1
|
Lara Aparicio SY, Laureani Fierro ÁDJ, Aranda Abreu GE, Toledo Cárdenas R, García Hernández LI, Coria Ávila GA, Rojas Durán F, Aguilar MEH, Manzo Denes J, Chi-Castañeda LD, Pérez Estudillo CA. Current Opinion on the Use of c-Fos in Neuroscience. NEUROSCI 2022; 3:687-702. [PMID: 39483772 PMCID: PMC11523728 DOI: 10.3390/neurosci3040050] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/16/2022] [Indexed: 11/03/2024] Open
Abstract
For years, the biochemical processes that are triggered by harmful and non-harmful stimuli at the central nervous system level have been extensively studied by the scientific community through numerous techniques and animal models. For example, one of these techniques is the use of immediate expression genes, which is a useful, accessible, and reliable method for observing and quantifying cell activation. It has been shown that both the c-fos gene and its protein c-Fos have rapid activation after stimulus, with the length of time that they remain active depending on the type of stimulus and the activation time depending on the stimulus and the structure studied. Fos requires the participation of other genes (such as c-jun) for its expression (during hetero-dimer forming). c-Fos dimerizes with c-Jun protein to form factor AP-1, which promotes the transcription of various genes. The production and removal of c-Fos is part of cellular homeostasis, but its overexpression results in increased cell proliferation. Although Fos has been used as a marker of cellular activity since the 1990s, which molecular mechanism participates in the regulation of the expression of this protein is still unknown because the gene and the protein are not specific to neurons or glial cells. For these reasons, this work has the objective of gathering information about this protein and its use in neuroscience.
Collapse
Affiliation(s)
- Sandra Yasbeth Lara Aparicio
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
- Laboratorio de Neurofisiología, Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Av. Luis Castelazo S/N, Col. Industrial Las Ánimas, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | | | | | - Rebeca Toledo Cárdenas
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | - Luis Isauro García Hernández
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | - Genaro Alfonso Coria Ávila
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | - Fausto Rojas Durán
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | | | - Jorge Manzo Denes
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | - Lizbeth Donají Chi-Castañeda
- Instituto en Investigaciones Cerebrales, Universidad Veracruzana, Xalapa de Enríquez, Veracruz C.P. 91190, Mexico
| | | |
Collapse
|
2
|
Albani A, Perez-Rivas LG, Tang S, Simon J, Lucia KE, Colón-Bolea P, Schopohl J, Roeber S, Buchfelder M, Rotermund R, Flitsch J, Thorsteinsdottir J, Herms J, Stalla G, Reincke M, Theodoropoulou M. Improved pasireotide response in USP8 mutant corticotroph tumours in vitro. Endocr Relat Cancer 2022; 29:503-511. [PMID: 35686696 DOI: 10.1530/erc-22-0088] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/10/2022] [Indexed: 12/13/2022]
Abstract
Cushing's disease is a rare but devastating and difficult to manage condition. The somatostatin analogue pasireotide is the only pituitary-targeting pharmaceutical approved for the treatment of Cushing's disease but is accompanied by varying efficacy and potentially severe side effects. Finding means to predict which patients are more likely to benefit from this treatment may improve their management. More than half of corticotroph tumours harbour mutations in the USP8 gene, and there is evidence of higher somatostatin receptor 5 (SSTR5) expression in the USP8-mutant tumours. Pasireotide has a high affinity for SSTR5, indicating that these tumours may be more sensitive to treatment. To test this hypothesis, we examined the inhibitory action of pasireotide on adrenocorticotrophic hormone synthesis in primary cultures of human corticotroph tumour with assessed USP8 mutational status and in immortalized murine corticotroph tumour cells overexpressing human USP8 mutants frequent in Cushing's disease. Our in vitro results demonstrate that pasireotide exerts a higher antisecretory response in USP8-mutant corticotroph tumours. Overexpressing USP8 mutants in a murine corticotroph tumour cell model increased endogenous somatostatin receptor 5 (Sstr5) transcription. The murine Sstr5 promoter has two binding sites for the activating protein 1 (AP-1) and USP8 mutants possibly to mediate their action by stimulating AP-1 transcriptional activity. Our data corroborate the USP8 mutational status as a potential marker of pasireotide response and describe a potential mechanism through which USP8 mutants may regulate SSTR5 gene expression.
Collapse
Affiliation(s)
- Adriana Albani
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Sicheng Tang
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julia Simon
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Kristin Elisabeth Lucia
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Paula Colón-Bolea
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jochen Schopohl
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sigrun Roeber
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Michael Buchfelder
- Department of Neurosurgery, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Roman Rotermund
- Department of Neurosurgery, Universitätskrankenhaus Hamburg-Eppendorf, Hamburg, Germany
| | - Jörg Flitsch
- Department of Neurosurgery, Universitätskrankenhaus Hamburg-Eppendorf, Hamburg, Germany
| | | | - Jochen Herms
- Center for Neuropathology and Prion Research, LMU Munich, Munich, Germany
| | - Günter Stalla
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
- Medicover Neuroendocrinology, Munich, Germany
| | - Martin Reincke
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Marily Theodoropoulou
- Medizinische Klinik und Poliklinik IV, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
3
|
Murillo-Rodríguez E, Millán-Aldaco D, Arankowsky-Sandoval G, Yamamoto T, Pertwee RG, Parker L, Mechoulam R. Assessing the treatment of cannabidiolic acid methyl ester: a stable synthetic analogue of cannabidiolic acid on c-Fos and NeuN expression in the hypothalamus of rats. J Cannabis Res 2021; 3:31. [PMID: 34253253 PMCID: PMC8276432 DOI: 10.1186/s42238-021-00081-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 06/14/2021] [Indexed: 04/03/2024] Open
Abstract
BACKGROUND Cannabidiol (CBD), the non-psychotropic compound from Cannabis sativa, shows positive results on controlling several health disturbances; however, comparable data regarding additional chemical from C. sativa, such as cannabidiolic acid (CBDA), is scarce due to its instability. To address this limitation, a stable CBDA analogue, CBDA methyl ester (HU-580), was synthetized and showed CBDA-like effects. Recently, we described that HU-580 increased wakefulness and wake-related neurochemicals. OBJECTIVE To extend the comprehension of HU-580´s properties on waking, the c-Fos and NeuN expression in a wake-linked brain area, the hypothalamus was evaluated. METHODS c-Fos and NeuN expression in hypothalamic sections were analyzed after the injections of HU-580 (0.1 or 100 μg/kg, i.p.). RESULTS Systemic administrations of HU-580 increased c-Fos and neuronal nuclei (NeuN) expression in hypothalamic nuclei, including the dorsomedial hypothalamic nucleus dorsal part, dorsomedial hypothalamic nucleus compact part, and dorsomedial hypothalamic nucleus ventral part. CONCLUSION HU-580 increased c-Fos and NeuN immunoreactivity in hypothalamus nuclei suggesting that this drug might modulate the sleep-wake cycle by engaging the hypothalamus.
Collapse
Affiliation(s)
- Eric Murillo-Rodríguez
- Laboratorio de Neurociencias Moleculares e Integrativas Escuela de Medicina, División Ciencias de la Salud, Universidad Anáhuac Mayab Mérida, Km. 15.5 Carretera Mérida-Progreso, Int. Km. 2 Carretera a Chablekal, Yucatán, C.P. 97,308, Mérida, México.
- Intercontinental Neuroscience Research Group, Mérida, Yucatán, México.
| | - Diana Millán-Aldaco
- Depto. de Neurociencia Cognitiva. División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Gloria Arankowsky-Sandoval
- Centro de Investigaciones Regionales "Dr. Hideyo Noguchi", Universidad Autónoma de Yucatán, Mérida, Yucatán, México
| | - Tetsuya Yamamoto
- Intercontinental Neuroscience Research Group, Mérida, Yucatán, México
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima, Japan
| | - Roger G Pertwee
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Linda Parker
- Department of Psychology and Neuroscience Graduate Program, University of Guelph, Guelph, Ontario, Canada
| | - Raphael Mechoulam
- Institute for Drug Research, Medical Faculty, Hebrew University, Jerusalem, Israel
| |
Collapse
|
4
|
Chang R, Hernandez J, Gastelum C, Guadagno K, Perez L, Wagner EJ. Pituitary Adenylate Cyclase-Activating Polypeptide Excites Proopiomelanocortin Neurons: Implications for the Regulation of Energy Homeostasis. Neuroendocrinology 2021; 111:45-69. [PMID: 32028278 DOI: 10.1159/000506367] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 01/30/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE We examined whether pituitary adenylate cyclase-activating polypeptide (PACAP) excites proopiomelanocortin (POMC) neurons via PAC1 receptor mediation and transient receptor potential cation (TRPC) channel activation. METHODS Electrophysiological recordings were done in slices from both intact male and ovariectomized (OVX) female PACAP-Cre mice and eGFP-POMC mice. RESULTS In recordings from POMC neurons in eGFP-POMC mice, PACAP induced a robust inward current and increase in conductance in voltage clamp, and a depolarization and increase in firing in current clamp. These postsynaptic actions were abolished by inhibitors of the PAC1 receptor, TRPC channels, phospholipase C, phosphatidylinositol-3-kinase, and protein kinase C. Estradiol augmented the PACAP-induced inward current, depolarization, and increased firing, which was abrogated by estrogen receptor (ER) antagonists. In optogenetic recordings from POMC neurons in PACAP-Cre mice, high-frequency photostimulation induced inward currents, depolarizations, and increased firing that were significantly enhanced by Gq-coupled membrane ER signaling in an ER antagonist-sensitive manner. Importantly, the PACAP-induced excitation of POMC neurons was notably reduced in obese, high-fat (HFD)-fed males. In vivo experiments revealed that intra-arcuate nucleus (ARC) PACAP as well as chemogenetic and optogenetic stimulation of ventromedial nucleus (VMN) PACAP neurons produced a significant decrease in energy intake accompanied by an increase in energy expenditure, effects blunted by HFD in males and partially potentiated by estradiol in OVX females. CONCLUSIONS These findings reveal that the PACAP-induced activation of PAC1 receptor and TRPC5 channels at VMN PACAP/ARC POMC synapses is potentiated by estradiol and attenuated under conditions of diet-induced obesity/insulin resistance. As such, they advance our understanding of how PACAP regulates the homeostatic energy balance circuitry under normal and pathophysiological circumstances.
Collapse
Affiliation(s)
- Rachel Chang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Jennifer Hernandez
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Cassandra Gastelum
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Kaitlyn Guadagno
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Lynnea Perez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Edward J Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, California, USA,
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA,
| |
Collapse
|
5
|
Baron M, Tagore M, Hunter MV, Kim IS, Moncada R, Yan Y, Campbell NR, White RM, Yanai I. The Stress-Like Cancer Cell State Is a Consistent Component of Tumorigenesis. Cell Syst 2020; 11:536-546.e7. [PMID: 32910905 DOI: 10.1016/j.cels.2020.08.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/23/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023]
Abstract
Transcriptional profiling of tumors has revealed a stress-like state among the cancer cells with the concerted expression of genes such as fos, jun, and heat-shock proteins, though this has been controversial given possible dissociation-effects associated with single-cell RNA sequencing. Here, we validate the existence of this state using a combination of zebrafish melanoma modeling, spatial transcriptomics, and human samples. We found that the stress-like subpopulation of cancer cells is present from the early stages of tumorigenesis. Comparing with previously reported single-cell RNA sequencing datasets from diverse cancer types, including triple-negative breast cancer, oligodendroglioma, and pancreatic adenocarcinoma, indicated the conservation of this state during tumorigenesis. We also provide evidence that this state has higher tumor-seeding capabilities and that its induction leads to increased growth under both MEK and BRAF inhibitors. Collectively, our study supports the stress-like cells as a cancer cell state expressing a coherent set of genes and exhibiting drug-resistance properties.
Collapse
Affiliation(s)
- Maayan Baron
- Institute for Computational Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Mohita Tagore
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miranda V Hunter
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Isabella S Kim
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Reuben Moncada
- Institute for Computational Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Yun Yan
- Institute for Computational Medicine, NYU Grossman School of Medicine, New York, NY, USA
| | - Nathaniel R Campbell
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard M White
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Itai Yanai
- Institute for Computational Medicine, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
6
|
Ansari MZ, Swaminathan R. Structure and dynamics at N- and C-terminal regions of intrinsically disordered human c-Myc PEST degron reveal a pH-induced transition. Proteins 2020; 88:889-909. [PMID: 31999378 DOI: 10.1002/prot.25880] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/09/2019] [Accepted: 01/25/2020] [Indexed: 12/11/2022]
Abstract
We investigated the structure and Brownian rotational motion of the PEST region (201-268) from human c-Myc oncoprotein, whose overexpression/dysregulation is associated with various types of cancer. The 77-residue PEST fragment revealed a large Stokes radius (~3.1 nm) and CD spectrum highlighting abundance of disordered structure. Changes in structure/dynamics at two specific sites in PEST degron were observed using time-resolved fluorescence spectroscopy by labeling Cys9 near N-terminal with dansyl probe and inserting a Trp70 near C-terminal (PEST M1). Trp in PEST M1 at pH 3 was inaccessible to quencher, showed hindered segmental motion and slow global rotation (~30 ns) in contrast to N-terminal where the dansyl probe was free, exposed with fast global rotation (~5 ns). Remarkably, this large monomeric structure at acidic pH was retained irrespective of ionic strength (0.03-0.25 M) and partially so in presence of 6 M Gdn.HCl. With gradual increase in pH, a structural transition (~pH 4.8) into a more exposed and freely rotating Trp was noticeable. Interestingly, the induced structure at C-terminal also influenced the dynamics of dansyl probe near N-terminal, which otherwise remained unstructured at pH > 5. FRET measurements confirmed a 11 Å decrease in distance between dansyl and indole at pH 4 compared to pH 9, coinciding with enhanced ANS binding and increase in strand/helix population in both PEST fragments. The protonation of glutamate/aspartate residues in C-terminal region of PEST is implicated in this disorder-order transition. This may have a bearing on the role of PEST in endocytic trafficking of eukaryotic proteins.
Collapse
Affiliation(s)
- Mohd Ziauddin Ansari
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Rajaram Swaminathan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
7
|
Xie Y, Ostriker AC, Jin Y, Hu H, Sizer AJ, Peng G, Morris AH, Ryu C, Herzog EL, Kyriakides T, Zhao H, Dardik A, Yu J, Hwa J, Martin KA. LMO7 Is a Negative Feedback Regulator of Transforming Growth Factor β Signaling and Fibrosis. Circulation 2019; 139:679-693. [PMID: 30586711 PMCID: PMC6371979 DOI: 10.1161/circulationaha.118.034615] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Vascular smooth muscle cells (SMCs) synthesize extracellular matrix (ECM) that contributes to tissue remodeling after revascularization interventions. The cytokine transforming growth factor β (TGF-β) is induced on tissue injury and regulates tissue remodeling and wound healing, but dysregulated signaling results in excess ECM deposition and fibrosis. The LIM (Lin11, Isl-1 & Mec-3) domain protein LIM domain only 7 (LMO7) is a TGF-β1 target gene in hepatoma cells, but its role in vascular physiology and fibrosis is unknown. METHODS We use carotid ligation and femoral artery denudation models in mice with global or inducible smooth muscle-specific deletion of LMO7, and knockout, knockdown, overexpression, and mutagenesis approaches in mouse and human SMC, and human arteriovenous fistula and cardiac allograft vasculopathy samples to assess the role of LMO7 in neointima and fibrosis. RESULTS We demonstrate that LMO7 is induced postinjury and by TGF-β in SMC in vitro. Global or SMC-specific LMO7 deletion enhanced neointimal formation, TGF-β signaling, ECM deposition, and proliferation in vascular injury models. LMO7 loss of function in human and mouse SMC enhanced ECM protein expression at baseline and after TGF-β treatment. TGF-β neutralization or receptor antagonism prevented the exacerbated neointimal formation and ECM synthesis conferred by loss of LMO7. Notably, loss of LMO7 coordinately amplified TGF-β signaling by inducing expression of Tgfb1 mRNA, TGF-β protein, αv and β3 integrins that promote activation of latent TGF-β, and downstream effectors SMAD3 phosphorylation and connective tissue growth factor. Mechanistically, the LMO7 LIM domain interacts with activator protein 1 transcription factor subunits c-FOS and c-JUN and promotes their ubiquitination and degradation, disrupting activator protein 1-dependent TGF-β autoinduction. Importantly, preliminary studies suggest that LMO7 is upregulated in human intimal hyperplastic arteriovenous fistula and cardiac allograft vasculopathy samples, and inversely correlates with SMAD3 phosphorylation in cardiac allograft vasculopathy. CONCLUSIONS LMO7 is induced by TGF-β and serves to limit vascular fibrotic responses through negative feedback regulation of the TGF-β pathway. This mechanism has important implications for intimal hyperplasia, wound healing, and fibrotic diseases.
Collapse
Affiliation(s)
- Yi Xie
- Departments of Medicine (Cardiovascular Medicine) (Y.X., A.C.O., Y.J., K.A.M., J.H.), Yale University, New Haven, CT.,Pharmacology (Y.X., A.C.O., Y.J., K.A.M.), Yale University, New Haven, CT
| | - Allison C Ostriker
- Departments of Medicine (Cardiovascular Medicine) (Y.X., A.C.O., Y.J., K.A.M., J.H.), Yale University, New Haven, CT.,Pharmacology (Y.X., A.C.O., Y.J., K.A.M.), Yale University, New Haven, CT
| | - Yu Jin
- Departments of Medicine (Cardiovascular Medicine) (Y.X., A.C.O., Y.J., K.A.M., J.H.), Yale University, New Haven, CT.,Pharmacology (Y.X., A.C.O., Y.J., K.A.M.), Yale University, New Haven, CT
| | - Haidi Hu
- Surgery (Vascular) (H.H., A.D.), Yale University, New Haven, CT
| | | | - Gang Peng
- Biostatistics (G.P., H.Z.), Yale University, New Haven, CT
| | - Aaron H Morris
- Pathology (A.H.M., T.K.), Yale University, New Haven, CT.,Department of Biomedical Engineering (A.H.M., T.K.), Yale University, New Haven, CT
| | - Changwan Ryu
- Medicine (Pulmonary) (C.R., E.L.H.), Yale University School of Medicine, Yale University, New Haven, CT
| | - Erica L Herzog
- Medicine (Pulmonary) (C.R., E.L.H.), Yale University School of Medicine, Yale University, New Haven, CT
| | - Themis Kyriakides
- Pathology (A.H.M., T.K.), Yale University, New Haven, CT.,Department of Biomedical Engineering (A.H.M., T.K.), Yale University, New Haven, CT
| | - Hongyu Zhao
- Biostatistics (G.P., H.Z.), Yale University, New Haven, CT
| | - Alan Dardik
- Surgery (Vascular) (H.H., A.D.), Yale University, New Haven, CT
| | - Jun Yu
- Department of Physiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (J.Y.)
| | - John Hwa
- Departments of Medicine (Cardiovascular Medicine) (Y.X., A.C.O., Y.J., K.A.M., J.H.), Yale University, New Haven, CT
| | - Kathleen A Martin
- Departments of Medicine (Cardiovascular Medicine) (Y.X., A.C.O., Y.J., K.A.M., J.H.), Yale University, New Haven, CT.,Pharmacology (Y.X., A.C.O., Y.J., K.A.M.), Yale University, New Haven, CT
| |
Collapse
|
8
|
The effects of proteasome on baseline and methamphetamine-dependent dopamine transmission. Neurosci Biobehav Rev 2019; 102:308-317. [DOI: 10.1016/j.neubiorev.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 12/16/2022]
|
9
|
Li B, Gou M, Han J, Yuan X, Li Y, Li T, Jiang Q, Xiao R, Li Q. Proteomic analysis of buccal gland secretion from fasting and feeding lampreys ( Lampetra morii). Proteome Sci 2018; 16:9. [PMID: 29796011 PMCID: PMC5964706 DOI: 10.1186/s12953-018-0137-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 05/14/2018] [Indexed: 12/05/2022] Open
Abstract
Background Previous studies have shown that lamprey buccal glands contain some regulators related to anticoagulation, nociception, and immune responses due to the blood sucking habit. Regrettably, the protein expression profile in the buccal glands of feeding lampreys has never been reported yet. The present study was performed in order to further identify more proteins which are closely associated with lamprey feeding process. Methods 2D-PAGE, NanoLC–MS/MS with higher resolution, Ensembl lamprey and NCBI protein databases, as well as western blot was used to compare the proteomics of buccal gland secretion from China northeast lampreys (Lampetra morii) which had been fed for 0, 10, and 60 min, respectively. Results In the present study, the number of identified protein species in the buccal glands of feeding groups (60 min) was increased significantly, nearly ten times of that in the fasting group. During the feeding stage, novel proteins emerged in the buccal gland secretion of lampreys. According to gene ontology (GO) analysis and function predictions, these proteins were summarized and discussed based on their potential roles during feeding process. Furthermore, some of the identified proteins were confirmed to express during the feeding time of lampreys. Conclusion When lampreys attack host fishes to suck blood and flesh, their buccal glands could secrete enough proteins to suppress blood coagulation, nociception, oxidative stress, immune response, as well as other adverse effects encountered during their parasitic lives. The present study would provide clues to clarify the feeding mechanism of the bloodsucking lampreys. Electronic supplementary material The online version of this article (10.1186/s12953-018-0137-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bowen Li
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| | - Meng Gou
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| | - Jianmei Han
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| | - Xiaofei Yuan
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| | - Yingying Li
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| | - Tiesong Li
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| | - Qi Jiang
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| | - Rong Xiao
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| | - Qingwei Li
- 1School of Life Sciences, Liaoning Normal University, Dalian, 116081 People's Republic of China.,2Lamprey Research Center, Liaoning Normal University, Dalian, 116081 People's Republic of China
| |
Collapse
|
10
|
Moon YM, Lee SY, Kwok SK, Lee SH, Kim D, Kim WK, Her YM, Son HJ, Kim EK, Ryu JG, Seo HB, Kwon JE, Hwang SY, Youn J, Seong RH, Jue DM, Park SH, Kim HY, Ahn SM, Cho ML. The Fos-Related Antigen 1-JUNB/Activator Protein 1 Transcription Complex, a Downstream Target of Signal Transducer and Activator of Transcription 3, Induces T Helper 17 Differentiation and Promotes Experimental Autoimmune Arthritis. Front Immunol 2017; 8:1793. [PMID: 29326694 PMCID: PMC5741610 DOI: 10.3389/fimmu.2017.01793] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/30/2017] [Indexed: 01/11/2023] Open
Abstract
Dysfunction of T helper 17 (Th17) cells leads to chronic inflammatory disorders. Signal transducer and activator of transcription 3 (STAT3) orchestrates the expression of proinflammatory cytokines and pathogenic cell differentiation from interleukin (IL)-17-producing Th17 cells. However, the pathways mediated by STAT3 signaling are not fully understood. Here, we observed that Fos-related antigen 1 (FRA1) and JUNB are directly involved in STAT3 binding to sites in the promoters of Fosl1 and Junb. Promoter binding increased expression of IL-17 and the development of Th17 cells. Overexpression of Fra1 and Junb in mice resulted in susceptibility to collagen-induced arthritis and an increase in Th17 cell numbers and inflammatory cytokine production. In patients with rheumatoid arthritis, FRA1 and JUNB were colocalized with STAT3 in the inflamed synovium. These observations suggest that FRA1 and JUNB are associated closely with STAT3 activation, and that this activation leads to Th17 cell differentiation in autoimmune diseases and inflammation.
Collapse
Affiliation(s)
- Young-Mee Moon
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Seon-Yeong Lee
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- Center for Rheumatic Disease, Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung Hoon Lee
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Deokhoon Kim
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea.,Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Woo Kyung Kim
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Yang-Mi Her
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Hea-Jin Son
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Kyung Kim
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Jun-Geol Ryu
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Hyeon-Beom Seo
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Jeong-Eun Kwon
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Sue-Yun Hwang
- Department of Chemical Engineering, Hankyong National University, Anseong, South Korea
| | - Jeehee Youn
- Department of Biomedical Sciences, College of Medicine, Hanyang University, Seoul, South Korea
| | - Rho H Seong
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Research Center for Functional Cellulomics, Seoul National University, Seoul, South Korea
| | - Dae-Myung Jue
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Center for Rheumatic Disease, Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ho-Youn Kim
- Center for Rheumatic Disease, Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Min Ahn
- Department of Hemato-oncology, Bioinformatics, Cancer Research, Systems Biology, Gachon University, Seongnam, South Korea
| | - Mi-La Cho
- Laboratory of Immune Network, The Rheumatism Research Center, College of Medicine, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
11
|
van IJzendoorn DGP, Forghany Z, Liebelt F, Vertegaal AC, Jochemsen AG, Bovée JVMG, Szuhai K, Baker DA. Functional analyses of a human vascular tumor FOS variant identify a novel degradation mechanism and a link to tumorigenesis. J Biol Chem 2017; 292:21282-21290. [PMID: 29150442 DOI: 10.1074/jbc.c117.815845] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/03/2017] [Indexed: 11/06/2022] Open
Abstract
Epithelioid hemangioma is a locally aggressive vascular neoplasm, found in bones and soft tissue, whose cause is currently unknown, but may involve oncogene activation. FOS is one of the earliest viral oncogenes to be characterized, and normal cellular FOS forms part of the activator protein 1 (AP-1) transcription factor complex, which plays a pivotal role in cell growth, differentiation, and survival as well as the DNA damage response. Despite this, a causal link between aberrant FOS function and naturally occurring tumors has not yet been established. Here, we describe a thorough molecular and biochemical analysis of a mutant FOS protein we identified in these vascular tumors. The mutant protein lacks a highly conserved helix consisting of the C-terminal four amino acids of FOS, which we show is indispensable for fast, ubiquitin-independent FOS degradation via the 20S proteasome. Our work reveals that FOS stimulates endothelial sprouting and that perturbation of normal FOS degradation could account for the abnormal vessel growth typical of epithelioid hemangioma. To the best of our knowledge, this is the first functional characterization of mutant FOS proteins found in tumors.
Collapse
Affiliation(s)
| | - Zary Forghany
- Molecular Cell Biology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | - Frauke Liebelt
- Molecular Cell Biology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | - Alfred C Vertegaal
- Molecular Cell Biology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | - Aart G Jochemsen
- Molecular Cell Biology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | | | - Karoly Szuhai
- Molecular Cell Biology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| | - David A Baker
- Molecular Cell Biology, Leiden University Medical Center (LUMC), 2300 RC Leiden, The Netherlands
| |
Collapse
|
12
|
A simple toolset to identify endogenous post-translational modifications for a target protein: a snapshot of the EGFR signaling pathway. Biosci Rep 2017; 37:BSR20170919. [PMID: 28724604 PMCID: PMC6192658 DOI: 10.1042/bsr20170919] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 06/22/2017] [Accepted: 07/18/2017] [Indexed: 12/17/2022] Open
Abstract
Identification of a novel post-translational modification (PTM) for a target protein, defining its physiologic role, and studying its potential crosstalk with other PTMs is a challenging process. A set of highly sensitive tools termed Signal-Seeker kits was developed, which enables rapid and simple detection of post-translational modifications on any target protein. The methodology for these tools utilizes affinity purification of modified proteins from a cell or tissue lysate and immunoblot analysis. These tools utilize a single lysis system that is effective at identifying endogenous, dynamic PTM changes, as well as the potential crosstalk between PTMs. As a proof-of-concept experiment, the acetylation, tyrosine phosphorylation, SUMOylation 2/3, and ubiquitination profiles of the EGFR - Ras - c-Fos axis were examined in response to EGF stimulation. All 10 previously identified PTMs of this signaling axis were confirmed using these tools, and it also identified acetylation as a novel modification of c-Fos. This axis in the EGF/EGFR signaling pathway was chosen because it is a well-established signaling pathway with proteins localized in the membrane, cytoplasmic, and nuclear compartments that ranged in abundance from 4.18x108 (EGFR) to 1.35x104 (c-Fos) molecules per A431 cell. These tools enabled the identification of low abundance PTMs, such as c-Fos Ac, at 17 molecules per cell. These studies highlight how pervasive PTMs are, and how stimulants like EGF induce multiple PTM changes on downstream signaling axis. Identification of endogenous changes and potential crosstalk between multiple PTMs for a target protein or signaling axis will provide regulatory mechanistic insight to investigators.
Collapse
|
13
|
Calò M, Licata P, Bitto A, Lo Cascio P, Giarratana F, Altavilla D. Effects of PCB-126 on aryl hydrocarbon receptor, ubiquitin and p53 expression levels in Sparus aurata. EXPERIMENTAL AND TOXICOLOGIC PATHOLOGY : OFFICIAL JOURNAL OF THE GESELLSCHAFT FUR TOXIKOLOGISCHE PATHOLOGIE 2017:S0940-2993(16)30221-4. [PMID: 28552628 DOI: 10.1016/j.etp.2017.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 04/06/2017] [Indexed: 06/07/2023]
Abstract
The aim of the present study is to determine if Ahr ligands as PCB-126, a dioxin-like, might contribute to inhibition of the tumor suppressor p53 by promoting its degradation through proteasome-ubiquitin system (UPS). The findings show, in the presence of PCB-126, a significant increase of p53 immunoreactivity in fish compared to the control. Subsequently, there is a decrease of p53 immunoreactivity at 24h which is maintained even at 72h. At the same time there is a slight decrease of ubiquitin immunoreactivity to 12h compared to the control and a marked decrease to 24 and 72h. The induction of ubiquitin expression is resulted very marked in the control and preserved at 12h. It's very important to underline as in our study we demonstrate a marked decrease of ubiquitin and p53 immunoreactivity at 24h and 72h. AHR activation, by ligands as PCB-126, increases p53 ubiquitation inhibiting its expression, in addition it decreases the free ubiquitin promoting disruption of Ub homeostasis; this is the first report that establishes a relationship between AhR, increases p53 ubiquitation, and reduction of free ubiquitin. Our result emphasize the need to deeply the role of this receptor in UPS regulation as potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- M Calò
- Department of Veterinary Science, University of Messina, Polo SS Annunziata, 98168 Messina, Italy.
| | - P Licata
- Department of Veterinary Science, University of Messina, Polo SS Annunziata, 98168 Messina, Italy
| | - A Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Torre Biologica, 5th Floor, AOU Policlinico "G. Martino", Via C. Valeria Gazzi, 98125, Messina, Italy
| | - P Lo Cascio
- Department of Biological and Environmental Sciences, University of Messina, Salita Sperone 31, S. Agata, Messina, 98166, Italy
| | - F Giarratana
- Department of Veterinary Science, University of Messina, Polo SS Annunziata, 98168 Messina, Italy
| | - D Altavilla
- Department of Clinical and Experimental Medicine, University of Messina, Torre Biologica, 5th Floor, AOU Policlinico "G. Martino", Via C. Valeria Gazzi, 98125, Messina, Italy
| |
Collapse
|
14
|
Calò M, Bitto A, Lo Cascio P, Giarratana F, Altavilla D, Gervasi T, Campone L, Cicero N, Licata P. PCB-126 effects on aryl hydrocarbon receptor, ubiquitin and p53 expression levels in a fish product (Sparus aurata L.). Nat Prod Res 2017; 32:1136-1144. [DOI: 10.1080/14786419.2017.1320794] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Margherita Calò
- Department of Veterinary Science, University of Messina, Messina, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Patrizia Lo Cascio
- Department of Chemical, Biological, Pharmacological and Environmental Sciences, University of Messina, Messina, Italy
| | | | - Domenica Altavilla
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Teresa Gervasi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Luca Campone
- Department of Pharmacy, University of Salerno, Fisciano SA, Italy
| | - Nicola Cicero
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- Science4Life, Spin Off Company, University of Messina, Messina, Italy
| | - Patrizia Licata
- Department of Veterinary Science, University of Messina, Messina, Italy
| |
Collapse
|
15
|
Kaplan GS, Torcun CC, Grune T, Ozer NK, Karademir B. Proteasome inhibitors in cancer therapy: Treatment regimen and peripheral neuropathy as a side effect. Free Radic Biol Med 2017; 103:1-13. [PMID: 27940347 DOI: 10.1016/j.freeradbiomed.2016.12.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/22/2016] [Accepted: 12/04/2016] [Indexed: 01/10/2023]
Abstract
Proteasomal system plays an important role in protein turnover, which is essential for homeostasis of cells. Besides degradation of oxidized proteins, it is involved in the regulation of many different signaling pathways. These pathways include mainly cell differentiation, proliferation, apoptosis, transcriptional activation and angiogenesis. Thus, proteasomal system is a crucial target for treatment of several diseases including neurodegenerative diseases, cystic fibrosis, atherosclerosis, autoimmune diseases, diabetes and cancer. Over the last fifteen years, proteasome inhibitors have been tested to highlight their mechanisms of action and used in the clinic to treat different types of cancer. Proteasome inhibitors are mainly used in combinational therapy along with classical chemo-radiotherapy. Several studies have proved their significant effects but serious side effects such as peripheral neuropathy, limits their use in required effective doses. Recent studies focus on peripheral neuropathy as the primary side effect of proteasome inhibitors. Therefore, it is important to delineate the underlying mechanisms of peripheral neuropathy and develop new inhibitors according to obtained data. This review will detail the role of proteasome inhibition in cancer therapy and development of peripheral neuropathy as a side effect. Additionally, new approaches to prevent treatment-limiting side effects will be discussed in order to help researchers in developing effective strategies to overcome side effects of proteasome inhibitors.
Collapse
Affiliation(s)
- Gulce Sari Kaplan
- Department of Biochemistry, School of Medicine/Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, 34854 Maltepe, Istanbul, Turkey
| | - Ceyda Corek Torcun
- Department of Biochemistry, School of Medicine/Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, 34854 Maltepe, Istanbul, Turkey
| | - Tilman Grune
- Department for Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - Nesrin Kartal Ozer
- Department of Biochemistry, School of Medicine/Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, 34854 Maltepe, Istanbul, Turkey
| | - Betul Karademir
- Department of Biochemistry, School of Medicine/Genetic and Metabolic Diseases Research and Investigation Center, Marmara University, 34854 Maltepe, Istanbul, Turkey.
| |
Collapse
|
16
|
Park SH, Kim JY, Cheon YH, Baek JM, Ahn SJ, Yoon KH, Lee MS, Oh J. Protocatechuic Acid Attenuates Osteoclastogenesis by Downregulating JNK/c-Fos/NFATc1 Signaling and Prevents Inflammatory Bone Loss in Mice. Phytother Res 2016; 30:604-12. [DOI: 10.1002/ptr.5565] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/09/2015] [Accepted: 12/10/2015] [Indexed: 11/08/2022]
Affiliation(s)
- Sun-Hyang Park
- Department of Anatomy, School of Medicine; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
| | - Ju-Young Kim
- Imaging Science-based Lung and Bone Diseases Research Center; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
| | - Yoon-Hee Cheon
- Center for Metabolic Function Regulation; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
| | - Jong Min Baek
- Department of Anatomy, School of Medicine; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
| | - Sung-Jun Ahn
- Department of Anatomy, School of Medicine; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
| | - Kwon-Ha Yoon
- Imaging Science-based Lung and Bone Diseases Research Center; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
- Department of Radiology, School of Medicine; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
| | - Myeung Su Lee
- Imaging Science-based Lung and Bone Diseases Research Center; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
- Division of Rheumatology, Department of Internal Medicine; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
- Institute for Skeletal Disease; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
| | - Jaemin Oh
- Department of Anatomy, School of Medicine; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
- Imaging Science-based Lung and Bone Diseases Research Center; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
- Institute for Skeletal Disease; Wonkwang University; Iksan Jeonbuk 570-749 Republic of Korea
| |
Collapse
|
17
|
Shankar E, Song K, Corum SL, Bane KL, Wang H, Kao HY, Danielpour D. A Signaling Network Controlling Androgenic Repression of c-Fos Protein in Prostate Adenocarcinoma Cells. J Biol Chem 2016; 291:5512-5526. [PMID: 26786102 DOI: 10.1074/jbc.m115.694877] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Indexed: 01/04/2023] Open
Abstract
The transcription factor c-Fos controls many important cellular processes, including cell growth and apoptosis. c-Fos expression is rapidly elevated in the prostate upon castration-mediated androgen withdrawal through an undefined mechanism. Here we show that androgens (5α-dihydrotestosterone and R1881) suppress c-Fos protein and mRNA expression induced by 12-O-tetradecanoylphorbol-13-acetate (TPA) or EGF in human prostate cancer (PCa) cell lines. Such suppression transpires through a transcriptional mechanism, predominantly at the proximal serum response element of the c-fos promoter. We show that androgen signaling suppresses TPA-induced c-Fos expression through repressing a PKC/MEK/ERK/ELK-1 signaling pathway. Moreover, our results support the hypothesis that p38(MAPK), PI3K, and PKCδ are involved in the androgenic regulation of c-Fos through controlling MEK/ERK. Stable silencing of c-Fos and PKCδ with shRNAs suggests that R1881 promotes cell death induced by low-dose TPA through a mechanism that is dependent on both PKCδ and loss of c-Fos expression. Reciprocally, loss of either PKCδ or c-Fos activates p38(MAPK) while suppressing the activation of ERK1/2. We also provide the first demonstration that R1881 permits cell death induced by low-dose TPA in the LNCaP androgen-dependent PCa cell line and that TPA-induced cell death is independent of exogenous androgen in the castration-resistant variants of LNCaP, C4-2 and C4-2B. Acquisition of androgen-independent killing by TPA correlates with activation of p38(MAPK), suppression of ERK1/2, and loss of c-Fos. These results provide new insights into androgenic control of c-Fos and use of PKC inhibitors in PCa therapy.
Collapse
Affiliation(s)
| | - Kyung Song
- Division of General Medical Sciences-Oncology
| | | | - Kara L Bane
- Division of General Medical Sciences-Oncology
| | | | - Hung-Ying Kao
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio 44106 and; From the Case Comprehensive Cancer Center
| | - David Danielpour
- Division of General Medical Sciences-Oncology,; Department of Pharmacology, and; the Department of Urology, University Hospitals of Cleveland, Cleveland, Ohio 44106; From the Case Comprehensive Cancer Center,.
| |
Collapse
|
18
|
KDM2B/FBXL10 targets c-Fos for ubiquitylation and degradation in response to mitogenic stimulation. Oncogene 2016; 35:4179-90. [PMID: 26725323 PMCID: PMC4931990 DOI: 10.1038/onc.2015.482] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 10/19/2015] [Accepted: 11/06/2015] [Indexed: 12/16/2022]
Abstract
KDM2B (also known as FBXL10) controls stem cell self-renewal, somatic cell reprogramming and senescence, and tumorigenesis. KDM2B contains multiple functional domains, including a JmjC domain that catalyzes H3K36 demethylation and a CxxC zing finger that recognizes CpG islands and recruits the polycomb repressive complex 1 (PRC1). Here, we report that KDM2B, via its F-box domain, functions as a subunit of the CUL1-RING ubiquitin ligase (CRL1/SCFKDM2B) complex. KDM2B targets c-Fos for polyubiquitylation and regulates c-Fos protein levels. Unlike the phosphorylation of other SCF/CRL1 substrates that promotes substrates binding to F-box, EGF-induced c-Fos S374 phosphorylation dissociates c-Fos from KDM2B and stabilizes c-Fos protein. Non-phosphorylatable and phosphomimetic mutations at S374 result in c-Fos protein which cannot be induced by EGF and accumulates constitutively and lead to decreased or increased cell proliferation, respectively. Multiple tumor-derived KDM2B mutations impaired the function of KDM2B to target c-Fos degradation and to suppress cell proliferation. These results reveal a novel function of KDM2B in the negative regulation of cell proliferation by assembling an E3 ligase to targeting c-Fos protein degradation that is antagonized by mitogenic stimulations.
Collapse
|
19
|
Activation of AHR mediates the ubiquitination and proteasome degradation of c-Fos through the induction of Ubcm4 gene expression. Toxicology 2015; 337:47-57. [PMID: 26318284 DOI: 10.1016/j.tox.2015.08.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 01/08/2023]
Abstract
The ubiquitin-proteasome system (UPS) is a specific, non-lysosomal pathway responsible for the controlled degradation of abnormal and short-half-life proteins. Despite its relevance in cell homeostasis, information regarding control of the UPS component gene expression is lacking. Data from a recent study suggest that the aryl hydrocarbon receptor (AHR), a ligand-dependent transcription factor, might control the expression of several genes encoding for UPS proteins. Here, we showed that activation of AHR by TCDD and β-naphthoflavone (β-NF) results in Ubcm4 gene induction accompanied by an increase in protein levels. UbcM4 is an ubiquitin-conjugating enzyme or E2 protein that in association with ubiquitin ligase enzymes or E3 ligases promotes the ubiquitination and 26S proteasome-mediated degradation of different proteins, including p53, c-Myc, and c-Fos. We also present data demonstrating increased c-Fos ubiquitination and proteasomal degradation through the AHR-mediated induction of UbcM4 expression. The present study shows that AHR modulates the degradation of proteins involved in cell cycle control, consistent with previous reports demonstrating an essential role of the AHR in cell cycle regulation.
Collapse
|
20
|
Abstract
Early growth response (Egr) is a member of the zinc finger family of transcription factors that reflects neuronal activity induced by various stimuli. Acute cocaine administration elicits rapid and transient induction of several immediate early genes in brain neurons. However, the mechanism regulating the degradation of the Egr-1 protein is not clearly understood. In this study, rats were injected with cocaine and the relationships among locomotor activity, Egr-1 protein level, phosphorylation of upstream kinase extracellular regulated kinase (ERK)1/2, Egr-1 mRNA expression, and ubiquitination of the Egr-1 protein were measured in the dorsal striatum and the frontal cortex. Locomotor activity reached a peak at about 15 min, and phosphorylation of ERK1/2 and Egr-1 mRNA level also increased at that time. However, the Egr-1 protein level decreased initially in the dorsal striatum, probably due to ubiquitination-mediated degradation. When locomotor activity decreased substantially at 30 min, the phosphorylation of ERKs and expression levels of Egr-1 mRNA and protein reached their peak levels and the protein level subsequently increased. These findings indicate that immediate early gene protein levels would not be a reliable indicator of increased regional activity in the brain. Thus, observations spanning multiple time periods or the examination of mRNA rather than protein would be recommended in these situations.
Collapse
|
21
|
Li Z, Qiao Z, Zheng W, Ma W. Network Cluster Analysis of Protein-Protein Interaction Network-Identified Biomarker for Type 2 Diabetes. Diabetes Technol Ther 2015; 17:475-81. [PMID: 25879401 PMCID: PMC4504429 DOI: 10.1089/dia.2014.0204] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a complex disease that is caused by an impairment in the secretion of β-cell insulin and by a peripheral resistance to insulin. Most patients suffering from T2DM and from obesity exhibit insulin resistance in the muscles, liver, and fat, resulting in a reduced response of these tissues to insulin. In healthy individuals, pancreatic islet β-cells secrete insulin to regulate the increase in blood glucose levels. Once these β-cells fail to function, T2DM develops. Despite the progress achieved in this field in recent years, the genetic causes for insulin resistance and for T2DM have not yet been fully discovered. The present study aims to characterize T2DM by comparing its gene expression with that of normal controls, as well as to identify biomarkers for early T2DM. Gene expression profiles were downloaded from the Gene Expression Omnibus, and differentially expressed genes (DEGs) were identified for type 2 diabetes. Furthermore, functional analyses were conducted for the gene ontology and for the pathway enrichment. In total, 781 DEGs were identified in the T2DM samples relative to healthy controls. These genes were found to be involved in several biological processes, including cell communication, cell proliferation, cell shape, and apoptosis. We constructed a protein-protein interaction (PPI) network, and the clusters in the PPI were analyzed by using ClusterONE. Six functional genes that may play important roles in the initiation of T2DM were identified within the network.
Collapse
Affiliation(s)
- Zhonghui Li
- Institute of Genetic Engineering, Southern Medical University , Guangzhou, Guangdong Province, China
| | - Zijun Qiao
- Institute of Genetic Engineering, Southern Medical University , Guangzhou, Guangdong Province, China
| | - Wenling Zheng
- Institute of Genetic Engineering, Southern Medical University , Guangzhou, Guangdong Province, China
| | - Wenli Ma
- Institute of Genetic Engineering, Southern Medical University , Guangzhou, Guangdong Province, China
| |
Collapse
|
22
|
Ji L, Zheng Z, Shi L, Huang Y, Lu B, Wang Z. Andrographolide decreased VEGFD expression in hepatoma cancer cells by inducing ubiquitin/proteasome-mediated cFos protein degradation. Biochim Biophys Acta Gen Subj 2015; 1850:750-8. [DOI: 10.1016/j.bbagen.2015.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 12/24/2014] [Accepted: 01/07/2015] [Indexed: 01/11/2023]
|
23
|
Grosicki S, Barchnicka A, Jurczyszyn A, Grosicka A. Bortezomib for the treatment of multiple myeloma. Expert Rev Hematol 2014; 7:173-85. [PMID: 24617331 DOI: 10.1586/17474086.2014.899144] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bortezomib is the first proteasome inhibitor drug tested in human patients. Bortezomib demonstrates a particular clinical utility in the treatment of multiple myeloma (MM), where it is the only one of the new drugs administered as mono-therapy that prolongs survival. The significant problem for the consistent pursuit of bortezomib was neurotoxicity, which has been significantly reduced by registering subcutaneous administration or being administered once per week. Bortezomib is currently approved for the treatment of patients with progressive MM in mono-therapy and in combination with prednisone and melphalan in cases of untreated patients who are not candidates for autologous hematopoietic stem cell transplantation (AHSCT) and in combination with dexamethasone or dexamethasone and thalidomide in untreated MM patients, who are candidates for treatment AHSCT. Clinical research is focused on the combination of bortezomib with other new drugs with the hope of further optimizing the treatment of patients with multiple myeloma.
Collapse
Affiliation(s)
- Sebastian Grosicki
- Department of Hematology, Community Hospital in Chorzow, Chorzow, Poland
| | | | | | | |
Collapse
|
24
|
Potential role of proteasome on c-jun related signaling in hypercholesterolemia induced atherosclerosis. Redox Biol 2014; 2:732-8. [PMID: 25009774 PMCID: PMC4085352 DOI: 10.1016/j.redox.2014.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 02/21/2014] [Accepted: 02/21/2014] [Indexed: 11/24/2022] Open
Abstract
Atherosclerosis and its complications are major causes of death all over the world. One of the major risks of atherosclerosis is hypercholesterolemia. During atherosclerosis, oxidized low density lipoprotein (oxLDL) regulates CD36-mediated activation of c-jun amino terminal kinase-1 (JNK1) and modulates matrix metalloproteinase (MMP) induction which stimulates inflammation with an invasion of monocytes. Additionally, inhibition of proteasome leads to an accumulation of c-jun and phosphorylated c-jun and activation of activator protein-1 (AP-1) related increase of MMP expression. We have previously reported a significant increase in cluster of differentiation 36 (CD36) mRNA levels in hypercholesterolemic rabbits and shown that vitamin E treatment prevented the cholesterol induced increase in CD36 mRNA expression. In the present study, our aim is to identify the signaling molecules/transcription factors involved in the progression of atherosclerosis following CD36 activation in an in vivo model of hypercholesterolemic (induced by 2% cholesterol containing diet) rabbits. In this direction, proteasomal activities by fluorometry and c-jun, phospo c-jun, JNK1, MMP-9 expressions by quantitative RT-PCR and immunoblotting were tested in aortic tissues. The effects of vitamin E on these changes were also investigated in this model. As a result, c-jun was phosphorylated following decreased proteasomal degradation in hypercholesterolemic group. MMP-9 expression was also increased in cholesterol group rabbits contributing to the development of atherosclerosis. In addition, vitamin E showed its effect by decreasing MMP-9 levels and phosphorylation of c-jun.
Collapse
Key Words
- AP-1
- AP-1, activator protein-1
- Atherosclerosis
- CD36, cluster of differentiation 36
- ERAD, endoplasmic-reticulum-associated protein degradation
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- HPLC, high-performance liquid chromatography
- Hypercholesterolemia
- JNK, c-Jun amino terminal kinase
- JNK1
- LDL, low density lipoprotein
- MAPK, mitogen-activated protein kinase
- MDA, malondialdehyde
- MMP, matrix metallo proteinase
- Proteasome
- TBA, thiobarbituric acid
- TNF a, tumor necrosis factor a
- UPS, ubiquitin-proteasome system
- Vitamin E
- oxLDL, oxidized low density lipoprotein
Collapse
|
25
|
Hoon Lee C, Kwak SC, Kim JY, Mee Oh H, Chual Rho M, Yoon KH, Yoo WH, Su Lee M, Oh J. Genipin Inhibits RANKL-Induced Osteoclast Differentiation Through Proteasome-Mediated Degradation of c-Fos Protein and Suppression of NF-κB Activation. J Pharmacol Sci 2014; 124:344-53. [DOI: 10.1254/jphs.13174fp] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
26
|
Chondrogianni N, Petropoulos I, Grimm S, Georgila K, Catalgol B, Friguet B, Grune T, Gonos ES. Protein damage, repair and proteolysis. Mol Aspects Med 2012; 35:1-71. [PMID: 23107776 DOI: 10.1016/j.mam.2012.09.001] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 09/26/2012] [Indexed: 01/10/2023]
Abstract
Proteins are continuously affected by various intrinsic and extrinsic factors. Damaged proteins influence several intracellular pathways and result in different disorders and diseases. Aggregation of damaged proteins depends on the balance between their generation and their reversal or elimination by protein repair systems and degradation, respectively. With regard to protein repair, only few repair mechanisms have been evidenced including the reduction of methionine sulfoxide residues by the methionine sulfoxide reductases, the conversion of isoaspartyl residues to L-aspartate by L-isoaspartate methyl transferase and deglycation by phosphorylation of protein-bound fructosamine by fructosamine-3-kinase. Protein degradation is orchestrated by two major proteolytic systems, namely the lysosome and the proteasome. Alteration of the function for both systems has been involved in all aspects of cellular metabolic networks linked to either normal or pathological processes. Given the importance of protein repair and degradation, great effort has recently been made regarding the modulation of these systems in various physiological conditions such as aging, as well as in diseases. Genetic modulation has produced promising results in the area of protein repair enzymes but there are not yet any identified potent inhibitors, and, to our knowledge, only one activating compound has been reported so far. In contrast, different drugs as well as natural compounds that interfere with proteolysis have been identified and/or developed resulting in homeostatic maintenance and/or the delay of disease progression.
Collapse
Affiliation(s)
- Niki Chondrogianni
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Helenic Research Foundation, 48 Vas. Constantinou Ave., 116 35 Athens, Greece.
| | - Isabelle Petropoulos
- Laboratoire de Biologie Cellulaire du Vieillissement, UR4-UPMC, IFR 83, Université Pierre et Marie Curie-Paris 6, 4 Place Jussieu, 75005 Paris, France
| | - Stefanie Grimm
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller University, Dornburger Straße 24, 07743 Jena, Germany
| | - Konstantina Georgila
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Helenic Research Foundation, 48 Vas. Constantinou Ave., 116 35 Athens, Greece
| | - Betul Catalgol
- Department of Biochemistry, Faculty of Medicine, Genetic and Metabolic Diseases Research Center (GEMHAM), Marmara University, Haydarpasa, Istanbul, Turkey
| | - Bertrand Friguet
- Laboratoire de Biologie Cellulaire du Vieillissement, UR4-UPMC, IFR 83, Université Pierre et Marie Curie-Paris 6, 4 Place Jussieu, 75005 Paris, France
| | - Tilman Grune
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller University, Dornburger Straße 24, 07743 Jena, Germany
| | - Efstathios S Gonos
- Institute of Biology, Medicinal Chemistry and Biotechnology, National Helenic Research Foundation, 48 Vas. Constantinou Ave., 116 35 Athens, Greece.
| |
Collapse
|
27
|
Abstract
Cancer is one of the most frightful diseases mostly resulting in mortality; it has recently become more possible to overcome with the help of new therapies. In this direction, carcinogenesis is defined as a complicated process that can include several different factors that contribute to its progress. Proteasome is implicated in cancer studies as it is the main degradation system for oxidatively damaged proteins and also for several proteins playing a role in the cell cycle and transcription, which are important for cancer improvement. Because of this crucial role of proteasome in cancer development, myriad in vitro and in vivo studies have focused on the proteasome in different cancer cases. In this chapter, the involvement of proteasome in the degradation of cancer-related proteins is explained with the results of representative studies. Related to these proteins, the use of proteasome inhibitors in cancer treatment is reviewed.
Collapse
|
28
|
Dong W, Li Y, Gao M, Hu M, Li X, Mai S, Guo N, Yuan S, Song L. IKKα contributes to UVB-induced VEGF expression by regulating AP-1 transactivation. Nucleic Acids Res 2011; 40:2940-55. [PMID: 22169952 PMCID: PMC3326327 DOI: 10.1093/nar/gkr1216] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Exposure to ultraviolet B (UVB) irradiation from sunlight induces the upregulation of VEGF, a potent angiogenic factor that is critical for mediating angiogenesis-associated photodamage. However, the molecular mechanisms related to UVB-induced VEGF expression have not been fully defined. Here, we demonstrate that one of the catalytic subunits of the IκB kinase complex (IKK), IKKα, plays a critical role in mediating UVB-induced VEGF expression in mouse embryonic fibroblasts (MEFs), which requires IKKα kinase activity but is independent of IKKβ, IKKγ and the transactivation of NF-κB. We further show that the transcriptional factor AP-1 functions as the downstream target of IKKα that is responsible for VEGF induction under UVB exposure. Both the accumulation of AP-1 component, c-Fos and the transactivation of AP-1 by UVB require the activated IKKα located within the nucleus. Moreover, nuclear IKKα can associate with c-Fos and recruit to the vegf promoter regions containing AP-1-responsive element and then trigger phosphorylation of the promoter-bound histone H3. Thus, our results have revealed a novel independent role for IKKα in controlling VEGF expression during the cellular UVB response by regulating the induction of the AP-1 component and phosphorylating histone H3 to facilitate AP-1 transactivation. Targeting IKKα shows promise for the prevention of UVB-induced angiogenesis and the associated photodamage.
Collapse
Affiliation(s)
- Wen Dong
- Department of Pathophysiology, Beijing Institute of Basic Medical Sciences, Beijing 100850, P R China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Brigelius-Flohé R, Flohé L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid Redox Signal 2011; 15:2335-81. [PMID: 21194351 PMCID: PMC3166203 DOI: 10.1089/ars.2010.3534] [Citation(s) in RCA: 434] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Convincing concepts of redox control of gene transcription have been worked out for prokaryotes and lower eukaryotes, whereas the knowledge on complex mammalian systems still resembles a patchwork of poorly connected findings. The article, therefore, reviews principles of redox regulation with special emphasis on chemical feasibility, kinetic requirements, specificity, and physiological context, taking well investigated mammalian transcription factor systems, nuclear transcription factor of bone marrow-derived lymphocytes (NF-κB), and kelch-like ECH-associated protein-1 (Keap1)/Nrf2, as paradigms. Major conclusions are that (i) direct signaling by free radicals is restricted to O(2)•- and •NO and can be excluded for fast reacting radicals such as •OH, •OR, or Cl•; (ii) oxidant signals are H(2)O(2), enzymatically generated lipid hydroperoxides, and peroxynitrite; (iii) free radical damage is sensed via generation of Michael acceptors; (iv) protein thiol oxidation/alkylation is the prominent mechanism to modulate function; (v) redox sensors must be thiol peroxidases by themselves or proteins with similarly reactive cysteine or selenocysteine (Sec) residues to kinetically compete with glutathione peroxidase (GPx)- and peroxiredoxin (Prx)-type peroxidases or glutathione-S-transferases, respectively, a postulate that still has to be verified for putative mammalian sensors. S-transferases and Prxs are considered for system complementation. The impact of NF-κB and Nrf2 on hormesis, management of inflammatory diseases, and cancer prevention is critically discussed.
Collapse
Affiliation(s)
- Regina Brigelius-Flohé
- Department Biochemistry of Micronutrients, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, Nuthetal, Germany.
| | | |
Collapse
|
30
|
Reyes-Hernández O, Mejía-García A, Sánchez-Ocampo E, Cabañas-Cortés M, Ramírez P, Chávez-González L, Gonzalez F, Elizondo G. Ube2l3 gene expression is modulated by activation of the aryl hydrocarbon receptor: implications for p53 ubiquitination. Biochem Pharmacol 2010; 80:932-40. [PMID: 20478272 PMCID: PMC6624428 DOI: 10.1016/j.bcp.2010.05.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 05/04/2010] [Accepted: 05/10/2010] [Indexed: 12/14/2022]
Abstract
Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a halogenated aromatic hydrocarbon and environmental contaminant, results in several deleterious effects, including fetal malformation and cancer. These effects are mediated by the aryl hydrocarbon receptor (AhR), a ligand-activated receptor that regulates the expression of genes encoding xenobiotic-metabolizing enzymes. Several reports suggest that AhR function is beyond the adaptive chemical response. In the present study, we analyzed and compared gene expression profiles of C57BL/6N wild-type (WT) and Ahr-null mice. DNA microarray and quantitative RT-PCR analyses revealed changes in the expression of genes involved in the ubiquitin-proteasome system (UPS). UPS has an important role in cellular homeostasis control and dysfunction of this pathway has been implicated in the development of several human pathologies. Protein ubiquitination is a multi-step enzymatic process that regulates the stability, function, and/or localization of the modified proteins. This system is highly regulated post-translationally by covalent modifications. However, little information regarding the transcriptional regulation of the genes encoding ubiquitin (Ub) proteins is available. Therefore, we investigated the role of the AhR in modulation of the UPS and regulation of Ube2l3 transcription, an E2 ubiquitin-conjugating enzyme, as well as the effects on p53 degradation. Our results indicate that AhR inactivation decreases on liver proteasome activity, probably due to a down-regulation on the expression of several proteasome subunits. On the other hand, AhR activation increases Ube2l3 mRNA and protein levels by controlling Ube2l3 gene expression, resulting in increased p53 ubiquitination and degradation. In agreement with this, induction of apoptosis was attenuated by the AhR activation.
Collapse
Affiliation(s)
- O.D. Reyes-Hernández
- Departamento de Toxicología, CINVESTAV-IPN, Zacatenco. México D.F., Av. IPN 2508, C.P. 07360, Mexico
| | - A. Mejía-García
- Departamento de Biología Celular, CINVESTAV-IPN, Zacatenco. México D.F., Av. IPN 2508, C.P. 07360, Mexico
| | - E.M. Sánchez-Ocampo
- Departamento de Biología Celular, CINVESTAV-IPN, Zacatenco. México D.F., Av. IPN 2508, C.P. 07360, Mexico
| | - M.A. Cabañas-Cortés
- Departamento de Biología Celular, CINVESTAV-IPN, Zacatenco. México D.F., Av. IPN 2508, C.P. 07360, Mexico
| | - P. Ramírez
- Unidad de Investigatión Multidisciplinaria, Laboratorio de Toxicología y Genética, Facultad de Estudios Superiores Cuautitlán, UNAM, Cuautitlán Izcallí, Mexico
| | | | - F.J. Gonzalez
- Laboratory of Metabolism, NCI, National Institutes of Health, Bethesda, MD, United States
| | - G. Elizondo
- Departamento de Biología Celular, CINVESTAV-IPN, Zacatenco. México D.F., Av. IPN 2508, C.P. 07360, Mexico
| |
Collapse
|
31
|
Sapin E, Bérod A, Léger L, Herman PA, Luppi PH, Peyron C. A very large number of GABAergic neurons are activated in the tuberal hypothalamus during paradoxical (REM) sleep hypersomnia. PLoS One 2010; 5:e11766. [PMID: 20668680 PMCID: PMC2909908 DOI: 10.1371/journal.pone.0011766] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 06/29/2010] [Indexed: 01/24/2023] Open
Abstract
We recently discovered, using Fos immunostaining, that the tuberal and mammillary hypothalamus contain a massive population of neurons specifically activated during paradoxical sleep (PS) hypersomnia. We further showed that some of the activated neurons of the tuberal hypothalamus express the melanin concentrating hormone (MCH) neuropeptide and that icv injection of MCH induces a strong increase in PS quantity. However, the chemical nature of the majority of the neurons activated during PS had not been characterized. To determine whether these neurons are GABAergic, we combined in situ hybridization of GAD67 mRNA with immunohistochemical detection of Fos in control, PS deprived and PS hypersomniac rats. We found that 74% of the very large population of Fos-labeled neurons located in the tuberal hypothalamus after PS hypersomnia were GAD-positive. We further demonstrated combining MCH immunohistochemistry and GAD67in situ hybridization that 85% of the MCH neurons were also GAD-positive. Finally, based on the number of Fos-ir/GAD+, Fos-ir/MCH+, and GAD+/MCH+ double-labeled neurons counted from three sets of double-staining, we uncovered that around 80% of the large number of the Fos-ir/GAD+ neurons located in the tuberal hypothalamus after PS hypersomnia do not contain MCH. Based on these and previous results, we propose that the non-MCH Fos/GABAergic neuronal population could be involved in PS induction and maintenance while the Fos/MCH/GABAergic neurons could be involved in the homeostatic regulation of PS. Further investigations will be needed to corroborate this original hypothesis.
Collapse
Affiliation(s)
- Emilie Sapin
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Université Claude Bernard-Lyon 1, Université de Lyon, Lyon, France
| | - Anne Bérod
- CNRS, EAC5006, Pharmacologie et Imagerie de la neurotransmission sérotoninergique, Université Claude Bernard-Lyon 1, Université de Lyon, Lyon, France
| | - Lucienne Léger
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Université Claude Bernard-Lyon 1, Université de Lyon, Lyon, France
| | - Paul A. Herman
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Université Claude Bernard-Lyon 1, Université de Lyon, Lyon, France
| | - Pierre-Hervé Luppi
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Université Claude Bernard-Lyon 1, Université de Lyon, Lyon, France
| | - Christelle Peyron
- CNRS, UMR5167, Physiopathologie des réseaux neuronaux du cycle veille-sommeil, Université Claude Bernard-Lyon 1, Université de Lyon, Lyon, France
- * E-mail:
| |
Collapse
|
32
|
c-Fos proteasomal degradation is activated by a default mechanism, and its regulation by NAD(P)H:quinone oxidoreductase 1 determines c-Fos serum response kinetics. Mol Cell Biol 2010; 30:3767-78. [PMID: 20498278 DOI: 10.1128/mcb.00899-09] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The short-lived proto-oncoprotein c-Fos is a component of the activator protein 1 (AP-1) transcription factor. A large region of c-Fos is intrinsically unstructured and susceptible to a recently characterized proteasomal ubiquitin-independent degradation (UID) pathway. UID is active by a default mechanism that is inhibited by NAD(P)H:quinone oxidoreductase 1 (NQO1), a 20S proteasome gatekeeper. Here, we show that NQO1 binds and induces robust c-Fos accumulation by blocking the UID pathway. c-Jun, a partner of c-Fos, also protects c-Fos from proteasomal degradation by default. Our findings suggest that NQO1 protects monomeric c-Fos from proteasomal UID, a function that is fulfilled later by c-Jun. We show that this process regulates c-Fos homeostasis (proteostasis) in response to serum stimulation, phosphorylation, nuclear translocation, and transcription activity. In addition, we show that NQO1 is important to ensure immediate c-Fos accumulation in response to serum, since a delayed response was observed under low NQO1 expression. These data suggest that in vivo, protein unstructured regions determine the kinetics and the homeostasis of regulatory proteins. Our data provide evidence for another layer of regulation of key regulatory proteins that functions at the level of protein degradation and is designed to ensure optimal formation of functional complexes such as AP-1.
Collapse
|
33
|
|
34
|
Cyclic Adenosine Monophosphate Suppresses the Transcription of Proinflammatory Cytokines via the Phosphorylated c-Fos Protein. Immunity 2009; 30:372-83. [DOI: 10.1016/j.immuni.2008.12.021] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 12/04/2008] [Accepted: 12/30/2008] [Indexed: 01/07/2023]
|
35
|
Sterz J, von Metzler I, Hahne JC, Lamottke B, Rademacher J, Heider U, Terpos E, Sezer O. The potential of proteasome inhibitors in cancer therapy. Expert Opin Investig Drugs 2008; 17:879-95. [PMID: 18491989 DOI: 10.1517/13543784.17.6.879] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND The ubiquitin-proteasome system has become a promising novel molecular target in cancer due to its critical role in cellular protein degradation, its interaction with cell cycle and apoptosis regulation and its unique mechanism of action. OBJECTIVE This review focuses both on preclinical results and on data from clinical trials with proteasome inhibitors in cancer. METHODS Results in hematological malignancies and solid tumors were included, and important data presented in abstract form were considered in this review. RESULTS/CONCLUSION Bortezomib as first-in-class proteasome inhibitor has proven to be highly effective in some hematological malignancies, overcomes conventional chemoresistance, directly induces cell cycle arrest and apoptosis, and also targets the tumor microenvironment. It has been granted approval by the FDA for relapsed multiple myeloma, and recently for relapsed mantle cell lymphoma. Combination chemotherapy regimens have been developed providing high remission rates and remission quality in frontline treatment or in the relapsed setting in multiple myeloma. The combination of proteasome inhibition with novel targeted therapies is an emerging field in oncology. Moreover, novel proteasome inhibitors, such as NPI-0052 and carfilzomib, have been developed. This review summarizes our knowledge of the ubiquitin-proteasome system and recent data from cancer clinical trials.
Collapse
Affiliation(s)
- Jan Sterz
- Charité-Universitätsmedizin Berlin, Department of Hematology and Oncology, Charité-Platz 1, 10117 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Jariel-Encontre I, Bossis G, Piechaczyk M. Ubiquitin-independent degradation of proteins by the proteasome. Biochim Biophys Acta Rev Cancer 2008; 1786:153-77. [PMID: 18558098 DOI: 10.1016/j.bbcan.2008.05.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Revised: 05/15/2008] [Accepted: 05/15/2008] [Indexed: 02/08/2023]
Abstract
The proteasome is the main proteolytic machinery of the cell and constitutes a recognized drugable target, in particular for treating cancer. It is involved in the elimination of misfolded, altered or aged proteins as well as in the generation of antigenic peptides presented by MHC class I molecules. It is also responsible for the proteolytic maturation of diverse polypeptide precursors and for the spatial and temporal regulation of the degradation of many key cell regulators whose destruction is necessary for progression through essential processes, such as cell division, differentiation and, more generally, adaptation to environmental signals. It is generally believed that proteins must undergo prior modification by polyubiquitin chains to be addressed to, and recognized by, the proteasome. In reality, however, there is accumulating evidence that ubiquitin-independent proteasomal degradation may have been largely underestimated. In particular, a number of proto-oncoproteins and oncosuppressive proteins are privileged ubiquitin-independent proteasomal substrates, the altered degradation of which may have tumorigenic consequences. The identification of ubiquitin-independent mechanisms for proteasomal degradation also poses the paramount question of the multiplicity of catabolic pathways targeting each protein substrate. As this may help design novel therapeutic strategies, the underlying mechanisms are critically reviewed here.
Collapse
Affiliation(s)
- Isabelle Jariel-Encontre
- Institut de Génétique Moléculaire de Montpellier, CNRS, UMR5535, IFR122, 1919 Route de Mende, Montpellier, F-34293, France
| | | | | |
Collapse
|
37
|
Manzo J, Miquel M, Toledo R, Mayor-Mar JA, Garcia LI, Aranda-Abreu GE, Caba M, Hernandez ME. Fos expression at the cerebellum following non-contact arousal and mating behavior in male rats. Physiol Behav 2008; 93:357-63. [PMID: 17936859 PMCID: PMC2978247 DOI: 10.1016/j.physbeh.2007.09.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2007] [Revised: 07/25/2007] [Accepted: 09/18/2007] [Indexed: 02/05/2023]
Abstract
The cerebellum is considered a center underlying fine movements, cognition, memory and sexual responses. The latter feature led us to correlate sexual arousal and copulation in male rats with neural activity at the cerebellar cortex. Two behavioral paradigms were used in this investigation: the stimulation of males by distant receptive females (non-contact sexual stimulation), and the execution of up to three consecutive ejaculations. The vermis area of the cerebellum was removed following behavioral experiments, cut into sagittal sections, and analyzed with Fos immunohistochemistry to determine neuronal activation. At the mid-vermis region (sections from the midline to 0.1 mm laterally), non-contact stimulation significantly increased the activity of granule neurons. The number of activated cells increased in every lobule, but lobules 1 and 6 to 9 showed the greatest increment. In sexual behavior tests, males reaching one ejaculation had a high number of activated neurons similar to those counted after non-contact stimulation. However, two or three consecutive ejaculations showed a smaller number of Fos-ir cells. In contrast to the mid-vermis region, sections farthest from the midline (0.1 to 0.9 mm laterally) revealed that only lobule 7 expressed activated neurons. These data suggest that a well-delineated group of granule neurons have a sexual biphasic response at the cerebellar vermis, and that Fos in them is under an active degradation mechanism. Thus, they participate as a neural substrate for male rat sexual responses with an activation-deactivation process corresponding with the sensory stimulation and motor performance occurring during copulation.
Collapse
Affiliation(s)
- Jorge Manzo
- Instituto de Neuroetologia, Universidad Veracruzana, Xalapa, Ver., Mexico.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Carle TL, Ohnishi YN, Ohnishi YH, Alibhai IN, Wilkinson MB, Kumar A, Nestler EJ. Proteasome-dependent and -independent mechanisms for FosB destabilization: identification of FosB degron domains and implications for DeltaFosB stability. Eur J Neurosci 2007; 25:3009-19. [PMID: 17561814 DOI: 10.1111/j.1460-9568.2007.05575.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The transcription factor DeltaFosB (Delta FosB) accumulates in a region-specific manner in the brain during chronic exposure to stress, drugs of abuse or other chronic stimuli. Once induced, DeltaFosB persists in the brain for at least several weeks following cessation of the chronic stimulus. The biochemical basis of the persistent expression of DeltaFosB has remained unknown. Here, we show that the FosB C-terminus, absent in DeltaFosB as a result of alternative splicing, contains two degron domains. Pulse-chase experiments of C-terminal truncation mutants of full-length FosB indicate that removal of its most C-terminal degron increases its half-life approximately fourfold, and prevents its proteasome-mediated degradation and ubiquitylation, properties similar to DeltaFosB. In addition, removal of a second degron domain, which generates DeltaFosB, further stabilizes FosB approximately twofold, but in a proteasome-independent manner. These data indicate that alternative splicing specifically removes two destabilizing elements from FosB in order to generate a longer-lived transcription factor, DeltaFosB, in response to chronic perturbations to the brain.
Collapse
Affiliation(s)
- Tiffany L Carle
- Department of Psychiatry and Center for Basic Neuroscience, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, Texas, 75390-9070, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
von Metzler I, Krebbel H, Hecht M, Manz RA, Fleissner C, Mieth M, Kaiser M, Jakob C, Sterz J, Kleeberg L, Heider U, Sezer O. Bortezomib inhibits human osteoclastogenesis. Leukemia 2007; 21:2025-34. [PMID: 17581612 DOI: 10.1038/sj.leu.2404806] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In multiple myeloma, the overexpression of receptor activator of nuclear factor kappa B (NF-kappaB) ligand (RANKL) leads to the induction of NF-kappaB and activator protein-1 (AP-1)-related osteoclast activation and enhanced bone resorption. The purpose of this study was to examine the molecular and functional effects of proteasome inhibition in RANKL-induced osteoclastogenesis. Furthermore, we aimed to compare the outcome of proteasome versus selective NF-kappaB inhibition using bortezomib (PS-341) and I-kappaB kinase inhibitor PS-1145. Primary human osteoclasts were derived from CD14+ precursors in presence of RANKL and macrophage colony-stimulating factor (M-CSF). Both bortezomib and PS-1145 inhibited osteoclast differentiation in a dose- and time-dependent manner and furthermore, the bone resorption activity of osteoclasts. The mechanisms of action involved in early osteoclast differentiation were found to be related to the inhibition of p38 mitogen-activated protein kinase pathways, whereas the later phase of differentiation and activation occurred due to inhibition of p38, AP-1 and NF-kappaB activation. The AP-1 blockade contributed to significant reduction of osteoclastic vascular endothelial growth factor production. In conclusion, our data demonstrate that proteasomal inhibition should be considered as a novel therapeutic option of cancer-induced lytic bone disease.
Collapse
Affiliation(s)
- I von Metzler
- Department of Hematology and Oncology, Charité - Universitätsmedizin Berlin, and Deutsches Rheumaforschungszentrum, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zavrski I, Jakob C, Kaiser M, Fleissner C, Heider U, Sezer O. Molecular and clinical aspects of proteasome inhibition in the treatment of cancer. Recent Results Cancer Res 2007; 176:165-76. [PMID: 17607924 DOI: 10.1007/978-3-540-46091-6_14] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The proteasome is a multicatalytic threonine protease responsible for intracellular protein turnover in eukaryotic cells, including the processing and degradation of several proteins involved in cell cycle control and the regulation of apoptosis. Preclinical studies have shown that the treatment with proteasome inhibitors results in decreased proliferation, induction of apoptosis, and sensitization of tumor cells against conventional chemotherapeutic agents and irradiation. The effects were conferred to stabilization of p21, p27, Bax, p53, I-KB, and the resulting inhibition of the nuclear factor-KB (NF-KB) activation. Bortezomib is the first proteasome inhibitor that has entered clinical trials. In multiple myeloma, both the FDA (United States Food and Drug Administration) and EMEA (European Medicine Evaluation Agency) granted an approval for the use of bortezomib (Velcade, Millennium Pharmaceuticals, Cambridge, MA, USA) for the treatment of relapsed multiple myeloma. At present, clinical trials are examining the activity in a variety of solid tumors and hematological malignancies.
Collapse
Affiliation(s)
- Ivana Zavrski
- Department of Hematology and Oncology, Charité Universitätsmedizin Berlin, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Nilsson J, Bjursell G, Kannius-Janson M. Nuclear Jak2 and transcription factor NF1-C2: a novel mechanism of prolactin signaling in mammary epithelial cells. Mol Cell Biol 2006; 26:5663-74. [PMID: 16847321 PMCID: PMC1592781 DOI: 10.1128/mcb.02095-05] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The classical mechanism by which prolactin transduces its signal in mammary epithelial cells is by activation of cytosolic signal transducer and activator of transcription 5 (Stat5) via a plasma membrane-associated prolactin receptor-Janus kinase 2 (Jak2) complex. Here we describe an alternative pathway through which prolactin via Jak2 localized in the nucleus activates the transcription factor nuclear factor 1-C2 (NF1-C2). Previous reports have demonstrated a nuclear localization of Jak2, but the physiologic importance of nuclear Jak2 has not been clear. We demonstrate that nuclear Jak2 regulates the amount of active NF1-C2 through tyrosine phosphorylation and proteasomal degradation. Our data also demonstrate a link between prolactin and p53 as well as the milk gene carboxyl ester lipase through nuclear Jak2 and NF1-C2. Hence, we describe a novel pathway through which nuclear Jak2 is subject to regulation by prolactin in mammary epithelial cells.
Collapse
Affiliation(s)
- Jeanette Nilsson
- Department of CMB/Molecular Biology, Box 462, S-405 30 Göteborg, Sweden
| | | | | |
Collapse
|
42
|
Abstract
In eukaryotes, thousands of genes have to be organized and expressed in the cell nucleus. Conformational and kinetic instability of nuclear structure and components appear to enable cells to use the encoded information selectively. The ubiquitin-proteasome system is active in distinct nuclear domains and plays a major role controlling the initial steps of gene expression, DNA repair and nuclear quality-control mechanisms. Recent work indicates that a tuned balance of ubiquitylation and proteasome-dependent protein degradation of nuclear proteins is instrumental in nuclear function and, when deregulated, leads to the development of diseases such as polyQ disorders and other neurodegenerative conditions.
Collapse
Affiliation(s)
- Anna von Mikecz
- Institut für umweltmedizinische Forschung, Heinrich-Heine-Universität Düsseldorf, Auf'm Hennekamp 50, 40225 Düsseldorf, Germany.
| |
Collapse
|
43
|
Colas D, Wagstaff J, Fort P, Salvert D, Sarda N. Sleep disturbances in Ube3a maternal-deficient mice modeling Angelman syndrome. Neurobiol Dis 2006; 20:471-8. [PMID: 15921919 DOI: 10.1016/j.nbd.2005.04.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Revised: 03/23/2005] [Accepted: 04/10/2005] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Angelman syndrome (AS) is a severe neurodevelopmental disorder with electroencephalographic (EEG) abnormalities and sleep disturbances. It results from lack of the functional maternal allele of UBE3A, which encodes a ubiquitin-protein ligase. Different mechanisms of UBE3A inactivation correlate with clinical phenotypes of varying severity; the majority of cases of AS are due to a de novo maternal deletion of the 15q11-q13 region. METHODS Ube3a maternal-deficient mice (Ube3a m-/p+) were generated in a C57Bl/6J background. This study compares cortical EEG and architecture of the sleep-waking cycle in adult Ube3a m-/p+ mice compared with those of age-matched WT (m+/p+) mice, under baseline conditions or after 4-h sleep deprivation (SD). RESULTS Ube3a m-/p+ mice exhibited: reduced slow-wave sleep (SWS) amount with increase waking (W) at the dark/light transitions; increased SWS and W episode numbers; and deterioration of paradoxical sleep (PS) over 24 h [amount: -44%; episode duration: -46%; episode number: -40%; theta peak frequency (TPF) acceleration: 7.6 Hz vs. 7.0 Hz in WT mice]. Characteristic paroxysmal EEG discharges are observed during W and SWS associated with synchronous muscle bursting activity during hypoactive W. During the recovery period following SD, Ube3a m-/p+ mice exhibited no rebound either in slow-wave activity (+89% in WT) or in delta-power spectra but a slight rebound in PS amount (+20%). CONCLUSIONS These data validate the mouse model produced by null mutation of the maternal Ube3a gene and provide useful results to investigate and better understand the molecular basis of sleep disturbances in AS patients.
Collapse
Affiliation(s)
- Damien Colas
- EA 3734, Claude Bernard University, 8 av. Rockefeller, 69373 Lyon Cedex 08, France
| | | | | | | | | |
Collapse
|
44
|
Eriksson M, Taskinen M, Leppä S. Mitogen activated protein kinase-dependent activation of c-Jun and c-Fos is required for neuronal differentiation but not for growth and stress response in PC12 cells. J Cell Physiol 2006; 210:538-48. [PMID: 17111371 DOI: 10.1002/jcp.20907] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
MAPK-dependent activation of AP-1 protein c-Jun is involved in PC12 cell differentiation and apoptosis. However, the role of other AP-1 proteins and their connection to MAPKs during growth, differentiation and apoptosis has remained elusive. Here we studied the activation of AP-1 proteins in response to ERK, JNK, and p38 signaling upon NGF, EGF and anisomycin exposures. All treatments caused different kinetics and strength of MAPK and AP-1 activities. NGF induced persistent ERK and AP-1 activities, whereas upon EGF and anisomycin exposures, their activities were only weakly and transiently induced. The sustained AP-1 activity was associated with concomitant c-Fos and c-Jun expression and phoshorylation, which were JNK and ERK dependent. While inhibition of the ERK, JNK, and p38 activities partially prevented AP-1 activity and suppressed differentiation, none of them was required for anisomycin-induced apoptosis. The importance of c-Fos and c-Jun as mediators of differentiation was demonstrated by the findings that the corresponding siRNAs suppressed NGF-induced neurite outgrowth. However, the capacity of c-Fos to promote differentiation required cooperation with Jun proteins. In contrast, Fra-2 expression was not required for the differentiation response. Together, the results show that sustained c-Jun and c-Fos activities mediate MAPK signaling and are essential for differentiation of PC12 cells.
Collapse
Affiliation(s)
- Minna Eriksson
- Department of Oncology, Helsinki University Central Hospital, HUCH, Finland
| | | | | |
Collapse
|
45
|
Ito Y, Inoue D, Kido S, Matsumoto T. c-Fos degradation by the ubiquitin-proteasome proteolytic pathway in osteoclast progenitors. Bone 2005; 37:842-9. [PMID: 16172035 DOI: 10.1016/j.bone.2005.04.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2005] [Revised: 03/22/2005] [Accepted: 04/22/2005] [Indexed: 11/28/2022]
Abstract
c-Fos is an immediate early gene type proto-oncogene that belongs to the AP (activator protein)-1 transcription factor family. Gene knockout experiments have demonstrated that, among the Fos family, only c-Fos is indispensable for osteoclast differentiation but that c-Fos can be substituted for by other Fos family members including FosB/DeltaFosB, Fra-1 and Fra-2, in most other tissues and cells. To further understand a unique role of c-Fos in osteoclastogenesis, we investigated the temporal profile and regulatory mode of expression of c-Fos during the course of osteoclast differentiation. The results indicated that c-Fos protein gradually increased in preosteoclasts during differentiation to a greater extent than that of mRNA induction. We then determined the proteolytic pathway of c-Fos conferring unstable nature on c-Fos protein in a preosteoclastic cell line, RAW264.7. Proteasome inhibitors including MG132 and Z-LLF caused a rapid increase in c-Fos protein expression in these cells within several hours, but other inhibitors of cysteine protease (E-64), lysosome (chloroquine) and calpain (ALLM) did not. Moreover, the proteasome inhibitors caused an extensive accumulation of ubiquitinated c-Fos protein and an approximately three-fold extension of the c-Fos protein half-life. We therefore conclude that the ubiquitin-proteasome system is the major proteolytic pathway conferring instability on c-Fos protein in preosteoclasts. Our results further imply that c-Fos stabilization due to dynamic changes in the ubiquitin-proteasome-dependent degradation may be involved in the accumulation of c-Fos protein in differentiating preosteoclasts.
Collapse
Affiliation(s)
- Yuji Ito
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School, Institute of Health Biosciences, 3-18-15 Kuramoto-cho Tokushima 770-8503, Japan
| | | | | | | |
Collapse
|
46
|
Hipp MS, Kalveram B, Raasi S, Groettrup M, Schmidtke G. FAT10, a ubiquitin-independent signal for proteasomal degradation. Mol Cell Biol 2005; 25:3483-91. [PMID: 15831455 PMCID: PMC1084302 DOI: 10.1128/mcb.25.9.3483-3491.2005] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
FAT10 is a small ubiquitin-like modifier that is encoded in the major histocompatibility complex and is synergistically inducible by tumor necrosis factor alpha and gamma interferon. It is composed of two ubiquitin-like domains and possesses a free C-terminal diglycine motif that is required for the formation of FAT10 conjugates. Here we show that unconjugated FAT10 and a FAT10 conjugate were rapidly degraded by the proteasome at a similar rate. Fusion of FAT10 to the N terminus of very long-lived proteins enhanced their degradation rate as potently as fusion with ubiquitin did. FAT10-green fluorescent protein fusion proteins were not cleaved but entirely degraded, suggesting that FAT10-specific deconjugating enzymes were not present in the analyzed cell lines. Interestingly, the prevention of ubiquitylation of FAT10 by mutation of all lysines or by expression in ubiquitylation-deficient cells did not affect FAT10 degradation. Thus, conjugation with FAT10 is an alternative and ubiquitin-independent targeting mechanism for degradation by the proteasome, which, in contrast to polyubiquitylation, is cytokine inducible and irreversible.
Collapse
Affiliation(s)
- Mark Steffen Hipp
- Universität Konstanz, Fachbereich Biologie/Immunologie, P1101, Universitätsstrasse 10, D-78457 Konstanz, Germany
| | | | | | | | | |
Collapse
|
47
|
Kalogeris T, Gray L, Laroux FS, Cockrell A, Fuseler J, Conner EM, Brand S, Grisham MB. Selective proteasome inhibitors as anti-inflammatory agents. Expert Opin Investig Drugs 2005; 8:1397-407. [PMID: 15992157 DOI: 10.1517/13543784.8.9.1397] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Nuclear factor kappaB (NF-kappaB) is an ubiquitous transcription factor and pleiotropic regulator of numerous inflammatory and immune responses. Once activated, NF-kappaB translocates from the cytosol to the nucleus of the cell, where it binds to its consensus sequence on the promoter-enhancer region of different genes. By so doing, this activates the transcription of a variety of different pro-inflammatory cytokines, adhesion molecules and specific enzymes, such as the inducible forms of nitric oxide synthase and cyclooxygenase. A number of different cytokines, bacterial products and oxidants activate NF-kappaB via selective phosphorlyation, polyubiquitination and degradation of the inhibitor protein, IkappaB. Since the 26S proteasome complex degrades the post-translationally modified IkappaB, thereby liberating the transcriptionally active p50/p65 heterodimeric NF-kappaB, this proteolytic complex represents a critical step in the activation of NF-kappaB. This review discusses the basic biology of the ubiquitin-proteasome pathway as it relates to the inflammatory response, and highlights those studies demonstrating that selective proteasome inhibitors are effective anti-inflammatory agents in vivo.
Collapse
Affiliation(s)
- T Kalogeris
- Department of Surgery, Louisiana State University Medical Center, PO Box 33932, 1501 Kings Highway, Shreveport, LA 71130 , USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Kim DJ, Murray IA, Burns AM, Gonzalez FJ, Perdew GH, Peters JM. Peroxisome proliferator-activated receptor-beta/delta inhibits epidermal cell proliferation by down-regulation of kinase activity. J Biol Chem 2005; 280:9519-27. [PMID: 15632134 DOI: 10.1074/jbc.m413808200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recent work has shown that peroxisome proliferator-activated receptor beta (PPARbeta) attenuates cell proliferation and skin carcinogenesis, and this is due in part to regulation of ubiquitin C expression. In these studies, the role of PPARbeta in modulating ubiquitin-dependent protein kinase Calpha (PKCalpha) levels and phosphorylation signaling pathways was evaluated. Intracellular phosphorylation analysis showed that phosphorylated PKCalpha and other kinases were lower in wild-type mouse skin treated with 12-O-tetradecanoylphorbol-13-acetate (TPA) as compared with PPARbeta-null mouse skin. No differences in expression levels of other PKC isoforms present in skin were observed. Lower ubiquitination of PKCalpha was found in TPA-treated PPARbeta-null skin as compared with wild-type, and inhibition of ubiquitin-dependent proteasome degradation prevented TPA-induced down-regulation of PKCalpha. The activity of PKCalpha and downstream signaling kinases is enhanced, and expression of cyclooxygenase-2 (COX-2) is significantly greater, in PPARbeta-null mouse skin in response to TPA compared with wild-type mouse skin. Inhibition of PKCalpha or COX-2 reduced cell proliferation in TPA-treated PPARbeta-null keratinocytes in a dose-dependent manner, whereas it only slightly influenced cell proliferation in wild-type keratinocytes. Combined, these studies provide strong evidence that PPARbeta attenuates cell proliferation by modulating PKCalpha/Raf1/MEK/ERK activity that may be due in part to reduced ubiquitin-dependent turnover of PKCalpha.
Collapse
Affiliation(s)
- Dae J Kim
- Department of Veterinary Science and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
| | | | | | | | | | | |
Collapse
|
49
|
Plafker SM, Plafker KS, Weissman AM, Macara IG. Ubiquitin charging of human class III ubiquitin-conjugating enzymes triggers their nuclear import. ACTA ACUST UNITED AC 2004; 167:649-59. [PMID: 15545318 PMCID: PMC2172591 DOI: 10.1083/jcb.200406001] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Ubiquitin is a small polypeptide that is conjugated to proteins and commonly serves as a degradation signal. The attachment of ubiquitin (Ub) to a substrate proceeds through a multi-enzyme cascade involving an activating enzyme (E1), a conjugating enzyme (E2), and a protein ligase (E3). We previously demonstrated that a murine E2, UbcM2, is imported into nuclei by the transport receptor importin-11. We now show that the import mechanism for UbcM2 and two other human class III E2s (UbcH6 and UBE2E2) uniquely requires the covalent attachment of Ub to the active site cysteine of these enzymes. This coupling of E2 activation and transport arises from the selective interaction of importin-11 with the Ub-loaded forms of these enzymes. Together, these findings reveal that Ub charging can function as a nuclear import trigger, and identify a novel link between E2 regulation and karyopherin-mediated transport.
Collapse
Affiliation(s)
- Scott M Plafker
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | | | |
Collapse
|
50
|
Houben K, Dominguez C, van Schaik FMA, Timmers HTM, Bonvin AMJJ, Boelens R. Solution Structure of the Ubiquitin-conjugating Enzyme UbcH5B. J Mol Biol 2004; 344:513-26. [PMID: 15522302 DOI: 10.1016/j.jmb.2004.09.054] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Revised: 09/17/2004] [Accepted: 09/20/2004] [Indexed: 11/19/2022]
Abstract
The ubiquitination pathway is the main pathway for protein degradation in eukaryotic cells. The attachment of ubiquitin to a substrate protein is catalyzed by three types of enzymes, namely a ubiquitin activating enzyme (E1), a ubiquitin-conjugating enzyme (E2), and a ubiquitin ligase (E3). Here, the structure of the human ubiquitin-conjugating enzyme (E2) UbcH5B has been solved by a combination of homology modeling, NMR relaxation data and automated NOE assignments. Comparison to E2 structures solved previously by X-ray crystallography or NMR shows in all cases the same compact fold, but differences are observed in the orientation of both N and C-terminal alpha-helices. The N-terminal helix that is involved in binding to ubiquitin ligases (E3) displays a different position, which could have consequences for precise E2-E3 recognition. In addition, multiple conformations of the side-chain of Asn77 are found in solution, which contrasts the single hydrogen-bonded conformation in the crystal structures of E2 enzymes. The possible implication of this conformational freedom of Asn77 for its catalytic function is discussed.
Collapse
Affiliation(s)
- Klaartje Houben
- Department of NMR Spectroscopy, Bijvoet Center for Biomolecular Research, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|