1
|
Abdelsam SS, Ghanem SK, Zahid MA, Abunada HH, Bader L, Raïq H, Khan A, Parray A, Djouhri L, Agouni A. Human antigen R: Exploring its inflammatory response impact and significance in cardiometabolic disorders. J Cell Physiol 2024; 239:e31229. [PMID: 38426269 DOI: 10.1002/jcp.31229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/30/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
RNA-binding proteins (RBPs) play a crucial role in the regulation of posttranscriptional RNA networks, which can undergo dysregulation in many pathological conditions. Human antigen R (HuR) is a highly researched RBP that plays a crucial role as a posttranscriptional regulator. HuR plays a crucial role in the amplification of inflammatory signals by stabilizing the messenger RNA of diverse inflammatory mediators and key molecular players. The noteworthy correlations between HuR and its target molecules, coupled with the remarkable impacts reported on the pathogenesis and advancement of multiple diseases, position HuR as a promising candidate for therapeutic intervention in diverse inflammatory conditions. This review article examines the significance of HuR as a member of the RBP family, its regulatory mechanisms, and its implications in the pathophysiology of inflammation and cardiometabolic illnesses. Our objective is to illuminate potential directions for future research and drug development by conducting a comprehensive analysis of the existing body of research on HuR.
Collapse
Affiliation(s)
- Shahenda Salah Abdelsam
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Sarah Khalaf Ghanem
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Muhammad Ammar Zahid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Hanan H Abunada
- Office of Vice President for Research and Graduate Studies, Qatar University, Doha, Qatar
| | - Loulia Bader
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Hicham Raïq
- Department of Social Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Abbas Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Aijaz Parray
- The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Laiche Djouhri
- Department of Basic Medical Science, College of Medicine, QU health, Qatar University, Doha, Qatar
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
- Office of Vice President for Medical & Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
2
|
Schroeder HT, De Lemos Muller CH, Heck TG, Krause M, Homem de Bittencourt PI. Heat shock response during the resolution of inflammation and its progressive suppression in chronic-degenerative inflammatory diseases. Cell Stress Chaperones 2024; 29:116-142. [PMID: 38244765 PMCID: PMC10939074 DOI: 10.1016/j.cstres.2024.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024] Open
Abstract
The heat shock response (HSR) is a crucial biochemical pathway that orchestrates the resolution of inflammation, primarily under proteotoxic stress conditions. This process hinges on the upregulation of heat shock proteins (HSPs) and other chaperones, notably the 70 kDa family of heat shock proteins, under the command of the heat shock transcription factor-1. However, in the context of chronic degenerative disorders characterized by persistent low-grade inflammation (such as insulin resistance, obesity, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular diseases) a gradual suppression of the HSR does occur. This work delves into the mechanisms behind this phenomenon. It explores how the Western diet and sedentary lifestyle, culminating in the endoplasmic reticulum stress within adipose tissue cells, trigger a cascade of events. This cascade includes the unfolded protein response and activation of the NOD-like receptor pyrin domain-containing protein-3 inflammasome, leading to the emergence of the senescence-associated secretory phenotype and the propagation of inflammation throughout the body. Notably, the activation of the NOD-like receptor pyrin domain-containing protein-3 inflammasome not only fuels inflammation but also sabotages the HSR by degrading human antigen R, a crucial mRNA-binding protein responsible for maintaining heat shock transcription factor-1 mRNA expression and stability on heat shock gene promoters. This paper underscores the imperative need to comprehend how chronic inflammation stifles the HSR and the clinical significance of evaluating the HSR using cost-effective and accessible tools. Such understanding is pivotal in the development of innovative strategies aimed at the prevention and treatment of these chronic inflammatory ailments, which continue to take a heavy toll on global health and well-being.
Collapse
Affiliation(s)
- Helena Trevisan Schroeder
- Laboratory of Cellular Physiology (FisCel), Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Henrique De Lemos Muller
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Thiago Gomes Heck
- Post Graduate Program in Integral Health Care (PPGAIS-UNIJUÍ/UNICRUZ/URI), Regional University of Northwestern Rio Grande Do Sul State (UNIJUI) and Post Graduate Program in Mathematical and Computational Modeling (PPGMMC), UNIJUI, Ijuí, Rio Grande do Sul, Brazil
| | - Mauricio Krause
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology (FisCel), Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
3
|
Schroeder HT, De Lemos Muller CH, Heck TG, Krause M, Homem de Bittencourt PI. The dance of proteostasis and metabolism: Unveiling the caloristatic controlling switch. Cell Stress Chaperones 2024; 29:175-200. [PMID: 38331164 PMCID: PMC10939077 DOI: 10.1016/j.cstres.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/10/2024] Open
Abstract
The heat shock response (HSR) is an ancient and evolutionarily conserved mechanism designed to restore cellular homeostasis following proteotoxic challenges. However, it has become increasingly evident that disruptions in energy metabolism also trigger the HSR. This interplay between proteostasis and energy regulation is rooted in the fundamental need for ATP to fuel protein synthesis and repair, making the HSR an essential component of cellular energy management. Recent findings suggest that the origins of proteostasis-defending systems can be traced back over 3.6 billion years, aligning with the emergence of sugar kinases that optimized glycolysis around 3.594 billion years ago. This evolutionary connection is underscored by the spatial similarities between the nucleotide-binding domain of HSP70, the key player in protein chaperone machinery, and hexokinases. The HSR serves as a hub that integrates energy metabolism and resolution of inflammation, further highlighting its role in maintaining cellular homeostasis. Notably, 5'-adenosine monophosphate-activated protein kinase emerges as a central regulator, promoting the HSR during predominantly proteotoxic stress while suppressing it in response to predominantly metabolic stress. The complex relationship between 5'-adenosine monophosphate-activated protein kinase and the HSR is finely tuned, with paradoxical effects observed under different stress conditions. This delicate equilibrium, known as caloristasis, ensures that cellular homeostasis is maintained despite shifting environmental and intracellular conditions. Understanding the caloristatic controlling switch at the heart of this interplay is crucial. It offers insights into a wide range of conditions, including glycemic control, obesity, type 2 diabetes, cardiovascular and neurodegenerative diseases, reproductive abnormalities, and the optimization of exercise routines. These findings highlight the profound interconnectedness of proteostasis and energy metabolism in cellular function and adaptation.
Collapse
Affiliation(s)
- Helena Trevisan Schroeder
- Laboratory of Cellular Physiology (FisCel) Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Henrique De Lemos Muller
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Thiago Gomes Heck
- Post Graduate Program in Integral Health Care (PPGAIS-UNIJUÍ/UNICRUZ/URI), Regional University of Northwestern Rio Grande Do Sul State (UNIJUI) and Post Graduate Program in Mathematical and Computational Modeling (PPGMMC), UNIJUI, Ijuí, Rio Grande do Sul, Brazil
| | - Mauricio Krause
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology (FisCel) Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
4
|
Raheja H, George B, Tripathi SK, Saha S, Maiti TK, Das S. Hepatitis C virus non-structural proteins modulate cellular kinases for increased cytoplasmic abundance of host factor HuR and facilitate viral replication. PLoS Pathog 2023; 19:e1011552. [PMID: 37540723 PMCID: PMC10431626 DOI: 10.1371/journal.ppat.1011552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/16/2023] [Accepted: 07/11/2023] [Indexed: 08/06/2023] Open
Abstract
Host protein HuR translocation from nucleus to cytoplasm following infection is crucial for the life cycle of several RNA viruses including hepatitis C virus (HCV), a major causative agent of hepatocellular carcinoma. HuR assists the assembly of replication-complex on the viral-3'UTR, and its depletion hampers viral replication. Although cytoplasmic HuR is crucial for HCV replication, little is known about how the virus orchestrates the mobilization of HuR into the cytoplasm from the nucleus. We show that two viral proteins, NS3 and NS5A, act co-ordinately to alter the equilibrium of the nucleo-cytoplasmic movement of HuR. NS3 activates protein kinase C (PKC)-δ, which in-turn phosphorylates HuR on S318 residue, triggering its export to the cytoplasm. NS5A inactivates AMP-activated kinase (AMPK) resulting in diminished nuclear import of HuR through blockade of AMPK-mediated phosphorylation and acetylation of importin-α1. Cytoplasmic retention or entry of HuR can be reversed by an AMPK activator or a PKC-δ inhibitor. Our findings suggest that efforts should be made to develop inhibitors of PKC-δ and activators of AMPK, either separately or in combination, to inhibit HCV infection.
Collapse
Affiliation(s)
- Harsha Raheja
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Biju George
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Sachin Kumar Tripathi
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | | | | | - Saumitra Das
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| |
Collapse
|
5
|
Wang J, Ren P, Zeng Z, Ma L, Li Y, Zhang H, Guo W. Inhibition of translocator protein 18 kDa suppressed the progression of glioma via the ELAV-like RNA-binding protein 1/MAPK-activated protein kinase 3 axis. Bioengineered 2022; 13:7457-7470. [PMID: 35285415 PMCID: PMC9208533 DOI: 10.1080/21655979.2022.2048992] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Jingya Wang
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, Dongcheng,China
- Department of Gastroenterology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Peng Ren
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, Dongcheng,China
| | - Zhirui Zeng
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| | - Li Ma
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, Dongcheng,China
| | - Yunjun Li
- Department of Neurosurgery, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, Dongcheng, China
| | - Hongmei Zhang
- Department of Gastroenterology, The Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Wenzhi Guo
- Department of Anesthesiology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, Dongcheng,China
| |
Collapse
|
6
|
Schaupp CM, Botta D, White CC, Scoville DK, Srinouanprachanh S, Bammler TK, MacDonald J, Kavanagh TJ. Persistence of improved glucose homeostasis in Gclm null mice with age and cadmium treatment. Redox Biol 2022; 49:102213. [PMID: 34953454 PMCID: PMC8715110 DOI: 10.1016/j.redox.2021.102213] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/23/2022] Open
Abstract
Antioxidant signaling/communication is among the most important cellular defense and survival pathways, and the importance of redox signaling and homeostasis in aging has been well-documented. Intracellular levels of glutathione (GSH), a very important endogenous antioxidant, both govern and are governed by the Nrf2 pathway through expression of genes involved in its biosynthesis, including the subunits of the rate-limiting enzyme (glutamate cysteine ligase, GCL) in GSH production, GCLC and GCLM. Mice homozygous null for the Gclm gene are severely deficient in GSH compared to wild-type controls, expressing approximately 10% of normal GSH levels. To compensate for GSH deficiency, Gclm null mice have upregulated redox-regulated genes, and, surprisingly, are less susceptible to certain types of oxidative damage. Furthermore, young Gclm null mice display an interesting lean phenotype, resistance to high fat diet-induced diabetes and obesity, improved insulin and glucose tolerance, and decreased expression of genes involved in lipogenesis. However, the persistence of this phenotype has not been investigated into old age, which is important in light of studies which suggest aging attenuates antioxidant signaling, particularly in response to exogenous stimuli. In this work, we addressed whether aging compromises the favorable phenotype of increased antioxidant activity and improved glucose homeostasis observed in younger Gclm null mice. We present data showing that under basal conditions and in response to cadmium exposure (2 mg/kg, dosed once via intraperitoneal injection), the phenotype previously described in young (<6 months) Gclm null mice persists into old age (24+ months). We also provide evidence that transcriptional activation of the Nrf2, AMPK, and PPARγ pathways underlie the favorable metabolic phenotype observed previously in young Gclm null mice.
Collapse
Affiliation(s)
- Christopher M Schaupp
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Dianne Botta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Collin C White
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - David K Scoville
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Sengkeo Srinouanprachanh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA
| | - Terrance J Kavanagh
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
7
|
Fattahi F, Ellis JS, Sylvester M, Bahleda K, Hietanen S, Correa L, Lugogo NL, Atasoy U. HuR-Targeted Inhibition Impairs Th2 Proinflammatory Responses in Asthmatic CD4 + T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:38-48. [PMID: 34862257 DOI: 10.4049/jimmunol.2100635] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/01/2021] [Indexed: 11/19/2022]
Abstract
RNA-binding protein HuR (ELAVL1) is a master regulator of gene expression in human pathophysiology. Its dysregulation plays an important role in many diseases. We hypothesized that HuR plays an important role in Th2 inflammation in asthma in both mouse and human. To address this, we used a model of airway inflammation in a T cell-specific knockout mouse model, distal lck-Cre HuRfl/fl, as well as small molecule inhibitors in human peripheral blood-derived CD4+ T cells. Peripheral CD4+ T cells were isolated from 26 healthy control subjects and 45 asthmatics (36 type 2 high and 9 non-type 2 high, determined by blood eosinophil levels and fraction of exhaled NO). Our mouse data showed conditional ablation of HuR in T cell-abrogated Th2 differentiation, cytokine production, and lung inflammation. Studies using human T cells showed that HuR protein levels in CD4+ T cells were significantly higher in asthmatics compared with healthy control subjects. The expression and secretion of Th2 cytokines were significantly higher in asthmatics compared with control subjects. AMP-activated protein kinase activator treatment reduced the expression of several cytokines in both type 2 high and non-type 2 high asthma groups. However, the effects of CMLD-2 (a HuR-specific inhibitor) were more specific to endotype-defining cytokines in type 2 high asthmatics. Taken together, these data suggest that HuR plays a permissive role in both allergen and non-allergen-driven airway inflammation by regulating key genes, and that interfering with its function may be a novel method of asthma treatment.
Collapse
Affiliation(s)
- Fatemeh Fattahi
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Jason S Ellis
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Michael Sylvester
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI.,Department of Otolaryngology, Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI
| | - Kristin Bahleda
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Samuel Hietanen
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Luis Correa
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| | - Njira L Lugogo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI; and
| | - Ulus Atasoy
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI; .,Division of Allergy-Immunology, Ann Arbor VA Health System, Ann Arbor, MI
| |
Collapse
|
8
|
Liu YJ, Kuo HC, Chern Y. A system-wide mislocalization of RNA-binding proteins in motor neurons is a new feature of ALS. Neurobiol Dis 2021; 160:105531. [PMID: 34634461 DOI: 10.1016/j.nbd.2021.105531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 09/28/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a motor neuron disease characterized by progressive degeneration of motor neurons. Mislocalization of TAR DNA-binding protein 43 (TDP-43) is an early event in the formation of cytoplasmic TDP-43-positive inclusions in motor neurons and a hallmark of ALS. However, the underlying mechanism and the pathogenic impact of this mislocalization are relatively unexplored. We previously reported that abnormal AMPK activation mediates TDP-43 mislocalization in motor neurons of humans and mice with ALS. In the present study, we hypothesized that other nuclear proteins are mislocalized in the cytoplasm of motor neurons due to the AMPK-mediated phosphorylation of importin-α1 and subsequently contribute to neuronal degeneration in ALS. To test this hypothesis, we analyzed motor neurons of sporadic ALS patients and found that when AMPK is activated, importin-α1 is abnormally located in the nucleus. Multiple integrative molecular and cellular approaches (including proteomics, immunoprecipitation/western blot analysis, immunohistological evaluations and gradient analysis of preribosomal complexes) were employed to demonstrate that numerous RNA binding proteins are mislocalized in a rodent motor neuron cell line (NSC34) and human motor neurons derived from iPSCs during AMPK activation. We used comparative proteomic analysis of importin-α1 complexes that were immunoprecipitated with a phosphorylation-deficient mutant of importin-α1 (importin-α1-S105A) and a phosphomimetic mutant of importin-α1 (importin-α1-S105D) to identify 194 proteins that have stronger affinity for the unphosphorylated form than the phosphorylated form of importin-α1. Furthermore, GO and STRING analyses suggested that RNA processing and protein translation is the major machinery affected by abnormalities in the AMPK-importin-α1 axis. Consistently, the expression of importin-α1-S105D alters the assembly of preribosomal complexes and increases cell apoptosis. Collectively, we propose that by impairing importin-α1-mediated nuclear import, abnormal AMPK activation in motor neurons alters the cellular distribution of many RNA-binding proteins, which pathogenically affect multiple cellular machineries in motor neurons and contribute to ALS pathogenesis.
Collapse
Affiliation(s)
- Yu-Ju Liu
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yijuang Chern
- Division of Neuroscience, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
9
|
Afinanisa Q, Cho MK, Seong HA. AMPK Localization: A Key to Differential Energy Regulation. Int J Mol Sci 2021; 22:10921. [PMID: 34681581 PMCID: PMC8535671 DOI: 10.3390/ijms222010921] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 11/23/2022] Open
Abstract
As the central node between nutrition signaling input and the metabolic pathway, AMP-activated protein kinase (AMPK) is tightly regulated to maintain energy homeostasis. Subcellular compartmentalization of AMPK is one of the critical regulations that enables AMPK to access proper targets and generate appropriate responses to specific perturbations and different levels of stress. One of the characterized localization mechanisms is RanGTPase-driven CRM1 that recognizes the nuclear export sequence (NES) on the α subunit to translocate AMPK into the cytoplasm. Nuclear localization putatively employs RanGTPase-driven importin that might recognize the nuclear localization signal (NLS) present on the AMPKα2 kinase domain. Nucleo-cytoplasmic shuttling of AMPK is influenced by multiple factors, such as starvation, exercise, heat shock, oxidant, cell density, and circadian rhythm. Tissue-specific localization, which distributes AMPK trimers with different combinations, has also been shown to be vital in maintaining tissue-specific metabolism. Tissue-specific and subcellular distribution of AMPK might be attributed to differences in the expression of the subunit, the stabilization by protein regulators, tissue activity, and the localization of AMPK activators. Considering the importance of AMPK localization in coordinating signaling and metabolism, further research is due to fully elucidate the largely unknown complex mechanism underlying this regulation.
Collapse
Affiliation(s)
| | | | - Hyun-A Seong
- Department of Biochemistry, School of Biological Sciences, Chungbuk National University, Cheongju 28644, Korea; (Q.A.); (M.K.C.)
| |
Collapse
|
10
|
Al-Habeeb F, Aloufi N, Traboulsi H, Liu X, Nair P, Haston C, Azuelos I, Huang SK, White ES, Gallouzi IE, Di Marco S, Eidelman DH, Baglole CJ. Human antigen R promotes lung fibroblast differentiation to myofibroblasts and increases extracellular matrix production. J Cell Physiol 2021; 236:6836-6851. [PMID: 33855709 DOI: 10.1002/jcp.30380] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 01/12/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease of progressive scarring caused by excessive extracellular matrix (ECM) deposition and activation of α-SMA-expressing myofibroblasts. Human antigen R (HuR) is an RNA binding protein that promotes protein translation. Upon translocation from the nucleus to the cytoplasm, HuR functions to stabilize messenger RNA (mRNA) to increase protein levels. However, the role of HuR in promoting ECM production, myofibroblast differentiation, and lung fibrosis is unknown. Human lung fibroblasts (HLFs) treated with transforming growth factor β1 (TGF-β1) showed a significant increase in translocation of HuR from the nucleus to the cytoplasm. TGF-β-treated HLFs that were transfected with HuR small interfering RNA had a significant reduction in α-SMA protein as well as the ECM proteins COL1A1, COL3A, and FN1. HuR was also bound to mRNA for ACTA2, COL1A1, COL3A1, and FN. HuR knockdown affected the mRNA stability of ACTA2 but not that of the ECM genes COL1A1, COL3A1, or FN. In mouse models of pulmonary fibrosis, there was higher cytoplasmic HuR in lung structural cells compared to control mice. In human IPF lungs, there was also more cytoplasmic HuR. This study is the first to show that HuR in lung fibroblasts controls their differentiation to myofibroblasts and consequent ECM production. Further research on HuR could assist in establishing the basis for the development of new target therapy for fibrotic diseases, such as IPF.
Collapse
Affiliation(s)
- Fatmah Al-Habeeb
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Noof Aloufi
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Departments of Pathology, McGill University, Montreal, Quebec, Canada
| | - Hussein Traboulsi
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Xingxing Liu
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Parameswaran Nair
- Department of Medicine, McMaster University & St Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Christina Haston
- Department of Computer Science, Mathematics, Physics and Statistics, University of British Columbia, British Columbia, Canada
| | - Ilan Azuelos
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Steven K Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Eric S White
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Imed E Gallouzi
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Faculty of Medicine, Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - Sergio Di Marco
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Faculty of Medicine, Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
| | - David H Eidelman
- Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Carolyn J Baglole
- Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada.,Departments of Pathology, McGill University, Montreal, Quebec, Canada.,Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
11
|
ELAV Proteins Bind and Stabilize C/EBP mRNA in the Induction of Long-Term Memory in Aplysia. J Neurosci 2020; 41:947-959. [PMID: 33298536 DOI: 10.1523/jneurosci.2284-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/22/2020] [Accepted: 11/23/2020] [Indexed: 12/27/2022] Open
Abstract
Long-term memory (LTM) formation is a critical survival process by which an animal retains information about prior experiences to guide future behavior. In the experimentally advantageous marine mollusk Aplysia, LTM for sensitization can be induced by the presentation of two aversive shocks to the animal's tail. Each of these training trials recruits distinct growth factor signaling systems that promote LTM formation. Specifically, whereas intact TrkB signaling during Trial 1 promotes an initial and transient increase of the immediate early gene apc/ebp mRNA, a prolonged increase in apc/ebp gene expression required for LTM formation requires the addition of TGFβ signaling during Trial 2. Here we explored the molecular mechanisms by which Trial 2 achieves the essential prolonged gene expression of apc/ebp We find that this prolonged gene expression is not dependent on de novo transcription, but that apc/ebp mRNA synthesized by Trial 1 is post-transcriptionally stabilized by interacting with the RNA-binding protein ApELAV. This interaction is promoted by p38 MAPK activation initiated by TGFβ. We further demonstrate that blocking the interaction of ApELAV with its target mRNA during Trial 2 blocks both the prolonged increase in apc/ebp gene expression and the behavioral induction of LTM. Collectively, our findings elucidate both when and how ELAV proteins are recruited for the stabilization of mRNA in LTM formation. Stabilization of a transiently expressed immediate early gene mRNA by a repeated training trial may therefore serve as a "filter" for learning, permitting only specific events to cause lasting transcriptional changes and behavioral LTM.SIGNIFICANCE STATEMENT: In the present paper, we significantly extend the general field of molecular processing in long-term memory (LTM) by describing a novel form of pretranslational processing required for LTM, which relies on the stabilization of a newly synthesized mRNA by a class of RNA binding proteins (ELAVs). There are now compelling data showing that important processing can occur after transcription of a gene, but before translation of the message into protein. Although the potential importance of ELAV proteins in LTM formation has previously been reported, the specific actions of ELAV proteins during LTM formation remained to be understood. Our new findings thus complement and extend this literature by demonstrating when and how this post-transcriptional gene regulation is mediated in the induction of LTM.
Collapse
|
12
|
Mattam U, Talari NK, Paripati AK, Krishnamoorthy T, Sepuri NBV. Kisspeptin preserves mitochondrial function by inducing mitophagy and autophagy in aging rat brain hippocampus and human neuronal cell line. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118852. [PMID: 32926943 DOI: 10.1016/j.bbamcr.2020.118852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 09/07/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022]
Abstract
It has become amply clear that mitochondrial function defined by quality, quantity, dynamics, homeostasis, and regulated by mitophagy and mitochondrial biogenesis is a critical metric of human aging and disease. As a consequence, therapeutic interventions that can improve mitochondrial function can have a profound impact on human health and longevity. Kisspeptins are neuropeptides belonging to the family of metastasis suppressors that are known to regulate functions like fertility, reproduction, and metabolism. Using SKNSH cell line, hippocampus explant cultures and hippocampus of aging Wistar rat models, we show that Kisspeptin-10 (Kp) induces autophagy and mitophagy via calcium, Ca2+/CaM-dependent protein kinase kinase β (CaMKKβ), AMP-activated protein kinase (AMPK), and Unc-51 like autophagy activating kinase (ULK1) signaling pathway that is independent of mammalian target of rapamycin (mTOR). Intriguingly, Kp administration in vivo also results in the enhancement of mitochondrial number, complex I activity, and Adenosine Triphosphate (ATP) levels. This study uncovers potential effects of Kp in protecting mitochondrial health and as a possible therapeutic intervention to hippocampus associated impairments such as memory, cognitive aging, and other diseases linked to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Ushodaya Mattam
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, T.S., India.
| | - Noble Kumar Talari
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, T.S., India
| | - Arun Kumar Paripati
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, T.S., India
| | - Thanuja Krishnamoorthy
- Vectrogen Biologicals Pvt Ltd, Hyderabad, BioNEST, School of Life Sciences, University of Hyderabad, Hyderabad 500046, T.S., India
| | - Naresh Babu V Sepuri
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, T.S., India.
| |
Collapse
|
13
|
Koepsell H. Glucose transporters in the small intestine in health and disease. Pflugers Arch 2020; 472:1207-1248. [PMID: 32829466 PMCID: PMC7462918 DOI: 10.1007/s00424-020-02439-5] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 07/11/2020] [Accepted: 07/17/2020] [Indexed: 12/23/2022]
Abstract
Absorption of monosaccharides is mainly mediated by Na+-D-glucose cotransporter SGLT1 and the facititative transporters GLUT2 and GLUT5. SGLT1 and GLUT2 are relevant for absorption of D-glucose and D-galactose while GLUT5 is relevant for D-fructose absorption. SGLT1 and GLUT5 are constantly localized in the brush border membrane (BBM) of enterocytes, whereas GLUT2 is localized in the basolateral membrane (BLM) or the BBM plus BLM at low and high luminal D-glucose concentrations, respectively. At high luminal D-glucose, the abundance SGLT1 in the BBM is increased. Hence, D-glucose absorption at low luminal glucose is mediated via SGLT1 in the BBM and GLUT2 in the BLM whereas high-capacity D-glucose absorption at high luminal glucose is mediated by SGLT1 plus GLUT2 in the BBM and GLUT2 in the BLM. The review describes functions and regulations of SGLT1, GLUT2, and GLUT5 in the small intestine including diurnal variations and carbohydrate-dependent regulations. Also, the roles of SGLT1 and GLUT2 for secretion of enterohormones are discussed. Furthermore, diseases are described that are caused by malfunctions of small intestinal monosaccharide transporters, such as glucose-galactose malabsorption, Fanconi syndrome, and fructose intolerance. Moreover, it is reported how diabetes, small intestinal inflammation, parental nutrition, bariatric surgery, and metformin treatment affect expression of monosaccharide transporters in the small intestine. Finally, food components that decrease D-glucose absorption and drugs in development that inhibit or downregulate SGLT1 in the small intestine are compiled. Models for regulations and combined functions of glucose transporters, and for interplay between D-fructose transport and metabolism, are discussed.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute for Anatomy and Cell Biology, University of Würzburg, Koellikerstr 6, 97070, Würzburg, Germany.
| |
Collapse
|
14
|
Wu M, Tong CWS, Yan W, To KKW, Cho WCS. The RNA Binding Protein HuR: A Promising Drug Target for Anticancer Therapy. Curr Cancer Drug Targets 2020; 19:382-399. [PMID: 30381077 DOI: 10.2174/1568009618666181031145953] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/24/2018] [Accepted: 10/18/2018] [Indexed: 02/07/2023]
Abstract
The stability of mRNA is one of the key factors governing the regulation of eukaryotic gene expression and function. Human antigen R (HuR) is an RNA-binding protein that regulates the stability, translation, and nucleus-to-cytoplasm shuttling of its target mRNAs. While HuR is normally localized within the nucleus, it has been shown that HuR binds mRNAs in the nucleus and then escorts the mRNAs to the cytoplasm where HuR protects them from degradation. It contains several RNA recognition motifs, which specifically bind to adenylate and uridylate-rich regions within the 3'-untranslated region of the target mRNA to mediate its effect. Many of the HuR target mRNAs encode proteins important for cell growth, tumorigenesis, angiogenesis, tumor inflammation, invasion and metastasis. HuR overexpression is known to correlate well with high-grade malignancy and poor prognosis in many tumor types. Thus, HuR has emerged as an attractive drug target for cancer therapy. Novel small molecule HuR inhibitors have been identified by high throughput screening and new formulations for targeted delivery of HuR siRNA to tumor cells have been developed with promising anticancer activity. This review summarizes the significant role of HuR in cancer development, progression, and poor treatment response. We will discuss the potential and challenges of targeting HuR therapeutically.
Collapse
Affiliation(s)
- Mingxia Wu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Christy W S Tong
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Wei Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - William C S Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong
| |
Collapse
|
15
|
Li Z, Agrawal V, Ramratnam M, Sharma RK, D'Auria S, Sincoular A, Jakubiak M, Music ML, Kutschke WJ, Huang XN, Gifford L, Ahmad F. Cardiac sodium-dependent glucose cotransporter 1 is a novel mediator of ischaemia/reperfusion injury. Cardiovasc Res 2020; 115:1646-1658. [PMID: 30715251 DOI: 10.1093/cvr/cvz037] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 12/11/2018] [Accepted: 02/01/2019] [Indexed: 01/07/2023] Open
Abstract
AIMS We previously reported that sodium-dependent glucose cotransporter 1 (SGLT1) is highly expressed in cardiomyocytes and is further up-regulated in ischaemia. This study aimed to determine the mechanisms by which SGLT1 contributes to ischaemia/reperfusion (I/R) injury. METHODS AND RESULTS Mice with cardiomyocyte-specific knockdown of SGLT1 (TGSGLT1-DOWN) and wild-type controls were studied. In vivo, the left anterior descending coronary artery was ligated for 30 min and reperfused for 48 h. Ex vivo, isolated perfused hearts were exposed to 20 min no-flow and up to 2 h reperfusion. In vitro, HL-1 cells and isolated adult murine ventricular cardiomyocytes were exposed to 1 h hypoxia and 24 h reoxygenation (H/R). We found that TGSGLT1-DOWN hearts were protected from I/R injury in vivo and ex vivo, with decreased infarct size, necrosis, dysfunction, and oxidative stress. 5'-AMP-activated protein kinase (AMPK) activation increased SGLT1 expression, which was abolished by extracellular signal-related kinase (ERK) inhibition. Co-immunoprecipitation studies showed that ERK, but not AMPK, interacts directly with SGLT1. AMPK activation increased binding of the hepatocyte nuclear factor 1 and specificity protein 1 transcription factors to the SGLT1 gene, and HuR to SGLT1 mRNA. In cells, up-regulation of SGLT1 during H/R was abrogated by AMPK inhibition. Co-immunoprecipitation studies showed that SGLT1 interacts with epidermal growth factor receptor (EGFR), and EGFR interacts with protein kinase C (PKC). SGLT1 overexpression activated PKC and NADPH oxidase 2 (Nox2), which was attenuated by PKC inhibition, EGFR inhibition, and/or disruption of the interaction between EGFR and SGLT1. CONCLUSION During ischaemia, AMPK up-regulates SGLT1 through ERK, and SGLT1 interacts with EGFR, which in turn increases PKC and Nox2 activity and oxidative stress. SGLT1 may represent a novel therapeutic target for mitigating I/R injury.
Collapse
Affiliation(s)
- Zhao Li
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine and Abboud Cardiovascular Research Center, University of Iowa, 100 Newton Road, 1191D ML, Iowa City, IA, USA
| | - Vineet Agrawal
- Division of Cardiology, Department of Medicine, UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mohun Ramratnam
- Division of Cardiology, Department of Medicine, UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA.,Cardiology Section, Medical Service, Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, William. S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ravi K Sharma
- Division of Cardiology, Department of Medicine, UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen D'Auria
- Division of Cardiology, Department of Medicine, UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Abigail Sincoular
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine and Abboud Cardiovascular Research Center, University of Iowa, 100 Newton Road, 1191D ML, Iowa City, IA, USA
| | - Margurite Jakubiak
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine and Abboud Cardiovascular Research Center, University of Iowa, 100 Newton Road, 1191D ML, Iowa City, IA, USA
| | - Meredith L Music
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine and Abboud Cardiovascular Research Center, University of Iowa, 100 Newton Road, 1191D ML, Iowa City, IA, USA
| | - William J Kutschke
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine and Abboud Cardiovascular Research Center, University of Iowa, 100 Newton Road, 1191D ML, Iowa City, IA, USA
| | - Xueyin N Huang
- Division of Cardiology, Department of Medicine, UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lindsey Gifford
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine and Abboud Cardiovascular Research Center, University of Iowa, 100 Newton Road, 1191D ML, Iowa City, IA, USA
| | - Ferhaan Ahmad
- Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine and Abboud Cardiovascular Research Center, University of Iowa, 100 Newton Road, 1191D ML, Iowa City, IA, USA.,Division of Cardiology, Department of Medicine, UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Radiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
16
|
Schultz CW, Preet R, Dhir T, Dixon DA, Brody JR. Understanding and targeting the disease-related RNA binding protein human antigen R (HuR). WILEY INTERDISCIPLINARY REVIEWS-RNA 2020; 11:e1581. [PMID: 31970930 DOI: 10.1002/wrna.1581] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/02/2019] [Accepted: 12/07/2019] [Indexed: 02/06/2023]
Abstract
Altered gene expression is a characteristic feature of many disease states such as tumorigenesis, and in most cancers, it facilitates cancer cell survival and adaptation. Alterations in global gene expression are strongly impacted by post-transcriptional gene regulation. The RNA binding protein (RBP) HuR (ELAVL1) is an established regulator of post-transcriptional gene regulation and is overexpressed in most human cancers. In many cancerous settings, HuR is not only overexpressed, but it is "overactive" as denoted by increased subcellular localization within the cytoplasm. This dysregulation of HuR expression and cytoplasmic localization allows HuR to stabilize and increase the translation of various prosurvival messenger RNA (mRNAs) involved in the pathogenesis of numerous cancers and various diseases. Based on almost 20 years of work, HuR is now recognized as a therapeutic target. Herein, we will review the role HuR plays in the pathophysiology of different diseases and ongoing therapeutic strategies to target HuR. We will focus on three ongoing-targeted strategies: (1) inhibiting HuR's translocation from the nucleus to the cytoplasm; (2) inhibiting the ability of HuR to bind target RNA; and (3) silencing HuR expression levels. In an oncologic setting, HuR has been demonstrated to be critical for a cancer cell's ability to survive a variety of cancer relevant stressors (including drugs and elements of the tumor microenvironment) and targeting this protein has been shown to sensitize cancer cells further to insult. We strongly believe that targeting HuR could be a powerful therapeutic target to treat different diseases, particularly cancer, in the near future. This article is categorized under: RNA in Disease and Development > RNA in Disease NRA Turnover and Surveillance > Regulation of RNA Stability Translation > Translation Regulation.
Collapse
Affiliation(s)
- Christopher W Schultz
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ranjan Preet
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas
| | - Teena Dhir
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Dan A Dixon
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas
| | - Jonathan R Brody
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Mynatt RL, Noland RC, Elks CM, Vandanmagsar B, Bayless DS, Stone AC, Ghosh S, Ravussin E, Warfel JD. The RNA binding protein HuR influences skeletal muscle metabolic flexibility in rodents and humans. Metabolism 2019; 97:40-49. [PMID: 31129047 PMCID: PMC6624076 DOI: 10.1016/j.metabol.2019.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/04/2019] [Accepted: 05/21/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Metabolic flexibility can be assessed by changes in respiratory exchange ratio (RER) following feeding. Though metabolic flexibility (difference in RER between fasted and fed state) is often impaired in individuals with obesity or type 2 diabetes, the cellular processes contributing to this impairment are unclear. MATERIALS AND METHODS From several clinical studies we identified the 16 most and 14 least metabolically flexible male and female subjects out of >100 participants based on differences between 24-hour and sleep RER measured in a whole-room indirect calorimeter. Global skeletal muscle gene expression profiles revealed that, in metabolically flexible subjects, transcripts regulated by the RNA binding protein, HuR, are enriched. We generated and characterized mice with a skeletal muscle-specific knockout of the HuR encoding gene, Elavl1 (HuRm-/-). RESULTS Male, but not female, HuRm-/- mice exhibit metabolic inflexibility, with mild obesity, impaired glucose tolerance, impaired fat oxidation and decreased in vitro palmitate oxidation compared to HuRfl/fl littermates. Expression levels of genes involved in mitochondrial fatty acid oxidation and oxidative phosphorylation are decreased in both mouse and human muscle when HuR is inhibited. CONCLUSIONS HuR inhibition results in impaired metabolic flexibility and decreased lipid oxidation, suggesting a role for HuR as an important regulator of skeletal muscle metabolism.
Collapse
Affiliation(s)
- Randall L Mynatt
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America
| | - Robert C Noland
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America
| | - Carrie M Elks
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America
| | - Bolormaa Vandanmagsar
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America
| | - David S Bayless
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America
| | - Allison C Stone
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America
| | - Sujoy Ghosh
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America; Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Eric Ravussin
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America
| | - Jaycob D Warfel
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States of America.
| |
Collapse
|
18
|
Ravel-Chapuis A, Al-Rewashdy A, Bélanger G, Jasmin BJ. Pharmacological and physiological activation of AMPK improves the spliceopathy in DM1 mouse muscles. Hum Mol Genet 2019; 27:3361-3376. [PMID: 29982462 DOI: 10.1093/hmg/ddy245] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/28/2018] [Indexed: 12/26/2022] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a debilitating multisystemic disorder caused by a triplet repeat expansion in the 3' untranslated region of dystrophia myotonica protein kinase mRNAs. Mutant mRNAs accumulate in the nucleus of affected cells and misregulate RNA-binding proteins, thereby promoting characteristic missplicing events. However, little is known about the signaling pathways that may be affected in DM1. Here, we investigated the status of activated protein kinase (AMPK) signaling in DM1 skeletal muscle and found that the AMPK pathway is markedly repressed in a DM1 mouse model (human skeletal actin-long repeat, HSALR) and patient-derived DM1 myoblasts. Chronic pharmacological activation of AMPK signaling in DM1 mice with 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR) has multiple beneficial effects on the DM1 phenotype. Indeed, a 6-week AICAR treatment of DM1 mice promoted expression of a slower, more oxidative phenotype, improved muscle histology and corrected several events associated with RNA toxicity. Importantly, AICAR also had a dose-dependent positive effect on the spliceopathy in patient-derived DM1 myoblasts. In separate experiments, we also show that chronic treatment of DM1 mice with resveratrol as well as voluntary wheel running also rescued missplicing events in muscle. Collectively, our findings demonstrate the therapeutic potential of chronic AMPK stimulation both physiologically and pharmacologically for DM1 patients.
Collapse
Affiliation(s)
- Aymeric Ravel-Chapuis
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Ali Al-Rewashdy
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Guy Bélanger
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
19
|
BAG3 regulates stability of IL-8 mRNA via interplay between HuR and miR-4312 in PDACs. Cell Death Dis 2018; 9:863. [PMID: 30154469 PMCID: PMC6113235 DOI: 10.1038/s41419-018-0874-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 12/13/2022]
Abstract
Bcl-2 associated athanogene 3 (BAG3) is highly expressed in pancreatic ductal adenocarcinoma (PDAC), and its high expression appears to be a poor prognostic factor for patients with PDAC. In this study, we show that BAG3 knockdown significantly decreases migration and invasion of PDACs via reduction of interleukine-8 (IL-8) production. BAG3 knockdown regulates IL-8 expression at the posttranscriptional levels via interplay between recruitment of RNA-binding protein HuR and miR-4312. HuR binds to the cis-elements located in the 3'-untranslational region (UTR) of the IL-8 transcript to stabilize it, whereas miR-4312-containing miRNA-induced silencing complex (miRISC) is recruited to the adjacent seed element to destabilize it. The binding of HuR prevents the recruitment of Argonaute (Ago2), overriding miR-4312-mediated translation inhibition of IL-8. BAG3 knockdown decreases cytoplasmic distribution of HuR via increasing its phosphorylation at Ser202, therefore compromising its recruitment while promoting recruitment of miR-4312 containing miRISC to IL-8 transcript. Furthermore, our data indicate that only phosphorylated Ago2 at Ser387 interacts with IL-8 transcript. BAG3 knockdown increases phosphorylation of Ago2 at Ser387, thereby further promoting loading of miR-4312 containing miRISC to IL-8 transcript. Taken together, we propose that BAG3 promotes invasion by stabilizing IL-8 transcript via HuR recruitment, and subsequently suppressing the loading of miR-4312 containing miRISC in PDACs. Our results reveal a novel pathway linking BAG3 expression to enhanced PDAC metastasis, thus making BAG3 a potential target for intervention in pancreatic cancer.
Collapse
|
20
|
Basu SK, Gonit M, Salotti J, Chen J, Bhat A, Gorospe M, Viollet B, Claffey KP, Johnson PF. A RAS-CaMKKβ-AMPKα2 pathway promotes senescence by licensing post-translational activation of C/EBPβ through a novel 3'UTR mechanism. Oncogene 2018; 37:3528-3548. [PMID: 29563610 PMCID: PMC6023738 DOI: 10.1038/s41388-018-0190-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/26/2018] [Accepted: 02/09/2018] [Indexed: 12/25/2022]
Abstract
Oncogene-induced senescence (OIS) is an intrinsic tumor suppression mechanism that requires the p53 and RB pathways and post-translational activation of C/EBPβ through the RAS-ERK cascade. We previously reported that in transformed/proliferating cells, C/EBPβ activation is inhibited by G/U-rich elements (GREs) in its 3′UTR. This mechanism, termed “3′UTR regulation of protein activity” (UPA), maintains C/EBPβ in a low-activity state in tumor cells and thus facilitates senescence bypass. Here we show that C/EBPβ UPA is overridden by AMPK signaling. AMPK activators decrease cytoplasmic levels of the GRE binding protein HuR, which is a key UPA component. Reduced cytoplasmic HuR disrupts 3′UTR-mediated trafficking of Cebpb transcripts to the peripheral cytoplasm – a fundamental feature of UPA – thereby stimulating C/EBPβ activation and growth arrest. In primary cells, oncogenic RAS triggers a Ca++-CaMKKβ-AMPKα2-HuR pathway, independent of AMPKα1, that is essential for C/EBPβ activation and OIS. This axis is disrupted in cancer cells through down-regulation of AMPKα2 and CaMKKβ. Thus, CaMKKβ-AMPKα2 signaling constitutes a key tumor suppressor pathway that activates a novel UPA-cancelling mechanism to unmask the cytostatic and pro-senescence functions of C/EBPβ.
Collapse
Affiliation(s)
- Sandip K Basu
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Mesfin Gonit
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA.,Lentigen Tech, Inc., Gaithersburg, MD, USA
| | - Jacqueline Salotti
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jiji Chen
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA.,Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, MD, USA
| | - Atharva Bhat
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Myriam Gorospe
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, 75014, France.,CNRS, UMR8104, Paris, 75014, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, 75014, France
| | - Kevin P Claffey
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT, USA
| | - Peter F Johnson
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
21
|
Dial AG, Rooprai P, Lally JS, Bujak AL, Steinberg GR, Ljubicic V. The role of AMP‐activated protein kinase in the expression of the dystrophin‐associated protein complex in skeletal muscle. FASEB J 2018; 32:2950-2965. [DOI: 10.1096/fj.201700868rrr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Athan G. Dial
- Department of KinesiologyMcMaster University Hamilton Ontario Canada
| | - Paul Rooprai
- Department of KinesiologyMcMaster University Hamilton Ontario Canada
| | - James S. Lally
- Department of MedicineMcMaster University Hamilton Ontario Canada
| | - Adam L. Bujak
- Department of MedicineMcMaster University Hamilton Ontario Canada
| | - Gregory R. Steinberg
- Department of MedicineMcMaster University Hamilton Ontario Canada
- Department of Biochemistry and Biomedical SciencesMcMaster University Hamilton Ontario Canada
| | - Vladimir Ljubicic
- Department of KinesiologyMcMaster University Hamilton Ontario Canada
| |
Collapse
|
22
|
Dhillon RS, Denu JM. Using comparative biology to understand how aging affects mitochondrial metabolism. Mol Cell Endocrinol 2017; 455:54-61. [PMID: 28025033 DOI: 10.1016/j.mce.2016.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/24/2016] [Accepted: 12/16/2016] [Indexed: 02/06/2023]
Abstract
Lifespan varies considerably among even closely related species, as exemplified by rodents and primates. Despite these disparities in lifespan, most studies have focused on intra-specific aging pathologies, primarily within a select few systems. While mice have provided much insight into aging biology, it is unclear if such a short-lived species lack defences against senescence that may have evolved in related longevous species. Many age-related diseases have been linked to mitochondrial dysfunction that are measured by decreased energy generation, structural damage to cellular components, and even cell death. Post translational modifications (PTMs) orchestrate many of the pathways associated with cellular metabolism, and are thought to be a key regulator in biological senescence. We propose hyperacylation as one such modification that may be implicated in numerous mitochondrial impairments affecting energy metabolism.
Collapse
Affiliation(s)
- Rashpal S Dhillon
- Department of Biomolecular Chemistry, University of Wisconsin- Madison, Madison, WI 53715, USA.
| | - John M Denu
- Department of Biomolecular Chemistry, University of Wisconsin- Madison, Madison, WI 53715, USA
| |
Collapse
|
23
|
Shang J, Zhao Z. Emerging role of HuR in inflammatory response in kidney diseases. Acta Biochim Biophys Sin (Shanghai) 2017; 49:753-763. [PMID: 28910975 DOI: 10.1093/abbs/gmx071] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 06/21/2017] [Indexed: 12/14/2022] Open
Abstract
Human antigen R (HuR) is a member of the embryonic lethal abnormal vision (ELAV) family which can bind to the A/U rich elements in 3' un-translated region of mRNA and regulate mRNA splicing, transportation, and stability. Unlike other members of the ELAV family, HuR is ubiquitously expressed. Early studies mainly focused on HuR function in malignant diseases. As researches proceed, more and more proofs demonstrate its relationship with inflammation. Since most kidney diseases involve pathological changes of inflammation, HuR is now suggested to play a pivotal role in glomerular nephropathy, tubular ischemia-reperfusion damage, renal fibrosis and even renal tumors. By regulating the mRNAs of target genes, HuR is causally linked to the onset and progression of kidney diseases. Reports on this topic are steadily increasing, however, the detailed function and mechanism of action of HuR are still not well understood. The aim of this review article is to summarize the present understanding of the role of HuR in inflammation in kidney diseases, and we anticipate that future research will ultimately elucidate the therapeutic value of this novel target.
Collapse
Affiliation(s)
- Jin Shang
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhanzheng Zhao
- Nephrology Hospital, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
24
|
Sato Y, Tsuyama T, Sato C, Karim MF, Yoshizawa T, Inoue M, Yamagata K. Hypoxia reduces HNF4α/MODY1 protein expression in pancreatic β-cells by activating AMP-activated protein kinase. J Biol Chem 2017; 292:8716-8728. [PMID: 28364040 DOI: 10.1074/jbc.m116.767574] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/29/2017] [Indexed: 12/12/2022] Open
Abstract
Hypoxia plays a role in the deterioration of β-cell function. Hepatocyte nuclear factor 4α (HNF4α) has an important role in pancreatic β-cells, and mutations of the human HNF4A gene cause a type of maturity-onset diabetes of the young (MODY1). However, it remains unclear whether hypoxia affects the expression of HNF4α in β-cells. Here, we report that hypoxia reduces HNF4α protein expression in β-cells. Hypoxia-inducible factor was not involved in the down-regulation of HNF4α under hypoxic conditions. The down-regulation of HNF4α was dependent on the activation of AMP-activated protein kinase (AMPK), and the reduction of HNF4α protein expression by metformin, an AMPK activator, and hypoxia was inhibited by the overexpression of a kinase-dead (KD) form of AMPKα2. In addition, hypoxia decreased the stability of the HNF4α protein, and the down-regulation of HNF4α was sensitive to proteasome inhibitors. Adenovirus-mediated overexpression of KD-AMPKα2 improved insulin secretion in metformin-treated islets, hypoxic islets, and ob/ob mouse islets. These results suggest that down-regulation of HNF4α could be of importance in β-cell dysfunction by hypoxia.
Collapse
Affiliation(s)
- Yoshifumi Sato
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Tomonori Tsuyama
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Chinami Sato
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Md Fazlul Karim
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Tatsuya Yoshizawa
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| | - Masahiro Inoue
- the Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka 537-8511, Japan
| | - Kazuya Yamagata
- From the Department of Medical Biochemistry, Faculty of Life Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo-ku, Kumamoto 860-8556 and
| |
Collapse
|
25
|
Brockhoff M, Rion N, Chojnowska K, Wiktorowicz T, Eickhorst C, Erne B, Frank S, Angelini C, Furling D, Rüegg MA, Sinnreich M, Castets P. Targeting deregulated AMPK/mTORC1 pathways improves muscle function in myotonic dystrophy type I. J Clin Invest 2017; 127:549-563. [PMID: 28067669 DOI: 10.1172/jci89616] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/17/2016] [Indexed: 12/13/2022] Open
Abstract
Myotonic dystrophy type I (DM1) is a disabling multisystemic disease that predominantly affects skeletal muscle. It is caused by expanded CTG repeats in the 3'-UTR of the dystrophia myotonica protein kinase (DMPK) gene. RNA hairpins formed by elongated DMPK transcripts sequester RNA-binding proteins, leading to mis-splicing of numerous pre-mRNAs. Here, we have investigated whether DM1-associated muscle pathology is related to deregulation of central metabolic pathways, which may identify potential therapeutic targets for the disease. In a well-characterized mouse model for DM1 (HSALR mice), activation of AMPK signaling in muscle was impaired under starved conditions, while mTORC1 signaling remained active. In parallel, autophagic flux was perturbed in HSALR muscle and in cultured human DM1 myotubes. Pharmacological approaches targeting AMPK/mTORC1 signaling greatly ameliorated muscle function in HSALR mice. AICAR, an AMPK activator, led to a strong reduction of myotonia, which was accompanied by partial correction of misregulated alternative splicing. Rapamycin, an mTORC1 inhibitor, improved muscle relaxation and increased muscle force in HSALR mice without affecting splicing. These findings highlight the involvement of AMPK/mTORC1 deregulation in DM1 muscle pathophysiology and may open potential avenues for the treatment of this disease.
Collapse
|
26
|
Mirisis AA, Alexandrescu A, Carew TJ, Kopec AM. The Contribution of Spatial and Temporal Molecular Networks in the Induction of Long-term Memory and Its Underlying Synaptic Plasticity. AIMS Neurosci 2016; 3:356-384. [PMID: 27819030 PMCID: PMC5096789 DOI: 10.3934/neuroscience.2016.3.356] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The ability to form long-lasting memories is critical to survival and thus is highly conserved across the animal kingdom. By virtue of its complexity, this same ability is vulnerable to disruption by a wide variety of neuronal traumas and pathologies. To identify effective therapies with which to treat memory disorders, it is critical to have a clear understanding of the cellular and molecular mechanisms which subserve normal learning and memory. A significant challenge to achieving this level of understanding is posed by the wide range of distinct temporal and spatial profiles of molecular signaling induced by learning-related stimuli. In this review we propose that a useful framework within which to address this challenge is to view the molecular foundation of long-lasting plasticity as composed of unique spatial and temporal molecular networks that mediate signaling both within neurons (such as via kinase signaling) as well as between neurons (such as via growth factor signaling). We propose that evaluating how cells integrate and interpret these concurrent and interacting molecular networks has the potential to significantly advance our understanding of the mechanisms underlying learning and memory formation.
Collapse
Affiliation(s)
- Anastasios A. Mirisis
- Center for Neural Science, New York University, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
| | - Anamaria Alexandrescu
- Center for Neural Science, New York University, New York, NY, USA
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Thomas J. Carew
- Center for Neural Science, New York University, New York, NY, USA
| | - Ashley M. Kopec
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Ganguly K, Giddaluru J, August A, Khan N. Post-transcriptional Regulation of Immunological Responses through Riboclustering. Front Immunol 2016; 7:161. [PMID: 27199986 PMCID: PMC4850162 DOI: 10.3389/fimmu.2016.00161] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 04/15/2016] [Indexed: 12/22/2022] Open
Abstract
Immunological programing of immune cells varies in response to changing environmental signals. This process is facilitated by modifiers that regulate the translational fate of mRNAs encoding various immune mediators, including cytokines and chemokines, which in turn determine the rapid activation, tolerance, and plasticity of the immune system. RNA-binding proteins (RBPs) recruited by the specific sequence elements in mRNA transcripts are one such modifiers. These RBPs form RBP-RNA complexes known as "riboclusters." These riboclusters serve as RNA sorting machinery, where depending upon the composition of the ribocluster, translation, degradation, or storage of mRNA is controlled. Recent findings suggest that this regulation of mRNA homeostasis is critical for controlling the immune response. Here, we present the current knowledge of the ribocluster-mediated post-transcriptional regulation of immune mediators and highlight recent findings regarding their implications for the pathogenesis of acute or chronic inflammatory diseases.
Collapse
Affiliation(s)
- Koelina Ganguly
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad , Hyderabad , India
| | - Jeevan Giddaluru
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad , Hyderabad , India
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University , New York, NY , USA
| | - Nooruddin Khan
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad , Hyderabad , India
| |
Collapse
|
28
|
Kim J, Abdelmohsen K, Yang X, De S, Grammatikakis I, Noh JH, Gorospe M. LncRNA OIP5-AS1/cyrano sponges RNA-binding protein HuR. Nucleic Acids Res 2016; 44:2378-92. [PMID: 26819413 PMCID: PMC4797289 DOI: 10.1093/nar/gkw017] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 01/07/2016] [Indexed: 12/20/2022] Open
Abstract
The function of the vast majority of mammalian long noncoding (lnc) RNAs remains unknown. Here, analysis of a highly abundant mammalian lncRNA, OIP5-AS1, known as cyrano in zebrafish, revealed that OIP5-AS1 reduces cell proliferation. In human cervical carcinoma HeLa cells, the RNA-binding protein HuR, which enhances cell proliferation, associated with OIP5-AS1 and stabilized it. Tagging OIP5-AS1 with MS2 hairpins to identify associated microRNAs revealed that miR-424 interacted with OIP5-AS1 and competed with HuR for binding to OIP5-AS1. We further identified a ‘sponge’ function for OIP5-AS1, as high levels of OIP5-AS1 increased HuR-OIP5-AS1 complexes and prevented HuR interaction with target mRNAs, including those that encoded proliferative proteins, while conversely, lowering OIP5-AS1 increased the abundance of HuR complexes with target mRNAs. We propose that OIP5-AS1 serves as a sponge or a competing endogenous (ce)RNA for HuR, restricting its availability to HuR target mRNAs and thereby repressing HuR-elicited proliferative phenotypes.
Collapse
Affiliation(s)
- Jiyoung Kim
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, MD 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, MD 21224, USA
| | - Xiaoling Yang
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, MD 21224, USA
| | - Supriyo De
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, MD 21224, USA
| | - Ioannis Grammatikakis
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, MD 21224, USA
| | - Ji Heon Noh
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
29
|
Mitochondria: Are they causal players in cellular senescence? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:1373-9. [DOI: 10.1016/j.bbabio.2015.05.017] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 12/25/2022]
|
30
|
Global translation variations in host cells upon attack of lytic and sublytic Staphylococcus aureus α-haemolysin1. Biochem J 2015; 472:83-95. [DOI: 10.1042/bj20150284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 09/11/2015] [Indexed: 02/07/2023]
Abstract
Staphylococcal alpha-hemolysin (AHL) is a clinically relevant toxin, whose effects on host translation are poorly understood. We characterized genome-wide alterations induced at transcriptional and transational levels by lytic and sublytic AHL, pinpointing the importance of translational control during host-pathogen interaction.
Collapse
|
31
|
Garcin G, Guiraud I, Lacroix M, Genthon C, Rialle S, Joujoux JM, Meunier L, Lavabre-Bertrand T, Stoebner PE, Le Gallic L. AMPK/HuR-Driven IL-20 Post-Transcriptional Regulation in Psoriatic Skin. J Invest Dermatol 2015; 135:2732-2741. [PMID: 26176762 DOI: 10.1038/jid.2015.282] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 05/18/2015] [Accepted: 06/23/2015] [Indexed: 11/09/2022]
Abstract
IL-20 is involved in the development of skin psoriasis. The molecular mechanisms underlying IL-20 overexpression in psoriatic epidermis remain to be elucidated. We showed that IL-20 was primarily upregulated in psoriatic skin at the post-transcriptional level. The RNA-binding protein HuR relocalized to the cytoplasm of keratinocytes (KCs) of psoriatic patients, suggesting that it stabilizes numerous transcripts, as observed in the human KC cell lines used to assess IL-20 mRNA. We characterized epidermal HuR RNA targets in psoriatic skin using ribonucleoprotein immunoprecipitation analyzed via high-throughput sequencing. Numerous transcripts that are upregulated in psoriasis were targeted by HuR, supporting the participation of HuR in pathogenic processes such as morphological changes, innate and adaptive immune responses, and metabolic inflammatory responses. Finally, we identified the metabolic sensor AMP-activated protein kinase (AMPK) as being responsible for HuR cytoplasmic relocalization because its activity was severely impaired in human psoriatic epidermis, and in vivo drug-mediated AMPK inhibition in mouse epidermis promoted HuR cytoplasmic localization, IL-20 overproduction, acanthosis, and hyperkeratosis. These results provide insights into the molecular links between metabolism and post-transcriptional networks during chronic inflammation.
Collapse
Affiliation(s)
- Geneviève Garcin
- Dynamique des Interactions Membranaires Normales et Pathologiques (DIMNP), CNRS UMR 5235, Université de Montpellier, Montpellier, France
| | - Isabelle Guiraud
- Laboratoire d'Histologie-Embryologie-Cytogénétique, Institut des Biomolécules Max Mousseron (IBMM), CNRS UMR 5247, Faculté de Médecine Montpellier-Nîmes, Université de Montpellier, Nîmes, France
| | - Matthieu Lacroix
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Institut régional du Cancer Montpellier (ICM), INSERM U896, Université de Montpellier, Montpellier, France
| | - Clémence Genthon
- Montpellier Genomix (MGX), Institut de Génomique Fonctionnelle (IGF), CNRS INSERM UMS3426, Université de Montpellier, Montpellier, France
| | - Stéphanie Rialle
- Montpellier Genomix (MGX), Institut de Génomique Fonctionnelle (IGF), CNRS INSERM UMS3426, Université de Montpellier, Montpellier, France
| | - Jean-Marie Joujoux
- Service d'Anatomopathologie, Centre Hospitalier Universitaire de Nîmes, Nîmes, France
| | - Laurent Meunier
- Service de Dermatologie, Centre Hospitalier Universitaire de Nîmes, Nîmes, France
| | - Thierry Lavabre-Bertrand
- Laboratoire d'Histologie-Embryologie-Cytogénétique, Institut des Biomolécules Max Mousseron (IBMM), CNRS UMR 5247, Faculté de Médecine Montpellier-Nîmes, Université de Montpellier, Nîmes, France
| | | | - Lionel Le Gallic
- Laboratoire d'Histologie-Embryologie-Cytogénétique, Institut des Biomolécules Max Mousseron (IBMM), CNRS UMR 5247, Faculté de Médecine Montpellier-Nîmes, Université de Montpellier, Nîmes, France.
| |
Collapse
|
32
|
Panganiban RP, Vonakis BM, Ishmael FT, Stellato C. Coordinated post-transcriptional regulation of the chemokine system: messages from CCL2. J Interferon Cytokine Res 2015; 34:255-66. [PMID: 24697203 DOI: 10.1089/jir.2013.0149] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The molecular cross-talk between epithelium and immune cells in the airway mucosa is a key regulator of homeostatic immune surveillance and is crucially involved in the development of chronic lung inflammatory diseases. The patterns of gene expression that follow the sensitization process occurring in allergic asthma and chronic rhinosinusitis and those present in the neutrophilic response of other chronic inflammatory lung diseases such as chronic obstructive pulmonary disease (COPD) are tightly regulated in their specificity. Studies exploring the global transcript profiles associated with determinants of post-transcriptional gene regulation (PTR) such as RNA-binding proteins (RBP) and microRNAs identified several of these factors as being crucially involved in controlling the expression of chemokines upon airway epithelial cell stimulation with cytokines prototypic of Th1- or Th2-driven responses. These studies also uncovered the participation of these pathways to glucocorticoids' inhibitory effect on the epithelial chemokine network. Unmasking the molecular mechanisms of chemokine PTR may likely uncover novel therapeutic strategies for the blockade of proinflammatory pathways that are pathogenetic for asthma, COPD, and other lung inflammatory diseases.
Collapse
Affiliation(s)
- Ronaldo P Panganiban
- 1 Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine , Hershey, Pennsylvania
| | | | | | | |
Collapse
|
33
|
Chuang HC, Chou CC, Kulp SK, Chen CS. AMPK as a potential anticancer target - friend or foe? Curr Pharm Des 2015; 20:2607-18. [PMID: 23859619 DOI: 10.2174/13816128113199990485] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 06/24/2013] [Indexed: 02/08/2023]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a key player in maintaining energy homeostasis in response to metabolic stress. Beyond diabetes and metabolic syndrome, there is a growing interest in the therapeutic exploitation of the AMPK pathway in cancer treatment in light of its unique ability to regulate cancer cell proliferation through the reprogramming of cell metabolism. Although many studies support the tumor-suppressive role of AMPK, emerging evidence suggests that the metabolic checkpoint function of AMPK might be overridden by stress or oncogenic signals so that tumor cells use AMPK activation as a survival strategy to gain growth advantage. These findings underscore the complexity in the cellular function of AMPK in maintaining energy homeostasis under physiological versus pathological conditions. Thus, this review aims to provide an overview of recent findings on the functional interplay of AMPK with different cell metabolic and signaling effectors, particularly histone deacetylases, in mediating downstream tumor suppressive or promoting mechanisms in different cell systems. Although AMPK activation inhibits tumor growth by targeting multiple signaling pathways relevant to tumorigenesis, under certain cellular contexts or certain stages of tumor development, AMPK might act as a protective response to metabolic stresses, such as nutrient deprivation, low oxygen, and low pH, or as downstream effectors of oncogenic proteins, including androgen receptor, hypoxia-inducible factor-1α, c-Src, and MYC. Thus, investigations to define at which stage(s) of tumorigenesis and cancer progression or for which genetic aberrations AMPK inhibition might represent a more relevant strategy than AMPK activation for cancer treatment are clearly warranted.
Collapse
Affiliation(s)
| | | | | | - Ching-Shih Chen
- Rm 336, Parks Hall, College of Pharmacy, The Ohio State University, 500 W. 12th Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
34
|
Ziegler DV, Wiley CD, Velarde MC. Mitochondrial effectors of cellular senescence: beyond the free radical theory of aging. Aging Cell 2015; 14:1-7. [PMID: 25399755 PMCID: PMC4310776 DOI: 10.1111/acel.12287] [Citation(s) in RCA: 260] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2014] [Indexed: 12/22/2022] Open
Abstract
Cellular senescence is a process that results from a variety of stresses, leading to a state of irreversible growth arrest. Senescent cells accumulate during aging and have been implicated in promoting a variety of age-related diseases. Mitochondrial stress is an effective inducer of cellular senescence, but the mechanisms by which mitochondria regulate permanent cell growth arrest are largely unexplored. Here, we review some of the mitochondrial signaling pathways that participate in establishing cellular senescence. We discuss the role of mitochondrial reactive oxygen species (ROS), mitochondrial dynamics (fission and fusion), the electron transport chain (ETC), bioenergetic balance, redox state, metabolic signature, and calcium homeostasis in controlling cellular growth arrest. We emphasize that multiple mitochondrial signaling pathways, besides mitochondrial ROS, can induce cellular senescence. Together, these pathways provide a broader perspective for studying the contribution of mitochondrial stress to aging, linking mitochondrial dysfunction and aging through the process of cellular senescence.
Collapse
Affiliation(s)
- Dorian V. Ziegler
- Département de Biologie Ecole Normale Supérieure de Lyon 46 allée d'Italie Lyon 69007 France
- Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| | | | - Michael C. Velarde
- Buck Institute for Research on Aging 8001 Redwood Blvd. Novato CA 94945 USA
| |
Collapse
|
35
|
Liu YJ, Lee LM, Lai HL, Chern Y. Aberrant activation of AMP-activated protein kinase contributes to the abnormal distribution of HuR in amyotrophic lateral sclerosis. FEBS Lett 2015; 589:432-9. [DOI: 10.1016/j.febslet.2014.12.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/08/2014] [Accepted: 12/26/2014] [Indexed: 10/24/2022]
|
36
|
Ming M, Han W, Zhao B, Sundaresan NR, Deng CX, Gupta MP, He YY. SIRT6 promotes COX-2 expression and acts as an oncogene in skin cancer. Cancer Res 2014; 74:5925-33. [PMID: 25320180 DOI: 10.1158/0008-5472.can-14-1308] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
SIRT6 is a SIR2 family member that regulates multiple molecular pathways involved in metabolism, genomic stability, and aging. It has been proposed previously that SIRT6 is a tumor suppressor in cancer. Here, we challenge this concept by presenting evidence that skin-specific deletion of SIRT6 in the mouse inhibits skin tumorigenesis. SIRT6 promoted expression of COX-2 by repressing AMPK signaling, thereby increasing cell proliferation and survival in the skin epidermis. SIRT6 expression in skin keratinocytes was increased by exposure to UVB light through activation of the AKT pathway. Clinically, we found that SIRT6 was upregulated in human skin squamous cell carcinoma. Taken together, our results provide evidence that SIRT6 functions as an oncogene in the epidermis and suggest greater complexity to its role in epithelial carcinogenesis.
Collapse
Affiliation(s)
- Mei Ming
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois
| | - Weinong Han
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois
| | - Baozhong Zhao
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois
| | - Nagalingam R Sundaresan
- Department of Surgery, Committee on Cellular and Molecular Physiology, University of Chicago, Chicago, Illinois. Division of Biological Sciences, Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, Karnataka, India
| | - Chu-Xia Deng
- National Institute of Diabetes, Digestive and Kidney Diseases, US NIH, Bethesda, Maryland
| | - Mahesh P Gupta
- Department of Surgery, Committee on Cellular and Molecular Physiology, University of Chicago, Chicago, Illinois
| | - Yu-Ying He
- Department of Medicine, Section of Dermatology, University of Chicago, Chicago, Illinois.
| |
Collapse
|
37
|
HuR represses Wnt/β-catenin-mediated transcriptional activity by promoting cytoplasmic localization of β-catenin. Biochem Biophys Res Commun 2014; 457:65-70. [PMID: 25534855 DOI: 10.1016/j.bbrc.2014.12.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 12/10/2014] [Indexed: 11/20/2022]
Abstract
β-Catenin is the key transcriptional activator of canonical Wnt signaling in the nucleus; thus, nuclear accumulation of β-catenin is a critical step for expressing target genes. β-Catenin accumulates in the nucleus of cancer cells where it activates oncogenic target genes. Hu antigen R (HuR) is a RNA binding protein that regulates multiple post-transcriptional processes including RNA stability. Thus, cytoplasmic HuR protein may be involved in tumorigenesis by stabilizing oncogenic transcripts, but the molecular mechanism remains unclear. Here, we observed that Wnt/β-catenin signaling induced export of the HuR protein, whereas HuR overexpression promoted accumulation of the β-catenin protein in the cytoplasm. Thus, Wnt/β-catenin-mediated transcriptional activity in the nucleus was reduced by overexpressing HuR. These results suggest novel and uncharacterized cytoplasmic β-catenin functions related to HuR-mediated RNA metabolism in cancer cells.
Collapse
|
38
|
Moderate hypoxia induces β-cell dysfunction with HIF-1-independent gene expression changes. PLoS One 2014; 9:e114868. [PMID: 25503986 PMCID: PMC4264765 DOI: 10.1371/journal.pone.0114868] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 11/14/2014] [Indexed: 01/09/2023] Open
Abstract
Pancreatic β-cell failure is central to the development and progression of type 2 diabetes. We recently demonstrated that β-cells become hypoxic under high glucose conditions due to increased oxygen consumption and that the pancreatic islets of diabetic mice but not those of control mice are moderately hypoxic. However, the impact of moderate hypoxia on β-cell number and function is unknown. In the present study, moderate hypoxia induced a hypoxic response in MIN6 cells, as evidenced by increased levels of HIF-1α protein and target genes. Under these conditions, a selective downregulation of Mafa, Pdx1, Slc2a2, Ndufa5, Kcnj11, Ins1, Wfs1, Foxa2, and Neurod1, which play important roles in β-cells, was also observed in both MIN6 cells and isolated pancreatic islets. Consistent with the altered expression of these genes, abnormal insulin secretion was detected in hypoxic MIN6 cells. Most of the hypoxia-induced gene downregulation in MIN6 cells was not affected by the suppression of HIF-1α, suggesting a HIF-1–independent mechanism. Moderate hypoxia also induced apoptosis in MIN6 cells. These results suggest that hypoxia is a novel stressor of β-cells and that hypoxic stress may play a role in the deterioration of β-cell function.
Collapse
|
39
|
Gupta R, Liu AY, Glazer PM, Wajapeyee N. LKB1 preserves genome integrity by stimulating BRCA1 expression. Nucleic Acids Res 2014; 43:259-71. [PMID: 25488815 PMCID: PMC4288185 DOI: 10.1093/nar/gku1294] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Serine/threonine kinase 11 (STK11, also known as LKB1) functions as a tumor suppressor in many human cancers. However, paradoxically loss of LKB1 in mouse embryonic fibroblast results in resistance to oncogene-induced transformation. Therefore, it is unclear why loss of LKB1 leads to increased predisposition to develop a wide variety of cancers. Here, we show that LKB1 protects cells from genotoxic stress. Cells lacking LKB1 display increased sensitivity to irradiation, accumulates more DNA double-strand breaks, display defective homology-directed DNA repair (HDR) and exhibit increased mutation rate, compared with that of LKB1-expressing cells. Conversely, the ectopic expression of LKB1 in cells lacking LKB1 protects them against genotoxic stress-induced DNA damage and prevents the accumulation of mutations. We find that LKB1 post-transcriptionally stimulates HDR gene BRCA1 expression by inhibiting the cytoplasmic localization of the RNA-binding protein, HU antigen R, in an AMP kinase-dependent manner and stabilizes BRCA1 mRNA. Cells lacking BRCA1 similar to the cell lacking LKB1 display increased genomic instability and ectopic expression of BRCA1 rescues LKB1 loss-induced sensitivity to genotoxic stress. Collectively, our results demonstrate that LKB1 is a crucial regulator of genome integrity and reveal a novel mechanism for LKB1-mediated tumor suppression with direct therapeutic implications for cancer prevention.
Collapse
Affiliation(s)
- Romi Gupta
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Alex Y Liu
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Peter M Glazer
- Department of Therapeutic Radiology and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Narendra Wajapeyee
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
40
|
Post-transcriptional up-regulation of PDGF-C by HuR in advanced and stressed breast cancer. Int J Mol Sci 2014; 15:20306-20. [PMID: 25383675 PMCID: PMC4264168 DOI: 10.3390/ijms151120306] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/16/2014] [Accepted: 10/28/2014] [Indexed: 01/20/2023] Open
Abstract
Breast cancer is a heterogeneous disease characterized by multiple genetic alterations leading to the activation of growth factor signaling pathways that promote cell proliferation. Platelet-derived growth factor-C (PDGF-C) is overexpressed in various malignancies; however, the involvement of PDGF-C in breast cancers and the mechanisms underlying PDGF-C deregulation remain unclear. Here, we show that PDGF-C is overexpressed in clinical breast cancers and correlates with poor prognosis. PDGF-C up-regulation was mediated by the human embryonic lethal abnormal vision-like protein HuR, which stabilizes the PDGF-C transcript by binding to two predicted AU-rich elements (AREs) in the 3'-untranslated region (3'-UTR). HuR is up-regulated in hydrogen peroxide-treated or ultraviolet-irradiated breast cancer cells. Clinically, HuR levels are correlated with PDGF-C expression and histological grade or pathological tumor-node-metastasis (pTNM) stage. Our findings reveal a novel mechanism underlying HuR-mediated breast cancer progression, and suggest that HuR and PDGF-C are potential molecular candidates for targeted therapy of breast cancers.
Collapse
|
41
|
Liu YJ, Ju TC, Chen HM, Jang YS, Lee LM, Lai HL, Tai HC, Fang JM, Lin YL, Tu PH, Chern Y. Activation of AMP-activated protein kinase α1 mediates mislocalization of TDP-43 in amyotrophic lateral sclerosis. Hum Mol Genet 2014; 24:787-801. [DOI: 10.1093/hmg/ddu497] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
42
|
Kafasla P, Skliris A, Kontoyiannis DL. Post-transcriptional coordination of immunological responses by RNA-binding proteins. Nat Immunol 2014; 15:492-502. [PMID: 24840980 DOI: 10.1038/ni.2884] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Accepted: 04/01/2014] [Indexed: 12/22/2022]
Abstract
Immunological reactions are propelled by ever-changing signals that alter the translational ability of the RNA in the cells involved. Such alterations are considered to be consequential modifications in the transcriptomic decoding of the genetic blueprint. The identification of RNA-binding protein (RBP) assemblies engaged in the coordinative regulation of state-specific RNAs indicates alternative and exclusive means for determining the activation, plasticity and tolerance of cells of the immune system. Here we review current knowledge about RBP-regulated post-transcriptional events involved in the reactivity of cells of the immune system and the importance of their alteration during chronic inflammatory pathology and autoimmunity.
Collapse
Affiliation(s)
- Panagiota Kafasla
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Antonis Skliris
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Dimitris L Kontoyiannis
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| |
Collapse
|
43
|
Xiao L, Wang JY. RNA-binding proteins and microRNAs in gastrointestinal epithelial homeostasis and diseases. Curr Opin Pharmacol 2014; 19:46-53. [PMID: 25063919 DOI: 10.1016/j.coph.2014.07.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 10/25/2022]
Abstract
The epithelium of gastrointestinal (GI) mucosa is a rapidly self-renewing tissue in the body, and its homeostasis is preserved through strict regulation of cell proliferation and apoptosis. Epithelial cells originate from a small number of pluripotent stem cells, which divide to either renew themselves or become committed crypt cells. RNA-binding proteins (RBPs) and microRNAs (miRNAs) regulate gene expression at the posttranscriptional level and are recently shown to modulate GI mucosal growth and repair after injury. Here we highlight the roles of RBPs HuR, CUG-binding protein 1, AU-binding factor 1, and several GI epithelial-specific miRNAs in gut mucosal homeostasis and diseases and also further analyze the mechanisms through which RBPs and miRNAs modulate the stability and translation of target mRNAs.
Collapse
Affiliation(s)
- Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, USA; Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, USA; Department of Pathology, University of Maryland School of Medicine, USA; Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA.
| |
Collapse
|
44
|
McCarty MF. AMPK activation--protean potential for boosting healthspan. AGE (DORDRECHT, NETHERLANDS) 2014; 36:641-663. [PMID: 24248330 PMCID: PMC4039279 DOI: 10.1007/s11357-013-9595-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 10/22/2013] [Indexed: 06/01/2023]
Abstract
AMP-activated kinase (AMPK) is activated when the cellular (AMP+ADP)/ATP ratio rises; it therefore serves as a detector of cellular "fuel deficiency." AMPK activation is suspected to mediate some of the health-protective effects of long-term calorie restriction. Several drugs and nutraceuticals which slightly and safely impede the efficiency of mitochondrial ATP generation-most notably metformin and berberine-can be employed as clinical AMPK activators and, hence, may have potential as calorie restriction mimetics for extending healthspan. Indeed, current evidence indicates that AMPK activators may reduce risk for atherosclerosis, heart attack, and stroke; help to prevent ventricular hypertrophy and manage congestive failure; ameliorate metabolic syndrome, reduce risk for type 2 diabetes, and aid glycemic control in diabetics; reduce risk for weight gain; decrease risk for a number of common cancers while improving prognosis in cancer therapy; decrease risk for dementia and possibly other neurodegenerative disorders; help to preserve the proper structure of bone and cartilage; and possibly aid in the prevention and control of autoimmunity. While metformin and berberine appear to have the greatest utility as clinical AMPK activators-as reflected by their efficacy in diabetes management-regular ingestion of vinegar, as well as moderate alcohol consumption, may also achieve a modest degree of health-protective AMPK activation. The activation of AMPK achievable with any of these measures may be potentiated by clinical doses of the drug salicylate, which can bind to AMPK and activate it allosterically.
Collapse
Affiliation(s)
- Mark F McCarty
- Catalytic Longevity, 7831 Rush Rose Dr., Apt. 316, Carlsbad, CA, 92009, USA,
| |
Collapse
|
45
|
González-Feliciano JA, Hernández-Pérez M, Estrella LA, Colón-López DD, López A, Martínez M, Maurás-Rivera KR, Lasalde C, Martínez D, Araujo-Pérez F, González CI. The role of HuR in the post-transcriptional regulation of interleukin-3 in T cells. PLoS One 2014; 9:e92457. [PMID: 24658545 PMCID: PMC3962401 DOI: 10.1371/journal.pone.0092457] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 02/21/2014] [Indexed: 02/05/2023] Open
Abstract
Human Interleukin-3 (IL-3) is a lymphokine member of a class of transiently expressed mRNAs harboring Adenosine/Uridine-Rich Elements (ARE) in their 3' untranslated regions (3'-UTRs). The regulatory effects of AREs are often mediated by specific ARE-binding proteins (ARE-BPs). In this report, we show that the human IL-3 3'-UTR plays a post-transcriptional regulation role in two human transformed cell lines. More specifically, we demonstrate that the hIL-3 3'-UTR represses the translation of a luciferase reporter both in HeLa and Jurkat T-cells. These results also revealed that the hIL-3 3'-UTR-mediated translational repression is exerted by an 83 nt region comprised mainly by AREs and some non-ARE sequences. Moreover, electrophoretic mobility shift assays (EMSAs) and UV-crosslinking analysis show that this hIL-3 ARE-rich region recruits five specific protein complexes, including the ARE-BPs HuR and TIA-1. HuR binding to this ARE-rich region appears to be spatially modulated during T-cell activation. Together, these results suggest that HuR recognizes the ARE-rich region and plays a role in the IL-3 3'-UTR-mediated post-transcriptional control in T-cells.
Collapse
Affiliation(s)
- José A. González-Feliciano
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
| | - Marimar Hernández-Pérez
- Department of Biochemistry, University of Puerto Rico-Medical Sciences, San Juan, Puerto Rico
| | - Luis A. Estrella
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
| | - Daisy D. Colón-López
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
| | - Armando López
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
| | - Marina Martínez
- Department of Biochemistry, University of Puerto Rico-Medical Sciences, San Juan, Puerto Rico
| | - Kirla R. Maurás-Rivera
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
| | - Clarivel Lasalde
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
| | - Daviana Martínez
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
| | - Félix Araujo-Pérez
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
| | - Carlos I. González
- University of Puerto Rico-Río Piedras, Department of Biology, College of Natural Sciences, San Juan, Puerto Rico
- Department of Biochemistry, University of Puerto Rico-Medical Sciences, San Juan, Puerto Rico
- Molecular Sciences Research Building, San Juan, Puerto Rico
| |
Collapse
|
46
|
Zhang W, Wang Q, Wu Y, Moriasi C, Liu Z, Dai X, Wang Q, Liu W, Yuan ZY, Zou MH. Endothelial cell-specific liver kinase B1 deletion causes endothelial dysfunction and hypertension in mice in vivo. Circulation 2014; 129:1428-39. [PMID: 24637557 DOI: 10.1161/circulationaha.113.004146] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Liver kinase B1 (LKB1), a tumor suppressor, is a central regulator of cell polarity and energy homeostasis. The role of LKB1 in endothelial function in vivo has not been explored. METHODS AND RESULTS Endothelium-specific LKB1 knockout (LKB1(endo-/-)) mice were generated by cross-breeding LKB1(flox/flox) mice with VE-Cadherin-Cre mice. LKB1(endo-/-) mice exhibited hypertension, cardiac hypertrophy, and impaired endothelium-dependent relaxation. LKB1(endo-/-) endothelial cells exhibited reduced endothelial nitric oxide synthase activity and AMP kinase (a downstream enzyme of LKB1) phosphorylation at Thr172 compared with wild-type (WT) cells. In addition, the levels of caveolin-1 were higher in the endothelial cells of LKB1(endo-/-) mice, and knockdown of caveolin-1 by siRNA normalized endothelial nitric oxide synthase activity. Human antigen R bound with the adenylate-uridylate-rich elements of caveolin-1 mRNA 3' untranslated region, resulting in the increased stability of caveolin-1, and genetic knockdown of human antigen R decreased the expression of caveolin-1 in LKB1-deficient endothelial cells. Finally, adenoviral overexpression of constitutively active AMP kinase, but not green fluorescent protein, decreased caveolin-1, lowered blood pressure, and improved endothelial function in LKB1(endo-/-) mice in vivo. CONCLUSIONS Our findings indicate that endothelial LKB1 regulates endothelial nitric oxide synthase activity, endothelial function, and blood pressure by modulating AMP kinase-mediated caveolin-1 expression.
Collapse
Affiliation(s)
- Wencheng Zhang
- Section of Molecular Medicine, Department of Medicine (W.Z., Q.W., Y.W., C.M., Z.L., X.D., Q.W., M.-H.Z.) and Department of Biochemistry and Molecular Biology (Z.L., M.-H.Z.), University of Oklahoma Health Sciences Center, Oklahoma City; Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, China (Y.W., W.L., Z.-Y.Y., M.-H.Z.); and Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Jinan, Shandong, China (M.-H.Z.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Saks V, Schlattner U, Tokarska-Schlattner M, Wallimann T, Bagur R, Zorman S, Pelosse M, Santos PD, Boucher F, Kaambre T, Guzun R. Systems Level Regulation of Cardiac Energy Fluxes Via Metabolic Cycles: Role of Creatine, Phosphotransfer Pathways, and AMPK Signaling. SYSTEMS BIOLOGY OF METABOLIC AND SIGNALING NETWORKS 2014. [DOI: 10.1007/978-3-642-38505-6_11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
48
|
Park SH, Do KH, Choi HJ, Kim J, Kim KH, Park J, Oh CG, Moon Y. Novel regulatory action of ribosomal inactivation on epithelial Nod2-linked proinflammatory signals in two convergent ATF3-associated pathways. THE JOURNAL OF IMMUNOLOGY 2013; 191:5170-81. [PMID: 24098051 DOI: 10.4049/jimmunol.1301145] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In response to excessive nucleotide-binding oligomerization domain-containing protein 2 (Nod2) stimulation caused by mucosal bacterial components, gut epithelia need to activate regulatory machinery to maintain epithelial homeostasis. Activating transcription factor 3 (ATF3) is a representative regulator in the negative feedback loop that modulates TLR-associated inflammatory responses. In the current study, the regulatory effects of ribosomal stress-induced ATF3 on Nod2-stimulated proinflammatory signals were assessed. Ribosomal inactivation caused persistent ATF3 expression that in turn suppressed proinflammatory chemokine production facilitated by Nod2. Decreased chemokine production was due to attenuation of Nod2-activated NF-κB and early growth response protein 1 (EGR-1) signals by ATF3. However, the underlying molecular mechanisms involve two convergent regulatory pathways. Although ATF3 induced by ribosomal inactivation regulated Nod2-induced EGR-1 expression epigenetically through the recruitment of histone deacetylase 1, NF-κB regulation was associated with posttranscriptional regulation by ATF3 rather than epigenetic modification. ATF3 induced by ribosomal inactivation led to the destabilization of p65 mRNA caused by nuclear entrapment of transcript-stabilizing human Ag R protein via direct interaction with ATF3. These findings demonstrate that ribosomal stress-induced ATF3 is a critical regulator in the convergent pathways between EGR-1 and NF-κB, which contributes to the suppression of Nod2-activated proinflammatory gene expression.
Collapse
Affiliation(s)
- Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Microbiology and Immunology, Medical Research Institute, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Kim SJ, Lee JH, Chung HS, Song JH, Ha J, Bae H. Neuroprotective Effects of AMP-Activated Protein Kinase on Scopolamine Induced Memory Impairment. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2013; 17:331-8. [PMID: 23946693 PMCID: PMC3741490 DOI: 10.4196/kjpp.2013.17.4.331] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 06/27/2013] [Accepted: 07/02/2013] [Indexed: 12/18/2022]
Abstract
AMP-activated protein kinase (AMPK), an important regulator of energy metabolism, is activated in response to cellular stress when intracellular levels of AMP increase. We investigated the neuroprotective effects of AMPK against scopolamine-induced memory impairment in vivo and glutamate-induced cytotoxicity in vitro. An adenovirus expressing AMPK wild type alpha subunit (WT) or a dominant negative form (DN) was injected into the hippocampus of rats using a stereotaxic apparatus. The AMPK WT-injected rats showed significant reversal of the scopolamine induced cognitive deficit as evaluated by escape latency in the Morris water maze. In addition, they showed enhanced acetylcholinesterase (AChE)-reactive neurons in the hippocampus, implying increased cholinergic activity in response to AMPK. We also studied the cellular mechanism by which AMPK protects against glutamate-induced cell death in primary cultured rat hippocampal neurons. We further demonstrated that AMPK WT-infected cells increased cell viability and reduced Annexin V positive hippocampal neurons. Western blot analysis indicated that AMPK WT-infected cells reduced the expression of Bax and had no effects on Bcl-2, which resulted in a decreased Bax/Bcl-2 ratio. These data suggest that AMPK is a useful cognitive impairment treatment target, and that its beneficial effects are mediated via the protective capacity of hippocampal neurons.
Collapse
Affiliation(s)
- Soo-Jeong Kim
- College of Korean Medicine, Kyung Hee University, Seoul 130-701, Korea
| | | | | | | | | | | |
Collapse
|
50
|
Yu E, Calvert PA, Mercer JR, Harrison J, Baker L, Figg NL, Kumar S, Wang JC, Hurst LA, Obaid DR, Logan A, West NEJ, Clarke MCH, Vidal-Puig A, Murphy MP, Bennett MR. Mitochondrial DNA damage can promote atherosclerosis independently of reactive oxygen species through effects on smooth muscle cells and monocytes and correlates with higher-risk plaques in humans. Circulation 2013; 128:702-12. [PMID: 23841983 DOI: 10.1161/circulationaha.113.002271] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Mitochondrial DNA (mtDNA) damage occurs in both circulating cells and the vessel wall in human atherosclerosis. However, it is unclear whether mtDNA damage directly promotes atherogenesis or is a consequence of tissue damage, which cell types are involved, and whether its effects are mediated only through reactive oxygen species. METHODS AND RESULTS mtDNA damage occurred early in the vessel wall in apolipoprotein E-null (ApoE(-/-)) mice, before significant atherosclerosis developed. mtDNA defects were also identified in circulating monocytes and liver and were associated with mitochondrial dysfunction. To determine whether mtDNA damage directly promotes atherosclerosis, we studied ApoE(-/-) mice deficient for mitochondrial polymerase-γ proofreading activity (polG(-/-)/ApoE(-/-)). polG(-/-)/ApoE(-/-) mice showed extensive mtDNA damage and defects in oxidative phosphorylation but no increase in reactive oxygen species. polG(-/-)/ApoE(-/-) mice showed increased atherosclerosis, associated with impaired proliferation and apoptosis of vascular smooth muscle cells, and hyperlipidemia. Transplantation with polG(-/-)/ApoE(-/-) bone marrow increased the features of plaque vulnerability, and polG(-/-)/ApoE(-/-) monocytes showed increased apoptosis and inflammatory cytokine release. To examine mtDNA damage in human atherosclerosis, we assessed mtDNA adducts in plaques and in leukocytes from patients who had undergone virtual histology intravascular ultrasound characterization of coronary plaques. Human atherosclerotic plaques showed increased mtDNA damage compared with normal vessels; in contrast, leukocyte mtDNA damage was associated with higher-risk plaques but not plaque burden. CONCLUSIONS We show that mtDNA damage in vessel wall and circulating cells is widespread and causative and indicates higher risk in atherosclerosis. Protection against mtDNA damage and improvement of mitochondrial function are potential areas for new therapeutics.
Collapse
Affiliation(s)
- Emma Yu
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Centre for Clinical Investigation, P.O. Box 110, Addenbrooke's Hospital, Cambridge, CB2 2QQ, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|