28 results on '"MINK1"'
Search Results
2. Clinical Potential of Misshapen/NIKs-Related Kinase (MINK) 1—A Many-Sided Element of Cell Physiology and Pathology
- Author
-
Anna Kot, Dominika Koszewska, Błażej Ochman, and Elżbieta Świętochowska
- Subjects
MINK1 ,STRIPAK ,striatins ,TNIK ,MAP4Ks ,Wnt ,Biology (General) ,QH301-705.5 - Abstract
Misshapen/NIKs-related kinase (MINK) 1 belongs to the mammalian germinal center kinase (GCK) family. It contains the N-terminal, conserved kinase domain, a coiled-coil region, a proline-rich region, and a GCK, C-terminal domain with the Citron-NIK-Homology (CNH) domain. The kinase is an essential component of cellular signaling pathways, which include Wnt signaling, JNK signaling, pathways engaging Ras proteins, the Hippo pathway, and STRIPAK complexes. It thus contributes to regulating the cell cycle, apoptosis, cytoskeleton organization, cell migration, embryogenesis, or tissue homeostasis. MINK1 plays an important role in immunological responses, inhibiting Th17 and Th1 cell differentiation and regulating NLRP3 inflammasome function. It may be considered a link between ROS and the immunological system, and a potential antiviral target for human enteroviruses. The kinase has been implicated in the pathogenesis of sepsis, rheumatoid arthritis, asthma, SLE, and more. It is also involved in tumorigenesis and drug resistance in cancer. Silencing MINK1 reduces cancer cell migration, suggesting potential for new therapeutic approaches. Targeting MINK1 could be a promising treatment strategy for patients insensitive to current chemotherapies, and could improve their prognosis. Moreover, MINK1 plays an important role in the nervous system and the cardiovascular system development and function. The modulation of MINK1 activity could influence the course of neurodegenerative diseases, including Alzheimer’s disease. Further exploration of the activity of the kinase could also help in gaining more insight into factors involved in thrombosis or congenital heart disease. This review aims to summarize the current knowledge on MINK1, highlight its therapeutic and prognostic potential, and encourage more studies in this area.
- Published
- 2024
- Full Text
- View/download PDF
3. Regulation of MINK1 by APC, PKA and MARK3/4 and consequences for epithelial cell biology
- Author
-
Volk, Melanie, Nathke, Inke, MacKintosh, Carol, and Sknepnek, Rastko
- Subjects
571.5 ,APC ,MINK1 - Published
- 2021
4. Mink1 regulates spemann organizer cell fate in the xenopus gastrula via Hmga2.
- Author
-
Colleluori, Vaughn and Khokha, Mustafa K.
- Subjects
- *
WNT signal transduction , *GASTRULATION , *XENOPUS , *CONGENITAL heart disease , *HEART development , *INFANT mortality - Abstract
Congenital Heart Disease (CHD) is the most common birth defect and leading cause of infant mortality, yet molecular mechanisms explaining CHD remain mostly unknown. Sequencing studies are identifying CHD candidate genes at a brisk rate including MINK1 , a serine/threonine kinase. However, a plausible molecular mechanism connecting CHD and MINK1 is unknown. Here, we reveal that mink1 is required for proper heart development due to its role in left-right patterning. Mink1 regulates canonical Wnt signaling to define the cell fates of the Spemann Organizer and the Left-Right Organizer, a ciliated structure that breaks bilateral symmetry in the vertebrate embryo. To identify Mink1 targets, we applied an unbiased proteomics approach and identified the high mobility group architectural transcription factor, Hmga2. We report that Hmga2 is necessary and sufficient for regulating Spemann's Organizer. Indeed, we demonstrate that Hmga2 can induce Spemann Organizer cell fates even when β-catenin, a critical effector of the Wnt signaling pathway, is depleted. In summary, we discover a transcription factor, Hmga2, downstream of Mink1 that is critical for the regulation of Spemann's Organizer, as well as the LRO, defining a plausible mechanism for CHD. [Display omitted] • MINK1 is a novel regulator of left-right patterning and heart development. • MINK1 regulates canonical Wnt signaling during induction of the Spemann Organizer. • HMGA2 is an indirect target of MINK1 phosphorylation. • HMGA2 functions downstream of b-catenin during induction of Spemann Organizer. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Multi-Omic Investigations of a 17–19 Translocation Links MINK1 Disruption to Autism, Epilepsy and Osteoporosis.
- Author
-
Eisfeldt, Jesper, Schuy, Jakob, Stattin, Eva-Lena, Kvarnung, Malin, Falk, Anna, Feuk, Lars, and Lindstrand, Anna
- Subjects
- *
INDUCED pluripotent stem cells , *AUTISM , *EPILEPSY , *OSTEOPOROSIS , *NUCLEOTIDE sequencing , *ANAPLASTIC lymphoma kinase - Abstract
Balanced structural variants, such as reciprocal translocations, are sometimes hard to detect with sequencing, especially when the breakpoints are located in repetitive or insufficiently mapped regions of the genome. In such cases, long-range information is required to resolve the rearrangement, identify disrupted genes and, in symptomatic carriers, pinpoint the disease-causing mechanisms. Here, we report an individual with autism, epilepsy and osteoporosis and a de novo balanced reciprocal translocation: t(17;19) (p13;p11). The genomic DNA was analyzed by short-, linked- and long-read genome sequencing, as well as optical mapping. Transcriptional consequences were assessed by transcriptome sequencing of patient-specific neuroepithelial stem cells derived from induced pluripotent stem cells (iPSC). The translocation breakpoints were only detected by long-read sequencing, the first on 17p13, located between exon 1 and exon 2 of MINK1 (Misshapen-like kinase 1), and the second in the chromosome 19 centromere. Functional validation in induced neural cells showed that MINK1 expression was reduced by >50% in the patient's cells compared to healthy control cells. Furthermore, pathway analysis revealed an enrichment of changed neural pathways in the patient's cells. Altogether, our multi-omics experiments highlight MINK1 as a candidate monogenic disease gene and show the advantages of long-read genome sequencing in capturing centromeric translocations. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. LncRNA SNHG14 contributes to proinflammatory cytokine production in rheumatoid arthritis via the regulation of the miR‐17‐5p/MINK1‐JNK pathway.
- Author
-
Zhang, Jihui, Lei, Hongwei, and Li, Xiu
- Subjects
RHEUMATOID arthritis ,LINCRNA ,CYTOKINES ,NON-coding RNA ,JOINT diseases ,CIRCULAR RNA - Abstract
Rheumatoid arthritis (RA) is a widespread autoimmune disorder of the joints. Long noncoding RNAs (lncRNAs) have been reported to participate in the pathogenesis of RA by serving as competitive endogenous RNAs. LncRNA small nucleolar RNA host gene 14 (SNHG14) is involved in the development of various diseases. Here, we found that high expression of SNHG14 in RA was closely related to the disease activity. Functional assays indicated that SNHG14 knockdown obviously hampered phorbol myristate acetate‐activated THP‐1 (pTHP‐1) cell proliferation and proinflammatory cytokines production. In mechanism, SNHG14 served as a sponge of microRNA‐17‐5p (miR‐17‐5p), and misshapen like kinase 1 (MINK1) was a target of miR‐17‐5p. SNHG14 depletion‐induced inhibitory effects on cell proliferation and inflammatory response were reversed by MINK1 overexpression in macrophages. Moreover, SNHG14 promoted the jun N‐terminal kinase (JNK) signaling via the miR‐17‐5p/MINK1 axis. Overall, SNHG14 boosted the process of RA by MINK1 activating the JNK pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
7. Dual roles of misshapen/NIK-related kinase (MINK1) in osteoarthritis subtypes through the activation of TGFβ signaling.
- Author
-
Yu, D., Hu, J., Sheng, Z., Fu, G., Wang, Y., Chen, Y., Pan, Z., Zhang, X., Wu, Y., Sun, H., Dai, J., Lu, L., and Ouyang, H.
- Abstract
Objective: To identify the role of misshapen/NIK-related kinase (MINK1) in age-related Osteoarthritis (OA) and injury-induced OA, and the effects of enhanced TGFβ signaling in these progresses.Design: The effect of MINK1 was analyzed with MINK1 knock out (Mink1-/-) mice and C57BL/6J mice. OA progress was studied in age-related OA and instability-associated OA (destabilization of the medial meniscus, DMM) models. The murine knee joint was evaluated through histological staining, Osteoarthritis Research Society International (OARSI) scores, immunohistochemistry, and μCT analysis. Primary chondrocytes were isolated from wild type and Mink1-/- mice and subjected to osteogenic induction and Western blot analysis.Results: MINK1 is highly expressed during cartilage development and in normal cartilage. Mink1-/- mice displayed markedly lower OARSI scores, aggrecan degradation neoepitope positive cells and increased Safranin O and pSMAD2 staining in aging-related OA model. However, in injury-induced OA, loss of MINK1 accelerates extracellular matrix (ECM) destruction, osteophyte formation, and subchondral bone sclerosis. Accelerated subchondral bone remodeling in Mink1-/- mice was accompanied with increased numbers of nestin-positive mesenchymal stem cells (MSCs) and osterix-positive osteoprogenitors. pSMAD2 staining was increased in the subchondral bone marrow of Mink1-/- mice and overexpression of MINK1 inhibited SMAD2 phosphorylation in vitro.Conclusions: This study shows for the first time that activation of TGFβ/SMAD2 by MINK1 deficiency plays opposite roles in aging-related and injury-induced OA. MINK1 deficiency protects cartilage from degeneration in aging joints through increased SMAD2 activation in chondrocytes, while accelerating OA progress in injury-induced model through enhanced osteogenesis of MSCs in the subchondral bone. These findings provide insights for developing precision OA therapeutics targeting TGFβ/SMAD2 signaling. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
8. The Misshapen subfamily of Ste20 kinases regulate proliferation in the aging mammalian intestinal epithelium.
- Author
-
Li, Qi, Nirala, Niraj K., Chen, Hsi‐Ju, Nie, Yingchao, Wang, Wei, Zhang, Biliang, Czech, Michael P., Wang, Qi, Xu, Lan, Mao, Junhao, and Ip, Y. Tony
- Subjects
- *
KINASES , *MITOGEN-activated protein kinases , *EPITHELIUM , *GASTROINTESTINAL system , *INTESTINES , *TISSUE culture - Abstract
The intestinal epithelium has a high rate of cell turn over and is an excellent system to study stem cell‐mediated tissue homeostasis. The Misshapen subfamily of the Ste20 kinases in mammals consists of misshapen like kinase 1 (MINK1), mitogen‐activated protein kinase kinase kinase kinase 4 (MAP4K4), and TRAF2 and NCK interacting kinase (TNIK). Recent reports suggest that this subfamily has a novel function equal to the Hippo/MST subfamily as upstream kinases for Warts/Large tumor suppressor kinase (LATS) to suppress tissue growth. To study the in vivo functions of Mink1, Map4k4, and Tnik, we generated a compound knockout of these three genes in the mouse intestinal epithelium. The intestinal epithelia of the mutant animals were phenotypically normal up to approximately 12 months. The older animals then exhibited mildly increased proliferation throughout the lower GI tract. We also observed that the normally spatially organized Paneth cells in the crypt base became dispersed. The expression of one of the YAP pathway target genes Sox9 was increased while other target genes including CTGF did not show a significant change. Therefore, the Misshapen and Hippo subfamilies may have highly redundant functions to regulate growth in the intestinal epithelium, as illustrated in recent tissue culture models. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
9. <scp>LncRNA SNHG14</scp> contributes to proinflammatory cytokine production in rheumatoid arthritis via the regulation of the <scp>miR</scp> ‐17‐5p/ <scp>MINK1‐JNK</scp> pathway
- Author
-
Xiu Li, Jihui Zhang, and Hongwei Lei
- Subjects
Gene knockdown ,MAP Kinase Signaling System ,Cell growth ,Kinase ,Health, Toxicology and Mutagenesis ,General Medicine ,Protein Serine-Threonine Kinases ,Management, Monitoring, Policy and Law ,Biology ,Toxicology ,Proinflammatory cytokine ,Arthritis, Rheumatoid ,Pathogenesis ,MicroRNAs ,chemistry.chemical_compound ,MINK1 ,chemistry ,Phorbol ,Cancer research ,Cytokines ,Humans ,RNA, Long Noncoding ,Small nucleolar RNA ,Cell Proliferation - Abstract
Rheumatoid arthritis (RA) is a widespread autoimmune disorder of the joints. Long noncoding RNAs (lncRNAs) have been reported to participate in the pathogenesis of RA by serving as competitive endogenous RNAs. LncRNA small nucleolar RNA host gene 14 (SNHG14) is involved in the development of various diseases. Here, we found that high expression of SNHG14 in RA was closely related to the disease activity. Functional assays indicated that SNHG14 knockdown obviously hampered phorbol myristate acetate-activated THP-1 (pTHP-1) cell proliferation and proinflammatory cytokines production. In mechanism, SNHG14 served as a sponge of microRNA-17-5p (miR-17-5p), and misshapen like kinase 1 (MINK1) was a target of miR-17-5p. SNHG14 depletion-induced inhibitory effects on cell proliferation and inflammatory response were reversed by MINK1 overexpression in macrophages. Moreover, SNHG14 promoted the jun N-terminal kinase (JNK) signaling via the miR-17-5p/MINK1 axis. Overall, SNHG14 boosted the process of RA by MINK1 activating the JNK pathway.
- Published
- 2021
- Full Text
- View/download PDF
10. Priming of NLRP3 inflammasome activation by Msn kinase MINK1 in macrophages
- Author
-
Dante Neculai, Linrong Lu, Songquan Wu, Xuexiao Jin, Kaixiang Zhu, Xuai Lin, Hu Hu, Zhexu Chi, Sheng Chen, Di Wang, and Richard D Sloan
- Subjects
MINK1 kinase ,Inflammasomes ,Immunology ,Priming (immunology) ,Inflammation ,Pyrin domain ,Article ,Inflammasome ,Mice ,MINK1 ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Immunology and Allergy ,Phosphorylation ,Kinase activity ,Monocytes and macrophages ,integumentary system ,Kinase ,Chemistry ,Macrophages ,ROS ,NLRP3 inflammasome ,Cell biology ,Infectious Diseases ,medicine.symptom ,Reactive Oxygen Species ,medicine.drug - Abstract
The nucleotide-binding domain, leucine-rich-repeat containing family, pyrin domain-containing 3 (NLRP3) inflammasome is essential in inflammation and inflammatory disorders. Phosphorylation at various sites on NLRP3 differentially regulates inflammasome activation. The Ser725 phosphorylation site on NLRP3 is depicted in multiple inflammasome activation scenarios, but the importance and regulation of this site has not been clarified. The present study revealed that the phosphorylation of Ser725 was an essential step for the priming of the NLRP3 inflammasome in macrophages. We also showed that Ser725 was directly phosphorylated by misshapen (Msn)/NIK-related kinase 1 (MINK1), depending on the direct interaction between MINK1 and the NLRP3 LRR domain. MINK1 deficiency reduced NLRP3 activation and suppressed inflammatory responses in mouse models of acute sepsis and peritonitis. Reactive oxygen species (ROS) upregulated the kinase activity of MINK1 and subsequently promoted inflammasome priming via NLRP3 Ser725 phosphorylation. Eliminating ROS suppressed NLRP3 activation and reduced sepsis and peritonitis symptoms in a MINK1-dependent manner. Altogether, our study reveals a direct regulation of the NLRP3 inflammasome by Msn family kinase MINK1 and suggests that modulation of MINK1 activity is a potential intervention strategy for inflammasome-related diseases.
- Published
- 2021
- Full Text
- View/download PDF
11. The Ste20 Family Kinases MAP4K4, MINK1, and TNIK Converge to Regulate Stress-Induced JNK Signaling in Neurons.
- Author
-
Larhammar, Martin, Huntwork-Rodriguez, Sarah, Rudhard, York, Sengupta-Ghosh, Arundhati, and Lewcock, Joseph W.
- Subjects
- *
C-Jun N-terminal kinases , *NEURONS , *NERVOUS system development , *NEURODEGENERATION , *PHOSPHORYLATION , *LEUCINE zippers - Abstract
The c-Jun-iV-terminal kinase (JNK) signaling pathway regulates nervous system development, axon regeneration, and neuronal degeneration after acute injury or in chronic neurodegenerative disease. Dual leucine zipper kinase (DLK) is required for stress-induced JNK signaling in neurons, yet the factors that initiate DLK/JNK pathway activity remain poorly defined. In the present study, we identify the Ste20 kinases MAP4K4, misshapen-like kinase 1 (MINK1 or MAP4K6) and TNIK Traf2- and Nck-interacting kinase (TNIK or MAP4K7), as upstream regulators of DLK/JNK signaling in neurons. Using a trophic factor withdrawal-based model of neurodegeneration in both male and female embryonic mouse dorsal root ganglion neurons, we show that MAP4K4, MINK1, and TNIK act redundantly to regulate DLK activation and downstream JNK-dependent phosphorylation of c-Jun in response to stress. Targeting MAP4K4, MINK1, and TNIK, but not any of these kinases individually, is sufficient to protect neurons potently from degeneration. Pharmacological inhibition of MAP4Ks blocks stabilization and phosphorylation of DLK within axons and subsequent retrograde translocation of the JNK signaling complex to the nucleus. These results position MAP4Ks as important regulators of the DLK/JNK signaling pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
12. PRICKLE1 Contributes to Cancer Cell Dissemination through Its Interaction with mTORC2.
- Author
-
Daulat, Avais M., Bertucci, François, Audebert, Stéphane, Sergé, Arnauld, Finetti, Pascal, Josselin, Emmanuelle, Castellano, Rémy, Birnbaum, Daniel, Angers, Stéphane, and Borg, Jean-Paul
- Subjects
- *
CANCER cells , *CELL communication , *CANCER invasiveness , *PRICKLE1 gene , *MTOR protein , *PROGRESSION-free survival - Abstract
Summary Components of the evolutionarily conserved developmental planar cell polarity (PCP) pathway were recently described to play a prominent role in cancer cell dissemination. However, the molecular mechanisms by which PCP molecules drive the spread of cancer cells remain largely unknown. PRICKLE1 encodes a PCP protein bound to the promigratory serine/threonine kinase MINK1. We identify RICTOR, a member of the mTORC2 complex, as a PRICKLE1-binding partner and show that the integrity of the PRICKLE1-MINK1-RICTOR complex is required for activation of AKT, regulation of focal adhesions, and cancer cell migration. Disruption of the PRICKLE1-RICTOR interaction results in a strong impairment of breast cancer cell dissemination in xenograft assays. Finally, we show that upregulation of PRICKLE1 in basal breast cancers, a subtype characterized by high metastatic potential, is associated with poor metastasis-free survival. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
13. The serine/threonine kinase MINK1 directly regulates the function of promigratory proteins
- Author
-
Jeremy Ariey-Bonnet, Mônica Silveira Wagner, Pascal Finetti, Malgorzata Kowalczewska, Stéphane Audebert, François Bertucci, Luc Camoin, Jean-Paul Borg, Avais M. Daulat, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC), and Bertucci, François
- Subjects
Serine/threonine-specific protein kinase ,Chemistry ,Kinase ,Wnt signaling pathway ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Triple Negative Breast Neoplasms ,Cell Biology ,Protein complex assembly ,Protein Serine-Threonine Kinases ,Microtubules ,Cell biology ,Focal adhesion ,MINK1 ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Cell Movement ,Cell Line, Tumor ,Cell cortex ,Serine ,Phosphorylation ,Humans ,[INFO.INFO-BI]Computer Science [cs]/Bioinformatics [q-bio.QM] - Abstract
Upregulation of the developmental Wnt/planar cell polarity pathway is observed in many cancers and is associated with cancer development at early and late stages. We recently showed that PRICKLE1 and VANGL2, two core Wnt/PCP components, are overexpressed in triple negative breast cancer and associated with poor prognosis. PRICKLE1 is a cytoplasmic protein phosphorylated by the poorly described serine/threonine kinase MINK1 which triggers its localization at the plasma membrane, a key step for its function. Knockdown experiments have demonstrated that MINK1 and PRICKLE1 contribute to TNBC cell motility and spreading in vitro and in vivo. However, the identity of MINK1 substrates and the role of MINK1 enzymatic activity in this process have not yet been addressed issues.We carried out a phosphoproteomic strategy and identified novel MINK1 substrates including LL5β. LL5β is a membrane scaffold molecule that anchors microtubules at the cell cortex through its association with the plus-end MT proteins CLASPs to trigger focal adhesion disassembly. LL5β is a prominent member of the MINK1-PRICKLE1 protein complex and is directly phosphorylated by MINK1 that promotes its interaction with CLASP. Using a kinase inhibitor, we demonstrate that the enzymatic activity of MINK1 is involved in the protein complex assembly and localization, and cell migration. Analysis of gene expression data show that the concomitant up-regulation of PRICKLE1 and LL5β mRNA levels encoding MINK1 substrates is associated with a poor metastasis-free survival for TNBC patients. Altogether, our results suggest that MINK1 may represent a potential target in TNBC.
- Published
- 2021
- Full Text
- View/download PDF
14. Abstract 2290: The serine-threonine kinase Mink1 as a potential therapeutic target in triple negative breast cancer
- Author
-
Pascal Finetti, Rémy Castellano, Avais M. Daulat, Mônica Silveira Wagner, Luc Camoin, Malgorzata Kowalczewska, François Bertucci, Jean-Paul Borg, Stéphane Audebert, Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, and Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Serine/threonine-specific protein kinase ,Cancer Research ,Kinase ,Wnt signaling pathway ,Cancer ,Biology ,medicine.disease ,Phosphorylation cascade ,MINK1 ,Oncology ,Cancer research ,medicine ,Phosphorylation ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Triple-negative breast cancer - Abstract
The developmental Wnt/planar cell polarity (PCP) pathway is the most recently described branch of the Wnt signaling pathways and is strongly implicated in cancer development at early and late stages (PMID: 28718442 and PMID: 30952630). Upregulation of the Wnt/PCP components is observed in many cancers and is associated with cancer progression. However, how their molecular functions are regulated remains an open question. Recent data from our laboratory showed that Prickle1 and Vangl2, two core Wnt/PCP components, are overexpressed in Triple Negative Breast Cancers (TNBC) and associated with poor prognosis (PMID: 27184734 and 26754771). Prickle1 is a cytoplasmic protein phosphorylated by the serine/threonine kinase Mink1, which triggers Prickle1 localization at the plasma membrane and regulates its activity (PMID: 22037766). Activation of this axis contributes to breast cancer cell motility and metastatic spreading in vitro and in vivo (PMID: 27184734). To extend our knowledge of the Mink1 substrates, we carried out a phosphoproteomic strategy and identified LL5β. LL5β is a membrane scaffold molecule that anchors microtubules (MTs) at the cell cortex through its association with CLASP, a plus-end MT protein, to trigger focal adhesion disassembly. We found that LL5β is a prominent member of the Prickle1-associated protein complex and harbors a consensus motif for Mink1 phosphorylation. At the molecular level, we demonstrated a two-step phosphorylation cascade carried out by Mink1 that phosphorylates sequentially Prickle1 to mediate its association with LL5β, then LL5β to promote its interaction with CLASP. Finally, analysis of gene expression data suggests that the concomitant up-regulation of Prickle1 and LL5β mRNA levels is associated with a poor metastasis-free survival for TNBC patients. In an effort to counteract the Mink1-Prickle1-LL5β prometastatic pathway, we selected a Mink1 inhibitor that recapitulates all the phenotypes observed following the knockdown of Mink1 expression. This chemical compound is currently under investigation in preclinical assays using xenografted TNBC cell lines and patient-derived xenografts in nude mice. Citation Format: Avais Daulat, Monica Silveira Wagner, Malgorzata Kowalczewska, Pascal Finetti, Rémy Castellano, Stéphane Audebert, François Bertucci, Luc Camoin, Jean-Paul Borg. The serine-threonine kinase Mink1 as a potential therapeutic target in triple negative breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2290.
- Published
- 2021
- Full Text
- View/download PDF
15. Glucocorticoid receptor Thr524 phosphorylation by MINK1 induces interactions with 14-3-3 protein regulators
- Author
-
Anders Gunnarsson, Arjan Snijder, Christian Ottmann, Saleha Patel, Leonardo De Maria, Luc Brunsveld, Claire C. Munier, Carol MacKintosh, Karl Edman, Lisa Wissler, Perry Matthew, Marianna Longo, Chemical Biology, and ICMS Core
- Subjects
Threonine ,0301 basic medicine ,MINK1, misshapen-like kinase 1 ,Biochemistry ,LBD, ligand-binding domain ,Glucocorticoid receptor ,MINK1 ,HATU, 1-[bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-b]pyridinium 3-oxid hexafluorophosphate ,glucocorticoid receptor ,nuclear receptor ,ROCK1 ,Phosphorylation ,Kinase ,Chemistry ,TBST, tris-buffered saline with Polysorbate 20 ,DCM, dichloromethane ,Cell biology ,ROCK1, Rho-associated coiled-coil containing protein kinase 1 ,DMF, dimethylformamide ,FP, fluorescence polarization ,Protein Binding ,Research Article ,Transcriptional Activation ,MINK1 kinase ,TCEP, tris(2-carboxyethyl)phosphine ,SPR, surface plasmon resonance ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,Receptors, Glucocorticoid ,PDB, Protein Data Bank ,DIPEA, N,N-diisopropylethylamine ,Humans ,NTD, N-terminal domain ,NMP, N-methyl-2-pyrrolidone ,Protein kinase A ,Molecular Biology ,Transcription factor ,GR, glucocorticoid receptor ,030102 biochemistry & molecular biology ,14-3-3 protein ,ERα, estrogen receptor α ,Cell Biology ,KPSI, kilopounds per square inch ,HEK293 Cells ,protein–protein interaction ,030104 developmental biology ,14-3-3 Proteins ,Gene Expression Regulation ,Nuclear receptor ,DBD, DNA-binding domain ,Mutation ,ERRγ, estrogen-related receptor γ ,Co-IP, co-immunoprecipitation - Abstract
The glucocorticoid receptor (GR) is a ligand-dependent transcription factor that plays a central role in inflammation. The GR activity is also modulated via protein-protein interactions, including binding of 14-3-3 proteins induced by GR phosphorylation. However, the specific phosphorylation sites on the GR that trigger these interactions and their functional consequences are less clear. Hence, we sought to examine this system in more detail. We used phosphorylated GR peptides, biophysical studies, and X-ray crystallography to identify key residues within the ligand-binding domain of the GR, T524 and S617, whose phosphorylation results in binding of the representative 14-3-3 protein 14-3-3ζ. A kinase screen identified misshapen-like kinase 1 (MINK1) as responsible for phosphorylating T524 and Rho-associated protein kinase 1 for phosphorylating S617; cell-based approaches confirmed the importance of both GR phosphosites and MINK1 but not Rhoassociated protein kinase 1 alone in inducing GR-14-3-3 binding. Together our results provide molecular-level insight into 14-3-3-mediated regulation of the GR and highlight both MINK1 and the GR-14-3-3 axis as potential targets for future therapeutic intervention.
- Published
- 2021
- Full Text
- View/download PDF
16. KCNE1 is an auxiliary subunit of two distinct ion channel superfamilies
- Author
-
Pablo Ávalos Prado, Guillaume Sandoz, Bernard Attali, Christophe Duranton, Stephanie Häfner, Brigitte Wdziekonski, Jacques Barhanin, Yannick Comoglio, sandoz, guillaume, Institut de Biologie Valrose (IBV), Université Nice Sophia Antipolis (1965 - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), Laboratoire de PhysioMédecine Moléculaire (LP2M), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), Tel Aviv University (TAU), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Côte d'Azur (UCA)-Centre National de la Recherche Scientifique (CNRS), and Tel Aviv University [Tel Aviv]
- Subjects
Protein subunit ,voltage-dependent ion channels ,Biology ,anoctamin ,General Biochemistry, Genetics and Molecular Biology ,Kidney Tubules, Proximal ,Renin-Angiotensin System ,03 medical and health sciences ,Mice ,0302 clinical medicine ,MINK1 ,MinK1 ,Protein Domains ,Chloride Channels ,[SDV.BBM] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Animals ,Humans ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Gene ,Ion channel ,Anoctamin-1 ,030304 developmental biology ,0303 health sciences ,protein complexes ,single molecule fluorescence ,Polymorphism, Genetic ,Cardiac arrhythmia ,ancillary subunits ,Single-molecule experiment ,Potassium channel ,Cell biology ,Protein Subunits ,HEK293 Cells ,Potassium Channels, Voltage-Gated ,Chloride channel ,cardiovascular system ,KCNE1 ,Calcium ,Mutant Proteins ,IsK ,Peptides ,proximal convoluted tubule cells ,030217 neurology & neurosurgery ,potassium channel ,Protein Binding - Abstract
International audience; Determination of what is the specificity of subunits composing a protein complex is essential when studying gene variants on human pathophysiology. The pore-forming α-subunit KCNQ1, which belongs to the voltage-gated ion channel superfamily, associates to its β-auxiliary subunit KCNE1 to generate the slow cardiac potassium IKs current, whose dysfunction leads to cardiac arrhythmia. Using pharmacology, gene invalidation and single molecule fluorescence assays, we found that KCNE1 fulfils all criteria of a bona fide auxiliary subunit of the TMEM16A chloride channel, which belongs to the anoctamin superfamily. Strikingly, assembly with KCNE1 switches TMEM16A from a calcium-dependent to a voltage-dependent ion channel. Importantly, clinically relevant inherited mutations within the TMEM16A-regulating domain of KCNE1 abolish the TMEM16A modulation, suggesting that the TMEM16A-KCNE1 current may contribute to inherited pathologies. Altogether, these findings challenge the dogma of the specificity of auxiliary subunits regarding protein complexes and questions ion channel classification.
- Published
- 2020
- Full Text
- View/download PDF
17. The serine/threonine kinase MINK1 directly regulates the function of promigratory proteins.
- Author
-
Daulat AM, Wagner MS, Audebert S, Kowalczewska M, Ariey-Bonnet J, Finetti P, Bertucci F, Camoin L, and Borg JP
- Subjects
- Cell Line, Tumor, Cell Movement, Humans, Microtubules metabolism, Serine metabolism, Protein Serine-Threonine Kinases genetics, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism
- Abstract
Upregulation of the developmental Wnt planar cell polarity (Wnt/PCP) pathway is observed in many cancers and is associated with cancer development. We have recently shown that PRICKLE1, a core Wnt/PCP pathway component, is a marker of poor prognosis in triple-negative breast cancer (TNBC). PRICKLE1 is phosphorylated by the serine/threonine kinase MINK1 and contributes to TNBC cell motility and invasiveness. However, the identity of the substrates of MINK1 and the role of MINK1 enzymatic activity in this process remain to be addressed. We used a phosphoproteomic strategy to identify MINK1 substrates, including LL5β (also known as PHLDB2). LL5β anchors microtubules at the cell cortex through its association with CLASP proteins to trigger focal adhesion disassembly. LL5β is phosphorylated by MINK1, promoting its interaction with CLASP proteins. Using a kinase inhibitor, we demonstrate that the enzymatic activity of MINK1 is involved in PRICKLE1-LL5β complex assembly and localization, as well as in cell migration. Analysis of gene expression data reveals that the concomitant upregulation of levels of mRNA encoding PRICKLE1 and LL5β, which are MINK1 substrates, is associated with poor metastasis-free survival in TNBC patients. Taken together, our results suggest that MINK1 may represent a potential target for treatment of TNBC., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2022. Published by The Company of Biologists Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
18. The Ste20 Family Kinases MAP4K4, MINK1, and TNIK Converge to Regulate Stress-Induced JNK Signaling in Neurons
- Author
-
Joseph W. Lewcock, Sarah Huntwork-Rodriguez, York Rudhard, Martin Larhammar, and Arundhati Sengupta-Ghosh
- Subjects
Male ,0301 basic medicine ,MAP Kinase Signaling System ,Regulator ,Protein Serine-Threonine Kinases ,Biology ,Mice ,03 medical and health sciences ,MINK1 ,Stress, Physiological ,Ganglia, Spinal ,medicine ,Animals ,Axon ,Protein Kinase Inhibitors ,Cells, Cultured ,Research Articles ,Neurons ,Kinase ,General Neuroscience ,Neurodegeneration ,medicine.disease ,Rats ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,TNIK ,Phosphorylation ,Female ,Signal transduction - Abstract
The c-Jun-N-terminal kinase (JNK) signaling pathway regulates nervous system development, axon regeneration, and neuronal degeneration after acute injury or in chronic neurodegenerative disease. Dual leucine zipper kinase (DLK) is required for stress-induced JNK signaling in neurons, yet the factors that initiate DLK/JNK pathway activity remain poorly defined. In the present study, we identify the Ste20 kinases MAP4K4, misshapen-like kinase 1 (MINK1 or MAP4K6) and TNIK Traf2- and Nck-interacting kinase (TNIK or MAP4K7), as upstream regulators of DLK/JNK signaling in neurons. Using a trophic factor withdrawal-based model of neurodegeneration in both male and female embryonic mouse dorsal root ganglion neurons, we show that MAP4K4, MINK1, and TNIK act redundantly to regulate DLK activation and downstream JNK-dependent phosphorylation of c-Jun in response to stress. Targeting MAP4K4, MINK1, and TNIK, but not any of these kinases individually, is sufficient to protect neurons potently from degeneration. Pharmacological inhibition of MAP4Ks blocks stabilization and phosphorylation of DLK within axons and subsequent retrograde translocation of the JNK signaling complex to the nucleus. These results position MAP4Ks as important regulators of the DLK/JNK signaling pathway.SIGNIFICANCE STATEMENTNeuronal degeneration occurs in disparate circumstances: during development to refine neuronal connections, after injury to clear damaged neurons, or pathologically during disease. The dual leucine zipper kinase (DLK)/c-Jun-N-terminal kinase (JNK) pathway represents a conserved regulator of neuronal injury signaling that drives both neurodegeneration and axon regeneration, yet little is known about the factors that initiate DLK activity. Here, we uncover a novel role for a subfamily of MAP4 kinases consisting of MAP4K4, Traf2- and Nck-interacting kinase (TNIK or MAP4K7), and misshapen-like kinase 1 (MINK1 or MAP4K6) in regulating DLK/JNK signaling in neurons. Inhibition of these MAP4Ks blocks stress-induced retrograde JNK signaling and protects from neurodegeneration, suggesting that these kinases may represent attractive therapeutic targets.
- Published
- 2017
- Full Text
- View/download PDF
19. Flavone-based arylamides as potential anticancers: Design, synthesis and in vitro cell-based/cell-free evaluations
- Author
-
Ahmed H.E. Hassan, Kyung-Tae Lee, and Yong Sup Lee
- Subjects
Cell cycle checkpoint ,Colorectal cancer ,Antineoplastic Agents ,Flavones ,chemistry.chemical_compound ,Structure-Activity Relationship ,MINK1 ,Drug Discovery ,medicine ,Humans ,Cell Proliferation ,Pharmacology ,chemistry.chemical_classification ,Dose-Response Relationship, Drug ,Molecular Structure ,Kinase ,Organic Chemistry ,Masitinib ,General Medicine ,Cell Cycle Checkpoints ,medicine.disease ,Amides ,In vitro ,Molecular Docking Simulation ,chemistry ,TNIK ,Drug Design ,Cancer research ,Drug Screening Assays, Antitumor ,HT29 Cells - Abstract
Several arylamide-based antiproliferative agents are known and some of them are currently FDA-approved anticancer drugs. Provoked by the need to fill the existing room with new drugs, 31 compounds constituting a series of flavone-based arylamide derivatives were synthesized and biologically evaluated. Towards extensive evaluation, sixty diverse cancer cell lines representing nine cancer diseases of various origins have been used for evaluation of their efficacy, spectrum and potency. Two compounds 2aw and 2ax emerged as effective, broad-spectrum and potent anticancer agents that outperformed masitinibt and imatininb, which are FDA-approved anticancer drugs. Kinases profiling as possible targets for the potent compound 2aw showed that it might be a hit compound offering a starting point to develop inhibitors of STE20/GCK-IV kinase family members including HGK, TNIK and MINK1 kinases. Mechanistic study showed that compounds 2aw triggers cell cycle arrest in HT29 colon cancer cells. In conclusion, this work presents compound 2aw as a new broad-spectrum anticancer agent for further development of promising treatment of diverse cancers.
- Published
- 2019
20. Tool inhibitors and assays to interrogate the biology of the TRAF2 and NCK interacting kinase
- Author
-
I. Collie, James Robinson, Jon Read, Roland Bürli, Nicholas J. Brandon, Carly I. Dix, Bernard G. Van den Hoven, Leslie C. Conway, Elizabeth Flavell, Dean G. Brown, Gerard Donohoe, Juli Warwicker, Jayanta Mukherjee, Stephen J. Haggarty, Eileen McCall, Michelle Nguyen-McCarty, Christopher Lucaj, Stephen J. Moss, Andrew Madin, and Karolina Peciak
- Subjects
TRAF2 ,Clinical Biochemistry ,Pharmaceutical Science ,Context (language use) ,Computational biology ,Protein Serine-Threonine Kinases ,01 natural sciences ,Biochemistry ,MINK1 ,Drug Discovery ,Humans ,Molecular Biology ,Molecular Structure ,010405 organic chemistry ,Kinase ,Chemistry ,Organic Chemistry ,TNF Receptor-Associated Factor 2 ,Small molecule ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,TNIK ,Synaptic plasticity ,Schizophrenia ,Molecular Medicine ,Biological Assay ,Function (biology) - Abstract
The TRAF2 and NCK interacting kinase (TNIK) has been proposed to play a role in cytoskeletal organization and synaptic plasticity and has been linked, among others, to neurological disorders. However, target validation efforts for TNIK have been hampered by the limited kinase selectivity of small molecule probes and possible functional compensation in mouse models. Both issues are at least in part due to its close homology to the kinases MINK1 (or MAP4K6) and MAP4K4 (or HGK). As part of our interest in validating TNIK as a therapeutic target for neurological diseases, we set up a panel of biochemical and cellular assays, which are described herein. We then examined the activity of known amino-pyridine-based TNIK inhibitors (1, 3) and prepared structurally very close analogs that lack the ability to inhibit the target. We also developed a structurally orthogonal, naphthyridine-based TNIK inhibitor (9) and an inactive control molecule of the same chemical series. These validated small-molecule probes will enable dissection of the function of TNIK family in the context of human disease biology.
- Published
- 2019
21. Suppression of Th17 cell differentiation by misshapen/NIK-related kinase MINK1
- Author
-
Di Wang, Shunli Dong, Xuetao Cao, Linrong Lu, Xuexiao Jin, Yuanjun Qiu, Guotong Fu, Yuehai Ke, Hu Hu, Qin Xu, Harvey Cantor, Xiang Gao, Ting Xu, and Jianli Wang
- Subjects
0301 basic medicine ,animal structures ,Cellular differentiation ,Immunology ,Regulator ,Biology ,Article ,03 medical and health sciences ,MINK1 ,Transforming Growth Factor beta ,medicine ,Immunology and Allergy ,Research Articles ,chemistry.chemical_classification ,Reactive oxygen species ,Kinase ,Experimental autoimmune encephalomyelitis ,Germinal center ,Cell Differentiation ,medicine.disease ,Molecular biology ,Cell biology ,030104 developmental biology ,chemistry ,Phosphorylation ,Th17 Cells ,Reactive Oxygen Species - Abstract
Fu et al. demonstrate that reactive oxygen species (ROS)–sensing molecule misshapen/NIK-related kinase 1 (MINK1) can specifically suppress Th 17 cell differentiation through direct phosphorylation of SMAD2. This study provides the mechanism of how ROS limit inflammatory response and unveils the potential health risk of antioxidant supplementation., T helper type 17 cells (Th17 cells) are major contributors to many autoimmune diseases. In this study, we demonstrate that the germinal center kinase family member MINK1 (misshapen/NIK-related kinase 1) negatively regulates Th17 cell differentiation. The suppressive effect of MINK1 on induction of Th17 cells is mediated by the inhibition of SMAD2 activation through direct phosphorylation of SMAD2 at the T324 residue. The importance of MINK1 to Th17 cell differentiation was strengthened in the animal model of experimental autoimmune encephalomyelitis (EAE). Moreover, we show that the reactive oxygen species (ROS) scavenger N-acetyl cysteine boosts Th17 cell differentiation in a MINK1-dependent manner and exacerbates the severity of EAE. Thus, we have not only established MINK1 as a critical regulator of Th17 cell differentiation, but also clarified that accumulation of ROS may limit the generation of Th17 cells. The contribution of MINK1 to ROS-regulated Th17 cell differentiation may suggest an important mechanism for the development of autoimmune diseases influenced by antioxidant dietary supplements.
- Published
- 2017
22. Identification of endogenous Adenomatous polyposis coli interaction partners and β-catenin-independent targets by proteomics
- Author
-
Joao A. Paulo, Alejandro Rojaa, Inke S. Näthke, Nicholas Loyer, Olesja Popow, Ian P. Newton, Jens Januschke, Michael H. Tatham, and Kevin M. Haigis
- Subjects
MINK1 ,biology ,Adenomatous polyposis coli ,Kinase ,Colorectal cancer ,Catenin ,Mutant ,biology.protein ,medicine ,Wnt signaling pathway ,medicine.disease ,Proteomics ,Cell biology - Abstract
SummaryAdenomatous polyposis coli(APC) is the most frequently mutated gene in colorectal cancer. APC negatively regulates the pro-proliferative Wnt signaling pathway by promoting the degradation of β-catenin, but the extent to which APC exerts Wnt/β-catenin-independent tumor suppressive activity is unclear. To identify interaction partners and β-catenin-independent targets of endogenous, full-length APC, we applied label-free and multiplexed TMT mass spectrometry. Affinity enrichment-mass spectrometry revealed over 150 previously unidentified APC interaction partners. Moreover, our global proteomic analysis revealed that roughly half of the protein expression changes that occur in response to APC loss are independent of β-catenin. By combining these two analyses, we identified Misshapen-like kinase 1 (MINK1) as a putative substrate of an alternative APC-containing destruction complex and provide evidence for the potential contribution of MINK1 toAPCmutant phenotypes. Collectively, our results highlight the extent and importance of Wnt-independent APC functions in epithelial biology and disease.
- Published
- 2018
- Full Text
- View/download PDF
23. Identification of Eendogenous Adenomatous Polyposis Coli Interaction Partners and β-Catenin-Independent Targets by Proteomics
- Author
-
Kevin M. Haigis, Joao A. Paulo, Olesja Popow, Michael H. Tatham, Ian P. Newton, Jens Januschke, Nicolas Loyer, Alejandro Rojas-Fernandez, and Inke S. Näthke
- Subjects
MINK1 ,biology ,Colorectal cancer ,Adenomatous polyposis coli ,Kinase ,Catenin ,Mutant ,medicine ,Wnt signaling pathway ,biology.protein ,Cancer research ,medicine.disease ,Proteomics - Abstract
Adenomatous polyposis coli (APC) is the most frequently mutated gene in colorectal cancer. APC negatively regulates the pro-proliferative Wnt signaling pathway by promoting the degradation of β-catenin, but the extent to which APC exerts Wnt/β-catenin-independent tumor suppressive activity is unclear. To identify interaction partners and β-catenin-independent targets of endogenous, full-length APC, we applied label-free and multiplexed TMT mass spectrometry. Affinity enrichment-mass spectrometry revealed over 150 previously unidentified APC interaction partners. Moreover, our global proteomic analysis revealed that roughly half of the protein expression changes that occur in response to APC loss are independent of β-catenin. By combining these two analyses, we identified Misshapen-like kinase 1 (MINK1) as a putative substrate of an alternative APC-containing destruction complex and provide evidence for the potential contribution of MINK1 to APC mutant phenotypes. Collectively, our results highlight the extent and importance of Wntindependent APC functions in epithelial biology and disease.
- Published
- 2018
- Full Text
- View/download PDF
24. KCNE1 is an auxiliary subunit of two distinct ion channel superfamilies.
- Author
-
Ávalos Prado, Pablo, Häfner, Stephanie, Comoglio, Yannick, Wdziekonski, Brigitte, Duranton, Christophe, Attali, Bernard, Barhanin, Jacques, and Sandoz, Guillaume
- Subjects
- *
ION channels , *CHLORIDE channels , *VOLTAGE-gated ion channels , *INTRACELLULAR calcium , *ARRHYTHMIA , *ANGIOTENSIN II , *SINGLE molecules - Abstract
Determination of what is the specificity of subunits composing a protein complex is essential when studying gene variants on human pathophysiology. The pore-forming α-subunit KCNQ1, which belongs to the voltage-gated ion channel superfamily, associates to its β-auxiliary subunit KCNE1 to generate the slow cardiac potassium I Ks current, whose dysfunction leads to cardiac arrhythmia. Using pharmacology, gene invalidation, and single-molecule fluorescence assays, we found that KCNE1 fulfils all criteria of a bona fide auxiliary subunit of the TMEM16A chloride channel, which belongs to the anoctamin superfamily. Strikingly, assembly with KCNE1 switches TMEM16A from a calcium-dependent to a voltage-dependent ion channel. Importantly, clinically relevant inherited mutations within the TMEM16A-regulating domain of KCNE1 abolish the TMEM16A modulation, suggesting that the TMEM16A-KCNE1 current may contribute to inherited pathologies. Altogether, these findings challenge the dogma of the specificity of auxiliary subunits regarding protein complexes and questions ion channel classification. • KCNE1 can serve as an auxiliary subunit of two superfamilies of ion channels • KCNE1 is an auxiliary subunit of the TMEM16A chloride channel • TMEM16A-KCNE1 channel mediates a voltage-gated Cl− current independently of Ca2+ • KCNE1 S38G and R32H polymorphisms abolish the KCNE1-TMEM16A Cl− current KCNE1, a β-subunit of the cardiac KCNQ1 channel belonging to the voltage-dependent channel superfamily, serves also as an auxiliary subunit of the anoctamin superfamily member TMEM16A, a calcium-activated chloride channel. By interacting with TMEM16A, KCNE1 induces a voltage-dependent current in the absence of intracellular calcium elevation dynamically regulated by angiotensin II in native tissue. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. PRICKLE1 contributes to cancer cell dissemination through its interaction with mTORC2
- Author
-
Pascal Finetti, Rémy Castellano, Jean-Paul Borg, Emmanuelle Josselin, François Bertucci, Avais M. Daulat, Stéphane Audebert, Daniel Birnbaum, Stephane Angers, Arnauld Sergé, Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Aix Marseille Université (AMU), Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Canada, 9Department of Biochemistry, Faculty of Medicine, University of Toronto, This work was funded by La Ligue Nationale Contre le Cancer (Label Ligue JPB and DB, and post-doctoral fellowship to AMD), Fondation de France (post-doctoral fellowship to AMD), Fondation ARC pour la Recherche sur le Cancer (grant to JPB and AS), and SIRIC (INCa-DGOS-Inserm 6038, fellowship to AMD). The Marseille Proteomics (IBiSA) and TrGET platforms are supported by Institut Paoli-Calmettes (IPC) and Canceropôle PACA., Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), and BORG, Jean-Paul
- Subjects
0301 basic medicine ,MINK1 ,mTORC2 ,Cell Movement ,Protein Interaction Mapping ,Neoplasm Metastasis ,Phosphorylation ,Genes, Dominant ,Kinase ,TOR Serine-Threonine Kinases ,cancer cell migration ,LIM Domain Proteins ,Prognosis ,3. Good health ,Cell biology ,Up-Regulation ,Female ,Protein Binding ,Breast Neoplasms ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Mechanistic Target of Rapamycin Complex 2 ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Protein Serine-Threonine Kinases ,General Biochemistry, Genetics and Molecular Biology ,Focal adhesion ,03 medical and health sciences ,Downregulation and upregulation ,Protein Domains ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Cell Line, Tumor ,Humans ,Molecular Biology ,Protein kinase B ,[SDV.BC] Life Sciences [q-bio]/Cellular Biology ,Cell Proliferation ,Focal Adhesions ,PRICKLE1 ,Cell growth ,Tumor Suppressor Proteins ,Cell Biology ,030104 developmental biology ,Rapamycin-Insensitive Companion of mTOR Protein ,Multiprotein Complexes ,Cancer cell ,Cancer research ,Carrier Proteins ,Proto-Oncogene Proteins c-akt ,Developmental Biology - Abstract
International audience; Components of the evolutionarily conserved developmental planar cell polarity (PCP) pathway were recently described to play a prominent role in cancer cell dissemination. However, the molecular mechanisms by which PCP molecules drive the spread of cancer cells remain largely unknown. PRICKLE1 encodes a PCP protein bound to the promigratory serine/threonine kinase MINK1. We identify RICTOR, a member of the mTORC2 complex, as a PRICKLE1-binding partner and show that the integrity of the PRICKLE1-MINK1-RICTOR complex is required for activation of AKT, regulation of focal adhesions, and cancer cell migration. Disruption of the PRICKLE1-RICTOR interaction results in a strong impairment of breast cancer cell dissemination in xenograft assays. Finally, we show that upregulation of PRICKLE1 in basal breast cancers, a subtype characterized by high metastatic potential, is associated with poor metastasis-free survival.
- Published
- 2016
- Full Text
- View/download PDF
26. Glomerular Immune Complex Injury in Aleutian Disease of Mink1
- Author
-
G. Trautwein and R. Müller-Peddinghaus
- Subjects
MINK1 ,business.industry ,Immunology ,medicine ,Glomerulonephritis ,medicine.disease ,Aleutian disease ,business ,Immune complex - Published
- 2015
- Full Text
- View/download PDF
27. MINK1: The missing link between ROS and its inhibition of Th17 cells
- Author
-
Gustavo J. Martinez
- Subjects
0301 basic medicine ,Male ,Encephalomyelitis, Autoimmune, Experimental ,Cellular differentiation ,Immunology ,Cell generation ,Smad2 Protein ,Biology ,News ,Protein Serine-Threonine Kinases ,Insights ,Negative regulator ,03 medical and health sciences ,Mice ,MINK1 ,Immunology and Allergy ,Animals ,chemistry.chemical_classification ,Mice, Knockout ,Reactive oxygen species ,Kinase ,Cell Differentiation ,Transforming growth factor beta ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,chemistry ,biology.protein ,Th17 Cells ,Reactive Oxygen Species ,Transforming growth factor - Abstract
In this issue of JEM, Fu et al. identified the kinase Mink1 as a novel negative regulator of Th17 cell generation. Mink1, activated by reactive oxygen species (ROS), prevents TGF-β activation of Smad2, therefore limiting Th17 cell differentiation., In this issue of JEM, Fu et al. (https://doi.org/10.1084/jem.20161120) identified the kinase Mink1 as a novel negative regulator of Th17 cell generation. Mink1, activated by reactive oxygen species (ROS), prevents TGF-β activation of Smad2, therefore limiting Th17 cell differentiation.
- Published
- 2017
28. Abstract 4899: The regulation by kinases of the expression of human major histocompatibility class I molecules
- Author
-
George Mo, Eusebio Manchado, Elliott J. Brea, Neal Rosen, David A. Scheinberg, Ron S. Gejman, Claire Y. Oh, Ralph Garippa, and Patrizia Mondello
- Subjects
Cancer Research ,MINK1 ,Oncology ,biology ,Kinase ,MAP2K1 ,T-cell receptor ,Antigen presentation ,biology.protein ,Cancer research ,Kinome ,Human leukocyte antigen ,Major histocompatibility complex - Abstract
The major histocompatability complex (MHC) is a central receptor in the adaptive immune response and is the underlying target of several effective therapies for cancer. Druggable kinases may provide the opportunity to modulate the immune response toward MHC. However, the regulation of MHC-I by kinases is largely unstudied, even though many patients with cancer are receiving therapeutic kinase inhibitors. The entire human kinome was screened using a pooled shRNA interference-based approach in a human mesothelioma cell line to uncover kinase regulators of MHC-I. Negative and positive regulators of cell surface HLA levels were discovered. A subset of highly scoring positive and negative kinase hits were subsequently validated by additional RNAi, and pharmacologic inhibitors when available. MAP2K1 (MEK), EGFR, and RET were validated as negative regulators of HLA expression in multiple cancer types. We mapped the pathways responsible for increased HLA upon kinase inhibition. Interestingly, inhibition of the MAP Kinase pathway broadly influenced expression of other components of the antigen presentation machinery. Moreover, DDR2 and MINK1 were shown to positively regulate HLA-A*02:01. This had therapeutic relevance, as shown with a therapeutic TCR mimic antibody to a MHC/peptide complex. Druggable kinases may thus serve as immediately applicable targets for modulating immunotherapy for many diseases. Citation Format: Elliott J. Brea, Claire Oh, Eusebio Manchado, Ron Gejman, George Mo, Patrizia Mondello, Ralph Garippa, Neal Rosen, David A. Scheinberg. The regulation by kinases of the expression of human major histocompatibility class I molecules. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4899.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.