1
|
Huynh TN, Toperzer J, Scherer A, Gumina A, Brunetti T, Mansour MK, Markovitz DM, Russo BC. Vimentin regulates mitochondrial ROS production and inflammatory responses of neutrophils. Front Immunol 2024; 15:1416275. [PMID: 39139560 PMCID: PMC11319119 DOI: 10.3389/fimmu.2024.1416275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
The intermediate filament vimentin is present in immune cells and is implicated in proinflammatory immune responses. Whether and how it supports antimicrobial activities of neutrophils are not well established. Here, we developed an immortalized neutrophil model to examine the requirement of vimentin. We demonstrate that vimentin restricts the production of proinflammatory cytokines and reactive oxygen species (ROS), but enhances phagocytosis and swarming. We observe that vimentin is dispensable for neutrophil extracellular trap (NET) formation, degranulation, and inflammasome activation. Moreover, gene expression analysis demonstrated that the presence of vimentin was associated with changes in expression of multiple genes required for mitochondrial function and ROS overproduction. Treatment of wild-type cells with rotenone, an inhibitor for complex I of the electron transport chain, increases the ROS levels. Likewise, treatment with mitoTEMPO, a SOD mimetic, rescues the ROS production in cells lacking vimentin. Together, these data show vimentin regulates neutrophil antimicrobial functions and alters ROS levels through regulation of mitochondrial activity.
Collapse
Affiliation(s)
- Thao Ngoc Huynh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Jody Toperzer
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Allison Scherer
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Anne Gumina
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Tonya Brunetti
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Michael K. Mansour
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - David M. Markovitz
- Department of Internal Medicine, Division of Infectious Diseases, University of Michigan, Ann Arbor, MI, United States
| | - Brian C. Russo
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
2
|
Dong Z, Han W, Jiang P, Hao L, Fu X. Regulation of mitochondrial network architecture and function in mesenchymal stem cells by micropatterned surfaces. Regen Biomater 2024; 11:rbae052. [PMID: 38854681 PMCID: PMC11162196 DOI: 10.1093/rb/rbae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/04/2024] [Accepted: 04/25/2024] [Indexed: 06/11/2024] Open
Abstract
Mitochondrial network architecture, which is closely related to mitochondrial function, is mechanically sensitive and regulated by multiple stimuli. However, the effects of microtopographic cues on mitochondria remain poorly defined. Herein, polycaprolactone (PCL) surfaces were used as models to investigate how micropatterns regulate mitochondrial network architecture and function in rat adipose-derived stem cells (rASCs). It was found that large pit (LP)-induced rASCs to form larger and more complex mitochondrial networks. Consistently, the expression of key genes related to mitochondrial dynamics revealed that mitochondrial fusion (MFN1 and MFN2) and midzone fission (DRP1 and MFF) were increased in rASCs on LP. In contrast, the middle pit (MP)-enhanced mitochondrial biogenesis, as evidenced by the larger mitochondrial area and higher expression of PGC-1. Both LP and MP promoted ATP production in rASCs. It is likely that LP increased ATP levels through modulating mitochondrial network architecture while MP stimulated mitochondria biogenesis to do so. Our study clarified the regulation of micropatterned surfaces on mitochondria, highlighting the potential of LP and MP as a simple platform to stimulate mitochondria and the subsequent cellular function of MSCs.
Collapse
Affiliation(s)
- Zixuan Dong
- The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Weiju Han
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Panyu Jiang
- The Second Affiliated Hospital, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| | - Lijing Hao
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
- School of Materials Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Xiaoling Fu
- National Engineering Research Center for Tissue Restoration and Reconstruction and Innovation Center for Tissue Restoration and Reconstruction, Guangzhou 510006, China
- Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
3
|
Huynh TN, Toperzer J, Scherer A, Gumina A, Brunetti T, Mansour MK, Markovitz DM, Russo BC. Vimentin regulates mitochondrial ROS production and inflammatory responses of neutrophils. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.589146. [PMID: 38659904 PMCID: PMC11042233 DOI: 10.1101/2024.04.11.589146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The intermediate filament vimentin is present in immune cells and is implicated in proinflammatory immune responses. Whether and how it supports antimicrobial activities of neutrophils is not well established. Here, we developed an immortalized neutrophil model to examine the requirement of vimentin. We demonstrate that vimentin restricts the production of proinflammatory cytokines and reactive oxygen species (ROS), but enhances phagocytosis and swarming. We observe that vimentin is dispensable for neutrophil extracellular trap (NET) formation, degranulation, and inflammasome activation. Moreover, gene expression analysis demonstrated that the presence of vimentin was associated with changes in expression of multiple genes required for mitochondrial function and ROS overproduction. Treatment of wild-type cells with rotenone, an inhibitor for complex I of the electron transport chain, increases the ROS levels. Likewise, treatment with mitoTEMPO, a SOD mimetic, rescues the ROS production in cells lacking vimentin. Together, these data show vimentin regulates neutrophil antimicrobial functions and alters ROS levels through regulation of mitochondrial activity.
Collapse
|
4
|
Jiang H, Sun Y, Li F, Yu X, Lei S, Du S, Wu T, Jiang X, Zhu J, Wang J, Ji Y, Li N, Feng X, Gu J, Han W, Zeng L, Lei L. Enolase of Streptococcus suis serotype 2 promotes biomolecular condensation of ribosomal protein SA for HBMECs apoptosis. BMC Biol 2024; 22:33. [PMID: 38331785 PMCID: PMC10854124 DOI: 10.1186/s12915-024-01835-y] [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: 06/27/2023] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Ribosomal protein SA (RPSA) of human brain microvascular endothelial cells (HBMECs) can transfer from the cytosol to the cell surface and act as a receptor for some pathogens, including Streptococcus suis serotype 2 (SS2), a zoonotic pathogen causing meningitis in pigs and humans. We previously reported that SS2 virulence factor enolase (ENO) binds to RPSA on the cell surface of HBMECs and induces apoptosis. However, the mechanism that activates RPSA translocation to the cell surface and induces ENO-mediated HBMEC apoptosis is unclear. RESULTS Here, we show that RPSA localization and condensation on the host cell surface depend on its internally disordered region (IDR). ENO binds to the IDR of RPSA and promotes its interaction with RPSA and vimentin (VIM), which is significantly suppressed after 1,6-Hexanediol (1,6-Hex, a widely used tool to disrupt phase separation) treatment, indicating that ENO incorporation and thus the concentration of RPSA/VIM complexes via co-condensation. Furthermore, increasing intracellular calcium ions (Ca2+) in response to SS2 infection further facilitates the liquid-like condensation of RPSA and aggravates ENO-induced HBMEC cell apoptosis. CONCLUSIONS Together, our study provides a previously underappreciated molecular mechanism illuminating that ENO-induced RPSA condensation activates the migration of RPSA to the bacterial cell surface and stimulates SS2-infected HBMEC death and, potentially, disease progression. This study offers a fresh avenue for investigation into the mechanism by which other harmful bacteria infect hosts via cell surfaces' RPSA.
Collapse
Affiliation(s)
- Hexiang Jiang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Yi Sun
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Fengyang Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xibing Yu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Siyu Lei
- Department of Respiratory Medicine, Center for Infectious Diseases and Pathogen Biology, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, The First Hospital of Jilin University, Changchun, 130021, China
| | - Sulan Du
- Department of Veterinary Medicine, College of Animal Science, Yangtze University, Jingzhou, 434023, China
| | - Tong Wu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xuan Jiang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Junhui Zhu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jun Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Yalu Ji
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Na Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xin Feng
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jingmin Gu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Wenyu Han
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Lei Zeng
- Bethune Institute of Epigenetic Medicine, The First Hospital of Jilin University, Changchun, Jilin, China.
- International Center of Future Science, Jillin University, Changchun, Jilin, China.
| | - Liancheng Lei
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, 130062, China
- Department of Veterinary Medicine, College of Animal Science, Yangtze University, Jingzhou, 434023, China
| |
Collapse
|
5
|
Ekenstedt KJ, Minor KM, Shelton GD, Hammond JJ, Miller AD, Taylor SM, Huang Y, Mickelson JR. A SACS deletion variant in Great Pyrenees dogs causes autosomal recessive neuronal degeneration. Hum Genet 2023; 142:1587-1601. [PMID: 37758910 PMCID: PMC10602964 DOI: 10.1007/s00439-023-02599-1] [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: 08/20/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023]
Abstract
ARSACS (autosomal recessive spastic ataxia of Charlevoix-Saguenay) is a human neurological disorder characterized by progressive cerebellar ataxia and peripheral neuropathy. A recently recognized disorder in Great Pyrenees dogs is similarly characterized by widespread central nervous system degeneration leading to progressive cerebellar ataxia and spasticity, combined with peripheral neuropathy. Onset of clinical signs occurred in puppies as young as 4 months of age, with slow progression over several years. A multi-generation pedigree suggested an autosomal recessive mode of inheritance. Histopathology revealed consistent cerebellar Purkinje cell degeneration, neuronal degeneration in brainstem nuclei, widespread spinal cord white matter degeneration, ganglion cell degeneration, inappropriately thin myelin sheaths or fully demyelinated peripheral nerve fibers, and normal or only mild patterns of denervation atrophy in skeletal muscles. Genome-wide single nucleotide polymorphism (SNP) genotype data was collected from 6 cases and 26 controls, where homozygosity mapping identified a 3.3 Mb region on CFA25 in which all cases were homozygous and all controls were either heterozygous or homozygous for alternate haplotypes. This region tagged the SACS gene where variants are known to cause ARSACS. Sanger sequencing of SACS in affected dogs identified a 4 bp deletion that causes a frame shift and truncates 343 amino acids from the C terminus of the encoded sacsin protein (p.Val4244AlafsTer32). Our clinical and histopathological descriptions of this canine disorder contribute to the description of human ARSACS and represents the first naturally occurring large animal model of this disorder.
Collapse
Affiliation(s)
- Kari J Ekenstedt
- Department of Basic Medical Sciences, College of Veterinary Medicine, Purdue University, Lynn Hall, 625 Harrison Street, West Lafayette, IN, 47907, USA.
| | - Katie M Minor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, 55108, USA
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - James J Hammond
- Department of Neurology, Pieper Memorial Veterinary Center, Middletown, CT, 06457, USA
| | - Andrew D Miller
- Section of Anatomic Pathology, Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Susan M Taylor
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5B4, Canada
| | - Yanyun Huang
- Prairie Diagnostic Services, Inc., Saskatoon, SK, S7N 5B4, Canada
| | - James R Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, 55108, USA
| |
Collapse
|
6
|
Cremer T, Voortman LM, Bos E, Jongsma MLM, ter Haar LR, Akkermans JJLL, Talavera Ormeño CMP, Wijdeven RHM, de Vries J, Kim RQ, Janssen GMC, van Veelen PA, Koning RI, Neefjes J, Berlin I. RNF26 binds perinuclear vimentin filaments to integrate ER and endolysosomal responses to proteotoxic stress. EMBO J 2023; 42:e111252. [PMID: 37519262 PMCID: PMC10505911 DOI: 10.15252/embj.2022111252] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/28/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Proteotoxic stress causes profound endoplasmic reticulum (ER) membrane remodeling into a perinuclear quality control compartment (ERQC) for the degradation of misfolded proteins. Subsequent return to homeostasis involves clearance of the ERQC by endolysosomes. However, the factors that control perinuclear ER integrity and dynamics remain unclear. Here, we identify vimentin intermediate filaments as perinuclear anchors for the ER and endolysosomes. We show that perinuclear vimentin filaments engage the ER-embedded RING finger protein 26 (RNF26) at the C-terminus of its RING domain. This restricts RNF26 to perinuclear ER subdomains and enables the corresponding spatial retention of endolysosomes through RNF26-mediated membrane contact sites (MCS). We find that both RNF26 and vimentin are required for the perinuclear coalescence of the ERQC and its juxtaposition with proteolytic compartments, which facilitates efficient recovery from ER stress via the Sec62-mediated ER-phagy pathway. Collectively, our findings reveal a scaffolding mechanism that underpins the spatiotemporal integration of organelles during cellular proteostasis.
Collapse
Affiliation(s)
- Tom Cremer
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| | - Lenard M Voortman
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Erik Bos
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Marlieke LM Jongsma
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| | - Laurens R ter Haar
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jimmy JLL Akkermans
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| | - Cami MP Talavera Ormeño
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Ruud HM Wijdeven
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam NeuroscienceAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Jelle de Vries
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Robbert Q Kim
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - George MC Janssen
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Peter A van Veelen
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Roman I Koning
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| | - Ilana Berlin
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
- Oncode Institute, Leiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
7
|
Udi Y, Zhang W, Stein ME, Ricardo-Lax I, Pasolli HA, Chait BT, Rout MP. A general method for quantitative fractionation of mammalian cells. J Cell Biol 2023; 222:213941. [PMID: 36920247 PMCID: PMC10040634 DOI: 10.1083/jcb.202209062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 01/11/2023] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
Subcellular fractionation in combination with mass spectrometry-based proteomics is a powerful tool to study localization of key proteins in health and disease. Here we offered a reliable and rapid method for mammalian cell fractionation, tuned for such proteomic analyses. This method proves readily applicable to different cell lines in which all the cellular contents are accounted for, while maintaining nuclear and nuclear envelope integrity. We demonstrated the method's utility by quantifying the effects of a nuclear export inhibitor on nucleoplasmic and cytoplasmic proteomes.
Collapse
Affiliation(s)
- Yael Udi
- Laboratory of Cellular and Structural Biology, The Rockefeller University , New York, NY, USA
| | - Wenzhu Zhang
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University , New York, NY, USA
| | - Milana E Stein
- Laboratory of Cellular and Structural Biology, The Rockefeller University , New York, NY, USA
| | - Inna Ricardo-Lax
- Laboratory of Virology and Infectious Disease, The Rockefeller University , New York, NY, USA
| | - Hilda A Pasolli
- Electron Microscopy Resource Center, The Rockefeller University , New York, NY, USA
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller University , New York, NY, USA
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University , New York, NY, USA
| |
Collapse
|
8
|
Cheng Y, Lou JX, Liu YY, Liu CC, Chen J, Yang MC, Ye YB, Go YY, Zhou B. Intracellular Vimentin Regulates the Formation of Classical Swine Fever Virus Replication Complex through Interaction with NS5A Protein. J Virol 2023; 97:e0177022. [PMID: 37129496 PMCID: PMC10231149 DOI: 10.1128/jvi.01770-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/08/2023] [Indexed: 05/03/2023] Open
Abstract
Vimentin (VIM), an indispensable protein, is responsible for the formation of intermediate filament structures within cells and plays a crucial role in viral infections. However, the precise role of VIM in classical swine fever virus (CSFV) infection remains unclear. Herein, we systematically investigated the function of VIM in CSFV replication. We demonstrated that both knockdown and overexpression of VIM affected CSFV replication. Furthermore, we observed by confocal microscopy the rearrangement of cellular VIM into a cage-like structure during CSFV infection. Three-dimensional (3D) imaging indicated that the cage-like structures were localized in the endoplasmic reticulum (ER) and ringed around the double-stranded RNA (dsRNA), thereby suggesting that VIM was associated with the formation of the viral replication complex (VRC). Mechanistically, phosphorylation of VIM at serine 72 (Ser72), regulated by the RhoA/ROCK signaling pathway, induced VIM rearrangement upon CSFV infection. Confocal microscopy and coimmunoprecipitation assays revealed that VIM colocalized and interacted with CSFV NS5A. Structurally, it was determined that amino acids 96 to 407 of VIM and amino acids 251 to 416 of NS5A were the respective important domains for this interaction. Importantly, both VIM knockdown and disruption of VIM rearrangement inhibited the localization of NS5A in the ER, implying that VIM rearrangement recruited NS5A to the ER for VRC formation. Collectively, our results suggest that VIM recruits NS5A to form a stable VRC that is protected by the cage-like structure formed by VIM rearrangement, ultimately leading to enhanced virus replication. These findings highlight the critical role of VIM in the formation and stabilization of VRC, which provides alternative strategies for the development of antiviral drugs. IMPORTANCE Classical swine fever (CSF), caused by classical swine fever virus (CSFV), is a highly infectious disease that poses a significant threat to the global pig industry. Therefore, gaining insights into the virus and its interaction with host cells is crucial for developing effective antiviral measures and controlling the spread of CSF. Previous studies have shown that CSFV infection induces rearrangement of the endoplasmic reticulum, leading to the formation of small vesicular organelles containing nonstructural protein and double-stranded RNA of CSFV, as well as some host factors. These organelles then assemble into viral replication complexes (VRCs). In this study, we have discovered that VIM recruited CSFV NS5A to form a stable VRC that was protected by a cage-like structure formed by rearranged VIM. This enhanced viral replication. Our findings not only shed light on the molecular mechanism of CSFV replication but also offer new insights into the development of antiviral strategies for controlling CSFV.
Collapse
Affiliation(s)
- Yan Cheng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jin-xiu Lou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ya-yun Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Chun-chun Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jing Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ming-chuan Yang
- The State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science, Anhui Agricultural University, Hefei, China
| | - Yin-bo Ye
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yun Young Go
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong SAR, China
| | - Bin Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
9
|
Fernández Casafuz AB, De Rossi MC, Bruno L. Mitochondrial cellular organization and shape fluctuations are differentially modulated by cytoskeletal networks. Sci Rep 2023; 13:4065. [PMID: 36906690 PMCID: PMC10008531 DOI: 10.1038/s41598-023-31121-w] [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: 12/20/2022] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
The interactions between mitochondria and the cytoskeleton have been found to alter mitochondrial function; however, the mechanisms underlying this phenomenon are largely unknown. Here, we explored how the integrity of the cytoskeleton affects the cellular organization, morphology and mobility of mitochondria in Xenopus laevis melanocytes. Cells were imaged in control condition and after different treatments that selectively affect specific cytoskeletal networks (microtubules, F-actin and vimentin filaments). We observed that mitochondria cellular distribution and local orientation rely mostly on microtubules, positioning these filaments as the main scaffolding of mitochondrial organization. We also found that cytoskeletal networks mold mitochondria shapes in distinct ways: while microtubules favor more elongated organelles, vimentin and actin filaments increase mitochondrial bending, suggesting the presence of mechanical interactions between these filaments and mitochondria. Finally, we identified that microtubule and F-actin networks play opposite roles in mitochondria shape fluctuations and mobility, with microtubules transmitting their jittering to the organelles and F-actin restricting the organelles motion. All our results support that cytoskeleton filaments interact mechanically with mitochondria and transmit forces to these organelles molding their movements and shapes.
Collapse
Affiliation(s)
- Agustina Belén Fernández Casafuz
- CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Cálculo (IC), Buenos Aires, 1428, Argentina
| | - María Cecilia De Rossi
- CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, 1428, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| | - Luciana Bruno
- CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Cálculo (IC), Buenos Aires, 1428, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| |
Collapse
|
10
|
Kuburich NA, den Hollander P, Pietz JT, Mani SA. Vimentin and cytokeratin: Good alone, bad together. Semin Cancer Biol 2022; 86:816-826. [PMID: 34953942 PMCID: PMC9213573 DOI: 10.1016/j.semcancer.2021.12.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/26/2021] [Accepted: 12/14/2021] [Indexed: 01/27/2023]
Abstract
The cytoskeleton plays an integral role in maintaining the integrity of epithelial cells. Epithelial cells primarily employ cytokeratin in their cytoskeleton, whereas mesenchymal cells use vimentin. During the epithelial-mesenchymal transition (EMT), cytokeratin-positive epithelial cells begin to express vimentin. EMT induces stem cell properties and drives metastasis, chemoresistance, and tumor relapse. Most studies of the functions of cytokeratin and vimentin have relied on the use of either epithelial or mesenchymal cell types. However, it is important to understand how these two cytoskeleton intermediate filaments function when co-expressed in cells undergoing EMT. Here, we discuss the individual and shared functions of cytokeratin and vimentin that coalesce during EMT and how alterations in intermediate filament expression influence carcinoma progression.
Collapse
Affiliation(s)
- Nick A Kuburich
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Jordan T Pietz
- Department of Creative Services, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States.
| |
Collapse
|
11
|
Viedma-Poyatos Á, González-Jiménez P, Pajares MA, Pérez-Sala D. Alexander disease GFAP R239C mutant shows increased susceptibility to lipoxidation and elicits mitochondrial dysfunction and oxidative stress. Redox Biol 2022; 55:102415. [PMID: 35933901 PMCID: PMC9364016 DOI: 10.1016/j.redox.2022.102415] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 01/04/2023] Open
Abstract
Alexander disease is a fatal neurological disorder caused by mutations in the intermediate filament protein Glial Fibrillary Acidic Protein (GFAP), which is key for astrocyte homeostasis. These mutations cause GFAP aggregation, astrocyte dysfunction and neurodegeneration. Remarkably, most of the known GFAP mutations imply a change by more nucleophilic amino acids, mainly cysteine or histidine, which are more susceptible to oxidation and lipoxidation. Therefore, we hypothesized that a higher susceptibility of Alexander disease GFAP mutants to oxidative or electrophilic damage, which frequently occurs during neurodegeneration, could contribute to disease pathogenesis. To address this point, we have expressed GFP-GFAP wild type or the harmful Alexander disease GFP-GFAP R239C mutant in astrocytic cells. Interestingly, GFAP R239C appears more oxidized than the wild type under control conditions, as indicated both by its lower cysteine residue accessibility and increased presence of disulfide-bonded oligomers. Moreover, GFP-GFAP R239C undergoes lipoxidation to a higher extent than GFAP wild type upon treatment with the electrophilic mediator 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2). Importantly, GFAP R239C filament organization is altered in untreated cells and is earlier and more severely disrupted than GFAP wild type upon exposure to oxidants (diamide, H2O2) or electrophiles (4-hydroxynonenal, 15d-PGJ2), which exacerbate GFAP R239C aggregation. Furthermore, H2O2 causes reversible alterations in GFAP wild type, but irreversible damage in GFAP R239C expressing cells. Finally, we show that GFAP R239C expression induces a more oxidized cellular status, with decreased free thiol content and increased mitochondrial superoxide generation. In addition, mitochondria show decreased mass, increased colocalization with GFAP and altered morphology. Notably, a GFP-GFAP R239H mutant recapitulates R239C-elicited alterations whereas an R239G mutant induces a milder phenotype. Together, our results outline a deleterious cycle involving altered GFAP R239C organization, mitochondrial dysfunction, oxidative stress, and further GFAP R239C protein damage and network disruption, which could contribute to astrocyte derangement in Alexander disease.
Collapse
Affiliation(s)
- Álvaro Viedma-Poyatos
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040, Madrid, Spain
| | - Patricia González-Jiménez
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040, Madrid, Spain
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040, Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040, Madrid, Spain.
| |
Collapse
|
12
|
Barmaver SN, Muthaiyan Shanmugam M, Chang Y, Bayansan O, Bhan P, Wu GH, Wagner OI. Loss of intermediate filament IFB-1 reduces mobility, density and physiological function of mitochondria in C. elegans sensory neurons. Traffic 2022; 23:270-286. [PMID: 35261124 DOI: 10.1111/tra.12838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 03/03/2022] [Accepted: 03/06/2022] [Indexed: 11/26/2022]
Abstract
Mitochondria and intermediate filament (IF) accumulations often occur during imbalanced axonal transport leading to various types of neurological diseases. It is still poorly understood whether a link between neuronal IFs and mitochondrial mobility exist. In C. elegans, among the 11 cytoplasmic IF family proteins, IFB-1 is of particular interest as it is expressed in a subset of sensory neurons. Depletion of IFB-1 leads to mild dye-filling and significant chemotaxis defects as well as reduced life span. Sensory neuron development is affected and mitochondria transport is slowed down leading to reduced densities of these organelles. Mitochondria tend to cluster in neurons of IFB-1 mutants likely independent of the fission and fusion machinery. Oxygen consumption and mitochondrial membrane potential is measurably reduced in worms carrying mutations in the ifb-1 gene. Membrane potential also seems to play a role in transport such as FCCP treatment led to increased directional switching of mitochondria. Mitochondria colocalize with IFB-1 in worm neurons and appear in a complex with IFB-1 in pull-down assays. In summary, we propose a model in which neuronal intermediate filaments may serve as critical (transient) anchor points for mitochondria during their long-range transport in neurons for steady and balanced transport. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Syed Nooruzuha Barmaver
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu, Taiwan (R.O.C.)
| | - Muniesh Muthaiyan Shanmugam
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu, Taiwan (R.O.C.)
| | - Yen Chang
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu, Taiwan (R.O.C.)
| | - Odvogmed Bayansan
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu, Taiwan (R.O.C.)
| | - Prerana Bhan
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu, Taiwan (R.O.C.).,Research Center for Healthy Aging, China Medical University, Taichung, Taiwan (R.O.C.)
| | - Gong-Her Wu
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu, Taiwan (R.O.C.)
| | - Oliver I Wagner
- National Tsing Hua University, Institute of Molecular and Cellular Biology, Department of Life Science, Hsinchu, Taiwan (R.O.C.)
| |
Collapse
|
13
|
Shah M, Chacko LA, Joseph JP, Ananthanarayanan V. Mitochondrial dynamics, positioning and function mediated by cytoskeletal interactions. Cell Mol Life Sci 2021; 78:3969-3986. [PMID: 33576841 PMCID: PMC11071877 DOI: 10.1007/s00018-021-03762-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/27/2020] [Accepted: 01/15/2021] [Indexed: 12/22/2022]
Abstract
The ability of a mitochondrion to undergo fission and fusion, and to be transported and localized within a cell are central not just to proper functioning of mitochondria, but also to that of the cell. The cytoskeletal filaments, namely microtubules, F-actin and intermediate filaments, have emerged as prime movers in these dynamic mitochondrial shape and position transitions. In this review, we explore the complex relationship between the cytoskeleton and the mitochondrion, by delving into: (i) how the cytoskeleton helps shape mitochondria via fission and fusion events, (ii) how the cytoskeleton facilitates the translocation and anchoring of mitochondria with the activity of motor proteins, and (iii) how these changes in form and position of mitochondria translate into functioning of the cell.
Collapse
Affiliation(s)
- Mitali Shah
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Leeba Ann Chacko
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Joel P Joseph
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Vaishnavi Ananthanarayanan
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India.
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia.
| |
Collapse
|
14
|
Prasch J, Bernhart E, Reicher H, Kollroser M, Rechberger GN, Koyani CN, Trummer C, Rech L, Rainer PP, Hammer A, Malle E, Sattler W. Myeloperoxidase-Derived 2-Chlorohexadecanal Is Generated in Mouse Heart during Endotoxemia and Induces Modification of Distinct Cardiomyocyte Protein Subsets In Vitro. Int J Mol Sci 2020; 21:ijms21239235. [PMID: 33287422 PMCID: PMC7730634 DOI: 10.3390/ijms21239235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023] Open
Abstract
Sepsis is a major cause of mortality in critically ill patients and associated with cardiac dysfunction, a complication linked to immunological and metabolic aberrations. Cardiac neutrophil infiltration and subsequent release of myeloperoxidase (MPO) leads to the formation of the oxidant hypochlorous acid (HOCl) that is able to chemically modify plasmalogens (ether-phospholipids) abundantly present in the heart. This reaction gives rise to the formation of reactive lipid species including aldehydes and chlorinated fatty acids. During the present study, we tested whether endotoxemia increases MPO-dependent lipid oxidation/modification in the mouse heart. In hearts of lipopolysaccharide-injected mice, we observed significantly higher infiltration of MPO-positive cells, increased fatty acid content, and formation of 2-chlorohexadecanal (2-ClHDA), an MPO-derived plasmalogen modification product. Using murine HL-1 cardiomyocytes as in vitro model, we show that exogenously added HOCl attacks the cellular plasmalogen pool and gives rise to the formation of 2-ClHDA. Addition of 2-ClHDA to HL-1 cardiomyocytes resulted in conversion to 2-chlorohexadecanoic acid and 2-chlorohexadecanol, indicating fatty aldehyde dehydrogenase-mediated redox metabolism. However, a recovery of only 40% indicated the formation of non-extractable (protein) adducts. To identify protein targets, we used a clickable alkynyl analog, 2-chlorohexadec-15-yn-1-al (2-ClHDyA). After Huisgen 1,3-dipolar cycloaddition of 5-tetramethylrhodamine azide (N3-TAMRA) and two dimensional-gel electrophoresis (2D-GE), we were able to identify 51 proteins that form adducts with 2-ClHDyA. Gene ontology enrichment analyses revealed an overrepresentation of heat shock and chaperone, energy metabolism, and cytoskeletal proteins as major targets. Our observations in a murine endotoxemia model demonstrate formation of HOCl-modified lipids in the heart, while pathway analysis in vitro revealed that the chlorinated aldehyde targets specific protein subsets, which are central to cardiac function.
Collapse
Affiliation(s)
- Jürgen Prasch
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | - Eva Bernhart
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | - Helga Reicher
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | | | - Gerald N. Rechberger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria;
- Center for Explorative Lipidomics, BioTechMed Graz, 8010 Graz, Austria
| | - Chintan N. Koyani
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8010 Graz, Austria; (L.R.); (P.P.R.)
| | - Christopher Trummer
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | - Lavinia Rech
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8010 Graz, Austria; (L.R.); (P.P.R.)
| | - Peter P. Rainer
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, 8010 Graz, Austria; (L.R.); (P.P.R.)
| | - Astrid Hammer
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria;
| | - Ernst Malle
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
| | - Wolfgang Sattler
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria; (J.P.); (E.B.); (H.R.); (C.N.K.); (C.T.); (E.M.)
- Center for Explorative Lipidomics, BioTechMed Graz, 8010 Graz, Austria
- Correspondence: ; Tel.: +43-316-385-71950
| |
Collapse
|
15
|
Type III intermediate filaments as targets and effectors of electrophiles and oxidants. Redox Biol 2020; 36:101582. [PMID: 32711378 PMCID: PMC7381704 DOI: 10.1016/j.redox.2020.101582] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 05/05/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
Intermediate filaments (IFs) play key roles in cell mechanics, signaling and homeostasis. Their assembly and dynamics are finely regulated by posttranslational modifications. The type III IFs, vimentin, desmin, peripherin and glial fibrillary acidic protein (GFAP), are targets for diverse modifications by oxidants and electrophiles, for which their conserved cysteine residue emerges as a hot spot. Pathophysiological examples of these modifications include lipoxidation in cell senescence and rheumatoid arthritis, disulfide formation in cataracts and nitrosation in endothelial shear stress, although some oxidative modifications can also be detected under basal conditions. We previously proposed that cysteine residues of vimentin and GFAP act as sensors for oxidative and electrophilic stress, and as hinges influencing filament assembly. Accumulating evidence indicates that the structurally diverse cysteine modifications, either per se or in combination with other posttranslational modifications, elicit specific functional outcomes inducing distinct assemblies or network rearrangements, including filament stabilization, bundling or fragmentation. Cysteine-deficient mutants are protected from these alterations but show compromised cellular performance in network assembly and expansion, organelle positioning and aggresome formation, revealing the importance of this residue. Therefore, the high susceptibility to modification of the conserved cysteine of type III IFs and its cornerstone position in filament architecture sustains their role in redox sensing and integration of cellular responses. This has deep pathophysiological implications and supports the potential of this residue as a drug target. Type III intermediate filaments can be modified by many oxidants and electrophiles. Oxidative modifications of type III IFs occur in normal and pathological conditions. The conserved cysteine residue acts as a hub for redox/electrophilic modifications. Cysteine modifications elicit structure-dependent type III IF rearrangements. Type III intermediate filaments act as sensors for oxidative and electrophilic stress.
Collapse
|
16
|
Alessandra G, Algerta M, Paola M, Carsten S, Cristina L, Paolo M, Elisa M, Gabriella T, Carla P. Shaping Pancreatic β-Cell Differentiation and Functioning: The Influence of Mechanotransduction. Cells 2020; 9:E413. [PMID: 32053947 PMCID: PMC7072458 DOI: 10.3390/cells9020413] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/08/2023] Open
Abstract
Embryonic and pluripotent stem cells hold great promise in generating β-cells for both replacing medicine and novel therapeutic discoveries in diabetes mellitus. However, their differentiation in vitro is still inefficient, and functional studies reveal that most of these β-like cells still fail to fully mirror the adult β-cell physiology. For their proper growth and functioning, β-cells require a very specific environment, the islet niche, which provides a myriad of chemical and physical signals. While the nature and effects of chemical stimuli have been widely characterized, less is known about the mechanical signals. We here review the current status of knowledge of biophysical cues provided by the niche where β-cells normally live and differentiate, and we underline the possible machinery designated for mechanotransduction in β-cells. Although the regulatory mechanisms remain poorly understood, the analysis reveals that β-cells are equipped with all mechanosensors and signaling proteins actively involved in mechanotransduction in other cell types, and they respond to mechanical cues by changing their behavior. By engineering microenvironments mirroring the biophysical niche properties it is possible to elucidate the β-cell mechanotransductive-regulatory mechanisms and to harness them for the promotion of β-cell differentiation capacity in vitro.
Collapse
Affiliation(s)
- Galli Alessandra
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Marku Algerta
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Marciani Paola
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| | - Schulte Carsten
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Lenardi Cristina
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Milani Paolo
- CIMAINA, Department of Physics, Università degli Studi di Milano, 20133 Milan, Italy
| | - Maffioli Elisa
- Department of Veterinary Medicine, Università degli Studi di Milano, 20133 Milan, Italy
| | - Tedeschi Gabriella
- Department of Veterinary Medicine, Università degli Studi di Milano, 20133 Milan, Italy
| | - Perego Carla
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20134 Milan, Italy
| |
Collapse
|
17
|
Kuznetsov AV, Javadov S, Grimm M, Margreiter R, Ausserlechner MJ, Hagenbuchner J. Crosstalk between Mitochondria and Cytoskeleton in Cardiac Cells. Cells 2020; 9:cells9010222. [PMID: 31963121 PMCID: PMC7017221 DOI: 10.3390/cells9010222] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/28/2022] Open
Abstract
Elucidation of the mitochondrial regulatory mechanisms for the understanding of muscle bioenergetics and the role of mitochondria is a fundamental problem in cellular physiology and pathophysiology. The cytoskeleton (microtubules, intermediate filaments, microfilaments) plays a central role in the maintenance of mitochondrial shape, location, and motility. In addition, numerous interactions between cytoskeletal proteins and mitochondria can actively participate in the regulation of mitochondrial respiration and oxidative phosphorylation. In cardiac and skeletal muscles, mitochondrial positions are tightly fixed, providing their regular arrangement and numerous interactions with other cellular structures such as sarcoplasmic reticulum and cytoskeleton. This can involve association of cytoskeletal proteins with voltage-dependent anion channel (VDAC), thereby, governing the permeability of the outer mitochondrial membrane (OMM) to metabolites, and regulating cell energy metabolism. Cardiomyocytes and myocardial fibers demonstrate regular arrangement of tubulin beta-II isoform entirely co-localized with mitochondria, in contrast to other isoforms of tubulin. This observation suggests the participation of tubulin beta-II in the regulation of OMM permeability through interaction with VDAC. The OMM permeability is also regulated by the specific isoform of cytolinker protein plectin. This review summarizes and discusses previous studies on the role of cytoskeletal proteins in the regulation of energy metabolism and mitochondrial function, adenosine triphosphate (ATP) production, and energy transfer.
Collapse
Affiliation(s)
- Andrey V. Kuznetsov
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University, 6020 Innsbruck, Austria;
- Department of Paediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria;
- Correspondence: (A.V.K.); (J.H.); Tel.: +43-512-504-27815 (A.V.K.); +43-512-504-81578 (J.H.)
| | - Sabzali Javadov
- Department of Physiology, School of Medicine, University of Puerto Rico, San Juan, PR 00936-5067, USA;
| | - Michael Grimm
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University, 6020 Innsbruck, Austria;
| | - Raimund Margreiter
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | | | - Judith Hagenbuchner
- Department of Paediatrics II, Medical University of Innsbruck, 6020 Innsbruck, Austria
- Correspondence: (A.V.K.); (J.H.); Tel.: +43-512-504-27815 (A.V.K.); +43-512-504-81578 (J.H.)
| |
Collapse
|
18
|
Bhan P, Muthaiyan Shanmugam M, Wang D, Bayansan O, Chen C, Wagner OI. Characterization of TAG‐63 and its role on axonal transport inC.elegans. Traffic 2019; 21:231-249. [DOI: 10.1111/tra.12706] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 10/13/2019] [Accepted: 10/13/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Prerana Bhan
- Department of Life ScienceNational Tsing Hua University, Institute of Molecular and Cellular Biology Hsinchu Taiwan, ROC
- Research Center for Healthy AgingChina Medical University Taichung Taiwan, ROC
| | - Muniesh Muthaiyan Shanmugam
- Department of Life ScienceNational Tsing Hua University, Institute of Molecular and Cellular Biology Hsinchu Taiwan, ROC
| | - Ding Wang
- Department of Life ScienceNational Tsing Hua University, Institute of Molecular and Cellular Biology Hsinchu Taiwan, ROC
| | - Odvogmed Bayansan
- Department of Life ScienceNational Tsing Hua University, Institute of Molecular and Cellular Biology Hsinchu Taiwan, ROC
| | - Chih‐Wei Chen
- Department of Life ScienceNational Tsing Hua University, Institute of Molecular and Cellular Biology Hsinchu Taiwan, ROC
| | - Oliver I. Wagner
- Department of Life ScienceNational Tsing Hua University, Institute of Molecular and Cellular Biology Hsinchu Taiwan, ROC
| |
Collapse
|
19
|
The Role of Mitochondria in the Mechanisms of Cardiac Ischemia-Reperfusion Injury. Antioxidants (Basel) 2019; 8:antiox8100454. [PMID: 31590423 PMCID: PMC6826663 DOI: 10.3390/antiox8100454] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 01/11/2023] Open
Abstract
Mitochondria play a critical role in maintaining cellular function by ATP production. They are also a source of reactive oxygen species (ROS) and proapoptotic factors. The role of mitochondria has been established in many aspects of cell physiology/pathophysiology, including cell signaling. Mitochondria may deteriorate under various pathological conditions, including ischemia-reperfusion (IR) injury. Mitochondrial injury can be one of the main causes for cardiac and other tissue injuries by energy stress and overproduction of toxic reactive oxygen species, leading to oxidative stress, elevated calcium and apoptotic and necrotic cell death. However, the interplay among these processes in normal and pathological conditions is still poorly understood. Mitochondria play a critical role in cardiac IR injury, where they are directly involved in several pathophysiological mechanisms. We also discuss the role of mitochondria in the context of mitochondrial dynamics, specializations and heterogeneity. Also, we wanted to stress the existence of morphologically and functionally different mitochondrial subpopulations in the heart that may have different sensitivities to diseases and IR injury. Therefore, various cardioprotective interventions that modulate mitochondrial stability, dynamics and turnover, including various pharmacologic agents, specific mitochondrial antioxidants and uncouplers, and ischemic preconditioning can be considered as the main strategies to protect mitochondrial and cardiovascular function and thus enhance longevity.
Collapse
|
20
|
de Pablo Y, Marasek P, Pozo-Rodrigálvarez A, Wilhelmsson U, Inagaki M, Pekna M, Pekny M. Vimentin Phosphorylation Is Required for Normal Cell Division of Immature Astrocytes. Cells 2019; 8:cells8091016. [PMID: 31480524 PMCID: PMC6769829 DOI: 10.3390/cells8091016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/20/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022] Open
Abstract
Vimentin (VIM) is an intermediate filament (nanofilament) protein expressed in multiple cell types, including astrocytes. Mice with VIM mutations of serine sites phosphorylated during mitosis (VIMSA/SA) show cytokinetic failure in fibroblasts and lens epithelial cells, chromosomal instability, facilitated cell senescence, and increased neuronal differentiation of neural progenitor cells. Here we report that in vitro immature VIMSA/SA astrocytes exhibit cytokinetic failure and contain vimentin accumulations that co-localize with mitochondria. This phenotype is transient and disappears with VIMSA/SA astrocyte maturation and expression of glial fibrillary acidic protein (GFAP); it is also alleviated by the inhibition of cell proliferation. To test the hypothesis that GFAP compensates for the effect of VIMSA/SA in astrocytes, we crossed the VIMSA/SA and GFAP−/− mice. Surprisingly, the fraction of VIMSA/SA immature astrocytes with abundant vimentin accumulations was reduced when on GFAP−/− background. This indicates that the disappearance of vimentin accumulations and cytokinetic failure in mature astrocyte cultures are independent of GFAP expression. Both VIMSA/SA and VIMSA/SAGFAP−/− astrocytes showed normal mitochondrial membrane potential and vulnerability to H2O2, oxygen/glucose deprivation, and chemical ischemia. Thus, mutation of mitotic phosphorylation sites in vimentin triggers formation of vimentin accumulations and cytokinetic failure in immature astrocytes without altering their vulnerability to oxidative stress.
Collapse
Affiliation(s)
- Yolanda de Pablo
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden
| | - Pavel Marasek
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden
| | - Andrea Pozo-Rodrigálvarez
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden
| | - Ulrika Wilhelmsson
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden
| | - Masaki Inagaki
- Department of Physiology, Mie University Graduate School of Medicine, Mie 5148507, Japan
| | - Marcela Pekna
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
- University of Newcastle, New South Wales 2308, Australia
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 40530 Gothenburg, Sweden.
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia.
- University of Newcastle, New South Wales 2308, Australia.
| |
Collapse
|
21
|
Wang Z, Jiang S, Cao J, Liu K, Xu S, Arfat Y, Guo Q, Chang H, Goswami N, Hinghofer‐Szalkay H, Gao Y. Novel findings on ultrastructural protection of skeletal muscle fibers during hibernation of Daurian ground squirrels: Mitochondria, nuclei, cytoskeleton, glycogen. J Cell Physiol 2019; 234:13318-13331. [DOI: 10.1002/jcp.28008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 12/18/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Zhe Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education Xi'an China
| | - Shan‐Feng Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education Xi'an China
- Laboratory for Bone Metabolism, Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University Xi'an Shaanxi People's Republic of China
| | - Jin Cao
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education Xi'an China
| | - Kun Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education Xi'an China
| | - Shen‐Hui Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education Xi'an China
| | - Yasir Arfat
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education Xi'an China
| | - Quan‐Ling Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education Xi'an China
| | - Hui Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Sciences, Northwest University, Ministry of Education Xi'an China
| | - Nandu Goswami
- Physiology Unit, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz Graz Austria
| | - Helmut Hinghofer‐Szalkay
- Physiology Unit, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz Graz Austria
| | | |
Collapse
|
22
|
Danielsson F, Peterson MK, Caldeira Araújo H, Lautenschläger F, Gad AKB. Vimentin Diversity in Health and Disease. Cells 2018; 7:E147. [PMID: 30248895 PMCID: PMC6210396 DOI: 10.3390/cells7100147] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022] Open
Abstract
Vimentin is a protein that has been linked to a large variety of pathophysiological conditions, including cataracts, Crohn's disease, rheumatoid arthritis, HIV and cancer. Vimentin has also been shown to regulate a wide spectrum of basic cellular functions. In cells, vimentin assembles into a network of filaments that spans the cytoplasm. It can also be found in smaller, non-filamentous forms that can localise both within cells and within the extracellular microenvironment. The vimentin structure can be altered by subunit exchange, cleavage into different sizes, re-annealing, post-translational modifications and interacting proteins. Together with the observation that different domains of vimentin might have evolved under different selection pressures that defined distinct biological functions for different parts of the protein, the many diverse variants of vimentin might be the cause of its functional diversity. A number of review articles have focussed on the biology and medical aspects of intermediate filament proteins without particular commitment to vimentin, and other reviews have focussed on intermediate filaments in an in vitro context. In contrast, the present review focusses almost exclusively on vimentin, and covers both ex vivo and in vivo data from tissue culture and from living organisms, including a summary of the many phenotypes of vimentin knockout animals. Our aim is to provide a comprehensive overview of the current understanding of the many diverse aspects of vimentin, from biochemical, mechanical, cellular, systems biology and medical perspectives.
Collapse
Affiliation(s)
- Frida Danielsson
- Science for Life Laboratory, Royal Institute of Technology, 17165 Stockholm, Sweden.
| | | | | | - Franziska Lautenschläger
- Campus D2 2, Leibniz-Institut für Neue Materialien gGmbH (INM) and Experimental Physics, NT Faculty, E 2 6, Saarland University, 66123 Saarbrücken, Germany.
| | - Annica Karin Britt Gad
- Centro de Química da Madeira, Universidade da Madeira, 9020105 Funchal, Portugal.
- Department of Medical Biochemistry and Microbiology, Uppsala University, 75237 Uppsala, Sweden.
| |
Collapse
|
23
|
Chen M, Lee HK, Moo L, Hanlon E, Stein T, Xia W. Common proteomic profiles of induced pluripotent stem cell-derived three-dimensional neurons and brain tissue from Alzheimer patients. J Proteomics 2018; 182:21-33. [PMID: 29709615 DOI: 10.1016/j.jprot.2018.04.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/03/2018] [Accepted: 04/24/2018] [Indexed: 01/21/2023]
Abstract
We established a unique platform for proteomic analysis of cultured three-dimensional (3D) neurons and brain tissue from Alzheimer's disease (AD) patients. We collected peripheral blood mononuclear cells (PBMC), converted PBMC to induced pluripotent stem cell (iPSC) lines, and differentiated the iPSC into human 3D neuro-spheroids. The postmortem brain tissue from the superior frontal cortex, inferior frontal cortex and cerebellum area of the AD patients was compared to the same regions from the control subjects. Proteomic analysis of 3D neuro-spheroids derived from AD subjects revealed the alteration of a number of proteins involved in axon growth, mitochondrial function, and antioxidant defense. Similar analysis of post-mortem AD brain tissue revealed significant alteration in proteins involved in oxidative stress, neuro-inflammation, along with proteins related to axonal injury. These results clearly indicate that the dysfunction of 3D neurons from AD patients in our in vitro environment is comparable to the post-mortem AD brain tissue in vivo. In conclusion, our study revealed a number of candidate proteins that have important implications in AD pathogenesis and supports the notion that the iPSC-derived 3D neuronal system functions as a model to examine novel aspects of AD pathology. SIGNIFICANCE In this study, we present a unique platform for proteomic analysis of induced pluripotent stem cell-derived three dimensional (3D) neurons and compare the results to those from three regions of post-mortem brain tissue from Alzheimer's disease patients and normal control subjects. Our results show that the dysfunction of 3D neurons from AD patients in our in vitro environment is comparable to the post-mortem AD brain tissue in vivo. Our results revealed several candidate proteins that have important implications in AD pathogenesis.
Collapse
Affiliation(s)
- Mei Chen
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States; Department of Environmental Health, Harvard T H Chan School of Public Health, Boston, MA, United States
| | - Han-Kyu Lee
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States
| | - Lauren Moo
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States
| | - Eugene Hanlon
- Office of Research and Development, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States
| | - Thor Stein
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States; Department of Pathology, Boston University School of Medicine, Boston, MA, United States
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, MA, United States; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States.
| |
Collapse
|
24
|
Trogden KP, Battaglia RA, Kabiraj P, Madden VJ, Herrmann H, Snider NT. An image-based small-molecule screen identifies vimentin as a pharmacologically relevant target of simvastatin in cancer cells. FASEB J 2018; 32:2841-2854. [PMID: 29401610 DOI: 10.1096/fj.201700663r] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Vimentin is a cytoskeletal intermediate filament protein that is expressed in mesenchymal cells and cancer cells during the epithelial-mesenchymal transition. The goal of this study was to identify vimentin-targeting small molecules by using the Tocriscreen library of 1120 biochemically active compounds. We monitored vimentin filament reorganization and bundling in adrenal carcinoma SW13 vimentin-positive (SW13-vim+) cells via indirect immunofluorescence. The screen identified 18 pharmacologically diverse hits that included 2 statins-simvastatin and mevastatin. Simvastatin induced vimentin reorganization within 15-30 min and significant perinuclear bundling within 60 min (IC50 = 6.7 nM). Early filament reorganization coincided with increased vimentin solubility. Mevastatin produced similar effects at >1 µM, whereas the structurally related pravastatin and lovastatin did not affect vimentin. In vitro vimentin filament assembly assays revealed a direct targeting mechanism, as determined biochemically and by electron microscopy. In SW13-vim+ cells, simvastatin, but not pravastatin, reduced total cell numbers (IC50 = 48.1 nM) and promoted apoptosis after 24 h. In contrast, SW13-vim- cell viability was unaffected by simvastatin, unless vimentin was ectopically expressed. Simvastatin similarly targeted vimentin filaments and induced cell death in MDA-MB-231 (vim+), but lacked effect in MCF7 (vim-) breast cancer cells. In conclusion, this study identified vimentin as a direct molecular target that mediates simvastatin-induced cell death in 2 different cancer cell lines.-Trogden, K. P., Battaglia, R. A., Kabiraj, P., Madden, V. J., Herrmann, H., Snider, N. T. An image-based small-molecule screen identifies vimentin as a pharmacologically relevant target of simvastatin in cancer cells.
Collapse
Affiliation(s)
- Kathryn P Trogden
- Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rachel A Battaglia
- Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Parijat Kabiraj
- Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Victoria J Madden
- Department of Pathology and Laboratory Medicine, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Harald Herrmann
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany.,Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Natasha T Snider
- Department of Cell Biology and Physiology, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
25
|
Duncan EJ, Larivière R, Bradshaw TY, Longo F, Sgarioto N, Hayes MJ, Romano LEL, Nethisinghe S, Giunti P, Bruntraeger MB, Durham HD, Brais B, Maltecca F, Gentil BJ, Chapple JP. Altered organization of the intermediate filament cytoskeleton and relocalization of proteostasis modulators in cells lacking the ataxia protein sacsin. Hum Mol Genet 2018; 26:3130-3143. [PMID: 28535259 PMCID: PMC5886247 DOI: 10.1093/hmg/ddx197] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/16/2017] [Indexed: 01/09/2023] Open
Abstract
Autosomal Recessive Spastic Ataxia of Charlevoix-Saguenay (ARSACS) is caused by mutations in the gene SACS, encoding the 520 kDa protein sacsin. Although sacsin’s physiological role is largely unknown, its sequence domains suggest a molecular chaperone or protein quality control function. Consequences of its loss include neurofilament network abnormalities, specifically accumulation and bundling of perikaryal and dendritic neurofilaments. To investigate if loss of sacsin affects intermediate filaments more generally, the distribution of vimentin was analysed in ARSACS patient fibroblasts and in cells where sacsin expression was reduced. Abnormal perinuclear accumulation of vimentin filaments, which sometimes had a cage-like appearance, occurred in sacsin-deficient cells. Mitochondria and other organelles were displaced to the periphery of vimentin accumulations. Reorganization of the vimentin network occurs in vitro under stress conditions, including when misfolded proteins accumulate. In ARSACS patient fibroblasts HSP70, ubiquitin and the autophagy-lysosome pathway proteins Lamp2 and p62 relocalized to the area of the vimentin accumulation. There was no overall increase in ubiquitinated proteins, suggesting the ubiquitin–proteasome system was not impaired. There was evidence for alterations in the autophagy–lysosome pathway. Specifically, in ARSACS HDFs cellular levels of Lamp2 were elevated while levels of p62, which is degraded in autophagy, were decreased. Moreover, autophagic flux was increased in ARSACS HDFs under starvation conditions. These data show that loss of sacsin effects the organization of intermediate filaments in multiple cell types, which impacts the cellular distribution of other organelles and influences autophagic activity.
Collapse
Affiliation(s)
- Emma J Duncan
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Roxanne Larivière
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Teisha Y Bradshaw
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Fabiana Longo
- Università Vita-Salute San Raffaele and Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy.,Università degli Studi dell'Insubria, Varese, Italy
| | - Nicolas Sgarioto
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | | | - Lisa E L Romano
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Suran Nethisinghe
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Paola Giunti
- Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK
| | - Michaela B Bruntraeger
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Heather D Durham
- Laboratory of Neurogenetics of Motion, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Bernard Brais
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Francesca Maltecca
- Università Vita-Salute San Raffaele and Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Benoit J Gentil
- Laboratory of Neurogenetics of Motion, Montreal Neurological Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - J Paul Chapple
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
26
|
Baek YH, Lee WJ, Kim GJ. Mitochondria-Specific Monoclonal Antibodies in Eggs and Embryos of the Ascidian Halocynthia roretzi. Dev Reprod 2017; 21:467-473. [PMID: 29354792 PMCID: PMC5769141 DOI: 10.12717/dr.2017.21.4.467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/02/2017] [Accepted: 11/20/2017] [Indexed: 12/02/2022]
Abstract
Ascidian embryos have become an important model for embryological studies, offering a simple example for mechanisms of cytoplasmic components segregation. It is a well-known example that the asymmetric segregation of mitochondria into muscle lineage cells occurs during ascidian embryogenesis. However, it is still unclear which signaling pathway is involved in this process. To obtain molecular markers for studying mechanisms involved in the asymmetric distribution of mitochondria, we have produced monoclonal antibodies, Mito-1, Mito-2 and Mito-3, that specifically recognize mitochondriarich cytoplasm in cells of the ascidian Halocynthia roretzi embryos. These antibodies stained cytoplasm like reticular structure in epidermis cells, except for nuclei, at the early tailbud stage. Similar pattern was observed in vital staining of mitochondria with DiOC2, a fluorescent probe of mitochondria. Immunostaining with these antibodies showed that mitochondria are evenly distributed in the animal hemisphere blastomeres at cleavage stages, whereas not in the vegetal hemisphere blastomeres. Mitochondria were transferred to the presumptive muscle and nerve cord lineage cells of the marginal zone in the vegetal hemisphere more than to the presumptive mesenchyme, notochord and endoderm lineage of the central zone. Therefore, it is suggested that these antibodies will be useful markers for studying mechanisms involved in the polarized distribution of mitochondria during ascidian embryogenesis.
Collapse
Affiliation(s)
- Yong Han Baek
- Dept. of Marine Molecular Bioscience, Gangneung-Wonju National
University, Gangneung 25457, Republic of Korea
| | - Wang Jong Lee
- Dept. of Marine Molecular Bioscience, Gangneung-Wonju National
University, Gangneung 25457, Republic of Korea
| | - Gil Jung Kim
- Dept. of Marine Molecular Bioscience, Gangneung-Wonju National
University, Gangneung 25457, Republic of Korea
| |
Collapse
|
27
|
Jackson JG, Robinson MB. Regulation of mitochondrial dynamics in astrocytes: Mechanisms, consequences, and unknowns. Glia 2017; 66:1213-1234. [PMID: 29098734 DOI: 10.1002/glia.23252] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/20/2017] [Accepted: 10/09/2017] [Indexed: 12/15/2022]
Abstract
Astrocytes are the major glial cell in the central nervous system. These polarized cells possess numerous processes that ensheath the vasculature and contact synapses. Astrocytes play important roles in synaptic signaling, neurotransmitter synthesis and recycling, control of nutrient uptake, and control of local blood flow. Many of these processes depend on local metabolism and/or energy utilization. While astrocytes respond to increases in neuronal activity and metabolic demand by upregulating glycolysis and glycogenolysis, astrocytes also possess significant capacity for oxidative (mitochondrial) metabolism. Mitochondria mediate energy supply and metabolism, cellular survival, ionic homeostasis, and proliferation. These organelles are dynamic structures undergoing extensive fission and fusion, directed movement along cytoskeletal tracts, and degradation. While many of the mechanisms underlying the dynamics of these organelles and their physiologic roles have been characterized in neurons and other cells, the roles that mitochondrial dynamics play in glial physiology is less well understood. Recent work from several laboratories has demonstrated that mitochondria are present within the fine processes of astrocytes, that their movement is regulated, and that they contribute to local Ca2+ signaling within the astrocyte. They likely play a role in local ATP production and metabolism, particularly that of glutamate. Here we will review these and other findings describing the mechanism by which mitochondrial dynamics are regulated in astrocytes, how mitochondrial dynamics might influence astrocyte and brain metabolism, and draw parallels to mitochondrial dynamics in neurons. Additionally, we present new analyses of the size, distribution, and dynamics of mitochondria in astrocytes performed using in vivo using 2-photon microscopy.
Collapse
Affiliation(s)
- Joshua G Jackson
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, 19104.,Departments of Pediatrics, University of Pennsylvania, Philadelphia, PA, 19104
| | - Michael B Robinson
- Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, 19104.,Departments of Pediatrics, University of Pennsylvania, Philadelphia, PA, 19104.,Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, 19104
| |
Collapse
|
28
|
Bartolák-Suki E, Imsirovic J, Nishibori Y, Krishnan R, Suki B. Regulation of Mitochondrial Structure and Dynamics by the Cytoskeleton and Mechanical Factors. Int J Mol Sci 2017; 18:E1812. [PMID: 28825689 PMCID: PMC5578198 DOI: 10.3390/ijms18081812] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/09/2017] [Accepted: 08/18/2017] [Indexed: 12/21/2022] Open
Abstract
Mitochondria supply cells with energy in the form of ATP, guide apoptosis, and contribute to calcium buffering and reactive oxygen species production. To support these diverse functions, mitochondria form an extensive network with smaller clusters that are able to move along microtubules aided by motor proteins. Mitochondria are also associated with the actin network, which is involved in cellular responses to various mechanical factors. In this review, we discuss mitochondrial structure and function in relation to the cytoskeleton and various mechanical factors influencing cell functions. We first summarize the morphological features of mitochondria with an emphasis on fission and fusion as well as how network properties govern function. We then review the relationship between the mitochondria and the cytoskeletal structures, including mechanical interactions. We also discuss how stretch and its dynamic pattern affect mitochondrial structure and function. Finally, we present preliminary data on how extracellular matrix stiffness influences mitochondrial morphology and ATP generation. We conclude by discussing the more general role that mitochondria may play in mechanobiology and how the mechanosensitivity of mitochondria may contribute to the development of several diseases and aging.
Collapse
Affiliation(s)
| | - Jasmin Imsirovic
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| | - Yuichiro Nishibori
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan.
| | - Ramaswamy Krishnan
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA.
| |
Collapse
|
29
|
Biomarkers of chondriome topology and function: implications for the extension of healthy aging. Biogerontology 2016; 18:201-215. [PMID: 28028686 DOI: 10.1007/s10522-016-9673-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 12/19/2016] [Indexed: 12/27/2022]
Abstract
Multiple theories of aging (e.g., free radical, error catastrophe, mitochondrial) are complementary but fail to provide adequate models that comprehensively predict lifelong aging processes and that are valid across species. Hayflick (PLoS Genet 3(12):2351-2354, 2007) described six universal characteristics of aging that focus upon post-reproductive molecular entropy. Here we present a thermodynamic potential model of aging in which the energetic and topological properties of the mitochondrion drive functional and structural stabilities within living systems. Using multivariate regressions of physiological assessments from the National Health and Nutrition Examination Survey, VO2 max consistently declined with age regardless of gender or race, although it had a significantly greater decline for African American females. Percent fat (negative), hematocrit (negative), and urine creatinine (negative) were strongly and significantly associated with VO2 max and male aging, although cholesterol (positive) was an additional factor for African American males. Bioenergetic measures such as VO2 max can be useful for physical assessments to promote healthy aging.
Collapse
|
30
|
Pasqualini FS, Nesmith AP, Horton RE, Sheehy SP, Parker KK. Mechanotransduction and Metabolism in Cardiomyocyte Microdomains. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4081638. [PMID: 28044126 PMCID: PMC5164897 DOI: 10.1155/2016/4081638] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 01/11/2023]
Abstract
Efficient contractions of the left ventricle are ensured by the continuous transfer of adenosine triphosphate (ATP) from energy production sites, the mitochondria, to energy utilization sites, such as ionic pumps and the force-generating sarcomeres. To minimize the impact of intracellular ATP trafficking, sarcomeres and mitochondria are closely packed together and in proximity with other ultrastructures involved in excitation-contraction coupling, such as t-tubules and sarcoplasmic reticulum junctions. This complex microdomain has been referred to as the intracellular energetic unit. Here, we review the literature in support of the notion that cardiac homeostasis and disease are emergent properties of the hierarchical organization of these units. Specifically, we will focus on pathological alterations of this microdomain that result in cardiac diseases through energy imbalance and posttranslational modifications of the cytoskeletal proteins involved in mechanosensing and transduction.
Collapse
Affiliation(s)
- Francesco S. Pasqualini
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Institute for Regenerative Medicine (IREM), Wyss Translational Center, University and ETH Zurich, Zurich, Switzerland
| | - Alexander P. Nesmith
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Renita E. Horton
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- James Worth Bagley College of Engineering and College of Agriculture and Life Sciences, Mississippi State University, Starkville, MS, USA
| | - Sean P. Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
31
|
Pisani C, Onori A, Gabanella F, Delle Monache F, Borreca A, Ammassari-Teule M, Fanciulli M, Di Certo MG, Passananti C, Corbi N. eEF1Bγ binds the Che-1 and TP53 gene promoters and their transcripts. J Exp Clin Cancer Res 2016; 35:146. [PMID: 27639846 PMCID: PMC5027090 DOI: 10.1186/s13046-016-0424-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/10/2016] [Indexed: 11/25/2022] Open
Abstract
Background We have previously shown that the eukaryotic elongation factor subunit 1B gamma (eEF1Bγ) interacts with the RNA polymerase II (pol II) alpha-like subunit “C” (POLR2C), alone or complexed, in the pol II enzyme. Moreover, we demonstrated that eEF1Bγ binds the promoter region and the 3’ UTR mRNA of the vimentin gene. These events contribute to localize the vimentin transcript and consequentially its translation, promoting a proper mitochondrial network. Methods With the intent of identifying additional transcripts that complex with the eEF1Bγ protein, we performed a series of ribonucleoprotein immunoprecipitation (RIP) assays using a mitochondria-enriched heavy membrane (HM) fraction. Results Among the eEF1Bγ complexed transcripts, we found the mRNA encoding the Che-1/AATF multifunctional protein. As reported by other research groups, we found the tumor suppressor p53 transcript complexed with the eEF1Bγ protein. Here, we show for the first time that eEF1Bγ binds not only Che-1 and p53 transcripts but also their promoters. Remarkably, we demonstrate that both the Che-1 transcript and its translated product localize also to the mitochondria and that eEF1Bγ depletion strongly perturbs the mitochondrial network and the correct localization of Che-1. In a doxorubicin (Dox)-induced DNA damage assay we show that eEF1Bγ depletion significantly decreases p53 protein accumulation and slightly impacts on Che-1 accumulation. Importantly, Che-1 and p53 proteins are components of the DNA damage response machinery that maintains genome integrity and prevents tumorigenesis. Conclusions Our data support the notion that eEF1Bγ, besides its canonical role in translation, is an RNA-binding protein and a key player in cellular stress responses. We suggest for eEF1Bγ a role as primordial transcription/translation factor that links fundamental steps from transcription control to local translation. Electronic supplementary material The online version of this article (doi:10.1186/s13046-016-0424-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cinzia Pisani
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161, Rome, Italy.
| | - Annalisa Onori
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161, Rome, Italy
| | - Francesca Gabanella
- CNR -Institute of Cell Biology and Neurobiology, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Francesca Delle Monache
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161, Rome, Italy
| | - Antonella Borreca
- CNR -Institute of Cell Biology and Neurobiology, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Martine Ammassari-Teule
- CNR -Institute of Cell Biology and Neurobiology, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Maurizio Fanciulli
- Department of Research, Advanced Diagnostic, and Technological Innovation, SAFU Laboratory, Regina Elena Cancer Institute, Rome, Italy
| | - Maria Grazia Di Certo
- CNR -Institute of Cell Biology and Neurobiology, Rome, Italy.,IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Claudio Passananti
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161, Rome, Italy
| | - Nicoletta Corbi
- CNR-Institute of Molecular Biology and Pathology, Department of Molecular Medicine, Sapienza University, Viale Regina Elena 291, 00161, Rome, Italy.
| |
Collapse
|
32
|
Varghese M, Santa-Maria I, Ho L, Ward L, Yemul S, Dubner L, Księżak-Reding H, Pasinetti GM. Extracellular Tau Paired Helical Filaments Differentially Affect Tau Pathogenic Mechanisms in Mitotic and Post-Mitotic Cells: Implications for Mechanisms of Tau Propagation in the Brain. J Alzheimers Dis 2016; 54:477-96. [DOI: 10.3233/jad-160166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Merina Varghese
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Ismael Santa-Maria
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Lap Ho
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Libby Ward
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shrishailam Yemul
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Lauren Dubner
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanna Księżak-Reding
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| | - Giulio Maria Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| |
Collapse
|
33
|
Intermediate Filaments as Organizers of Cellular Space: How They Affect Mitochondrial Structure and Function. Cells 2016; 5:cells5030030. [PMID: 27399781 PMCID: PMC5040972 DOI: 10.3390/cells5030030] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/24/2016] [Accepted: 06/30/2016] [Indexed: 12/17/2022] Open
Abstract
Intermediate filaments together with actin filaments and microtubules form the cytoskeleton, which is a complex and highly dynamic 3D network. Intermediate filaments are the major mechanical stress protectors but also affect cell growth, differentiation, signal transduction, and migration. Using intermediate filament-mitochondrial crosstalk as a prominent example, this review emphasizes the importance of intermediate filaments as crucial organizers of cytoplasmic space to support these functions. We summarize observations in different mammalian cell types which demonstrate how intermediate filaments influence mitochondrial morphology, subcellular localization, and function through direct and indirect interactions and how perturbations of these interactions may lead to human diseases.
Collapse
|
34
|
Lowery J, Jain N, Kuczmarski ER, Mahammad S, Goldman A, Gelfand VI, Opal P, Goldman RD. Abnormal intermediate filament organization alters mitochondrial motility in giant axonal neuropathy fibroblasts. Mol Biol Cell 2015; 27:608-16. [PMID: 26700320 PMCID: PMC4750921 DOI: 10.1091/mbc.e15-09-0627] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 12/18/2015] [Indexed: 11/17/2022] Open
Abstract
GAN patient cells have abnormal aggregates of vimentin intermediate filaments, to which mitochondria appear to be tethered. Motility of mitochondria, but not lysosomes, is inhibited in these cells. Transfection with wild-type gigaxonin (the protein mutated in this disease) disperses these aggregates and bundles, and mitochondrial motility returns to normal. Giant axonal neuropathy (GAN) is a rare disease caused by mutations in the GAN gene, which encodes gigaxonin, an E3 ligase adapter that targets intermediate filament (IF) proteins for degradation in numerous cell types, including neurons and fibroblasts. The cellular hallmark of GAN pathology is the formation of large aggregates and bundles of IFs. In this study, we show that both the distribution and motility of mitochondria are altered in GAN fibroblasts and this is attributable to their association with vimentin IF aggregates and bundles. Transient expression of wild-type gigaxonin in GAN fibroblasts reduces the number of IF aggregates and bundles, restoring mitochondrial motility. Conversely, silencing the expression of gigaxonin in control fibroblasts leads to changes in IF organization similar to that of GAN patient fibroblasts and a coincident loss of mitochondrial motility. The inhibition of mitochondrial motility in GAN fibroblasts is not due to a global inhibition of organelle translocation, as lysosome motility is normal. Our findings demonstrate that it is the pathological changes in IF organization that cause the loss of mitochondrial motility.
Collapse
Affiliation(s)
- Jason Lowery
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Nikhil Jain
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611 Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Zurich 8093, Switzerland
| | - Edward R Kuczmarski
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Saleemulla Mahammad
- Stem Cell and Cancer Research Institute, Michael DeGroote Centre for Learning and Discovery, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Anne Goldman
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| | - Puneet Opal
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL 60611
| |
Collapse
|
35
|
Wang J, Cai J, Huang Y, Ke Q, Wu B, Wang S, Han X, Wang T, Wang Y, Li W, Lao C, Song W, Xiang AP. Nestin regulates proliferation and invasion of gastrointestinal stromal tumor cells by altering mitochondrial dynamics. Oncogene 2015; 35:3139-50. [PMID: 26434586 DOI: 10.1038/onc.2015.370] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 08/21/2015] [Accepted: 08/28/2015] [Indexed: 12/11/2022]
Abstract
Nestin is widely expressed in numerous tumors and has become a diagnostic and prognostic indicator. However, the exact mechanism by which nestin contributes to tumor malignancy remains poorly understood. Here, we found marked upregulation of nestin expression in highly proliferative and invasive gastrointestinal stromal tumor (GIST) specimens. Nestin knockdown in GIST cells reduced the proliferative and invasive activity owing to a decrease of mitochondrial intracellular reactive oxygen species (ROS) generation. Furthermore, nestin was co-localized with mitochondria, and knockdown of nestin increased mitochondrial elongation and influenced the mitochondrial function, including oxygen consumption rates, ATP generation and mitochondrial membrane potential and so on. In exploring the underlying mechanism, we demonstrated nestin knockdown inhibited the mitochondrial recruitment of Dynamin-related protein1 and induced the change of mitochondrial dynamics. Thus, nestin may have an important role in GIST malignancy by regulating mitochondrial dynamics and altering intracellular ROS levels. The findings provide new clues to reveal mechanisms by which nestin mediates the proliferation and invasion of GISTs.
Collapse
Affiliation(s)
- J Wang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - J Cai
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China.,Biotherapy Center, Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Y Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Q Ke
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China.,Department of Cell Biology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - B Wu
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - S Wang
- Department of Gastrointestinal-Pancreatic Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - X Han
- Department of Population Genetics and Prevention, Fuwai Hospital of Peking Union Medical College, Beijing, China
| | - T Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - Y Wang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - W Li
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - C Lao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
| | - W Song
- Department of Gastrointestinal-Pancreatic Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - A P Xiang
- Program of Stem Cells and Regenerative Medicine, Affiliated Guangzhou Women and Children's Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-Sen University, Guangzhou, China.,Biotherapy Center, Third Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China.,Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
36
|
Matveeva EA, Venkova LS, Chernoivanenko IS, Minin AA. Vimentin is involved in regulation of mitochondrial motility and membrane potential by Rac1. Biol Open 2015; 4:1290-7. [PMID: 26369929 PMCID: PMC4610213 DOI: 10.1242/bio.011874] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In this study we show that binding of mitochondria to vimentin intermediate filaments (VIF) is regulated by GTPase Rac1. The activation of Rac1 leads to a redoubling of mitochondrial motility in murine fibroblasts. Using double-mutants Rac1(G12V, F37L) and Rac1(G12V, Y40H) that are capable to activate different effectors of Rac1, we show that mitochondrial movements are regulated through PAK1 kinase. The involvement of PAK1 kinase is also confirmed by the fact that expression of its auto inhibitory domain (PID) blocks the effect of activated Rac1 on mitochondrial motility. The observed effect of Rac1 and PAK1 kinase on mitochondria depends on phosphorylation of the Ser-55 of vimentin. Besides the effect on motility Rac1 activation also decreases the mitochondrial membrane potential (MMP) which is detected by ∼20% drop of the fluorescence intensity of mitochondria stained with the potential sensitive dye TMRM. One of important consequences of the discovered regulation of MMP by Rac1 and PAK1 is a spatial differentiation of mitochondria in polarized fibroblasts: at the front of the cell they are less energized (by ∼25%) than at the rear part.
Collapse
Affiliation(s)
- Elena A Matveeva
- Institute of Protein Research, Russian Academy of Sciences, Department of Cell Biology, Moscow 119988, Russia
| | - Larisa S Venkova
- Institute of Protein Research, Russian Academy of Sciences, Department of Cell Biology, Moscow 119988, Russia
| | - Ivan S Chernoivanenko
- Institute of Protein Research, Russian Academy of Sciences, Department of Cell Biology, Moscow 119988, Russia
| | - Alexander A Minin
- Institute of Protein Research, Russian Academy of Sciences, Department of Cell Biology, Moscow 119988, Russia
| |
Collapse
|
37
|
Zheng P, Xiong Q, Wu Y, Chen Y, Chen Z, Fleming J, Gao D, Bi L, Ge F. Quantitative Proteomics Analysis Reveals Novel Insights into Mechanisms of Action of Long Noncoding RNA Hox Transcript Antisense Intergenic RNA (HOTAIR) in HeLa Cells. Mol Cell Proteomics 2015; 14:1447-63. [PMID: 25762744 DOI: 10.1074/mcp.m114.043984] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Indexed: 12/20/2022] Open
Abstract
Long noncoding RNAs (lncRNAs), which have emerged in recent years as a new and crucial layer of gene regulators, regulate various biological processes such as carcinogenesis and metastasis. HOTAIR (Hox transcript antisense intergenic RNA), a lncRNA overexpressed in most human cancers, has been shown to be an oncogenic lncRNA. Here, we explored the role of HOTAIR in HeLa cells and searched for proteins regulated by HOTAIR. To understand the mechanism of action of HOTAIR from a systems perspective, we employed a quantitative proteomic strategy to systematically identify potential targets of HOTAIR. The expression of 170 proteins was significantly dys-regulated after inhibition of HOTAIR, implying that they could be potential targets of HOTAIR. Analysis of this data at the systems level revealed major changes in proteins involved in diverse cellular components, including the cytoskeleton and the respiratory chain. Further functional studies on vimentin (VIM), a key protein involved in the cytoskeleton, revealed that HOTAIR exerts its effects on migration and invasion of HeLa cells, at least in part, through the regulation of VIM expression. Inhibition of HOTAIR leads to mitochondrial dysfunction and ultrastructural alterations, suggesting a novel role of HOTAIR in maintaining mitochondrial function in cancer cells. Our results provide novel insights into the mechanisms underlying the function of HOTAIR in cancer cells. We expect that the methods used in this study will become an integral part of functional studies of lncRNAs.
Collapse
Affiliation(s)
- Peng Zheng
- From the ‡Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Qian Xiong
- From the ‡Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Ying Wu
- From the ‡Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Ying Chen
- From the ‡Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Zhuo Chen
- From the ‡Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
| | - Joy Fleming
- §Key Laboratory of Noncoding RNA, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ding Gao
- ¶State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Lijun Bi
- §Key Laboratory of Noncoding RNA, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Ge
- From the ‡Key Laboratory of Algal Biology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China;
| |
Collapse
|
38
|
Kyriakou E, Kravariti L, Vasilopoulos T, Zouros E, Rodakis GC. A protein binding site in the M mitochondrial genome of Mytilus galloprovincialis may be responsible for its paternal transmission. Gene 2015; 562:83-94. [PMID: 25701604 DOI: 10.1016/j.gene.2015.02.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/19/2015] [Accepted: 02/16/2015] [Indexed: 10/24/2022]
Abstract
Sea mussels (genus Mytilus) have two mitochondrial genomes in obligatory co-existence, one that is transmitted through the egg and the other through the sperm. The phenomenon, known as Doubly Uniparental Inheritance (DUI) of mitochondrial DNA (mtDNA), is presently known to occur in more than 40 molluscan bivalve species. Females and the somatic tissues of males contain mainly the maternal (F) genome. In contrast, the sperm contains only the paternal (M) genome. Through electrophoretic mobility shift assay (EMSA) experiments we have identified a sequence element in the control region (CR) of the M genome that acts as a binding site for the formation of a complex with a protein factor that occurs in the male gonad. An adenine tract upstream to the element is also essential for the formation of the complex. The reaction is highly specific. It does not occur with protein extracts from the female gonad or from a male or female somatic tissue. Further experiments showed that the interaction takes place in mitochondria surrounding the nucleus of the cells of male gonads, suggesting a distinct role of perinuclear mitochondria. We propose that at a certain point during spermatogenesis mitochondria are subject to degradation and that perinuclear mitochondria with the M mtDNA-protein complex are protected from this degradation with the result that mature spermatozoa contain only the paternal mitochondrial genome.
Collapse
Affiliation(s)
- Eleni Kyriakou
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Panepistimioupolis, 15701 Athens, Greece
| | - Lara Kravariti
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Panepistimioupolis, 15701 Athens, Greece
| | - Themistoklis Vasilopoulos
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Panepistimioupolis, 15701 Athens, Greece
| | - Eleftherios Zouros
- Department of Biology, University of Crete, 71409 Heraklion, Crete, Greece
| | - George C Rodakis
- Department of Biochemistry and Molecular Biology, National and Kapodistrian University of Athens, Panepistimioupolis, 15701 Athens, Greece.
| |
Collapse
|
39
|
Peinado JR, Diaz-Ruiz A, Frühbeck G, Malagon MM. Mitochondria in metabolic disease: getting clues from proteomic studies. Proteomics 2014; 14:452-66. [PMID: 24339000 DOI: 10.1002/pmic.201300376] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 11/08/2013] [Accepted: 11/21/2013] [Indexed: 01/11/2023]
Abstract
Mitochondria play a key role as major regulators of cellular energy homeostasis, but in the context of mitochondrial dysfunction, mitochondria may generate reactive oxidative species and induce cellular apoptosis. Indeed, altered mitochondrial status has been linked to the pathogenesis of several metabolic disorders and specially disorders related to insulin resistance, such as obesity, type 2 diabetes, and other comorbidities comprising the metabolic syndrome. In the present review, we summarize information from various mitochondrial proteomic studies of insulin-sensitive tissues under different metabolic states. To that end, we first focus our attention on the pancreas, as mitochondrial malfunction has been shown to contribute to beta cell failure and impaired insulin release. Furthermore, proteomic studies of mitochondria obtained from liver, muscle, and adipose tissue are summarized, as these tissues constitute the primary insulin target metabolic tissues. Since recent advances in proteomic techniques have exposed the importance of PTMs in the development of metabolic disease, we also present information on specific PTMs that may directly affect mitochondria during the pathogenesis of metabolic disease. Specifically, mitochondrial protein acetylation, phosphorylation, and other PTMs related to oxidative damage, such as nitrosylation and carbonylation, are discussed.
Collapse
Affiliation(s)
- Juan R Peinado
- Department of Medical Sciences, Faculty of Medicine, Ciudad Real, Spain
| | | | | | | |
Collapse
|
40
|
Stress Conditions Increase Vimentin Cleavage by Omi/HtrA2 Protease in Human Primary Neurons and Differentiated Neuroblastoma Cells. Mol Neurobiol 2014; 52:1077-1092. [DOI: 10.1007/s12035-014-8906-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/24/2014] [Indexed: 10/24/2022]
|
41
|
The role of tubulin in the mitochondrial metabolism and arrangement in muscle cells. J Bioenerg Biomembr 2014; 46:421-34. [PMID: 25209018 DOI: 10.1007/s10863-014-9579-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/02/2014] [Indexed: 12/19/2022]
Abstract
Tubulin, a well-known component of the microtubule in the cytoskeleton, has an important role in the transport and positioning of mitochondria in a cell type dependent manner. This review describes different functional interactions of tubulin with cellular protein complexes and its functional interaction with the mitochondrial outer membrane. Tubulin is present in oxidative as well as glycolytic type muscle cells, but the kinetics of the in vivo regulation of mitochondrial respiration in these muscle types is drastically different. The interaction between VDAC and tubulin is probably influenced by such factors as isoformic patterns of VDAC and tubulin, post-translational modifications of tubulin and phosphorylation of VDAC. Important factor of the selective permeability of VDAC is the mitochondrial creatine kinase pathway which is present in oxidative cells, but is inactive or missing in glycolytic muscle and cancer cells. As the tubulin-VDAC interaction reduces the permeability of the channel by adenine nucleotides, energy transfer can then take place effectively only through the mitochondrial creatine kinase/phosphocreatine pathway. Therefore, closure of VDAC by tubulin may be one of the reasons of apoptosis in cells without the creatine kinase pathway. An important question in tubulin regulated interactions is whether other proteins are interacting with tubulin. The functional interaction may be direct, through other proteins like plectins, or influenced by simultaneous interaction of other complexes with VDAC.
Collapse
|
42
|
U KP, Subramanian V, Nicholas AP, Thompson PR, Ferretti P. Modulation of calcium-induced cell death in human neural stem cells by the novel peptidylarginine deiminase-AIF pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1162-71. [PMID: 24607566 PMCID: PMC3996523 DOI: 10.1016/j.bbamcr.2014.02.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 02/19/2014] [Accepted: 02/24/2014] [Indexed: 11/29/2022]
Abstract
PADs (peptidylarginine deiminases) are calcium-dependent enzymes that change protein-bound arginine to citrulline (citrullination/deimination) affecting protein conformation and function. PAD up-regulation following chick spinal cord injury has been linked to extensive tissue damage and loss of regenerative capability. Having found that human neural stem cells (hNSCs) expressed PAD2 and PAD3, we studied PAD function in these cells and investigated PAD3 as a potential target for neuroprotection by mimicking calcium-induced secondary injury responses. We show that PAD3, rather than PAD2 is a modulator of cell growth/death and that PAD activity is not associated with caspase-3-dependent cell death, but is required for AIF (apoptosis inducing factor)-mediated apoptosis. PAD inhibition prevents association of PAD3 with AIF and AIF cleavage required for its translocation to the nucleus. Finally, PAD inhibition also hinders calcium-induced cytoskeleton disassembly and association of PAD3 with vimentin, that we show to be associated also with AIF; together this suggests that PAD-dependent cytoskeleton disassembly may play a role in AIF translocation to the nucleus. This is the first study highlighting a role of PAD activity in balancing hNSC survival/death, identifying PAD3 as an important upstream regulator of calcium-induced apoptosis, which could be targeted to reduce neural loss, and shedding light on the mechanisms involved.
Collapse
Affiliation(s)
- Kin Pong U
- Developmental Biology Unit, UCL Institute of Child Health, London WC1N 1EH, UK
| | | | - Antony P Nicholas
- Department of Neurology, University of Alabama at Birmingham and Birmingham VA Medical Center, Birmingham, AL 35294, USA
| | - Paul R Thompson
- Department of Chemistry, TSRI, Scripps Florida, FL 33458, USA
| | - Patrizia Ferretti
- Developmental Biology Unit, UCL Institute of Child Health, London WC1N 1EH, UK.
| |
Collapse
|
43
|
Bereiter-Hahn J. Mitochondrial dynamics in aging and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 127:93-131. [PMID: 25149215 DOI: 10.1016/b978-0-12-394625-6.00004-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitochondria are self-replicating organelles but nevertheless strongly depend on supply coded in nuclear genes. They serve many physiological demands in living cells. Supply of the cytoplasm with ATP and engagement in Ca(2+) regulation belong to the main functions of mitochondria. In large eukaryotic cells, in particular in neurons, with their long dendrites and axons, mitochondria have to move to the sites of their action. This trafficking involves several motor molecules and mechanisms to sense the sites of requirements of mitochondria. With aging and as a consequence of some diseases, mitochondrial components may be rendered dysfunctional, and mtDNA mutations arise during the course of replication and by the action of reactive oxygen species. Mutants in motor molecules engaged in trafficking and in the machinery of fusion and fission are causing severe deficiencies on the cellular level; they support neurodegeneration and, thus, cause many diseases. Frequent fusion and fission events mediate the elimination of impaired parts from mitochondria which finally will be degraded by autophagosomes. Extensive fusion provides a basis for functional complementation. Mobility of proteins and small molecules within the mitochondria is necessary to reach the functional goals of fusion and fission, although cristae and a large fraction of proteins of the respiratory complexes proved to be stable for hours after fusion and perform slow exchange of material.
Collapse
Affiliation(s)
- Jürgen Bereiter-Hahn
- Institute for Cell Biology and Neurosciences, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
44
|
Sultana R, Baglioni M, Cecchetti R, Cai J, Klein JB, Bastiani P, Ruggiero C, Mecocci P, Butterfield DA. Lymphocyte mitochondria: toward identification of peripheral biomarkers in the progression of Alzheimer disease. Free Radic Biol Med 2013; 65:595-606. [PMID: 23933528 PMCID: PMC3849349 DOI: 10.1016/j.freeradbiomed.2013.08.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 07/23/2013] [Accepted: 08/02/2013] [Indexed: 11/18/2022]
Abstract
Alzheimer disease (AD) is an age-related neurodegenerative condition. AD is histopathologically characterized by the presence of three main hallmarks: senile plaques (rich in amyloid-β peptide), neuronal fibrillary tangles (rich in phosphorylated tau protein), and synapse loss. However, definitive biomarkers for this devastating disease in living people are still lacking. In this study, we show that levels of oxidative stress markers are significantly increased in the mitochondria isolated from lymphocytes of subjects with mild cognitive impairment (MCI) compared to cognitively normal individuals. Further, an increase in mitochondrial oxidative stress in MCI is associated with MMSE score, vitamin E components, and β-carotene. Further, a proteomics approach showed that alterations in the levels of thioredoxin-dependent peroxide reductase, myosin light polypeptide 6, and ATP synthase subunit β might be important in the progression and pathogenesis of AD. Increased understanding of oxidative stress and protein alterations in easily obtainable peripheral tissues will be helpful in developing biomarkers to combat this devastating disorder.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Chemistry, Center of Membrane Sciences, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
| | - Mauro Baglioni
- Institute of Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | - Roberta Cecchetti
- Institute of Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | - Jian Cai
- Department of Nephrology and Proteomics Center, University of Louisville, Louisville, KY 40292, USA
| | - Jon B Klein
- Department of Nephrology and Proteomics Center, University of Louisville, Louisville, KY 40292, USA
| | - Patrizia Bastiani
- Institute of Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | - Carmelinda Ruggiero
- Institute of Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | - Patrizia Mecocci
- Institute of Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy.
| | - D Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA.
| |
Collapse
|
45
|
Mallat Y, Tritsch E, Ladouce R, Winter DL, Friguet B, Li Z, Mericskay M. Proteome modulation in H9c2 cardiac cells by microRNAs miR-378 and miR-378. Mol Cell Proteomics 2013; 13:18-29. [PMID: 24068033 DOI: 10.1074/mcp.m113.030569] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
MicroRNAs are a novel class of powerful endogenous regulators of gene expression. MiR-378 and miR-378* are localized in the first intron of the Ppargc1b gene that codes the transcriptional co-activator PGC-1β. The latter regulates energy expenditure as well as mitochondrial biogenesis. The miR-378:miR-378* hairpin is highly expressed in cardiac cells. To better assess their role in cardiomyocytes, we identified miR-378 and miR-378* targets via a proteomic screen. We established H9c2 cellular models of overexpression of miR-378 and miR-378* and identified a total of 86 down-regulated proteins in the presence of either one of these miRs. Functional annotation clustering showed that miR-378 and miR-378* regulate related pathways in cardiomyocytes, including energy metabolism, notably glycolysis, cytoskeleton, notably actin filaments and muscle contraction. Using bioinformatics algorithms we found that 20 proteins were predicted as direct targets of the miRs. We validated eight of these targets by quantitative RT-PCR and luciferase reporter assay. We found that miR-378 targets lactate dehydrogenase A and impacts on cell proliferation and survival whereas miR-378* targets cytoskeleton proteins actin and vimentin. Proteins involved in endoplasmic reticulum stress response such as chaperone and/or calcium buffering proteins GRP78, PPIA (cyclophilin A), calumenin, and GMMPA involved in glycosylation are repressed by these miRs. Our results show that the miR-378/378* hairpin establishes a connection among energy metabolism, cytoskeleton remodeling, and endoplasmic reticulum function through post-transcriptional regulation of key proteins involved in theses pathways.
Collapse
|
46
|
Capanni C, Bruschi M, Columbaro M, Cuccarolo P, Ravera S, Dufour C, Candiano G, Petretto A, Degan P, Cappelli E. Changes in vimentin, lamin A/C and mitofilin induce aberrant cell organization in fibroblasts from Fanconi anemia complementation group A (FA-A) patients. Biochimie 2013; 95:1838-47. [PMID: 23831462 DOI: 10.1016/j.biochi.2013.06.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/21/2013] [Indexed: 01/03/2023]
Abstract
Growing number of publication has proved an increasing of cellular function of the Fanconi anemia proteins. To chromosome stability and DNA repair new roles have been attributed to FA proteins in oxidative stress response and homeostasis, immune response and cytokines sensibility, gene expression. Our work shows a new role for FA-A protein: the organization of the cellular structure. By 2D-PAGE of FA-A and correct fibroblasts treated and untreated with H2O2 we identify different expression of protein involved in the structural organization of nucleus, intermediate filaments and mitochondria. Immunofluorescence and electronic microscopy analysis clearly show an already altered cellular structure in normal culture condition and this worsted after oxidative stress. FA-A cell appears structurally prone to physiologic stress and this could explain part of the phenotype of FA cells.
Collapse
|
47
|
Kraemer FB, Khor VK, Shen WJ, Azhar S. Cholesterol ester droplets and steroidogenesis. Mol Cell Endocrinol 2013; 371:15-9. [PMID: 23089211 PMCID: PMC3584206 DOI: 10.1016/j.mce.2012.10.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 10/05/2012] [Accepted: 10/11/2012] [Indexed: 12/12/2022]
Abstract
Intracellular lipid droplets (LDs) are dynamic organelles that contain a number of associated proteins including perilipin (Plin) and vimentin. Cholesteryl ester (CE)-rich LDs normally accumulate in steroidogenic cells and their mobilization is the preferred initial source of cholesterol for steroidogenesis. Plin1a, 1b and 5 were found to preferentially associate with triacylglycerol-rich LDs and Plin1c and Plin4 to associate with CE-rich LDs, but the biological significance of this remains unanswered. Vimentin null mice were found to have decreased ACTH-stimulated corticosterone levels, and decreased progesterone levels in females, but normal hCG-stimulated testosterone levels in males. Smaller LDs were seen in null cells. Lipoprotein cholesterol delivery to adrenals and ovary was normal, as was the expression of steroidogenic genes; however, the movement of cholesterol to mitochondria was reduced in vimentin null mice. These results suggest that vimentin is important in the maintenance of CE-rich LDs and in the movement of cholesterol for steroidogenesis.
Collapse
|
48
|
Kuznetsov AV, Javadov S, Guzun R, Grimm M, Saks V. Cytoskeleton and regulation of mitochondrial function: the role of beta-tubulin II. Front Physiol 2013; 4:82. [PMID: 23630499 PMCID: PMC3631707 DOI: 10.3389/fphys.2013.00082] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/26/2013] [Indexed: 12/17/2022] Open
Abstract
The control of mitochondrial function is a cardinal issue in the field of cardiac bioenergetics, and the analysis of mitochondrial regulations is central to basic research and in the diagnosis of many diseases. Interaction between cytoskeletal proteins and mitochondria can actively participate in mitochondrial regulation. Potential candidates for the key roles in this regulation are the cytoskeletal proteins plectin and tubulin. Analysis of cardiac cells has revealed regular arrangement of β-tubulin II, fully co-localized with mitochondria. β-Tubulin IV demonstrated a characteristic staining of branched network, β-tubulin III was matched with Z-lines, and β-tubulin I was diffusely spotted and fragmentary polymerized. In contrast, HL-1 cells were characterized by the complete absence of β-tubulin II. Comparative analysis of cardiomyocytes and HL-1 cells revealed a dramatic difference in the mechanisms of mitochondrial regulation. In the heart, colocalization of β-tubulin isotype II with mitochondria suggests that it can participate in the coupling of ATP-ADP translocase (ANT), mitochondrial creatine kinase (MtCK), and VDAC (ANT-MtCK-VDAC). This mitochondrial supercomplex is responsible for the efficient intracellular energy transfer via the phosphocreatine pathway. Existing data suggest that cytoskeletal proteins may control the VDAC, contributing to maintenance of mitochondrial and cellular physiology.
Collapse
Affiliation(s)
- Andrey V Kuznetsov
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University Innsbruck, Tirol, Austria
| | | | | | | | | |
Collapse
|
49
|
Ionising radiation induces persistent alterations in the cardiac mitochondrial function of C57BL/6 mice 40 weeks after local heart exposure. Radiother Oncol 2013; 106:404-10. [PMID: 23522698 DOI: 10.1016/j.radonc.2013.01.017] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 12/19/2012] [Accepted: 01/29/2013] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND PURPOSE Radiotherapy of thoracic and chest-wall tumours increases the long-term risk of radiation-induced heart disease. The aim of this study was to investigate the long-term effect of local heart irradiation on cardiac mitochondria. METHODS C57BL/6 and atherosclerosis-prone ApoE(-/-) mice received local heart irradiation with a single X-ray dose of 2 Gy. To investigate the low-dose effect, C57BL/6 mice also received a single heart dose of 0.2 Gy. Functional and proteomic alterations of cardiac mitochondria were evaluated after 40 weeks, compared to age-matched controls. RESULTS The respiratory capacity of irradiated C57BL/6 cardiac mitochondria was significantly reduced at 40 weeks. In parallel, protein carbonylation was increased, suggesting enhanced oxidative stress. Considerable alterations were found in the levels of proteins of mitochondria-associated cytoskeleton, respiratory chain, ion transport and lipid metabolism. Radiation induced similar but less pronounced effects in the mitochondrial proteome of ApoE(-/-) mice. In ApoE(-/-), no significant change was observed in mitochondrial respiration or protein carbonylation. The dose of 0.2 Gy had no significant effects on cardiac mitochondria. CONCLUSION This study suggests that ionising radiation causes non-transient alterations in cardiac mitochondria, resulting in oxidative stress that may ultimately lead to malfunctioning of the heart muscle.
Collapse
|
50
|
Lee JS, Hou X, Bishop N, Wang S, Flack A, Cho WJ, Chen X, Mao G, Taatjes DJ, Sun F, Zhang K, Jena BP. Aquaporin-assisted and ER-mediated mitochondrial fission: a hypothesis. Micron 2013; 47:50-8. [PMID: 23416165 DOI: 10.1016/j.micron.2013.01.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 01/20/2013] [Accepted: 01/21/2013] [Indexed: 01/14/2023]
Abstract
It is well established that the status of the endoplasmic reticulum (ER) and mitochondria, and the interactions between them, is critical to numerous cellular functions including apoptosis. Mitochondrial dynamics is greatly influenced by cell stress, and recent studies implicate ER in mitochondrial fission. Although a number of proteins have been identified to participate in ER-induced mitochondrial fission, the molecular mechanism of the process is little understood. In the current study, we confirm the involvement of ER in mitochondrial fission and hypothesize the involvement of water channels or aquaporins (AQP) in the process. Previous studies demonstrate the presence of AQP both in the ER and mitochondrial membranes. Mitochondrial swelling has been observed following mitochondrial calcium overload, and studies report that chelation of cytosolic calcium induces extensive mitochondrial division at ER contact sites. Based on this information, the involvement of ER in mitochondrial division, possibly via water channels, is hypothesized. Utilizing a multi-faceted imaging approach consisting of atomic force microscopy on aldehyde-fixed and semi-dry cells, transmission electron microscopy, and immunofluorescence microscopy on live cells, the physical interactions between the two organelles are demonstrated. Mitochondrial fission following ER stress was abrogated with exposure of cells to the AQP inhibitor mercuric chloride, suggesting the involvement of AQP(s) especially AQP8 and AQP9 known to be present in the mitochondrial membrane, in mitochondrial fission.
Collapse
Affiliation(s)
- Jin-Sook Lee
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|