makoto Murakami, Julian Gomez-Cambronero, Karen M. Henkels, Christopher T. Cottage, Balaji Sundararaman, Shabana Din, Nirmala Hariharan, Mark A. Sussman, Dana Onica, David W. Litchfield, Peter Storz, Anthony John Sadler, Emanuel E. Strehler, Daryl L. Goad, Michael A. Grillo, Peter Koulen, Hisashi Tatebe, Kazuhiro Shiozaki, Giorgio Iotti, Francesco Blasi, Daisuke Urano, Hiroshi Itoh, Rafael Linden, Vilma R. Martins, Marco A. M. Prado, Lai N. Chan, Fuyuhiko Tamanoi, Amanda Harvey, Mingyao Liu, Melissa Rodriguez, Nicholas R. Leslie, Laura Spinelli, Georgios Zilidis, Nimmi R. Weerasinghe, Priyanka Tibarewal, Laura M. Westrate, Jeffrey P. MacKeigan, Ari Elson, Liat Rousso-Noori, Timothy J. Bauler, Philip D. King, Marina Tiemi Shio, and Martin Olivier
- Published
- 2016
24. Production of Osteoclasts for Studying Protein Tyrosine Phosphatase Signaling
- Author
-
Ari Elson, Eynat Finkelshtein, and Einat Levy-Apter
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Chemistry ,Cellular differentiation ,Osteopetrosis ,Protein tyrosine phosphatase ,Protein phosphatase 2 ,medicine.disease ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Osteoclast ,medicine ,Phosphorylation ,Bone marrow ,Tyrosine - Abstract
Osteoclasts, specialized cells that degrade bone, are key components of the cellular system that regulates and maintains bone homeostasis. Aberrant function of osteoclasts can lead to pathological loss or gain of bone mass, such as in osteopetrosis, osteoporosis, and several types of cancer that metastasize to bone. Phosphorylation of osteoclast proteins on tyrosine residues is critical for formation of osteoclasts and for their proper function and responses to physiological signals. Here we describe preparation and growth of osteoclasts from bone marrow of mice, use of viral vectors to downregulate expression of endogenous proteins and to express exogenous proteins in osteoclasts, and analysis of signaling processes triggered by M-CSF, estrogen, and physical contact with matrix in these cells.
- Published
- 2016
25. Expression Profiling during Mammary Epithelial Cell Three-Dimensional Morphogenesis Identifies PTPRO as a Novel Regulator of Morphogenesis and ErbB2-Mediated Transformation
- Author
-
Syed Haider, Irina Kalatskaya, Guang Lin, Ari Elson, Min Yu, Senthil K. Muthuswamy, Francis Nguyen, Paul C. Boutros, Nicholas K. Tonks, Lakshmi Muthuswamy, Bin Xue, and Niloofar Arshadi
- Subjects
Epithelial cell morphogenesis ,Receptor, ErbB-2 ,Morphogenesis ,Regulator ,Down-Regulation ,Breast Neoplasms ,Protein tyrosine phosphatase ,Biology ,medicine.disease_cause ,Receptor tyrosine kinase ,chemistry.chemical_compound ,Cell Line, Tumor ,medicine ,Humans ,Breast ,Mammary Glands, Human ,skin and connective tissue diseases ,Molecular Biology ,Cell Proliferation ,Cell Death ,Cell growth ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,Gene Expression Regulation, Developmental ,Tyrosine phosphorylation ,Articles ,Cell Biology ,Prognosis ,Protein Structure, Tertiary ,Up-Regulation ,Cell biology ,Gene Expression Regulation, Neoplastic ,chemistry ,Cancer research ,biology.protein ,Female ,Transcriptome ,Carcinogenesis - Abstract
Identification of genes that are upregulated during mammary epithelial cell morphogenesis may reveal novel regulators of tumorigenesis. We have demonstrated that gene expression programs in mammary epithelial cells grown in monolayer cultures differ significantly from those in three-dimensional (3D) cultures. We identify a protein tyrosine phosphate, PTPRO, that was upregulated in mature MCF-10A mammary epithelial 3D structures but had low to undetectable levels in monolayer cultures. Downregulation of PTPRO by RNA interference inhibited proliferation arrest during morphogenesis. Low levels of PTPRO expression correlated with reduced survival for breast cancer patients, suggesting a tumor suppressor function. Furthermore, we showed that the receptor tyrosine kinase ErbB2/HER2 is a direct substrate of PTPRO and that loss of PTPRO increased ErbB2-induced cell proliferation and transformation, together with tyrosine phosphorylation of ErbB2. Moreover, in patients with ErbB2-positive breast tumors, low PTPRO expression correlated with poor clinical prognosis compared to ErbB2-positive patients with high levels of PTPRO. Thus, PTPRO is a novel regulator of ErbB2 signaling, a potential tumor suppressor, and a novel prognostic marker for patients with ErbB2-positive breast cancers. We have identified the protein tyrosine phosphatase PTPRO as a regulator of three-dimensional epithelial morphogenesis of mammary epithelial cells and as a regulator of ErbB2-mediated transformation. In addition, we demonstrated that ErbB2 is a direct substrate of PTPRO and that decreased expression of PTPRO predicts poor prognosis for ErbB2-positive breast cancer patients. Thus, our results identify PTPRO as a novel regulator of mammary epithelial transformation, a potential tumor suppressor, and a predictive biomarker for breast cancer.
- Published
- 2012
26. Multifaceted Modulation of K+ Channels by Protein-tyrosine Phosphatase ϵ Tunes Neuronal Excitability
- Author
-
Polina Kornilov, Sharon Ebner-Bennatan, Zohar Tiran, Eti Patrich, Ari Elson, Asher Peretz, and Bernard Attali
- Subjects
medicine.medical_specialty ,animal structures ,Nerve Tissue Proteins ,CHO Cells ,Protein tyrosine phosphatase ,Biology ,environment and public health ,Biochemistry ,Membrane Potentials ,Dephosphorylation ,Mice ,chemistry.chemical_compound ,Cricetulus ,Neurobiology ,Cricetinae ,Internal medicine ,medicine ,Animals ,Phosphorylation ,Molecular Biology ,Cerebral Cortex ,Mice, Knockout ,Neurons ,Membrane potential ,Kinase ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Tyrosine phosphorylation ,Cell Biology ,Potassium channel ,Cell biology ,enzymes and coenzymes (carbohydrates) ,Endocrinology ,nervous system ,chemistry ,Potassium Channels, Voltage-Gated ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Non-receptor-tyrosine kinases (protein-tyrosine kinases) and non-receptor tyrosine phosphatases (PTPs) have been implicated in the regulation of ion channels, neuronal excitability, and synaptic plasticity. We previously showed that protein-tyrosine kinases such as Src kinase and PTPs such as PTPα and PTPε modulate the activity of delayed-rectifier K(+) channels (I(K)). Here we show cultured cortical neurons from PTPε knock-out (EKO) mice to exhibit increased excitability when compared with wild type (WT) mice, with larger spike discharge frequency, enhanced fast after-hyperpolarization, increased after-depolarization, and reduced spike width. A decrease in I(K) and a rise in large-conductance Ca(2+)-activated K(+) currents (mBK) were observed in EKO cortical neurons compared with WT. Parallel studies in transfected CHO cells indicate that Kv1.1, Kv1.2, Kv7.2/7.3, and mBK are plausible molecular correlates of this multifaceted modulation of K(+) channels by PTPε. In CHO cells, Kv1.1, Kv1.2, and Kv7.2/7.3 K(+) currents were up-regulated by PTPε, whereas mBK channel activity was reduced. The levels of tyrosine phosphorylation of Kv1.1, Kv1.2, Kv7.3, and mBK potassium channels were increased in the brain cortices of neonatal and adult EKO mice compared with WT, suggesting that PTPε in the brain modulates these channel proteins. Our data indicate that in EKO mice, the lack of PTPε-mediated dephosphorylation of Kv1.1, Kv1.2, and Kv7.3 leads to decreased I(K) density and enhanced after-depolarization. In addition, the deficient PTPε-mediated dephosphorylation of mBK channels likely contributes to enhanced mBK and fast after-hyperpolarization, spike shortening, and consequent increase in neuronal excitability observed in cortical neurons from EKO mice.
- Published
- 2012
27. Epidermal Growth Factor Receptor (EGFR)-mediated Positive Feedback of Protein-tyrosine Phosphatase ϵ (PTPϵ) on ERK1/2 and AKT Protein Pathways Is Required for Survival of Human Breast Cancer Cells
- Author
-
Rafael Pulido, Ari Elson, and Caroline E. Nunes-Xavier
- Subjects
Cell Survival ,MAP Kinase Signaling System ,Breast Neoplasms ,Protein tyrosine phosphatase ,Fibroblast growth factor ,Biochemistry ,Cell Line, Tumor ,Humans ,Protein phosphorylation ,Epidermal growth factor receptor ,skin and connective tissue diseases ,Molecular Biology ,Protein kinase B ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,biology ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Cell Biology ,Cell biology ,Enzyme Activation ,ErbB Receptors ,Fibroblast Growth Factors ,Cancer cell ,Carcinogens ,biology.protein ,Tetradecanoylphorbol Acetate ,Phosphorylation ,Female ,Proto-Oncogene Proteins c-akt ,Tyrosine kinase ,Signal Transduction - Abstract
Increased tyrosine phosphorylation has been correlated with human cancer, including breast cancer. In general, the activation of tyrosine kinases (TKs) can be antagonized by the action of protein-tyrosine phosphatases (PTPs). However, in some cases PTPs can potentiate the activation of TKs. In this study, we have investigated the functional role of PTPε in human breast cancer cell lines. We found the up-regulation and activation of receptor PTPε (RPTPε) in MCF-7 cells and MDA-MB-231 upon PMA, FGF, and serum stimulation, which depended on EGFR and ERK1/2 activity. Diminishing the expression of PTPε in human breast cancer cells abolished ERK1/2 and AKT activation, and decreased the viability and anchorage-independent growth of the cells. Conversely, stable MCF-7 cell lines expressing inducible high levels of ectopic PTPε displayed higher activation of ERK1/2 and anchorage-independent growth. Our results demonstrate that expression of PTPε is up-regulated and activated in breast cancer cell lines, through EGFR, by sustained activation of the ERK1/2 pathway, generating a positive feedback regulatory loop required for survival of human breast cancer cells.
- Published
- 2012
28. Protein Tyrosine Phosphatase Epsilon Affects Body Weight by Downregulating Leptin Signaling in a Phosphorylation-Dependent Manner
- Author
-
Liat Rousso-Noori, Adi Neufeld-Cohen, Alon Chen, Ari Elson, Vasudheva R. Akepati, Einat Levy-Apter, Yonat Keshet, Richard A. Klinghoffer, Yael Kuperman, and Hilla Knobler
- Subjects
Leptin ,medicine.medical_specialty ,Physiology ,Immunoblotting ,Hypothalamus ,Down-Regulation ,Protein tyrosine phosphatase ,Biology ,Mice ,Internal medicine ,medicine ,Glucose homeostasis ,Animals ,Homeostasis ,Humans ,Obesity ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,Janus kinase 2 ,Leptin receptor ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Body Weight ,digestive, oral, and skin physiology ,Cell Biology ,Janus Kinase 2 ,Endocrinology ,Glucose ,biology.protein ,Diet, Atherogenic ,Receptors, Leptin ,Female ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
SummaryMolecular-level understanding of body weight control is essential for combating obesity. We show that female mice lacking tyrosine phosphatase epsilon (RPTPe) are protected from weight gain induced by high-fat food, ovariectomy, or old age and exhibit increased whole-body energy expenditure and decreased adiposity. RPTPe-deficient mice, in particular males, exhibit improved glucose homeostasis. Female nonobese RPTPe-deficient mice are leptin hypersensitive and exhibit reduced circulating leptin concentrations, suggesting that RPTPe inhibits hypothalamic leptin signaling in vivo. Leptin hypersensitivity persists in aged, ovariectomized, and high-fat-fed RPTPe-deficient mice, indicating that RPTPe helps establish obesity-associated leptin resistance. RPTPe associates with and dephosphorylates JAK2, thereby downregulating leptin receptor signaling. Leptin stimulation induces phosphorylation of hypothalamic RPTPe at its C-terminal Y695, which drives RPTPe to downregulate JAK2. RPTPe is therefore an inhibitor of hypothalamic leptin signaling in vivo, and provides controlled negative-feedback regulation of this pathway following its activation.
- Published
- 2011
- Full Text
- View/download PDF
29. Protein Tyrosine Phosphatase Epsilon Regulates Integrin-mediated Podosome Stability in Osteoclasts by Activating Src
- Author
-
Chen Luxenburg, Eynat Finkelshtein, Shira Granot-Attas, and Ari Elson
- Subjects
rac1 GTP-Binding Protein ,rho GTP-Binding Proteins ,Cytoplasm ,Integrins ,Podosome ,Recombinant Fusion Proteins ,Proto-Oncogene Proteins pp60(c-src) ,Integrin ,Osteoclasts ,Protein tyrosine phosphatase ,Biology ,environment and public health ,Mice ,Cell Adhesion ,Animals ,Phosphorylation ,Cell adhesion ,Molecular Biology ,Rho-associated protein kinase ,Feedback, Physiological ,Mice, Knockout ,rho-Associated Kinases ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Neuropeptides ,Articles ,Cell Biology ,rac GTP-Binding Proteins ,Cell biology ,Enzyme Activation ,Rac GTP-Binding Proteins ,enzymes and coenzymes (carbohydrates) ,Focal Adhesion Kinase 2 ,src-Family Kinases ,Mutagenesis, Site-Directed ,biology.protein ,Female ,Cell Surface Extensions ,rhoA GTP-Binding Protein ,Protein Processing, Post-Translational ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The nonreceptor isoform of tyrosine phosphatase epsilon (cyt-PTPe) supports osteoclast adhesion and activity in vivo, leading to increased bone mass in female mice lacking PTPe (EKO mice). The structure and organization of the podosomal adhesion structures of EKO osteoclasts are abnormal; the molecular mechanism behind this is unknown. We show here that EKO podosomes are disorganized, unusually stable, and reorganize poorly in response to physical contact. Phosphorylation and activities of Src, Pyk2, and Rac are decreased and Rho activity is increased in EKO osteoclasts, suggesting that integrin signaling is defective in these cells. Integrin activation regulates cyt-PTPe by inducing Src-dependent phosphorylation of cyt-PTPe at Y638. This phosphorylation event is crucial because wild-type—but not Y638F—cyt-PTPe binds and further activates Src and restores normal stability to podosomes in EKO osteoclasts. Increasing Src activity or inhibiting Rho or its downstream effector Rho kinase in EKO osteoclasts rescues their podosomal stability phenotype, indicating that cyt-PTPe affects podosome stability by functioning upstream of these molecules. We conclude that cyt-PTPe participates in a feedback loop that ensures proper Src activation downstream of integrins, thus linking integrin signaling with Src activation and accurate organization and stability of podosomes in osteoclasts.
- Published
- 2009
30. PTPϵ has a critical role in signaling transduction pathways and phosphoprotein network topology in red cells
- Author
-
Loris Turretta, Carlo Brugnara, Lucia De Franceschi, Ari Elson, Francesco Michelangelo Turrini, Andrea Biondani, Achille Iolascon, Anna Maria Brunati, Renzo Deana, Silverio Perrotta, Carlo Laudanna, Cristina Bulato, Franco Carta, DE FRANCESCHI, L, Biondani, A, Carta, F, Turrini, F, Laudanna, C, Deana, R, Brunati, Am, Turretta, L, Iolascon, A, Perrotta, Silverio, Elson, A, Bulato, C, Brugnara, C., De Franceschi, L, Iolascon, Achille, and Perrotta, S
- Subjects
Erythrocytes ,Syk ,phsophoproteomics ,Protein tyrosine phosphatase ,Signal transduction ,Biology ,Fyn ,Gardos channel ,Tyrosine phosphorylation ,red blood cells ,proteomics ,network analysis ,Biochemistry ,Article ,Mice ,chemistry.chemical_compound ,FYN ,Animals ,Syk Kinase ,Clotrimazole ,Tyrosine ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Intracellular Signaling Peptides and Proteins ,hemic and immune systems ,Protein-Tyrosine Kinases ,Tyrosine-phosphorylation ,Cell biology ,src-Family Kinases ,chemistry ,Phosphoprotein ,Potassium ,Phosphorylation ,Calcium ,Calcium Channels ,Algorithms ,Metabolic Networks and Pathways - Abstract
Protein tyrosine phosphatases (PTPs) are crucial components of cellular signal transduction pathways. We report here that red blood cells (RBCs) from mice lacking PTPe (Ptpre −/− ) exhibit abnormal morphology and increased Ca 2+ -activated-K + channel activity, which was partially blocked by the Src-Family-Kinases (SFKs) inhibitor PP1. In Ptpre −/− mouse RBCs, the activity of Fyn and Yes, two SFKs, were increased, suggesting a functional relationship between SFKs, PTPe and Ca 2+ -activated-K + -channel. The absence of PTPe markedly affected the RBC membrane tyrosine (Tyr-) phosphoproteome, indicating a perturbation of RBCs signal transduction pathways. Using signaling network computational analysis of the Tyr-phosphoproteomic data, we identified 7 topological clusters. We studied cluster 1, containing Syk-Tyr-kinase: Syk-kinase activity was higher in wild-type than in Ptpre −/− RBCs, validating the network computational analysis and indicating a novel signaling pathway, which involves Fyn and Syk in regulation of red cell morphology.
- Published
- 2008
31. Protein tyrosine phosphatases in osteoclast differentiation, adhesion, and bone resorption
- Author
-
Ari Elson and Shira Granot-Attas
- Subjects
Histology ,Podosome ,Cellular differentiation ,Osteoclasts ,Protein tyrosine phosphatase ,Biology ,Models, Biological ,Bone resorption ,Pathology and Forensic Medicine ,chemistry.chemical_compound ,Osteoclast ,Cell Adhesion ,medicine ,Animals ,Humans ,Bone Resorption ,Protein Tyrosine Phosphatase, Non-Receptor Type 6 ,Receptor-Like Protein Tyrosine Phosphatases, Class 3 ,Cell Differentiation ,Tyrosine phosphorylation ,Cell Biology ,General Medicine ,Protein-Tyrosine Kinases ,medicine.anatomical_structure ,chemistry ,Biochemistry ,Receptor-Like Protein Tyrosine Phosphatases ,Protein Tyrosine Phosphatases ,Tyrosine kinase - Abstract
Osteoclasts are large cells derived from the monocyte-macrophage hematopoietic cell lineage. Their primary function is to degrade bone in various physiological contexts. Osteoclasts adhere to bone via podosomes, specialized adhesion structures whose structure and subcellular organization are affected by mechanical contact of the cell with bone matrix. Ample evidence indicates that reversible tyrosine phosphorylation of podosomal proteins plays a major role in determining the organization and dynamics of podosomes. Although roles of several tyrosine kinases are known in detail in this respect, little is known concerning the roles of protein tyrosine phosphatases (PTPs) in regulating osteoclast adhesion. Here we summarize available information concerning the known and hypothesized roles of the best-researched PTPs in osteoclasts - PTPRO, PTP epsilon, SHP-1, and PTP-PEST. Of these, PTPRO, PTP epsilon, and PTP-PEST appear to support osteoclast activity while SHP-1 inhibits it. Additional studies are required to provide full molecular details of the roles of these PTPs in regulating osteoclast adhesion, and to uncover additional PTPs that participate in this process.
- Published
- 2008
32. Protein tyrosine phosphatases: functional inferences from mouse models and human diseases
- Author
-
Andrew W. Stoker, Wiljan Hendriks, Ari Elson, and Sheila Harroch
- Subjects
Genetically modified mouse ,Cell Biology ,Protein tyrosine phosphatase ,Computational biology ,Disease ,Biology ,Biochemistry ,Glucose homeostasis ,Protein phosphorylation ,Signal transduction ,Molecular Biology ,Gene ,Calcium signaling - Abstract
Some 40-odd genes in mammals encode phosphotyrosine-specific, 'classical' protein tyrosine phosphatases. The generation of animal model systems and the study of various human disease states have begun to elucidate the important and diverse roles of protein tyrosine phosphatases in cellular signalling pathways, development and disease. Here, we provide an overview of those findings from mice and men, and indicate several novel approaches that are now being exploited to further our knowledge of this fascinating enzyme family.
- Published
- 2008
33. Protein-tyrosine Phosphatase ϵ Regulates Shc Signaling in a Kinase-specific Manner
- Author
-
Ari Elson, William J. Muller, and Judith Kraut-Cohen
- Subjects
biology ,Chemistry ,Phosphatase ,Signal transducing adaptor protein ,Context (language use) ,Cell Biology ,Protein tyrosine phosphatase ,environment and public health ,Biochemistry ,Cell biology ,biology.protein ,Phosphorylation ,GRB2 ,biological phenomena, cell phenomena, and immunity ,Phosphotyrosine-binding domain ,Molecular Biology ,hormones, hormone substitutes, and hormone antagonists ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Individual protein tyrosine kinases and phosphatases target multiple substrates; this may generate conflicting signals, possibly within a single pathway. Protein-tyrosine phosphatase ϵ (PTPϵ) performs two potentially opposing roles: in Neu-induced mammary tumors, PTPϵ activates Src downstream of Neu, whereas in other systems PTPϵ can indirectly down-regulate MAP kinase signaling. We now show that the latter effect is mediated at least in part via the adaptor protein Shc. PTPϵ binds and dephosphorylates Shc in vivo, reducing the association of Shc with Grb2 and inhibiting downstream ERK activation. PTPϵ binds Shc in a phosphotyrosine-independent manner mediated by the Shc PTB domain and aided by a sequence of 10 N-terminal residues in PTPϵ. Surprisingly, PTPϵ dephosphorylates Shc in a kinase-dependent manner; PTPϵ targets Shc in the presence of Src but not in the presence of Neu. Using a series of point mutants of Shc and Neu, we show that Neu protects Shc from dephosphorylation by binding the PTB domain of Shc, most likely competing against PTPϵ for binding the same domain. In agreement, PTPϵ dephosphorylates Shc in mouse embryo fibroblasts but not in Neu-induced mammary tumor cells. We conclude that in the context of Neu-induced mammary tumor cells, Neu prevents PTPϵ from targeting Shc and from reducing its promitogenic signal while phosphorylating PTPϵ and directing it to activate Src in support of mitogenesis. In so doing, Neu contributes to the coherence of the promitogenic role of PTPϵ in this system.
- Published
- 2008
34. Cytosolic Protein Tyrosine Phosphatase-ε Is a Negative Regulator of Insulin Signaling in Skeletal Muscle
- Author
-
Ari Elson, Shlomit Aga-Mizrachi, Sanford R. Sampson, Tamar Brutman-Barazani, Avraham I. Jacob, and Asia Bak
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Protein tyrosine phosphatase ,Biology ,Gene Expression Regulation, Enzymologic ,Cell Line ,Mice ,Cytosol ,Endocrinology ,Internal medicine ,medicine ,Animals ,Insulin ,Phosphorylation ,Tyrosine ,Muscle, Skeletal ,Mice, Knockout ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Cell Membrane ,Skeletal muscle ,Receptor, Insulin ,Up-Regulation ,RNA silencing ,Insulin receptor ,Glucose ,src-Family Kinases ,medicine.anatomical_structure ,biology.protein ,RNA Interference ,Signal transduction ,Signal Transduction - Abstract
Whereas positive regulatory events triggered by insulin binding to insulin receptor (IR) have been well documented, the mechanism by which the activated IR is returned to the basal status is not completely understood. Recently studies focused on the involvement of protein tyrosine phosphatases (PTPs) and how they might influence IR signaling. In this study, we examined the possibility that cytosolic PTPepsilon (cytPTPepsilon) is involved in IR signaling. Studies were performed on L6 skeletal muscle cells. cytPTPepsilon was overexpressed by using pBABE retroviral expression vectors. In addition, we inhibited cytPTPepsilon by RNA silencing. We found that insulin induced rapid association of cytPTPepsilon with IR. Interestingly, this association appeared to occur in the plasma membrane and on stimulation with insulin the two proteins internalized together. Moreover, it appeared that almost all internalized IR was associated with cytPTPepsilon. We found that knockdown of cytPTPepsilon by RNA silencing increased insulin-induced tyrosine phosphorylation of IR and IR substrate (IRS)-1 as well as phosphorylation of protein kinase B and glycogen synthase kinase-3 and insulin-induced stimulation of glucose uptake. Moreover, overexpression of wild-type cytPTPepsilon reduced insulin-induced tyrosine phosphorylation of IR, IRS-1, and phosphorylation of protein kinase B and glycogen synthase kinase-3 and insulin-induced stimulation of glucose uptake. Finally, insulin-induced tyrosine phosphorylation of IR and IRS-1 was greater in skeletal muscle from mice lacking the cytPTPepsilon gene than that from wild-type control animals. We conclude that cytPTPepsilon serves as another major candidate negative regulator of IR signaling in skeletal muscle.
- Published
- 2007
35. Neu-mediated phosphorylation of protein tyrosine phosphatase epsilon is critical for activation of Src in mammary tumor cells
- Author
-
D Berman-Golan and Ari Elson
- Subjects
Cancer Research ,Phosphoric monoester hydrolases ,Receptor, ErbB-2 ,Proto-Oncogene Proteins pp60(c-src) ,Fluorescent Antibody Technique ,Mice, Transgenic ,Protein tyrosine phosphatase ,Biology ,Proto-Oncogene Proteins c-fyn ,medicine.disease_cause ,Mice ,Genetics ,medicine ,Animals ,Immunoprecipitation ,Phosphorylation ,Tyrosine ,Molecular Biology ,Mice, Knockout ,Proto-Oncogene Proteins c-yes ,chemistry.chemical_classification ,Mammary tumor ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Mammary Neoplasms, Experimental ,Peptide Fragments ,Cell biology ,Phenotype ,Enzyme ,Mammary Tumor Virus, Mouse ,chemistry ,Cancer research ,Female ,Protein Tyrosine Phosphatases ,Carcinogenesis ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The receptor-type protein tyrosine phosphatase epsilon (RPTPepsilon) activates c-Src in mammary tumor cells induced in vivo by Neu. Tumor cells lacking RPTPepsilon exhibit reduced c-Src activity, appear less transformed morphologically and proliferate slower in vitro and in vivo. Expression of Src rescues most of these phenotypes, indicating that c-Src activity is important for maintaining the transformed phenotype. However, the molecular mechanisms that control activation of c-Src by RPTPepsilon are unknown. We show that Neu induces phosphorylation of RPTPepsilon exclusively at its C-terminal Y695, and that this phosphorylation is required for activation of c-Src by RPTPepsilon. Phosphorylation of RPTPepsilon does not affect its activity toward another substrate, the voltage-gated potassium channel Kv2.1, suggesting that phosphorylation directs RPTPepsilon activity toward c-Src. Phosphorylation of RPTPepsilon reduces its dimerization at the cell membrane, although this does not affect its activity significantly. RPTPepsilon is subject to strong auto- and trans-dephosphorylation, suggesting that dephosphorylation limits the activation of c-Src downstream of Neu. We conclude that an Neu-RPTPepsilon-Src signaling pathway exists in mammary tumor cells, in which phosphorylation of RPTPepsilon by Neu directs RPTPepsilon to activate c-Src. Reversible phosphorylation of RPTPepsilon at Y695 may thus function as a 'molecular switch', which affects the substrate specificity of the phosphatase.
- Published
- 2007
36. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells
- Author
-
Shoham Shivtiel, Polina Goichberg, Orit Kollet, Ayelet Dar, Yejezkel Sztainberg, Ari Elson, Asaf Spiegel, Robert M. Samstein, Kfir Lapid, Tsvee Lapidot, Alexander Kalinkovich, and Melania Tesio
- Subjects
musculoskeletal diseases ,Receptors, CXCR4 ,Hematopoietic stem cell niche ,Cathepsin K ,Osteoclasts ,Mice, Inbred Strains ,Stem cell factor ,Bone and Bones ,General Biochemistry, Genetics and Molecular Biology ,Bone resorption ,Cell Line ,Bone remodeling ,Mice ,Cell Movement ,Animals ,Homeostasis ,Humans ,Bone Resorption ,Progenitor cell ,Mice, Knockout ,Endosteum ,Stem Cell Factor ,Membrane Glycoproteins ,Receptor Activator of Nuclear Factor-kappa B ,biology ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,RANK Ligand ,Proteolytic enzymes ,General Medicine ,Hematopoietic Stem Cells ,Cathepsins ,Chemokine CXCL12 ,Cell biology ,Matrix Metalloproteinase 9 ,RANKL ,Immunology ,biology.protein ,Female ,Protein Tyrosine Phosphatases ,Carrier Proteins ,Chemokines, CXC - Abstract
Here we investigated the potential role of bone-resorbing osteoclasts in homeostasis and stress-induced mobilization of hematopoietic progenitors. Different stress situations induced activity of osteoclasts (OCLs) along the stem cell-rich endosteum region of bone, secretion of proteolytic enzymes and mobilization of progenitors. Specific stimulation of OCLs with RANKL recruited mainly immature progenitors to the circulation in a CXCR4- and MMP-9-dependent manner; however, RANKL did not induce mobilization in young female PTPepsilon-knockout mice with defective OCL bone adhesion and resorption. Inhibition of OCLs with calcitonin reduced progenitor egress in homeostasis, G-CSF mobilization and stress situations. RANKL-stimulated bone-resorbing OCLs also reduced the stem cell niche components SDF-1, stem cell factor (SCF) and osteopontin along the endosteum, which was associated with progenitor mobilization. Finally, the major bone-resorbing proteinase, cathepsin K, also cleaved SDF-1 and SCF. Our findings indicate involvement of OCLs in selective progenitor recruitment as part of homeostasis and host defense, linking bone remodeling with regulation of hematopoiesis.
- Published
- 2006
37. Site-Selective Regulation of Platelet-Derived Growth Factor β Receptor Tyrosine Phosphorylation by T-Cell Protein Tyrosine Phosphatase
- Author
-
Michel L. Tremblay, Camilla Persson, Frank-D. Böhmer, Ari Elson, Carl-Henrik Heldin, Carina Hellberg, Boyka Markova, Fawaz G. Haj, Lars Rönnstrand, Arne Östman, Annie Bourdeau, Benjamin G. Neel, and Catrine Sävenhed
- Subjects
animal structures ,Protein tyrosine phosphatase ,environment and public health ,Receptor tyrosine kinase ,Receptor, Platelet-Derived Growth Factor beta ,Mice ,chemistry.chemical_compound ,Cell Movement ,Animals ,Phosphorylation ,Cell Growth and Development ,Molecular Biology ,Mice, Knockout ,Protein Tyrosine Phosphatase, Non-Receptor Type 2 ,biology ,Autophosphorylation ,Tyrosine phosphorylation ,Cell Biology ,Fibroblasts ,Molecular biology ,Isoenzymes ,enzymes and coenzymes (carbohydrates) ,chemistry ,ROR1 ,biology.protein ,Tyrosine ,Protein Tyrosine Phosphatases ,Medicinal Chemistry ,Signal transduction ,Antibodies, Phospho-Specific ,Platelet-derived growth factor receptor ,Signal Transduction - Abstract
The platelet-derived growth factor (PDGF) ß receptor mediates mitogenic and chemotactic signals. Like other tyrosine kinase receptors, the PDGF ß receptor is negatively regulated by protein tyrosine phosphatases (PTPs). To explore whether T-cell PTP (TC-PTP) negatively regulates the PDGF ß receptor, we compared PDGF ß receptor tyrosine phosphorylation in wild-type and TC-PTP knockout (ko) mouse embryos. PDGF ß receptors were hyperphosphorylated in TC-PTP ko embryos. Fivefold-higher ligand-induced receptor phosphorylation was observed in TC-PTP ko mouse embryo fibroblasts (MEFs) as well. Reexpression of TC-PTP partly abolished this difference. As determined with site-specific phosphotyrosine antibodies, the extent of hyperphosphorylation varied among different autophosphorylation sites. The phospholipase C1 binding site Y1021, previously implicated in chemotaxis, displayed the largest increase in phosphorylation. The increase in Y1021 phosphorylation was accompanied by increased phospholipase C1 activity and migratory hyperresponsiveness to PDGF. PDGF ß receptor tyrosine phosphorylation in PTP-1B ko MEFs but not in PTP ko MEFs was also higher than that in control cells. This increase occurred with a site distribution different from that seen after TC-PTP depletion. PDGF-induced migration was not increased in PTP-1B ko cells. In summary, our findings identify TC-PTP as a previously unrecognized negative regulator of PDGF ß receptor signaling and support the general notion that PTPs display site selectivity in their action on tyrosine kinase receptors. Copyright © 2004, American Society for Microbiology. All Rights Reserved
- Published
- 2004
38. Protein tyrosine phosphatase epsilon activates Yes and Fyn in Neu-induced mammary tumor cells
- Author
-
Ari Elson and Shira Granot-Attas
- Subjects
Phosphatase ,Mammary Neoplasms, Animal ,Mice, Transgenic ,Protein tyrosine phosphatase ,Biology ,Proto-Oncogene Proteins c-fyn ,Mice ,FYN ,Proto-Oncogene Proteins ,Tumor Cells, Cultured ,Animals ,Phosphorylation ,Tyrosine ,Mice, Knockout ,Proto-Oncogene Proteins c-yes ,Mammary tumor ,Kinase ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Cell Biology ,Genes, erbB-2 ,Enzyme Activation ,src-Family Kinases ,Cancer research ,Female ,Protein Tyrosine Phosphatases ,Proto-oncogene tyrosine-protein kinase Src - Abstract
The receptor-type form of protein tyrosine phosphatase epsilon (RPTPe) is among the few tyrosine phosphatases that can support the transformed phenotype of tumor cells. Accordingly, cells from mammary epithelial tumors induced by activated Neu in mice genetically lacking RPTPe appear morphologically less transformed and exhibit reduced proliferation. The effect of RPTPe in these cells is mediated at least in part by its ability to activate Src, the prototypic member of a family of related kinases. We show here that RPTPe is a physiological activator of two additional Src family kinases, Yes and Fyn. Activities of both kinases are inhibited in mammary tumor cells lacking RPTPe, and phosphorylation at their C-terminal inhibitory tyrosines is increased. In agreement, opposite effects on activities and phosphorylation of Yes and Fyn are observed following increased expression of PTPe. RPTPe also forms stable complexes with either kinase, providing physical opportunity for their activation by RPTPe. Surprisingly, expression of Yes or of Fyn does not rescue the morphological phenotype of RPTPe-deficient tumor cells in contrast with the strong ability of Src to do so. We conclude that RPTPe activates Src, Yes, and Fyn, but that these related kinases play distinct roles in Neu-induced mammary tumor cells.
- Published
- 2004
39. Tyrosine Phosphatase Epsilon Is a Positive Regulator of Osteoclast Function in Vitro and In Vivo
- Author
-
Chen Luxenburg, Shira Granot-Attas, Archana Sanjay, Roland Baron, Riccardo Chiusaroli, Zohar Tiran, Alon Harmelin, Ari Elson, Tsuyoshi Miyazaki, and Hilla Knobler
- Subjects
musculoskeletal diseases ,medicine.medical_specialty ,Podosome ,Osteoclasts ,Protein tyrosine phosphatase ,Bone and Bones ,Bone resorption ,Bone remodeling ,Mice ,chemistry.chemical_compound ,Osteoclast ,Internal medicine ,medicine ,Animals ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Progesterone ,Mice, Knockout ,Membrane Glycoproteins ,Osteoblasts ,Receptor Activator of Nuclear Factor-kappa B ,biology ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,RANK Ligand ,Cell Polarity ,Cell Differentiation ,Estrogens ,Tyrosine phosphorylation ,Articles ,Cell Biology ,Enzyme Activation ,src-Family Kinases ,Endocrinology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Receptor-Like Protein Tyrosine Phosphatases ,chemistry ,RANKL ,biology.protein ,Collagen ,Protein Tyrosine Phosphatases ,Carrier Proteins ,Signal Transduction - Abstract
Protein tyrosine phosphorylation is a major regulator of bone metabolism. Tyrosine phosphatases participate in regulating phosphorylation, but roles of specific phosphatases in bone metabolism are largely unknown. We demonstrate that young (
- Published
- 2004
40. Dimerization In Vivo and Inhibition of the Nonreceptor Form of Protein Tyrosine Phosphatase Epsilon
- Author
-
Gidi Shani, Tal Sines, Jeroen den Hertog, Zohar Tiran, Ari Elson, Hila Toledano-Katchalski, and Shira Granot-Attas
- Subjects
DNA, Complementary ,Potassium Channels ,Time Factors ,Phosphatase ,Protein tyrosine phosphatase ,Biology ,Transfection ,Models, Biological ,environment and public health ,Cell Line ,Mice ,chemistry.chemical_compound ,Shab Potassium Channels ,Cell surface receptor ,Extracellular ,Animals ,Humans ,Phosphorylation ,Tyrosine ,Cell Growth and Development ,Molecular Biology ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Cell Membrane ,Temperature ,Tyrosine phosphorylation ,3T3 Cells ,Hydrogen Peroxide ,Cell Biology ,Precipitin Tests ,Protein Structure, Tertiary ,Cell biology ,enzymes and coenzymes (carbohydrates) ,chemistry ,Potassium Channels, Voltage-Gated ,embryonic structures ,cardiovascular system ,Chromatography, Gel ,Protein Tyrosine Phosphatases ,Signal transduction ,Dimerization ,Delayed Rectifier Potassium Channels ,Protein Binding ,Signal Transduction - Abstract
cyt-PTP epsilon is a naturally occurring nonreceptor form of the receptor-type protein tyrosine phosphatase (PTP) epsilon. As such, cyt-PTP epsilon enables analysis of phosphatase regulation in the absence of extracellular domains, which participate in dimerization and inactivation of the receptor-type phosphatases receptor-type protein tyrosine phosphatase alpha (RPTPalpha) and CD45. Using immunoprecipitation and gel filtration, we show that cyt-PTP epsilon forms dimers and higher-order associations in vivo, the first such demonstration among nonreceptor phosphatases. Although cyt-PTP epsilon readily dimerizes in the absence of exogenous stabilization, dimerization is increased by oxidative stress. Epidermal growth factor receptor stimulation can affect cyt-PTP epsilon dimerization and tyrosine phosphorylation in either direction, suggesting that cell surface receptors can relay extracellular signals to cyt-PTP epsilon, which lacks extracellular domains of its own. The inactive, membrane-distal (D2) phosphatase domain of cyt-PTP epsilon is a major contributor to intermolecular binding and strongly interacts in a homotypic manner; the presence of D2 and the interactions that it mediates inhibit cyt-PTP epsilon activity. Intermolecular binding is inhibited by the extreme C and N termini of D2. cyt-PTP epsilon lacking these regions constitutively dimerizes, and its activities in vitro towards para-nitrophenylphosphate and in vivo towards the Kv2.1 potassium channel are markedly reduced. We conclude that physiological signals can regulate dimerization and phosphorylation of cyt-PTP epsilon in the absence of direct interaction between the PTP and extracellular molecules. Furthermore, dimerization can be mediated by the D2 domain and does not strictly require the presence of PTP extracellular domains.
- Published
- 2003
41. Phosphorylation-dependent Regulation of Kv2.1 Channel Activity at Tyrosine 124 by Src and by Protein-tyrosine Phosphatase ε
- Author
-
Ari Elson, Asher Peretz, Zohar Tiran, and Bernard Attali
- Subjects
Potassium Channels ,Protein tyrosine phosphatase ,Biology ,SH2 domain ,environment and public health ,Biochemistry ,SH3 domain ,Receptor tyrosine kinase ,Cell Line ,Mice ,chemistry.chemical_compound ,Shab Potassium Channels ,Animals ,Humans ,Phosphorylation ,Molecular Biology ,Tyrosine-protein kinase CSK ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Tyrosine phosphorylation ,Cell Biology ,Rats ,Up-Regulation ,Cell biology ,enzymes and coenzymes (carbohydrates) ,src-Family Kinases ,chemistry ,Potassium Channels, Voltage-Gated ,biology.protein ,Tyrosine ,Protein Tyrosine Phosphatases ,Ion Channel Gating ,Delayed Rectifier Potassium Channels ,Signal Transduction ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Voltage-gated potassium (Kv) channels are a complex and heterogeneous family of proteins that play major roles in brain and cardiac excitability. Although Kv channels are activated by changes in cell membrane potential, tyrosine phosphorylation of channel subunits can modulate the extent of channel activation by depolarization. We have previously shown that dephosphorylation of Kv2.1 by the nonreceptor-type tyrosine phosphatase PTPepsilon (cyt-PTPepsilon) down-regulates channel activity and counters its phosphorylation and up-regulation by Src or Fyn. In the present study, we identify tyrosine 124 within the T1 cytosolic domain of Kv2.1 as a target site for the activities of Src and cyt-PTPepsilon. Tyr(124) is phosphorylated by Src in vitro; in whole cells, Y124F Kv2.1 is significantly less phosphorylated by Src and loses most of its ability to bind the D245A substrate-trapping mutant of cyt-PTPepsilon. Phosphorylation of Tyr(124) is critical for Src-mediated up-regulation of Kv2.1 channel activity, since Y124F Kv2.1-mediated K(+) currents are only marginally up-regulated by Src, in contrast with a 3-fold up-regulation of wild-type Kv2.1 channels by the kinase. Other properties of Kv2.1, such as expression levels, subcellular localization, and voltage dependence of channel activation, are unchanged in Y124F Kv2.1, indicating that the effects of the Y124F mutation are specific. Together, these results indicate that Tyr(124) is a significant site at which the mutually antagonistic activities of Src and cyt-PTPepsilon affect Kv2.1 phosphorylation and activity.
- Published
- 2003
42. Tyrosine Phosphatase-ε Activates Src and Supports the Transformed Phenotype of Neu-induced Mammary Tumor Cells
- Author
-
Hava Gil-Henn and Ari Elson
- Subjects
Receptor, ErbB-2 ,Molecular Sequence Data ,Phosphatase ,Receptors, Cell Surface ,Protein tyrosine phosphatase ,Biology ,Biochemistry ,Mice ,Enzyme activator ,Animals ,Phosphorylation ,Tyrosine ,Molecular Biology ,Mammary tumor ,Binding Sites ,Base Sequence ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Mammary Neoplasms, Experimental ,Cell Biology ,Cell biology ,Enzyme Activation ,Isoenzymes ,Mice, Inbred C57BL ,Phenotype ,src-Family Kinases ,Receptor-Like Protein Tyrosine Phosphatases ,Female ,Protein Tyrosine Phosphatases ,Cell Division ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Few tyrosine phosphatases support, rather than inhibit, survival of tumor cells. We present genetic evidence that receptor-type protein-tyrosine phosphatase (RPTP)-epsilon performs such a function, as cells from mammary epithelial tumors induced by activated Neu in mice genetically lacking RPTPepsilon appeared morphologically less transformed and exhibited reduced proliferation. We show that at the molecular level, RPTPepsilon activates Src, a known collaborator of Neu in mammary tumorigenesis. Lack of RPTPepsilon reduced Src activity and altered Src phosphorylation in tumor cells; RPTPepsilon dephosphorylated and activated Src; and Src bound a substrate-trapping mutant of RPTPepsilon. The altered morphology of tumor cells lacking RPTPepsilon was corrected by exogenous Src and exogenous RPTPepsilon or RPTPalpha; exogenous activated Src corrected also the growth rate phenotype. Together, these results suggest that the altered morphology of RPTPepsilon-deficient tumor cells is caused by reduced Src activity, caused, in turn, by lack of RPTPepsilon. Unexpectedly, the phenotype of RPTPepsilon-deficient tumor cells occurs despite expression of the related RPTPalpha, indicating that endogenous RPTPalpha does not compensate for the absence of RPTPepsilon in this case. We conclude that RPTPepsilon is a physiological activator of Src in Neu-induced mammary tumors and suggest that pharmacological inhibition of phosphatases that activate Src may be useful to augment direct pharmacological inhibition of Src.
- Published
- 2003
43. Nuclear Localization of Non-receptor Protein Tyrosine Phosphatase ε Is Regulated by Its Unique N-Terminal Domain
- Author
-
Judith Kraut, Ari Elson, Gloria Volohonsky, and Hila Toledano-Katchalski
- Subjects
Gene isoform ,Green Fluorescent Proteins ,Active Transport, Cell Nucleus ,Protein tyrosine phosphatase ,Biology ,environment and public health ,Gene Expression Regulation, Enzymologic ,Mice ,Animals ,Protein Isoforms ,Amino Acid Sequence ,Receptor ,Cell Nucleus ,Mutagenesis ,Cell Biology ,Subcellular localization ,Cell Compartmentation ,Protein Structure, Tertiary ,N-terminus ,Luminescent Proteins ,Oxidative Stress ,enzymes and coenzymes (carbohydrates) ,Cytosol ,Eukaryotic Cells ,Biochemistry ,Mutation ,embryonic structures ,cardiovascular system ,Protein Tyrosine Phosphatases ,Nuclear localization sequence - Abstract
Precise subcellular localization is an important factor in regulation of the functions of protein tyrosine phosphatases. The non-receptor form of protein tyrosine phosphatase epsilon (cyt-PTP(epsilon)) can be found in cell nuclei, among other cellular locations, while p67 PTP(epsilon), a naturally occurring isoform which lacks the 27 N terminal residues of cyt-PTP(epsilon), is exclusively cytosolic. Using deletion and scanning mutagenesis we report that the first 10 amino acid residues of cyt-PTP(epsilon), in particular residues R4, K5, and R9, are critical components for its nuclear localization. We also establish that increased oxidative stress enhances accumulation of cyt-PTP(epsilon) in cell nuclei. Of the four known protein forms of PTP(epsilon), cyt-PTP(epsilon) is the only one which includes the extreme N-terminal sequence containing R4, K5, and R9. The role of the unique N terminus of cyt-PTP(epsilon) is therefore to regulate its subcellular localization. The existence of naturally occurring forms of PTP(epsilon) which lack this sequence and which are generated by translational and posttranslational mechanisms, suggests that nuclear localization of cyt-PTP(epsilon) can be actively regulated by cells.
- Published
- 2002
44. A mutant EGF-receptor defective in ubiquitylation and endocytosis unveils a role for Grb2 in negative signaling
- Author
-
Ari Elson, Menachem Katz, Thomas M. Jovin, Sara Lavi, Chanan Rubin, Keren Shtiegman, Hadassa Waterman, and Yosef Yarden
- Subjects
Ubiquitin-Protein Ligases ,Transfection ,Endocytosis ,Article ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Proto-Oncogene Proteins ,Animals ,Humans ,Proto-Oncogene Proteins c-cbl ,Epidermal growth factor receptor ,Ubiquitins ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,GRB2 Adaptor Protein ,General Immunology and Microbiology ,biology ,General Neuroscience ,fungi ,Proteins ,Ubiquitin ligase ,Cell biology ,ErbB Receptors ,Mutation ,biology.protein ,Cancer research ,Rabbits ,GRB2 ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Tyrosine kinase ,Signal Transduction - Abstract
Ligand-induced desensitization of the epidermal growth factor receptor (EGFR) is controlled by c-Cbl, a ubiquitin ligase that binds multiple signaling proteins, including the Grb2 adaptor. Consistent with a negative role for c-Cbl, here we report that defective Tyr1045 of EGFR, an inducible c-Cbl docking site, enhances the mitogenic response to EGF. Signaling potentiation is due to accelerated recycling of the mutant receptor and a concomitant defect in ligand-induced ubiquitylation and endocytosis of EGFR. Kinetic as well as morphological analyses of the internalization-defective mutant receptor imply that c- Cbl-mediated ubiquitylation sorts EGFR to endocytosis and to subsequent degradation in lysosomes. Unexpectedly, however, the mutant receptor displayed significant residual ligand-induced ubiquitylation, especially in the presence of an overexpressed c-Cbl. The underlying mechanism seems to involve recruitment of a Grb2 c-Cbl complex to Grb2-specific docking sites of EGFR, and concurrent acceleration of receptor ubiquitylation and desensitization. Thus, in addition to its well-characterized role in mediating positive signals, Grb2 can terminate signal transduction by accelerating c-Cbl-dependent sorting of active tyrosine kinases to destruction.
- Published
- 2002
45. Receptor Protein Tyrosine Phosphatase α-Mediated Enhancement of Rheumatoid Synovial Fibroblast Signaling and Promotion of Arthritis in Mice
- Author
-
Stephanie M, Stanford, Mattias N D, Svensson, Cristiano, Sacchetti, Caila A, Pilo, Dennis J, Wu, William B, Kiosses, Annelie, Hellvard, Brith, Bergum, German R Aleman, Muench, Christian, Elly, Yun-Cai, Liu, Jeroen, den Hertog, Ari, Elson, Jan, Sap, Piotr, Mydel, David L, Boyle, Maripat, Corr, Gary S, Firestein, and Nunzio, Bottini
- Subjects
Cell Survival ,Blotting, Western ,Apoptosis ,Enzyme-Linked Immunosorbent Assay ,Polymerase Chain Reaction ,Article ,Arthritis, Rheumatoid ,Mice ,Cell Movement ,Cell Adhesion ,Animals ,Phosphorylation ,Mice, Knockout ,Platelet-Derived Growth Factor ,Tumor Necrosis Factor-alpha ,Gene Expression Profiling ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Synovial Membrane ,Fibroblasts ,Arthritis, Experimental ,src-Family Kinases ,Focal Adhesion Protein-Tyrosine Kinases ,Gene Knockdown Techniques ,Disease Progression ,Ankle Joint ,Interleukin-1 ,Signal Transduction - Abstract
During rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLS) critically promote disease pathogenesis by aggressively invading the extracellular matrix of the joint. The focal adhesion kinase (FAK) signaling pathway is emerging as a contributor to the anomalous behavior of RA FLS. The receptor protein tyrosine phosphatase α (RPTPα), which is encoded by the PTPRA gene, is a key promoter of FAK signaling. The aim of this study was to investigate whether RPTPα mediates FLS aggressiveness and RA pathogenesis.Through RPTPα knockdown, we assessed FLS gene expression by quantitative polymerase chain reaction analysis and enzyme-linked immunosorbent assay, invasion and migration by Transwell assays, survival by annexin V and propidium iodide staining, adhesion and spreading by immunofluorescence microscopy, and activation of signaling pathways by Western blotting of FLS lysates. Arthritis development was examined in RPTPα-knockout (KO) mice using the K/BxN serum-transfer model. The contribution of radiosensitive and radioresistant cells to disease was evaluated by reciprocal bone marrow transplantation.RPTPα was enriched in the RA synovial lining. RPTPα knockdown impaired RA FLS survival, spreading, migration, invasiveness, and responsiveness to platelet-derived growth factor, tumor necrosis factor, and interleukin-1 stimulation. These phenotypes correlated with increased phosphorylation of Src on inhibitory Y(527) and decreased phosphorylation of FAK on stimulatory Y(397) . Treatment of RA FLS with an inhibitor of FAK phenocopied the knockdown of RPTPα. RPTPα-KO mice were protected from arthritis development, which was due to radioresistant cells.By regulating the phosphorylation of Src and FAK, RPTPα mediates proinflammatory and proinvasive signaling in RA FLS, correlating with the promotion of disease in an FLS-dependent model of RA.
- Published
- 2014
46. Protein tyrosine phosphatases ε and α perform nonredundant roles in osteoclasts
- Author
-
Ari Elson, Esther Arman, Eynat Finkelshtein, Jeroen den Hertog, Sutada Lotinun, Roland Baron, Einat Levy-Apter, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
musculoskeletal diseases ,animal structures ,Podosome ,Molecular Sequence Data ,Receptor-Like Protein Tyrosine Phosphatases ,Osteoclasts ,Protein tyrosine phosphatase ,Biology ,Inbred C57BL ,environment and public health ,Collagen Type I ,Serine ,Mice ,Animals ,Amino Acid Sequence ,Phosphorylation ,Molecular Biology ,Peptide sequence ,Tibia ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Class 4 ,Cell Biology ,Adhesion ,Articles ,Signaling ,Resorption ,Mice, Inbred C57BL ,enzymes and coenzymes (carbohydrates) ,src-Family Kinases ,Biochemistry ,Female ,Peptides ,Function (biology) - Abstract
The closely related tyrosine phosphatases PTPa and PTPe fulfill distinct roles in osteoclasts. The various effects of each PTP on podosome organization in osteoclasts are caused by their distinct N-termini. The function of PTPe in these cells requires the presence of its 12 N-terminal residues, in particular serine 2., Female mice lacking protein tyrosine phosphatase ε (PTP ε) are mildly osteopetrotic. Osteoclasts from these mice resorb bone matrix poorly, and the structure, stability, and cellular organization of their podosomal adhesion structures are abnormal. Here we compare the role of PTP ε with that of the closely related PTP α in osteoclasts. We show that bone mass and bone production and resorption, as well as production, structure, function, and podosome organization of osteoclasts, are unchanged in mice lacking PTP α. The varying effects of either PTP on podosome organization in osteoclasts are caused by their distinct N-termini. Osteoclasts express the receptor-type PTP α (RPTPa), which is absent from podosomes, and the nonreceptor form of PTP ε (cyt-PTPe), which is present in these structures. The presence of the unique 12 N-terminal residues of cyt-PTPe is essential for podosome regulation; attaching this sequence to the catalytic domains of PTP α enables them to function in osteoclasts. Serine 2 within this sequence regulates cyt-PTPe activity and its effects on podosomes. We conclude that PTPs α and ε play distinct roles in osteoclasts and that the N-terminus of cyt-PTPe, in particular serine 2, is critical for its function in these cells.
- Published
- 2014
47. Comparative study of protein tyrosine phosphatase-ɛ isoforms: membrane localization confers specificity in cellular signalling
- Author
-
Jannik N⊘rgaard ANDERSEN, Ari ELSON, Reiner LAMMERS, John RØMER, Jes Thorn CLAUSEN, Karin Bach MØLLER, and Niels Peter Hundahl MØLLER
- Subjects
Cell Biology ,Molecular Biology ,Biochemistry - Abstract
To study the influence of subcellular localization as a determinant of signal transduction specificity, we assessed the effects of wild-type transmembrane and cytoplasmic protein tyrosine phosphatase (PTP) ε on tyrosine kinase signalling in baby hamster kidney (BHK) cells overexpressing the insulin receptor (BHK-IR). The efficiency by which differently localized PTPε and PTPα variants attenuated insulin-induced cell rounding and detachment was determined in a functional clonal-selection assay and in stable cell lines. Compared with the corresponding receptor-type PTPs, the cytoplasmic PTPs (cytPTPs) were considerably less efficient in generating insulin-resistant clones, and exceptionally high compensatory expression levels were required to counteract phosphotyrosine-based signal transduction. Targeting of cytPTPε to the plasma membrane via the Lck-tyrosine kinase dual acylation motif restored high rescue efficiency and abolished the need for high cytPTPε levels. Consistent with these results, expression levels and subcellular localization of PTPε were also found to determine the phosphorylation level of cellular proteins including focal adhesion kinase (FAK). Furthermore, PTPε stabilized binding of phosphorylated FAK to Src, suggesting this complex as a possible mediator of the PTPε inhibitory response to insulin-induced cell rounding and detachment in BHK-IR cells. Taken together, the present localization–function study indicates that transcriptional control of the subcellular localization of PTPε may provide a molecular mechanism that determines PTPε substrate selectivity and isoform-specific function.
- Published
- 2001
48. Hypomyelination and increased activity of voltage-gated K+channels in mice lacking protein tyrosine phosphatase ϵ
- Author
-
Ari Elson, Alexander Sobko, Asher Peretz, Vera Shinder, Bernard Attali, and Hava Gil-Henn
- Subjects
medicine.medical_specialty ,Potassium Channels ,Down-Regulation ,Hyperphosphorylation ,Schwann cell ,Protein tyrosine phosphatase ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Kv1.5 Potassium Channel ,Mice ,chemistry.chemical_compound ,Shab Potassium Channels ,Internal medicine ,Peripheral Nervous System ,medicine ,Animals ,Molecular Biology ,Cells, Cultured ,Myelin Sheath ,General Immunology and Microbiology ,Voltage-gated ion channel ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,General Neuroscience ,Tyrosine phosphorylation ,Articles ,Precipitin Tests ,Mice, Mutant Strains ,Cell biology ,Electrophysiology ,Endocrinology ,medicine.anatomical_structure ,nervous system ,chemistry ,Receptor-Like Protein Tyrosine Phosphatases ,Potassium Channels, Voltage-Gated ,Schwann Cells ,Protein Tyrosine Phosphatases ,Ion Channel Gating ,Tyrosine kinase ,Delayed Rectifier Potassium Channels ,Protein Binding ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Protein tyrosine phosphatase epsilon (PTP epsilon) is strongly expressed in the nervous system; however, little is known about its physiological role. We report that mice lacking PTP epsilon exhibit hypomyelination of sciatic nerve axons at an early post-natal age. This occurs together with increased activity of delayed- rectifier, voltage-gated potassium (Kv) channels and with hyperphosphorylation of Kv1.5 and Kv2.1 Kv channel alpha-subunits in sciatic nerve tissue and in primary Schwann cells. PTP epsilon markedly reduces Kv1.5 or Kv2.1 current amplitudes in Xenopus oocytes. Kv2.1 associates with a substrate-trapping mutant of PTP epsilon, and PTP epsilon profoundly reduces Src- or Fyn-stimulated Kv2.1 currents and tyrosine phosphorylation in transfected HEK 293 cells. In all, PTP epsilon antagonizes activation of Kv channels by tyrosine kinases in vivo, and affects Schwann cell function during a critical period of Schwann cell growth and myelination.
- Published
- 2000
49. Protein tyrosine phosphatase ε increases the risk of mammary hyperplasia and mammary tumors in transgenic mice
- Author
-
Ari Elson
- Subjects
Genetically modified mouse ,Cancer Research ,medicine.medical_specialty ,Phosphoric monoester hydrolases ,Ratón ,Mammary gland ,Mice, Transgenic ,Protein tyrosine phosphatase ,Adenocarcinoma ,Biology ,medicine.disease_cause ,Mice ,Mammary Glands, Animal ,Pregnancy ,Internal medicine ,Gene expression ,Genetics ,medicine ,Animals ,Promoter Regions, Genetic ,Molecular Biology ,Hyperplasia ,Receptor-Like Protein Tyrosine Phosphatases, Class 4 ,Mammary Neoplasms, Experimental ,medicine.disease ,Molecular biology ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Endocrinology ,Mammary Tumor Virus, Mouse ,Female ,Protein Tyrosine Phosphatases ,Carcinogenesis - Abstract
Accurate phosphorylation of tyrosine residues in proteins plays a central role in regulation of cellular function. Although connections between aberrant tyrosine kinase activity and malignancy are well-established, significantly less is known about the roles of protein tyrosine phosphatases (PTPases) in tumorigenesis. We have previously shown that the transmembranal form of PTPase Epsilon (PTPepsilon) is upregulated in mouse mammary tumors initiated specifically by ras or neu, suggesting that PTPepsilon may play a role in transformation by these two oncogenes. In order to test this notion in vivo, we created transgenic mice that express elevated levels of PTPepsilon in their mammary epithelium by use of the MMTV promoter/enhancer. Following several cycles of pregnancy female MMTV-PTPepsilon mice uniformly developed pronounced and persistent mammary hyperplasia which was accompanied by residual milk production. Solitary mammary tumors were often detected secondary to mammary hyperplasia. The sporadic nature of the tumors, the long latency period prior to their development, and low levels of transgene expression in the tumors indicate that PTPepsilon provides a necessary, but insufficient, signal for oncogenesis. The results provide genetic evidence that PTPepsilon plays an accessory role in production of mammary tumors in a manner consistent with its upregulation in mammary tumors induced by ras or neu.
- Published
- 1999
50. Loss of p 21 increases sensitivity to ionizing radiation and delays the onset of lymphoma in atm -deficient mice
- Author
-
Philip Leder, Ari Elson, and Y. Alan Wang
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Lymphoma ,DNA damage ,Cell Cycle Proteins ,Context (language use) ,Ataxia Telangiectasia Mutated Proteins ,Protein Serine-Threonine Kinases ,Biology ,medicine.disease_cause ,Radiation Tolerance ,Ataxia Telangiectasia ,Mice ,Radiation sensitivity ,Cyclins ,medicine ,Animals ,Cells, Cultured ,Multidisciplinary ,Cerebellar ataxia ,Kinase ,Tumor Suppressor Proteins ,Proteins ,Fibroblasts ,Biological Sciences ,medicine.disease ,Mice, Mutant Strains ,DNA-Binding Proteins ,Ataxia-telangiectasia ,Cancer research ,medicine.symptom ,Signal transduction ,Carcinogenesis ,Gene Deletion ,Signal Transduction - Abstract
Ataxia telangiectasia (AT) is an autosomal recessive disorder characterized by growth retardation, cerebellar ataxia, oculocutaneous telangiectasias, and a high incidence of lymphomas and leukemias. In addition, AT patients are sensitive to ionizing radiation. Atm -deficient mice recapitulate most of the AT phenotype. p21 cip1/waf1 (p21 hereafter), an inhibitor of cyclin-dependent kinases, has been implicated in cellular senescence and response to γ-radiation-induced DNA damage. To study the role of p21 in ATM-mediated signal transduction pathways, we examined the combined effect of the genetic loss of atm and p21 on growth control, radiation sensitivity, and tumorigenesis. As might have been expected, our data provide evidence that p21 modifies the in vitro senescent response seen in AT fibroblasts. Further, it is a downstream effector of ATM-mediated growth control. In addition, however, we find that loss of p21 in the context of an atm -deficient mouse leads to a delay in thymic lymphomagenesis and an increase in acute radiation sensitivity in vivo (the latter principally because of effects on the gut epithelium). Modification of these two crucial aspects of the ATM phenotype can be related to an apparent increase in spontaneous apoptosis seen in tumor cells and in the irradiated intestinal epithelium of mice doubly null for atm and p21 . Thus, loss of p21 seems to contribute to tumor suppression by a mechanism that operates via a sensitized apoptotic response. These results have implications for cancer therapy in general and AT patients in particular.
- Published
- 1997
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.