1
|
Mahboubi H, Yu H, Malca M, McCusty D, Stochaj U. Pifithrin-µ Induces Stress Granule Formation, Regulates Cell Survival, and Rewires Cellular Signaling. Cells 2024; 13:885. [PMID: 38891018 PMCID: PMC11172192 DOI: 10.3390/cells13110885] [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: 12/20/2023] [Revised: 04/30/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
(1) Background: Stress granules (SGs) are cytoplasmic protein-RNA condensates that assemble in response to various insults. SG production is driven by signaling pathways that are relevant to human disease. Compounds that modulate SG characteristics are therefore of clinical interest. Pifithrin-µ is a candidate anti-tumor agent that inhibits members of the hsp70 chaperone family. While hsp70s are required for granulostasis, the impact of pifithrin-µ on SG formation is unknown. (2) Methods: Using HeLa cells as model system, cell-based assays evaluated the effects of pifithrin-µ on cell viability. Quantitative Western blotting assessed cell signaling events and SG proteins. Confocal microscopy combined with quantitative image analyses examined multiple SG parameters. (3) Results: Pifithrin-µ induced bona fide SGs in the absence of exogenous stress. These SGs were dynamic; their properties were determined by the duration of pifithrin-µ treatment. The phosphorylation of eIF2α was mandatory to generate SGs upon pifithrin-µ exposure. Moreover, the formation of pifithrin-µ SGs was accompanied by profound changes in cell signaling. Pifithrin-µ reduced the activation of 5'-AMP-activated protein kinase, whereas the pro-survival protein kinase Akt was activated. Long-term pifithrin-µ treatment caused a marked loss of cell viability. (4) Conclusions: Our study identified stress-related changes in cellular homeostasis that are elicited by pifithrin-µ. These insights are important knowledge for the appropriate therapeutic use of pifithrin-µ and related compounds.
Collapse
Affiliation(s)
- Hicham Mahboubi
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
| | - Henry Yu
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
| | - Michael Malca
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
| | - David McCusty
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada (H.Y.); (M.M.)
- Quantitative Life Sciences Program, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
2
|
Bietar K, Chu S, Mandl G, Zhang E, Chabaytah N, Sabelli R, Capobianco JA, Stochaj U. Silica-coated LiYF 4:Yb 3+, Tm 3+ upconverting nanoparticles are non-toxic and activate minor stress responses in mammalian cells. RSC Adv 2024; 14:8695-8708. [PMID: 38495986 PMCID: PMC10938293 DOI: 10.1039/d3ra08869c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
Lanthanide-doped upconverting nanoparticles (UCNPs) are ideal candidates for use in biomedicine. The interaction of nanomaterials with biological systems determines whether they are suitable for use in living cells. In-depth knowledge of the nano-bio interactions is therefore a pre-requisite for the development of biomedical applications. The current study evaluates fundamental aspects of the NP-cell interface for square bipyramidal UCNPs containing a LiYF4:Yb3+, Tm3+ core and two different silica surface coatings. Given their importance for mammalian physiology, fibroblast and renal proximal tubule epithelial cells were selected as cellular model systems. We have assessed the toxicity of the UCNPs and measured their impact on the homeostasis of living non-malignant cells. Rigorous analyses were conducted to identify possible toxic and sub-lethal effects of the UCNPs. To this end, we examined biomarkers that reveal if UCNPs induce cell killing or stress. Quantitative measurements demonstrate that short-term exposure to the UCNPs had no profound effects on cell viability, cell size or morphology. Indicators of oxidative, endoplasmic reticulum, or nucleolar stress, and the production of molecular chaperones varied with the surface modification of the UCNPs and the cell type analyzed. These differences emphasize the importance of evaluating cells of diverse origin that are relevant to the intended use of the nanomaterials. Taken together, we established that short-term, our square bipyramidal UCNPs are not toxic to non-malignant fibroblast and proximal renal epithelial cells. Compared with established inducers of cellular stress, these UCNPs have minor effects on cellular homeostasis. Our results build the foundation to explore square bipyramidal UCNPs for future in vivo applications.
Collapse
Affiliation(s)
- Kais Bietar
- Department of Physiology, McGill University Canada
| | - Siwei Chu
- Department of Physiology, McGill University Canada
| | - Gabrielle Mandl
- Department of Chemistry and Biochemistry, Centre for Nanoscience Research, Concordia University Canada
| | - Emma Zhang
- Department of Physiology, McGill University Canada
| | | | | | - John A Capobianco
- Department of Chemistry and Biochemistry, Centre for Nanoscience Research, Concordia University Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University Canada
- Quantitative Life Sciences Program, McGill University Montreal Canada
| |
Collapse
|
3
|
Stanczyk P, Tatekoshi Y, Shapiro JS, Nayudu K, Chen Y, Zilber Z, Schipma M, De Jesus A, Mahmoodzadeh A, Akrami A, Chang HC, Ardehali H. DNA Damage and Nuclear Morphological Changes in Cardiac Hypertrophy Are Mediated by SNRK Through Actin Depolymerization. Circulation 2023; 148:1582-1592. [PMID: 37721051 PMCID: PMC10840668 DOI: 10.1161/circulationaha.123.066002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/23/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND Proper nuclear organization is critical for cardiomyocyte function, because global structural remodeling of nuclear morphology and chromatin structure underpins the development and progression of cardiovascular disease. Previous reports have implicated a role for DNA damage in cardiac hypertrophy; however, the mechanism for this process is not well delineated. AMPK (AMP-activated protein kinase) family of proteins regulates metabolism and DNA damage response (DDR). Here, we examine whether a member of this family, SNRK (SNF1-related kinase), which plays a role in cardiac metabolism, is also involved in hypertrophic remodeling through changes in DDR and structural properties of the nucleus. METHODS We subjected cardiac-specific Snrk-/- mice to transaortic banding to assess the effect on cardiac function and DDR. In parallel, we modulated SNRK in vitro and assessed its effects on DDR and nuclear parameters. We also used phosphoproteomics to identify novel proteins that are phosphorylated by SNRK. Last, coimmunoprecipitation was used to verify Destrin (DSTN) as the binding partner of SNRK that modulates its effects on the nucleus and DDR. RESULTS Cardiac-specific Snrk-/- mice display worse cardiac function and cardiac hypertrophy in response to transaortic banding, and an increase in DDR marker pH2AX (phospho-histone 2AX) in their hearts. In addition, in vitro Snrk knockdown results in increased DNA damage and chromatin compaction, along with alterations in nuclear flatness and 3-dimensional volume. Phosphoproteomic studies identified a novel SNRK target, DSTN, a member of F-actin depolymerizing factor proteins that directly bind to and depolymerize F-actin. SNRK binds to DSTN, and DSTN downregulation reverses excess DNA damage and changes in nuclear parameters, in addition to cellular hypertrophy, with SNRK knockdown. We also demonstrate that SNRK knockdown promotes excessive actin depolymerization, measured by the increased ratio of G-actin to F-actin. Last, jasplakinolide, a pharmacological stabilizer of F-actin, rescues the increased DNA damage and aberrant nuclear morphology in SNRK-downregulated cells. CONCLUSIONS These results indicate that SNRK is a key player in cardiac hypertrophy and DNA damage through its interaction with DSTN. This interaction fine-tunes actin polymerization to reduce DDR and maintain proper cardiomyocyte nuclear shape and morphology.
Collapse
Affiliation(s)
- Paulina Stanczyk
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- These authors contributed equally
| | - Yuki Tatekoshi
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
- These authors contributed equally
| | - Jason S. Shapiro
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
- These authors contributed equally
| | - Krithika Nayudu
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Yihan Chen
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Zachary Zilber
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Matthew Schipma
- Department of Biochemistry and Molecular Genetics, Northwestern University School of Medicine, Chicago, IL, USA
| | - Adam De Jesus
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Amir Mahmoodzadeh
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Ashley Akrami
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Hsiang-Chun Chang
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Hossein Ardehali
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| |
Collapse
|
4
|
Stanczyk P, Tatekoshi Y, Shapiro JS, Nayudu K, Chen Y, Zilber Z, Schipma M, De Jesus A, Mahmoodzadeh A, Akrami A, Chang HC, Ardehali H. DNA damage and nuclear morphological changes in cardiac hypertrophy are mediated by SNRK through actin depolymerization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.14.549060. [PMID: 37503243 PMCID: PMC10370003 DOI: 10.1101/2023.07.14.549060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
BACKGROUND Proper nuclear organization is critical for cardiomyocyte (CM) function, as global structural remodeling of nuclear morphology and chromatin structure underpins the development and progression of cardiovascular disease. Previous reports have implicated a role for DNA damage in cardiac hypertrophy, however, the mechanism for this process is not well delineated. AMPK family of proteins regulate metabolism and DNA damage response (DDR). Here, we examine whether a member of this family, SNF1-related kinase (SNRK), which plays a role in cardiac metabolism, is also involved in hypertrophic remodeling through changes in DDR and structural properties of the nucleus. METHODS We subjected cardiac specific (cs)- Snrk -/- mice to trans-aortic banding (TAC) to assess the effect on cardiac function and DDR. In parallel, we modulated SNRK in vitro and assessed its effects on DDR and nuclear parameters. We also used phospho-proteomics to identify novel proteins that are phosphorylated by SNRK. Finally, co-immunoprecipitation (co-IP) was used to verify Destrin (DSTN) as the binding partner of SNRK that modulates its effects on the nucleus and DDR. RESULTS cs- Snrk -/- mice display worse cardiac function and cardiac hypertrophy in response to TAC, and an increase in DDR marker pH2AX in their hearts. Additionally, in vitro Snrk knockdown results in increased DNA damage and chromatin compaction, along with alterations in nuclear flatness and 3D volume. Phospho-proteomic studies identified a novel SNRK target, DSTN, a member of F-actin depolymerizing factor (ADF) proteins that directly binds to and depolymerize F-actin. SNRK binds to DSTN, and DSTN downregulation reverses excess DNA damage and changes in nuclear parameters, in addition to cellular hypertrophy, with SNRK knockdown. We also demonstrate that SNRK knockdown promotes excessive actin depolymerization, measured by the increased ratio of globular (G-) actin to F-actin. Finally, Jasplakinolide, a pharmacological stabilizer of F-actin, rescues the increased DNA damage and aberrant nuclear morphology in SNRK downregulated cells. CONCLUSIONS These results indicate that SNRK is a key player in cardiac hypertrophy and DNA damage through its interaction with DSTN. This interaction fine-tunes actin polymerization to reduce DDR and maintain proper CM nuclear shape and morphology. Clinical Perspective What is new? Animal hearts subjected to pressure overload display increased SNF1-related kinase (SNRK) protein expression levels and cardiomyocyte specific SNRK deletion leads to aggravated myocardial hypertrophy and heart failure.We have found that downregulation of SNRK impairs DSTN-mediated actin polymerization, leading to maladaptive changes in nuclear morphology, higher DNA damage response (DDR) and increased hypertrophy. What are the clinical implications? Our results suggest that disruption of DDR through genetic loss of SNRK results in an exaggerated pressure overload-induced cardiomyocyte hypertrophy.Targeting DDR, actin polymerization or SNRK/DSTN interaction represent promising therapeutic targets in pressure overload cardiac hypertrophy.
Collapse
Affiliation(s)
- Paulina Stanczyk
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
- These authors contributed equally
| | - Yuki Tatekoshi
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
- These authors contributed equally
| | - Jason S. Shapiro
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
- These authors contributed equally
| | - Krithika Nayudu
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Yihan Chen
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Zachary Zilber
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Matthew Schipma
- Department of Biochemistry and Molecular Genetics, Northwestern University School of Medicine, Chicago, IL, USA
| | - Adam De Jesus
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Amir Mahmoodzadeh
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Ashley Akrami
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Hsiang-Chun Chang
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| | - Hossein Ardehali
- Division of Cardiology, Department of Medicine, and Feinberg Cardiovascular and Renal Research Institute, Northwestern University School of Medicine, Chicago, IL, USA
| |
Collapse
|
5
|
Mesquita PHC, Vann CG, Phillips SM, McKendry J, Young KC, Kavazis AN, Roberts MD. Skeletal Muscle Ribosome and Mitochondrial Biogenesis in Response to Different Exercise Training Modalities. Front Physiol 2021; 12:725866. [PMID: 34646153 PMCID: PMC8504538 DOI: 10.3389/fphys.2021.725866] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/13/2021] [Indexed: 11/20/2022] Open
Abstract
Skeletal muscle adaptations to resistance and endurance training include increased ribosome and mitochondrial biogenesis, respectively. Such adaptations are believed to contribute to the notable increases in hypertrophy and aerobic capacity observed with each exercise mode. Data from multiple studies suggest the existence of a competition between ribosome and mitochondrial biogenesis, in which the first adaptation is prioritized with resistance training while the latter is prioritized with endurance training. In addition, reports have shown an interference effect when both exercise modes are performed concurrently. This prioritization/interference may be due to the interplay between the 5’ AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin complex 1 (mTORC1) signaling cascades and/or the high skeletal muscle energy requirements for the synthesis and maintenance of cellular organelles. Negative associations between ribosomal DNA and mitochondrial DNA copy number in human blood cells also provide evidence of potential competition in skeletal muscle. However, several lines of evidence suggest that ribosome and mitochondrial biogenesis can occur simultaneously in response to different types of exercise and that the AMPK-mTORC1 interaction is more complex than initially thought. The purpose of this review is to provide in-depth discussions of these topics. We discuss whether a curious competition between mitochondrial and ribosome biogenesis exists and show the available evidence both in favor and against it. Finally, we provide future research avenues in this area of exercise physiology.
Collapse
Affiliation(s)
| | | | - Stuart M Phillips
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - James McKendry
- Department of Kinesiology, McMaster University, Hamilton, ON, Canada
| | - Kaelin C Young
- School of Kinesiology, Auburn University, Auburn, AL, United States.,Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Auburn, AL, United States
| | | | - Michael D Roberts
- School of Kinesiology, Auburn University, Auburn, AL, United States.,Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Auburn, AL, United States
| |
Collapse
|
6
|
Saiz ML, DeDiego ML, López-García D, Corte-Iglesias V, Baragaño Raneros A, Astola I, Asensi V, López-Larrea C, Suarez-Alvarez B. Epigenetic targeting of the ACE2 and NRP1 viral receptors limits SARS-CoV-2 infectivity. Clin Epigenetics 2021; 13:187. [PMID: 34635175 PMCID: PMC8504098 DOI: 10.1186/s13148-021-01168-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND SARS-CoV-2 uses the angiotensin-converting enzyme 2 (ACE2) and neuropilin-1 (NRP1) receptors for entry into cells, and the serine protease TMPRSS2 for S protein priming. Inhibition of protease activity or the engagement with ACE2 and NRP1 receptors has been shown to be an effective strategy for blocking infectivity and viral spreading. Valproic acid (VPA; 2-propylpentanoic acid) is an epigenetic drug approved for clinical use. It produces potent antiviral and anti-inflammatory effects through its function as a histone deacetylase (HDAC) inhibitor. Here, we propose VPA as a potential candidate to tackle COVID-19, in which rapid viral spread and replication, and hyperinflammation are crucial elements. RESULTS We used diverse cell lines (HK-2, Huh-7, HUVEC, Caco-2, and BEAS-2B) to analyze the effect of VPA and other HDAC inhibitors on the expression of the ACE-2 and NRP-1 receptors and their ability to inhibit infectivity, viral production, and the inflammatory response. Treatment with VPA significantly reduced expression of the ACE2 and NRP1 host proteins in all cell lines through a mechanism mediated by its HDAC inhibitory activity. The effect is maintained after SARS-CoV-2 infection. Consequently, the treatment of cells with VPA before infection impairs production of SARS-CoV-2 infectious viruses, but not that of other ACE2- and NRP1-independent viruses (VSV and HCoV-229E). Moreover, the addition of VPA 1 h post-infection with SARS-CoV-2 reduces the production of infectious viruses in a dose-dependent manner without significantly modifying the genomic and subgenomic messenger RNAs (gRNA and sg mRNAs) or protein levels of N protein. The production of inflammatory cytokines (TNF-α and IL-6) induced by TNF-α and SARS-CoV-2 infection is diminished in the presence of VPA. CONCLUSIONS Our data showed that VPA blocks three essential processes determining the severity of COVID-19. It downregulates the expression of ACE2 and NRP1, reducing the infectivity of SARS-CoV-2; it decreases viral yields, probably because it affects virus budding or virions stability; and it dampens the triggered inflammatory response. Thus, administering VPA could be considered a safe treatment for COVID-19 patients until vaccines have been rolled out across the world.
Collapse
Affiliation(s)
- Maria Laura Saiz
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Marta L DeDiego
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Darío López-García
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Viviana Corte-Iglesias
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Aroa Baragaño Raneros
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Ivan Astola
- Intensive Care Department, Hospital Universitario Central de Asturias, Oviedo, Spain.,Translational Microbiology Research Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Victor Asensi
- Infectious Diseases Unit, Translational Research in Infectious Diseases Group, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Carlos López-Larrea
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain. .,Department of Immunology, Hospital Universitario Central De Asturias, Oviedo, Spain.
| | - Beatriz Suarez-Alvarez
- Translational Immunology Laboratory, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
7
|
Samhadaneh DM, Mandl GA, Han Z, Mahjoob M, Weber SC, Tuznik M, Rudko DA, Capobianco JA, Stochaj U. Evaluation of Lanthanide-Doped Upconverting Nanoparticles for in Vitro and in Vivo Applications. ACS APPLIED BIO MATERIALS 2020; 3:4358-4369. [DOI: 10.1021/acsabm.0c00381] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Dana M. Samhadaneh
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Gabrielle A. Mandl
- Department of Chemistry & Biochemistry and Centre for NanoScience Research, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Zhao Han
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Maryam Mahjoob
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Stephanie C. Weber
- Department of Biology, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Marius Tuznik
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - David A. Rudko
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 0G4, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 0G4, Canada
| | - John A. Capobianco
- Department of Chemistry & Biochemistry and Centre for NanoScience Research, Concordia University, Montreal, Quebec H4B 1R6, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
8
|
Samhadaneh DM, Alqarni KA, Smart A, Kuang M, Moujaber O, Maysinger D, Stochaj U. Gold nanourchins induce cellular stress, impair proteostasis and damage RNA. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 22:102083. [PMID: 31404650 DOI: 10.1016/j.nano.2019.102083] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/27/2019] [Accepted: 07/31/2019] [Indexed: 12/22/2022]
Abstract
Gold nanoparticles have excellent potential for theranostic applications, but their impact on living cells is only partially understood. Many gold nanoparticles enter cells through endosomes/lysosomes which are linked to different cell organelles and compartments. Our study focuses on the unfolded protein response (UPR) in the endoplasmic reticulum (ER), cytoplasmic RNA-granules and proteostasis, because they are established indicators of cell stress and key regulators of cellular homeostasis. Using HeLa and renal proximal tubule cells as model systems, we show that gold nanourchins reduce cell proliferation, cause ER stress and impair proteostasis. Specifically, gold nanourchins activate the PERK-branch of the UPR, promote RNA oxidation, enhance P-body formation, and accumulate the oxidative stress marker Nrf2 and NFκB in nuclei. Taken together, our study demonstrates that gold nanourchins compromise ER, redox, protein, and RNA homeostasis. These insights provide new information on the cellular responses and molecular changes that gold nanourchins elicit in mammalian cells.
Collapse
Affiliation(s)
| | | | - Adam Smart
- Department of Physiology, McGill University, Montreal, Canada
| | - Mohuizi Kuang
- Department of Physiology, McGill University, Montreal, Canada
| | - Ossama Moujaber
- Department of Physiology, McGill University, Montreal, Canada
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, Canada.
| |
Collapse
|
9
|
Steele HE, Guo Y, Li BY, Na S. Mechanotransduction of mitochondrial AMPK and its distinct role in flow-induced breast cancer cell migration. Biochem Biophys Res Commun 2019; 514:524-529. [PMID: 31060777 DOI: 10.1016/j.bbrc.2019.04.191] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 04/28/2019] [Indexed: 01/05/2023]
Abstract
The biophysical microenvironment of the tumor site has significant impact on breast cancer progression and metastasis. The importance of altered mechanotransduction in cancerous tissue has been documented, yet its role in the regulation of cellular metabolism and the potential link between cellular energy and cell migration remain poorly understood. In this study, we investigated the role of mechanotransduction in AMP-activated protein kinase (AMPK) activation in breast cancer cells in response to interstitial fluid flow (IFF). Additionally, we explored the involvement of AMPK in breast cancer cell migration. IFF was applied to the 3D cell-matrix construct. The subcellular signaling activity of Src, FAK, and AMPK was visualized in real-time using fluorescent resonance energy transfer (FRET). We observed that breast cancer cells (MDA-MB-231) are more sensitive to IFF than normal epithelial cells (MCF-10A). AMPK was activated at the mitochondria of MDA-MB-231 cells by IFF, but not in other subcellular compartments (i.e., cytosol, plasma membrane, and nucleus). The inhibition of FAK or Src abolished flow-induced AMPK activation in the mitochondria of MDA-MB-231 cells. We also observed that global AMPK activation reduced MDA-MB-231 cell migration. Interestingly, specific AMPK inhibition in the mitochondria reduced cell migration and blocked flow-induced cell migration. Our results suggest the linkage of FAK/Src and mitochondria-specific AMPK in mechanotransduction and the differential role of AMPK in breast cancer cell migration depending on its subcellular compartment-specific activation.
Collapse
Affiliation(s)
- Hannah E Steele
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA
| | - Yunxia Guo
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA; Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Bai-Yan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Sungsoo Na
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202, USA.
| |
Collapse
|
10
|
Tsogbadrakh B, Ryu H, Ju KD, Lee J, Yun S, Yu KS, Kim HJ, Ahn C, Oh KH. AICAR, an AMPK activator, protects against cisplatin-induced acute kidney injury through the JAK/STAT/SOCS pathway. Biochem Biophys Res Commun 2019; 509:680-686. [PMID: 30616891 DOI: 10.1016/j.bbrc.2018.12.159] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 12/23/2018] [Indexed: 12/19/2022]
Abstract
Cisplatin causes acute kidney injury (AKI) through proximal tubular injury. We investigated the protective effect of the adenosine monophosphate protein kinase (AMPK) activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) against cisplatin-induced AKI. We investigated whether the AMP-kinase activator AICAR ameliorates cisplatin-induced AKI through the JAK/STAT/SOCS pathway. Male Sprague-Dawley (SD) rats were randomly divided into four groups: control, AICAR, cisplatin, and cisplatin + AICAR. As appropriate to their treatment group, the rats were injected with a single dose of cisplatin (7 mg/kg, i.p.). AICAR was administered to the rats at 100 mg/kg i.p. daily. Blood urea nitrogen (BUN) and serum creatinine were measured. Renal damage was analyzed in sections stained with hematoxylin and eosin (H&E). Renal tissues were also examined by immunohistochemistry and western blot for p-AMPK, Kim-1, cleaved caspase 3, and JAK/STAT/SOCS. For in vitro studies, NRK-52E normal rat kidney cells were treated with cisplatin and/or AICAR. By western blot, we confirmed the expression of p-AMPK and the JAK/STAT/SOCS pathway in NRK-52E cells. AICAR was protective against cisplatin-induced acute tubular injury by up-regulating p-AMPK expression in NRK-52E cells. Protein expression levels of JAK2/STAT1 were markedly ameliorated in NRK-52E cells by AICAR. The protective mechanism of AICAR may be associated with suppression of the JAK2/STAT1 pathway and up-regulation of SOCS1, an inhibitor of the JAK2/STAT1 pathway. The present study demonstrates the protective effects of AICAR against cisplatin-induced AKI and shows a new renoprotective mechanism through the JAK2/STAT1/SOCS1 pathway and apoptosis inhibition. This study suggests that activation of the AMPK activator AICAR might ameliorate cisplatin-induced AKI.
Collapse
Affiliation(s)
- Bodokhsuren Tsogbadrakh
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Hyunjin Ryu
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung Don Ju
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jinho Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sohyun Yun
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Hyo Jin Kim
- Department of Internal Medicine, Dongkuk University, Kyungju, Republic of Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Transplantation Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
11
|
Stochaj U, Rodríguez Burbano DC, Cooper DR, Kodiha M, Capobianco JA. The effects of lanthanide-doped upconverting nanoparticles on cancer cell biomarkers. NANOSCALE 2018; 10:14464-14471. [PMID: 30022175 DOI: 10.1039/c8nr01451e] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Lanthanide-doped upconverting nanoparticles (Ln-UCNPs) possess optical and physicochemical properties that are promising for the design of new theranostic platforms. This applies in particular to the treatment of cancer. Towards this goal, oleate-capped-NaLuF4:Tm3+(0.5%)/Yb3+(20%)/Gd3+(30%) with an average size of 35 nm ± 2 nm were synthesized by co-precipitation. Due to their hydrophobic surface, these Ln-UCNPs produced agglomerates under cell culture conditions. To assess the cellular response to Ln-UCNPs at the molecular level, we evaluated several key aspects of tumor cell physiology. Using cancer lines of different origins, we demonstrated Ln-UCNP dependent changes of cancer cell biomarkers. Multiple cellular components that regulate tumorigenesis and cancer cell homeostasis were affected. In particular, Ln-UCNPs reduced the abundance of hsp70s, elevated DNA damage, and diminished nucleolin and B23/nucleophosmin, proteins required for the assembly of ribosomes. Treatment with Ln-UCNPs also decreased the concentration of paxillin, a focal adhesion protein that is involved in directed cell migration. Furthermore, epidermal growth factor (EGFR) levels were decreased by Ln-UCNPs for most cancer cell lines examined. Taken together, we identified several potential cancer cell targets that were affected by Ln-UCNPs. Our work thereby provides the foundation to optimize Ln-UCNPs for the targeted killing of tumor cells.
Collapse
|
12
|
Moujaber O, Omran N, Kodiha M, Pié B, Cooper E, Presley JF, Stochaj U. Data on the association of the nuclear envelope protein Sun1 with nucleoli. Data Brief 2017; 13:115-123. [PMID: 28580408 PMCID: PMC5447391 DOI: 10.1016/j.dib.2017.05.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 05/10/2017] [Accepted: 05/11/2017] [Indexed: 11/01/2022] Open
Abstract
SUN proteins participate in diverse cellular activities, many of which are connected to the nuclear envelope. Recently, the family member SUN1 has been linked to novel biological activities. These include the regulation of nucleoli, intranuclear compartments that assemble ribosomal subunits. We show that SUN1 associates with nucleoli in several mammalian epithelial cell lines. This nucleolar localization is not shared by all cell types, as SUN1 concentrates at the nuclear envelope in ganglionic neurons and non-neuronal satellite cells. Database analyses and Western blotting emphasize the complexity of SUN1 protein profiles in different mammalian cells. We constructed a STRING network which identifies SUN1-related proteins as part of a larger network that includes several nucleolar proteins. Taken together, the current data highlight the diversity of SUN1 proteins and emphasize the possible links between SUN1 and nucleoli.
Collapse
Affiliation(s)
| | - Nawal Omran
- Physiology, McGill University, Montreal, Canada
| | | | | | | | - John F Presley
- Anatomy & Cell Biology, McGill University, Montreal, Canada
| | | |
Collapse
|
13
|
Dissecting the molecular mechanisms that impair stress granule formation in aging cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:475-486. [DOI: 10.1016/j.bbamcr.2016.12.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 11/21/2016] [Accepted: 12/09/2016] [Indexed: 01/20/2023]
|
14
|
Salminen A, Kauppinen A, Kaarniranta K. AMPK/Snf1 signaling regulates histone acetylation: Impact on gene expression and epigenetic functions. Cell Signal 2016; 28:887-95. [PMID: 27010499 DOI: 10.1016/j.cellsig.2016.03.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 03/18/2016] [Indexed: 01/22/2023]
Abstract
AMP-activated protein kinase (AMPK) and its yeast homolog, Snf1, are critical regulators in the maintenance of energy metabolic balance not only stimulating energy production but also inhibiting energy-consuming processes. The AMPK/Snf1 signaling controls energy metabolism by specific phosphorylation of many metabolic enzymes and transcription factors, enhancing or suppressing their functions. The AMPK/Snf1 complexes can be translocated from cytoplasm into nuclei where they are involved in the regulation of transcription. Recent studies have indicated that AMPK/Snf1 activation can control histone acetylation through different mechanisms affecting not only gene transcription but also many other epigenetic functions. For instance, AMPK/Snf1 enzymes can phosphorylate the histone H3S10 (yeast) and H2BS36 (mammalian) sites which activate specific histone acetyltransferases (HAT), consequently enhancing histone acetylation. Moreover, nuclear AMPK can phosphorylate type 2A histone deacetylases (HDAC), e.g. HDAC4 and HDAC5, triggering their export from nuclei thus promoting histone acetylation reactions. AMPK activation can also increase the level of acetyl CoA, e.g. by inhibiting fatty acid and cholesterol syntheses. Acetyl CoA is a substrate for HATs, thus increasing their capacity for histone acetylation. On the other hand, AMPK can stimulate the activity of nicotinamide phosphoribosyltransferase (NAMPT) which increases the level of NAD(+). NAD(+) is a substrate for nuclear sirtuins, especially for SIRT1 and SIRT6, which deacetylate histones and transcription factors, e.g. those regulating ribosome synthesis and circadian clocks. Histone acetylation is an important epigenetic modification which subsequently can affect chromatin remodeling, e.g. via bromodomain proteins. We will review the signaling mechanisms of AMPK/Snf1 in the control of histone acetylation and subsequently clarify their role in the epigenetic regulation of ribosome synthesis and circadian clocks.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland.
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
15
|
Kodiha M, Mahboubi H, Maysinger D, Stochaj U. Gold Nanoparticles Impinge on Nucleoli and the Stress Response in MCF7 Breast Cancer Cells. Nanobiomedicine (Rij) 2016; 3:3. [PMID: 29942378 PMCID: PMC5998265 DOI: 10.5772/62337] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/01/2016] [Indexed: 01/15/2023] Open
Abstract
Cancer cells can take up gold nanoparticles of different morphologies. These particles interact with the plasma membrane and often travel to intracellular organelles. Among organelles, the nucleus is especially susceptible to the damage that is inflicted by gold nanoparticles. Located inside the nucleus, nucleoli are specialized compartments that transcribe ribosomal RNA genes, produce ribosomes and function as cellular stress sensors. Nucleoli are particularly prone to gold nanoparticle-induced injury. As such, small spherical gold nanoparticles and gold nanoflowers interfere with the transcription of ribosomal DNA. However, the underlying mechanisms are not fully understood. In this study, we examined the effects of gold nanoparticles on nucleolar proteins that are critical to ribosome biogenesis and other cellular functions. We show that B23/nucleophosmin, a nucleolar protein that is tightly linked to cancer, is significantly affected by gold nanoparticles. Furthermore, gold nanoparticles impinge on the cellular stress response, as they reduce the abundance of the molecular chaperone hsp70 and O-GlcNAc modified proteins in the nucleus and nucleoli. Together, our studies set the stage for the development of nanomedicines that target the nucleolus to eradicate proliferating cancer cells.
Collapse
Affiliation(s)
- Mohamed Kodiha
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Hicham Mahboubi
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Dusica Maysinger
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, QC, Canada
| |
Collapse
|
16
|
Mahboubi H, Stochaj U. Quantitative analysis of the interplay between hsc70 and its co-chaperone HspBP1. PeerJ 2015; 3:e1530. [PMID: 26713263 PMCID: PMC4690350 DOI: 10.7717/peerj.1530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/02/2015] [Indexed: 01/28/2023] Open
Abstract
Background. Chaperones and their co-factors are components of a cellular network; they collaborate to maintain proteostasis under normal and harmful conditions. In particular, hsp70 family members and their co-chaperones are essential to repair damaged proteins. Co-chaperones are present in different subcellular compartments, where they modulate chaperone activities. Methods and Results. Our studies assessed the relationship between hsc70 and its co-factor HspBP1 in human cancer cells. HspBP1 promotes nucleotide exchange on hsc70, but has also chaperone-independent functions. We characterized the interplay between hsc70 and HspBP1 by quantitative confocal microscopy combined with automated image analyses and statistical evaluation. Stress and the recovery from insult changed significantly the subcellular distribution of hsc70, but had little effect on HspBP1. Single-cell measurements and regression analysis revealed that the links between the chaperone and its co-factor relied on (i) the physiological state of the cell and (ii) the subcellular compartment. As such, we identified a linear relationship and strong correlation between hsc70 and HspBP1 distribution in control and heat-shocked cells; this correlation changed in a compartment-specific fashion during the recovery from stress. Furthermore, we uncovered significant stress-induced changes in the colocalization between hsc70 and HspBP1 in the nucleus and cytoplasm. Discussion. Our quantitative approach defined novel properties of the co-chaperone HspBP1 as they relate to its interplay with hsc70. We propose that changes in cell physiology promote chaperone redistribution and thereby stimulate chaperone-independent functions of HspBP1.
Collapse
Affiliation(s)
- Hicham Mahboubi
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Ursula Stochaj
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Chen KH, Hsu HH, Lee CC, Yen TH, Ko YC, Yang CW, Hung CC. The AMPK agonist AICAR inhibits TGF-β1 induced activation of kidney myofibroblasts. PLoS One 2014; 9:e106554. [PMID: 25188319 PMCID: PMC4154690 DOI: 10.1371/journal.pone.0106554] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 08/08/2014] [Indexed: 01/07/2023] Open
Abstract
Activation of interstitial myofibroblasts and excessive production of extracellular matrix proteins are common pathways that contribute to chronic kidney disease. In a number of tissues, AMP-activated kinase (AMPK) activation has been shown to inhibit fibrosis. Here, we examined the inhibitory effect of the AMPK activator, 5-aminoimidazole-4-carboxyamide ribonucleoside (AICAR), on renal fibrosis invivo and TGF-β1-induced renal fibroblasts activation invitro. A unilateral ureteral obstruction (UUO) model was induced in male BALB/c mice. Mice with UUO were administered AICAR (500 mg/Kg/day) or saline intraperitoneally 1 day before UUO surgery and daily thereafter. Both kidneys were harvested 7 days after surgery for further analysis. For the in vitro studies, NRK-49F rat fibroblasts were pre-incubated with AICAR before TGF-β1 stimulation. The inhibitory effects of AICAR on signaling pathways down-stream of TGF-β1 were analyzed. In UUO model mice, administration of AICAR attenuated extracellular matrix protein deposition and the expression of α-smooth muscle actin (α-SMA), type I collagen and fibronectin. Pre-incubation of NRK-49F cells with AICAR inhibited TGF-β1-induced myofibroblast activation. Silencing of AMPKα1 by siRNA or by blocking AMPK activation with Compound C diminished the inhibitory effect of AICAR. Moreover, the inhibitory effects of AICAR on TGF-β1-mediated myofibroblast activation were associated with down-regulation of ERK 1/2 and STAT3. Our results suggest that AICAR reduces tubulointerstitial fibrosis in UUO mice and inhibits TGF-β1-induced kidney myofibroblast activation. AMPK activation by AICAR may have therapeutic potential for the treatment of renal tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Kuan-Hsing Chen
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Hsiang-Hao Hsu
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Cheng-Chia Lee
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Tzu-Hai Yen
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Yi-Ching Ko
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Chih-Wei Yang
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
| | - Cheng-Chieh Hung
- Kidney Research Center, Chang Gung Memorial Hospital, Chang Gung University, School of Medicine, Taoyuan, Taiwan
- * E-mail:
| |
Collapse
|
18
|
Mahboubi H, Stochaj U. Nucleoli and Stress Granules: Connecting Distant Relatives. Traffic 2014; 15:1179-93. [DOI: 10.1111/tra.12191] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 06/30/2014] [Accepted: 06/30/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Hicham Mahboubi
- Department of Physiology; McGill University; 3655 Promenade Sir William Osler Montreal Quebec H3G 1Y6 Canada
| | - Ursula Stochaj
- Department of Physiology; McGill University; 3655 Promenade Sir William Osler Montreal Quebec H3G 1Y6 Canada
| |
Collapse
|