1
|
Shakya R, Suraneni P, Zaslavsky A, Rahi A, Magdongon CB, Gajjela R, Mattamana BB, Varma D. The Hexosamine Biosynthetic Pathway alters the cytoskeleton to modulate cell proliferation and migration in metastatic prostate cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.14.618283. [PMID: 39464080 PMCID: PMC11507681 DOI: 10.1101/2024.10.14.618283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Castration-resistant prostate cancer (CRPC) progresses despite androgen deprivation therapy, as cancer cells adapt to grow without testosterone, becoming more aggressive and prone to metastasis. CRPC biology complicates the development of effective therapies, posing challenges for patient care. Recent gene-expression and metabolomics studies highlight the Hexosamine Biosynthetic Pathway (HBP) as a critical player, with key components like GNPNAT1 and UAP1 being downregulated in metastatic CRPC. GNPNAT1 knockdown has been shown to increase cell proliferation and metastasis in CRPC cell lines, though the mechanisms remain unclear. To investigate the cellular basis of these CRPC phenotypes, we generated a CRISPR-Cas9 knockout model of GNPNAT1 in 22Rv1 CRPC cells, analyzing its impact on metabolomic, glycoproteomic, and transcriptomic profiles of cells. We hypothesize that HBP inhibition disrupts the cytoskeleton, altering mitotic progression and promoting uncontrolled growth. GNPNAT1 KO cells showed reduced levels of cytoskeletal filaments, such as actin and microtubules, leading to cell structure disorganization and chromosomal mis-segregation. GNPNAT1 inhibition also activated PI3K/AKT signaling, promoting proliferation, and impaired cell adhesion by mislocalizing EphB6, enhancing migration via the RhoA pathway and promoting epithelial-to-mesenchymal transition. These findings suggest that HBP plays a critical role in regulating CRPC cell behavior, and targeting this pathway could provide a novel therapeutic approach.
Collapse
Affiliation(s)
- Rajina Shakya
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Praveen Suraneni
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Alexander Zaslavsky
- Department of Urology, University of Michigan Medical School, Ann Harbor, MI 48108, USA
| | - Amit Rahi
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Christine B Magdongon
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Raju Gajjela
- Proteomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Basil B Mattamana
- Proteomics Core, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dileep Varma
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
2
|
Wolf L, Vogt J, Alber J, Franjic D, Feger M, Föller M. PKC regulates αKlotho gene expression in MDCK and NRK-52E cells. Pflugers Arch 2024; 476:75-86. [PMID: 37773536 PMCID: PMC10758369 DOI: 10.1007/s00424-023-02863-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 10/01/2023]
Abstract
Particularly expressed in the kidney, αKlotho is a transmembrane protein that acts together with bone hormone fibroblast growth factor 23 (FGF23) to regulate renal phosphate and vitamin D homeostasis. Soluble Klotho (sKL) is released from the transmembrane form and controls various cellular functions as a paracrine and endocrine factor. αKlotho deficiency accelerates aging, whereas its overexpression favors longevity. Higher αKlotho abundance confers a better prognosis in cardiovascular and renal disease owing to anti-inflammatory, antifibrotic, or antioxidant effects and tumor suppression. Serine/threonine protein kinase C (PKC) is ubiquitously expressed, affects several cellular responses, and is also implicated in heart or kidney disease as well as cancer. We explored whether PKC is a regulator of αKlotho. Experiments were performed in renal MDCK or NRK-52E cells and PKC isoform and αKlotho expression determined by qRT-PCR and Western Blotting. In both cell lines, PKC activation with phorbol ester phorbol-12-myristate-13-acetate (PMA) downregulated, while PKC inhibitor staurosporine enhanced αKlotho mRNA abundance. Further experiments with PKC inhibitor Gö6976 and RNA interference suggested that PKCγ is the major isoform for the regulation of αKlotho gene expression in the two cell lines. In conclusion, PKC is a negative regulator of αKlotho gene expression, an effect which may be relevant for the unfavorable effect of PKC on heart or kidney disease and tumorigenesis.
Collapse
Affiliation(s)
- Lisa Wolf
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany
| | - Julia Vogt
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany
| | - Jana Alber
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany
| | - Domenic Franjic
- Core Facility Hohenheim, Data and Statistical Consulting, University of Hohenheim, 70599, Stuttgart, Germany
| | - Martina Feger
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany.
| |
Collapse
|
3
|
Iglesias González PA, Valdivieso ÁG, Santa-Coloma TA. The G protein-coupled receptor GPRC5A-a phorbol ester and retinoic acid-induced orphan receptor with roles in cancer, inflammation, and immunity. Biochem Cell Biol 2023; 101:465-480. [PMID: 37467514 DOI: 10.1139/bcb-2022-0352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023] Open
Abstract
GPRC5A is the first member of a new class of orphan receptors coupled to G proteins, which also includes GPRC5B, GPRC5C, and GPRC5D. Since its cloning and identification in the 1990s, substantial progress has been made in understanding the possible functions of this receptor. GPRC5A has been implicated in a variety of cellular events, such as cytoskeleton reorganization, cell proliferation, cell cycle regulation, migration, and survival. It appears to be a central player in different pathological processes, including tumorigenesis, inflammation, immune response, and tissue damage. The levels of GPRC5A expression differ depending on the type of cancer, with increased expression in colon, pancreas, and prostate cancers; decreased expression in lung cancer; and varied results in breast cancer. In this review, we discuss the early discovery of GPRC5A as a phorbol ester-induced gene and later as a retinoic acid-induced gene, its regulation, and its participation in important canonical pathways related to numerous types of tumors and inflammatory processes. GPRC5A represents a potential new target for cancer, inflammation, and immunity therapies.
Collapse
Affiliation(s)
- Pablo A Iglesias González
- Laboratory of Cell and Molecular Biology, Institute for Biomedical Research (BIOMED), National Scientific and Technical Research Council (CONICET), Pontifical Catholic University of Argentina (UCA), Argentina
| | - Ángel G Valdivieso
- Laboratory of Cell and Molecular Biology, Institute for Biomedical Research (BIOMED), National Scientific and Technical Research Council (CONICET), Pontifical Catholic University of Argentina (UCA), Argentina
| | - Tomás A Santa-Coloma
- Laboratory of Cell and Molecular Biology, Institute for Biomedical Research (BIOMED), National Scientific and Technical Research Council (CONICET), Pontifical Catholic University of Argentina (UCA), Argentina
| |
Collapse
|
4
|
Gutiérrez-Casares JR, Quintero J, Segú-Vergés C, Rodríguez Monterde P, Pozo-Rubio T, Coma M, Montoto C. In silico clinical trial evaluating lisdexamfetamine's and methylphenidate's mechanism of action computational models in an attention-deficit/hyperactivity disorder virtual patients' population. Front Psychiatry 2023; 14:939650. [PMID: 37333910 PMCID: PMC10273406 DOI: 10.3389/fpsyt.2023.939650] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 04/21/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Attention-deficit/hyperactivity disorder (ADHD) is an impairing psychiatric condition with the stimulants, lisdexamfetamine (LDX), and methylphenidate (MPH), as the first lines pharmacological treatment. Methods Herein, we applied a novel in silico method to evaluate virtual LDX (vLDX) and vMPH as treatments for ADHD applying quantitative systems pharmacology (QSP) models. The objectives were to evaluate the model's output, considering the model characteristics and the information used to build them, to compare both virtual drugs' efficacy mechanisms, and to assess how demographic (age, body mass index, and sex) and clinical characteristics may affect vLDX's and vMPH's relative efficacies. Results and Discussion We molecularly characterized the drugs and pathologies based on a bibliographic search, and generated virtual populations of adults and children-adolescents totaling 2,600 individuals. For each virtual patient and virtual drug, we created physiologically based pharmacokinetic and QSP models applying the systems biology-based Therapeutic Performance Mapping System technology. The resulting models' predicted protein activity indicated that both virtual drugs modulated ADHD through similar mechanisms, albeit with some differences. vMPH induced several general synaptic, neurotransmitter, and nerve impulse-related processes, whereas vLDX seemed to modulate neural processes more specific to ADHD, such as GABAergic inhibitory synapses and regulation of the reward system. While both drugs' models were linked to an effect over neuroinflammation and altered neural viability, vLDX had a significant impact on neurotransmitter imbalance and vMPH on circadian system deregulation. Among demographic characteristics, age and body mass index affected the efficacy of both virtual treatments, although the effect was more marked for vLDX. Regarding comorbidities, only depression negatively impacted both virtual drugs' efficacy mechanisms and, while that of vLDX were more affected by the co-treatment of tic disorders, the efficacy mechanisms of vMPH were disturbed by wide-spectrum psychiatric drugs. Our in silico results suggested that both drugs could have similar efficacy mechanisms as ADHD treatment in adult and pediatric populations and allowed raising hypotheses for their differential impact in specific patient groups, although these results require prospective validation for clinical translatability.
Collapse
Affiliation(s)
- José Ramón Gutiérrez-Casares
- Unidad Ambulatoria de Psiquiatría y Salud Mental de la Infancia, Niñez y Adolescencia, Hospital Perpetuo Socorro, Badajoz, Spain
| | - Javier Quintero
- Servicio de Psiquiatría, Hospital Universitario Infanta Leonor, Universidad Complutense, Madrid, Spain
| | - Cristina Segú-Vergés
- Anaxomics Biotech, Barcelona, Spain
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | | | | | | | - Carmen Montoto
- Medical Department, Takeda Farmacéutica España, Madrid, Spain
| |
Collapse
|
5
|
An mTORC1 to HRI signaling axis promotes cytotoxicity of proteasome inhibitors in multiple myeloma. Cell Death Dis 2022; 13:969. [PMID: 36400754 PMCID: PMC9674573 DOI: 10.1038/s41419-022-05421-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 11/05/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
Multiple myeloma (MM) causes approximately 20% of deaths from blood cancers. Notwithstanding significant therapeutic progress, such as with proteasome inhibitors (PIs), MM remains incurable due to the development of resistance. mTORC1 is a key metabolic regulator, which frequently becomes dysregulated in cancer. While mTORC1 inhibitors reduce MM viability and synergize with other therapies in vitro, clinically, mTORC1 inhibitors are not effective for MM. Here we show that the inactivation of mTORC1 is an intrinsic response of MM to PI treatment. Genetically enforced hyperactivation of mTORC1 in MM was sufficient to compromise tumorigenicity in mice. In vitro, mTORC1-hyperactivated MM cells gained sensitivity to PIs and hypoxia. This was accompanied by increased mitochondrial stress and activation of the eIF2α kinase HRI, which initiates the integrated stress response. Deletion of HRI elevated the toxicity of PIs in wt and mTORC1-activated MM. Finally, we identified the drug PMA as a robust inducer of mTORC1 activity, which synergized with PIs in inducing MM cell death. These results help explain the clinical inefficacy of mTORC1 inhibitors in MM. Our data implicate mTORC1 induction and/or HRI inhibition as pharmacological strategies to enhance MM therapy by PIs.
Collapse
|
6
|
Jubaidi FF, Zainalabidin S, Taib IS, Abdul Hamid Z, Mohamad Anuar NN, Jalil J, Mohd Nor NA, Budin SB. The Role of PKC-MAPK Signalling Pathways in the Development of Hyperglycemia-Induced Cardiovascular Complications. Int J Mol Sci 2022; 23:ijms23158582. [PMID: 35955714 PMCID: PMC9369123 DOI: 10.3390/ijms23158582] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular disease is the most common cause of death among diabetic patients worldwide. Hence, cardiovascular wellbeing in diabetic patients requires utmost importance in disease management. Recent studies have demonstrated that protein kinase C activation plays a vital role in the development of cardiovascular complications via its activation of mitogen-activated protein kinase (MAPK) cascades, also known as PKC-MAPK pathways. In fact, persistent hyperglycaemia in diabetic conditions contribute to preserved PKC activation mediated by excessive production of diacylglycerol (DAG) and oxidative stress. PKC-MAPK pathways are involved in several cellular responses, including enhancing oxidative stress and activating signalling pathways that lead to uncontrolled cardiac and vascular remodelling and their subsequent dysfunction. In this review, we discuss the recent discovery on the role of PKC-MAPK pathways, the mechanisms involved in the development and progression of diabetic cardiovascular complications, and their potential as therapeutic targets for cardiovascular management in diabetic patients.
Collapse
Affiliation(s)
- Fatin Farhana Jubaidi
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Correspondence: (F.F.J.); (S.B.B.); Tel.: +603-9289-7645 (S.S.B.)
| | - Satirah Zainalabidin
- Center for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.Z.); (N.N.M.A.)
| | - Izatus Shima Taib
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
| | - Zariyantey Abdul Hamid
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
| | - Nur Najmi Mohamad Anuar
- Center for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (S.Z.); (N.N.M.A.)
| | - Juriyati Jalil
- Center for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Nor Anizah Mohd Nor
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Faculty of Health Sciences, University College MAIWP International, Kuala Lumpur 68100, Malaysia
| | - Siti Balkis Budin
- Center for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (I.S.T.); (Z.A.H.); (N.A.M.N.)
- Correspondence: (F.F.J.); (S.B.B.); Tel.: +603-9289-7645 (S.S.B.)
| |
Collapse
|
7
|
Sánchez-Alegría K, Bastián-Eugenio CE, Vaca L, Arias C. Palmitic acid induces insulin resistance by a mechanism associated with energy metabolism and calcium entry in neuronal cells. FASEB J 2021; 35:e21712. [PMID: 34110637 DOI: 10.1096/fj.202100243r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 01/05/2023]
Abstract
Palmitic acid (PA) is a saturated fatty acid whose high consumption has been largely associated with the development of different metabolic alterations, such as insulin resistance, metabolic syndrome, and type 2 diabetes. Particularly in the brain, insulin signaling disruption has been linked to cognitive decline and is considered a risk factor for Alzheimer's disease. Cumulative evidence has demonstrated the participation of PA in the molecular cascade underlying cellular insulin resistance in peripheral tissues, but its role in the development of neuronal insulin resistance and the mechanisms involved are not fully understood. It has generally been accepted that the brain does not utilize fatty acids as a primary energy source, but recent evidence shows that neurons possess the machinery for fatty acid β-oxidation. However, it is still unclear under what conditions neurons use fatty acids as energy substrates and the implications of their oxidative metabolism in modifying insulin-stimulated effects. In the present work, we have found that neurons differentiated from human neuroblastoma MSN exposed to high but nontoxic concentrations of PA generate ATP through mitochondrial metabolism, which is associated with an increase in the cytosolic Ca2+ and diminished insulin signaling in neurons. These findings reveal a novel mechanism by which saturated fatty acids produce Ca2+ entry and insulin resistance that may play a causal role in increasing neuronal vulnerability associated with metabolic diseases.
Collapse
Affiliation(s)
- Karina Sánchez-Alegría
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Carlos Ernesto Bastián-Eugenio
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Luis Vaca
- Departamento de Genética Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
8
|
Pathak P, Blech-Hermoni Y, Subedi K, Mpamugo J, Obeng-Nyarko C, Ohman R, Molloy I, Kates M, Hale J, Stauffer S, Sharan SK, Mankodi A. Myopathy associated LDB3 mutation causes Z-disc disassembly and protein aggregation through PKCα and TSC2-mTOR downregulation. Commun Biol 2021; 4:355. [PMID: 33742095 PMCID: PMC7979776 DOI: 10.1038/s42003-021-01864-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 02/17/2021] [Indexed: 12/18/2022] Open
Abstract
Mechanical stress induced by contractions constantly threatens the integrity of muscle Z-disc, a crucial force-bearing structure in striated muscle. The PDZ-LIM proteins have been proposed to function as adaptors in transducing mechanical signals to preserve the Z-disc structure, however the underlying mechanisms remain poorly understood. Here, we show that LDB3, a well-characterized striated muscle PDZ-LIM protein, modulates mechanical stress signaling through interactions with the mechanosensing domain in filamin C, its chaperone HSPA8, and PKCα in the Z-disc of skeletal muscle. Studies of Ldb3Ala165Val/+ mice indicate that the myopathy-associated LDB3 p.Ala165Val mutation triggers early aggregation of filamin C and its chaperones at muscle Z-disc before aggregation of the mutant protein. The mutation causes protein aggregation and eventually Z-disc myofibrillar disruption by impairing PKCα and TSC2-mTOR, two important signaling pathways regulating protein stability and disposal of damaged cytoskeletal components at a major mechanosensor hub in the Z-disc of skeletal muscle.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Animals
- Autophagy
- Disease Models, Animal
- Down-Regulation
- Filamins/metabolism
- HSC70 Heat-Shock Proteins/metabolism
- LIM Domain Proteins/genetics
- Mechanotransduction, Cellular
- Mice, Inbred C57BL
- Mice, Transgenic
- Muscle Contraction
- Muscle Strength
- Muscle, Skeletal/enzymology
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Myopathies, Structural, Congenital/enzymology
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/pathology
- Myopathies, Structural, Congenital/physiopathology
- Point Mutation
- Protein Aggregates
- Protein Aggregation, Pathological
- Protein Kinase C-alpha/genetics
- Protein Kinase C-alpha/metabolism
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Tuberous Sclerosis Complex 2 Protein/genetics
- Tuberous Sclerosis Complex 2 Protein/metabolism
- Mice
Collapse
Affiliation(s)
- Pankaj Pathak
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Yotam Blech-Hermoni
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Kalpana Subedi
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Jessica Mpamugo
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Charissa Obeng-Nyarko
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Rachel Ohman
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Ilda Molloy
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Malcolm Kates
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Jessica Hale
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Stacey Stauffer
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Shyam K Sharan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Ami Mankodi
- Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA.
| |
Collapse
|
9
|
Protein Kinase C Regulates ASIC1a Protein Expression and Channel Function via NF-kB Signaling Pathway. Mol Neurobiol 2020; 57:4754-4766. [PMID: 32783140 DOI: 10.1007/s12035-020-02056-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 08/03/2020] [Indexed: 10/23/2022]
Abstract
Tissue acidosis is a common feature in many pathological conditions. Activation of acid-sensing ion channel 1a (ASIC1a) plays a key role in acidosis-mediated neurotoxicity. Protein kinase C (PKC) activity has been proved to be associated with many physiological processes and pathological conditions; however, whether PKC activation regulates ASIC1a protein expression and channel function remains ill defined. In this study, we demonstrated that treatment with phorbol 12-myristate 13-acetate (PMA, a PKC activator) for 6 h significantly increased ASIC1a protein expression and ASIC currents in NS20Y cells, a neuronal cell line, and in primary cultured mouse cortical neurons. In contrast, treatment with Calphostin C (a nonselective PKC inhibitor) for 6 h or longer decreased ASIC1a protein expression and ASIC currents. Similar to Calphostin C, PKC α and βI inhibitor Go6976 exposure also reduced ASIC1a protein expression. The reduction in ASIC1a protein expression by PKC inhibition involves a change in ASIC1a protein degradation, which is mediated by ubiquitin-proteasome system (UPS)-dependent degradation pathway. In addition, we showed that PKC regulation of ASIC1a protein expression involves NF-κB signaling pathway. Consistent with their effects on ASIC1a protein expression and channel function, PKC inhibition protected NS20Y cells against acidosis-induced cytotoxicity, while PKC activation potentiated acidosis-induced cells injury. Together, these results indicate that ASIC1a protein expression and channel function are closely regulated by the activity of protein kinase C and its downstream signaling pathway(s).
Collapse
|
10
|
Gerbino A, De Zio R, Russo D, Milella L, Milano S, Procino G, Pusch M, Svelto M, Carmosino M. Role of PKC in the Regulation of the Human Kidney Chloride Channel ClC-Ka. Sci Rep 2020; 10:10268. [PMID: 32581267 PMCID: PMC7314819 DOI: 10.1038/s41598-020-67219-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 06/02/2020] [Indexed: 12/03/2022] Open
Abstract
The physiological role of the renal ClC-Ka/ClC-K1 channels is to confer a high Cl- permeability to the thin Ascending Limb of Henle (tAL), which in turn is essential for establishing the high osmolarity of the renal medulla that drives water reabsorption from collecting ducts. Here, we investigated by whole-cell patch-clamp measurements on HEK293 cells co-expressing ClC-Ka (tagged with GFP) and the accessory subunit barttin (tagged with m-Cherry) the effect of a natural diuretic extract from roots of Dandelion (DRE), and other compounds activating PKC, such as ATP, on ClC-Ka activity and its membrane localization. Treatment with 400 µg/ml DRE significantly inhibited Cl- currents time-dependently within several minutes. Of note, the same effect on Cl- currents was obtained upon treatment with 100 µM ATP. Pretreatment of cells with either the intracellular Ca2+ chelator BAPTA-AM (30 μM) or the PKC inhibitor Calphostin C (100 nM) reduced the inhibitory effect of DRE. Conversely, 1 µM of phorbol meristate acetate (PMA), a specific PKC activator, mimicked the inhibitory effect of DRE on ClC-Ka. Finally, we found that pretreatment with 30 µM Heclin, an E3 ubiquitin ligase inhibitor, did not revert DRE-induced Cl- current inhibition. In agreement with this, live-cell confocal analysis showed that DRE treatment did not induce ClC-Ka internalization. In conclusion, we demonstrate for the first time that the activity of ClC-Ka in renal cells could be significantly inhibited by the activation of PKC elicited by classical maneuvers, such as activation of purinergic receptors, or by exposure to herbal extracts that activates a PKC-dependent pathway. Overall, we provide both new information regarding the regulation of ClC-Ka and a proof-of-concept study for the use of DRE as new diuretic.
Collapse
Affiliation(s)
- Andrea Gerbino
- National Research Council, Institute of Biomembrane and Bioenergetics, Bari, IT, Italy.,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Roberta De Zio
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Daniela Russo
- Department of Sciences, University of Basilicata, Potenza, IT, Italy
| | - Luigi Milella
- Department of Sciences, University of Basilicata, Potenza, IT, Italy
| | - Serena Milano
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Giuseppe Procino
- Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Michael Pusch
- National Research Council, Institute of Biophysics, Genova, IT, Italy
| | - Maria Svelto
- National Research Council, Institute of Biomembrane and Bioenergetics, Bari, IT, Italy.,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy
| | - Monica Carmosino
- Department of Sciences, University of Basilicata, Potenza, IT, Italy. .,Department of Biosciences, Biotechnologies and Biopharamceutics, University of Bari, Bari, IT, Italy.
| |
Collapse
|
11
|
Barthelemy C, André B. Ubiquitylation and endocytosis of the human LAT1/SLC7A5 amino acid transporter. Sci Rep 2019; 9:16760. [PMID: 31728037 PMCID: PMC6856120 DOI: 10.1038/s41598-019-53065-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/22/2019] [Indexed: 12/11/2022] Open
Abstract
The human L-type amino acid transporter 1 (LAT1), also known as SLC7A5, catalyzes the transport of large neutral amino acids across the plasma membrane. As the main transporter of several essential amino acids, notably leucine, LAT1 plays an important role in mTORC1 activation. Furthermore, it is overexpressed in various types of cancer cells, where it contributes importantly to sustained growth. Despite the importance of LAT1 in normal and tumor cells, little is known about the mechanisms that might control its activity, for example by promoting its downregulation via endocytosis. Here we report that in HeLa cells, activation of protein kinase C by phorbol 12-myristate 13-acetate (PMA) triggers efficient endocytosis and degradation of LAT1. Under these conditions we found LAT1 downregulation to correlate with increased LAT1 ubiquitylation. This modification was considerably reduced in cells depleted of the Nedd4-2 ubiquitin ligase. By systematically mutagenizing the residues of the LAT1 cytosolic tails, we identified a group of three close lysines (K19, K25, K30) in the N-terminal tail that are important for PMA-induced ubiquitylation and downregulation. Our study thus unravels a mechanism of induced endocytosis of LAT1 elicited by Nedd4-2-mediated ubiquitylation of the transporter's N-terminal tail.
Collapse
Affiliation(s)
- Céline Barthelemy
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium
| | - Bruno André
- Molecular Physiology of the Cell, Université libre de Bruxelles (ULB), IBMM (Biopark), Gosselies, Belgium.
| |
Collapse
|
12
|
Katopodis P, Chudasama D, Wander G, Sales L, Kumar J, Pandhal M, Anikin V, Chatterjee J, Hall M, Karteris E. Kinase Inhibitors and Ovarian Cancer. Cancers (Basel) 2019; 11:E1357. [PMID: 31547471 PMCID: PMC6770231 DOI: 10.3390/cancers11091357] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/08/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Ovarian cancer is fifth in the rankings of cancer deaths among women, and accounts for more deaths than any other gynecological malignancy. Despite some improvement in overall-(OS) and progression-free survival (PFS) following surgery and first-line chemotherapy, there is a need for development of novel and more effective therapeutic strategies. In this mini review, we provide a summary of the current landscape of the clinical use of tyrosine kinase inhibitors (TKIs) and mechanistic target of rapamycin (mTOR) inhibitors in ovarian cancer. Emerging data from phase I and II trials reveals that a combinatorial treatment that includes TKIs and chemotherapy agents seems promising in terms of PFS despite some adverse effects recorded; whereas the use of mTOR inhibitors seems less effective. There is a need for further research into the inhibition of multiple signaling pathways in ovarian cancer and progression to phase III trials for drugs that seem most promising.
Collapse
Affiliation(s)
- Periklis Katopodis
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London UB9 6JH, UK.
| | - Dimple Chudasama
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Gurleen Wander
- Chelsea and Westminster Hospital NHS Trust, London UB9 6JH, UK.
| | - Louise Sales
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Juhi Kumar
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Manreen Pandhal
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| | - Vladimir Anikin
- Division of Thoracic Surgery, The Royal Brompton & Harefield NHS Foundation Trust, Harefield Hospital, London UB9 6JH, UK.
- Department of Oncology and Reconstructive Surgery, Sechenov First Moscow State Medical University, 119146 Moscow, Russia.
| | - Jayanta Chatterjee
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford GU2 7XH, UK.
| | - Marcia Hall
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
- Mount Vernon Cancer Centre, Rickmansworth Road, Northwood HA6 2RN, UK.
| | - Emmanouil Karteris
- Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK.
| |
Collapse
|
13
|
Bros M, Haas K, Moll L, Grabbe S. RhoA as a Key Regulator of Innate and Adaptive Immunity. Cells 2019; 8:cells8070733. [PMID: 31319592 PMCID: PMC6678964 DOI: 10.3390/cells8070733] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022] Open
Abstract
RhoA is a ubiquitously expressed cytoplasmic protein that belongs to the family of small GTPases. RhoA acts as a molecular switch that is activated in response to binding of chemokines, cytokines, and growth factors, and via mDia and the ROCK signaling cascade regulates the activation of cytoskeletal proteins, and other factors. This review aims to summarize our current knowledge on the role of RhoA as a general key regulator of immune cell differentiation and function. The contribution of RhoA for the primary functions of innate immune cell types, namely neutrophils, macrophages, and conventional dendritic cells (DC) to (i) get activated by pathogen-derived and endogenous danger signals, (ii) migrate to sites of infection and inflammation, and (iii) internalize pathogens has been fairly established. In activated DC, which constitute the most potent antigen-presenting cells of the immune system, RhoA is also important for the presentation of pathogen-derived antigen and the formation of an immunological synapse between DC and antigen-specific T cells as a prerequisite to induce adaptive T cell responses. In T cells and B cells as the effector cells of the adaptive immune system Rho signaling is pivotal for activation and migration. More recently, mutations of Rho and Rho-modulating factors have been identified to predispose for autoimmune diseases and as causative for hematopoietic malignancies.
Collapse
Affiliation(s)
- Matthias Bros
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Katharina Haas
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Lorna Moll
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stephan Grabbe
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
14
|
Shou X, Wang B, Zhou R, Wang L, Ren A, Xin S, Zhu L. Baicalin Suppresses Hypoxia-Reoxygenation-Induced Arterial Endothelial Cell Apoptosis via Suppressing PKCδ/p53 Signaling. Med Sci Monit 2017; 23:6057-6063. [PMID: 29272263 PMCID: PMC5747146 DOI: 10.12659/msm.907989] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background This study was aimed to investigate the protective role of baicalin on vascular endothelium exposed to ischemia reperfusion injury and the involved molecular mechanisms. Material/Methods Cultured human arterial endothelial cells (HAECs) were exposed to hypoxia/deoxygenation (H/R). Cells were also treated with baicalin at serially diluted concentrations. Cells were also treated with PKC activator PEP005 or specific siRNA against protein kinase Cδ (PKCδ). MTT assay was used to evaluate the cell viabilities. Flow cytometry was used to detect cell apoptosis. The protein phosphorylation and expression levels were determined by Western blotting. Results PKCδ-siRNA transfection increased cell viabilities and reduced cell apoptosis in HAECs exposed to H/R. Baicalin treatment preserved cell viabilities and reduced apoptosis of H/R-exposed HAECs in a concentration-dependent manner. Baicalin treatment reduced phosphorylation levels of PKCδ and p53, as well as the expression levels of active caspase3 and bax in HAECs exposed to H/R. The treatment of PKC activator PEP005 impaired the protective effects of baicalin in increasing cell viabilities and reducing apoptosis in HAECs exposed to H/R. Conclusions Baicalin exerts vascular a protective effect on HAECs exposed to H/R by reducing cell apoptosis. The PKCδ/p53 apoptotic signaling pathway was the pharmacological target of baicalin.
Collapse
Affiliation(s)
- Xiaoling Shou
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Bozhong Wang
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Rongfang Zhou
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Lei Wang
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Aihua Ren
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Shangping Xin
- Department of Cardiac Rehabilitation, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Liyue Zhu
- Rehabilitation Center, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|