1
|
Folahan JT, Fakir S, Barabutis N. Endothelial Unfolded Protein Response-Mediated Cytoskeletal Effects. Cell Biochem Funct 2024; 42:e70007. [PMID: 39449673 PMCID: PMC11528298 DOI: 10.1002/cbf.70007] [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: 05/28/2024] [Revised: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
The endothelial semipermeable monolayers ensure tissue homeostasis, are subjected to a plethora of stimuli, and their function depends on cytoskeletal integrity and remodeling. The permeability of those membranes can fluctuate to maintain organ homeostasis. In cases of severe injury, inflammation or disease, barrier hyperpermeability can cause irreparable damage of endothelium-dependent issues, and eventually death. Elucidation of the signaling regulating cytoskeletal structure and barrier integrity promotes the development of targeted pharmacotherapies towards disorders related to the impaired endothelium (e.g., acute respiratory distress syndrome, sepsis). Recent reports investigate the role of unfolded protein response in barrier function. Herein we review the cytoskeletal components, the unfolded protein response function; and their interrelations on health and disorder. Moreover, we emphasize on unfolded protein response modulators, since they ameliorate illness related to endothelial leak.
Collapse
Affiliation(s)
- Joy T Folahan
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Saikat Fakir
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| |
Collapse
|
2
|
Pelaez MC, Fiore F, Larochelle N, Dabbaghizadeh A, Comaduran MF, Arbour D, Minotti S, Marcadet L, Semaan M, Robitaille R, Nalbantoglu JN, Sephton CF, Durham HD. Reversal of cognitive deficits in FUS R521G amyotrophic lateral sclerosis mice by arimoclomol and a class I histone deacetylase inhibitor independent of heat shock protein induction. Neurotherapeutics 2024; 21:e00388. [PMID: 38972779 PMCID: PMC11579874 DOI: 10.1016/j.neurot.2024.e00388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/14/2024] [Accepted: 06/15/2024] [Indexed: 07/09/2024] Open
Abstract
Protein misfolding and mislocalization are common to both familial and sporadic forms of amyotrophic lateral sclerosis (ALS). Maintaining proteostasis through induction of heat shock proteins (HSP) to increase chaperoning capacity is a rational therapeutic strategy in the treatment of ALS. However, the threshold for upregulating stress-inducible HSPs remains high in neurons, presenting a therapeutic obstacle. This study used mouse models expressing the ALS variants FUSR521G or SOD1G93A to follow up on previous work in cultured motor neurons showing varied effects of the HSP co-inducer, arimoclomol, and class I histone deacetylase (HDAC) inhibitors on HSP expression depending on the ALS variant being expressed. As in cultured neurons, neither expression of the transgene nor drug treatments induced expression of HSPs in cortex, spinal cord or muscle of FUSR521G mice, indicating suppression of the heat shock response. Nonetheless, arimoclomol, and RGFP963, restored performance on cognitive tests and improved cortical dendritic spine densities. In SOD1G93A mice, multiple HSPs were upregulated in hindlimb skeletal muscle, but not in lumbar spinal cord with the exception of HSPB1 associated with astrocytosis. Drug treatments improved contractile force but reduced the increase in HSPs in muscle rather than facilitating their expression. The data point to mechanisms other than amplification of the heat shock response underlying recovery of cognitive function in ALS-FUS mice by arimoclomol and class I HDAC inhibition and suggest potential benefits in counteracting cognitive impairment in ALS, frontotemporal dementia and related disorders.
Collapse
Affiliation(s)
- Mari Carmen Pelaez
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC Canada.
| | - Frédéric Fiore
- Département de Neurosciences and Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, QC Canada.
| | - Nancy Larochelle
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Afrooz Dabbaghizadeh
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Mario Fernández Comaduran
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Danielle Arbour
- Département de Neurosciences and Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, QC Canada.
| | - Sandra Minotti
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Laetitia Marcadet
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC Canada.
| | - Martine Semaan
- Département de Neurosciences and Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, QC Canada.
| | - Richard Robitaille
- Département de Neurosciences and Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, and Centre Interdisciplinaire de Recherche sur le Cerveau et l'apprentissage, Montréal, QC Canada.
| | - Josephine N Nalbantoglu
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| | - Chantelle F Sephton
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, QC Canada.
| | - Heather D Durham
- Department of Neurology & Neurosurgery and Montreal Neurological Institute, McGill University, Montreal, QC Canada.
| |
Collapse
|
3
|
Servín Muñoz IV, Ortuño-Sahagún D, Griñán-Ferré C, Pallàs M, González-Castillo C. Alterations in Proteostasis Mechanisms in Niemann-Pick Type C Disease. Int J Mol Sci 2024; 25:3806. [PMID: 38612616 PMCID: PMC11011983 DOI: 10.3390/ijms25073806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 04/14/2024] Open
Abstract
Niemann-Pick Type C (NPC) represents an autosomal recessive disorder with an incidence rate of 1 in 150,000 live births, classified within lysosomal storage diseases (LSDs). The abnormal accumulation of unesterified cholesterol characterizes the pathophysiology of NPC. This phenomenon is not unique to NPC, as analogous accumulations have also been observed in Alzheimer's disease, Parkinson's disease, and other neurodegenerative disorders. Interestingly, disturbances in the folding of the mutant protein NPC1 I1061T are accompanied by the aggregation of proteins such as hyperphosphorylated tau, α-synuclein, TDP-43, and β-amyloid peptide. These accumulations suggest potential disruptions in proteostasis, a regulatory process encompassing four principal mechanisms: synthesis, folding, maintenance of folding, and protein degradation. The dysregulation of these processes leads to excessive accumulation of abnormal proteins that impair cell function and trigger cytotoxicity. This comprehensive review delineates reported alterations across proteostasis mechanisms in NPC, encompassing changes in processes from synthesis to degradation. Additionally, it discusses therapeutic interventions targeting pharmacological facets of proteostasis in NPC. Noteworthy among these interventions is valproic acid, a histone deacetylase inhibitor (HDACi) that modulates acetylation during NPC1 synthesis. In addition, various therapeutic options addressing protein folding modulation, such as abiraterone acetate, DHBP, calnexin, and arimoclomol, are examined. Additionally, treatments impeding NPC1 degradation, exemplified by bortezomib and MG132, are explored as potential strategies. This review consolidates current knowledge on proteostasis dysregulation in NPC and underscores the therapeutic landscape targeting diverse facets of this intricate process.
Collapse
Affiliation(s)
- Iris Valeria Servín Muñoz
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Daniel Ortuño-Sahagún
- Laboratorio de Neuroinmunobiología Molecular, Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Christian Griñán-Ferré
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain; (C.G.-F.); (M.P.)
- Centro de Investigación Biomédica en Red (CiberNed), Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28220 Madrid, Spain
| | - Mercè Pallàs
- Pharmacology Section, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Institute of Neuroscience, Universitat de Barcelona, 08028 Barcelona, Spain; (C.G.-F.); (M.P.)
- Centro de Investigación Biomédica en Red (CiberNed), Network Center for Neurodegenerative Diseases, National Spanish Health Institute Carlos III, 28220 Madrid, Spain
| | - Celia González-Castillo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Campus Guadalajara, Zapopan 45201, Mexico
| |
Collapse
|
4
|
A novel small molecule Hsp90 inhibitor, C-316-1, attenuates acute kidney injury by suppressing RIPK1-mediated inflammation and necroptosis. Int Immunopharmacol 2022; 108:108849. [PMID: 35588657 DOI: 10.1016/j.intimp.2022.108849] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/26/2022] [Accepted: 05/09/2022] [Indexed: 12/27/2022]
Abstract
Acute kidney injury (AKI) is marked by a fast deterioration of the kidney function that may be caused by a variety of factors. Recently, although our group found that PPBICA alleviated programmed cell death in AKI, poor water solubility limited its bioavailability. In this research, we screened a series of derivatives and found that C-316-1 had the best suppressive effect on preventing necroptosis and inflammation in cisplatin- and ischemia/reperfusion-induced AKI in vitro and in vivo with lower toxicity and better water solubility. Mass spectrometry results showed that C-316-1 bound to heat shock protein 90 (Hsp90), which was further confirmed by molecular docking and surface plasmon resonance. Additionally, the Hsp90 expression was upregulated in the blood and tissues of AKI patients. We discovered that C-316-1 decreased the RIPK1 protein level without affecting its mRNA expression. The proteasome inhibitor, MG132 restored the level of RIPK1 reduced by C-316-1, suggesting that C-316-1 limits necroptosis by promoting the degradation of RIPK1 rather than by reducing its production. Immunoprecipitation further showed that pretreatment with C-316-1 disrupted the Hsp90-Cdc37 protein-protein Interactions (PPIs). Thereby, C-316-1 inhibited the Hsp90-Cdc37 complex formation and led to a significant decrease in RIPK1, which in turn reduced necroptosis. Moreover, C-316-1 treatment did not protect against kidney injury in vivo and in vitro when Hsp90 was knocked down and R46, E47, and S50 in Cdc37 binding site of Hsp90 might form an important active pocket with C-316-1. These findings suggest that C-316-1 is a potential therapeutic agent against RIPK1-Mediated Necroptosis in AKI.
Collapse
|
5
|
Pipalia NH, Saad SZ, Subramanian K, Cross A, Al-Motawa A, Garg K, Blagg BSJ, Neckers L, Helquist P, Wiest O, Ory DS, Maxfield FR. HSP90 inhibitors reduce cholesterol storage in Niemann-Pick type C1 mutant fibroblasts. J Lipid Res 2021; 62:100114. [PMID: 34481829 PMCID: PMC8517605 DOI: 10.1016/j.jlr.2021.100114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/30/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
Niemann-Pick type C1 (NPC1) disease is a lysosomal lipid storage disorder caused by mutations of the NPC1 gene. More than 300 disease-associated mutations are reported in patients, resulting in abnormal accumulation of unesterified cholesterol, glycosphingolipids, and other lipids in late endosomes and lysosomes (LE/Ly) of many cell types. Previously, we showed that treatment of many different NPC1 mutant fibroblasts with histone deacetylase inhibitors resulted in reduction of cholesterol storage, and we found that this was associated with enhanced exit of the NPC1 protein from the endoplasmic reticulum and delivery to LE/Ly. This suggested that histone deacetylase inhibitors may work through changes in protein chaperones to enhance the folding of NPC1 mutants, allowing them to be delivered to LE/Ly. In this study, we evaluated the effect of several HSP90 inhibitors on NPC1I1061T skin fibroblasts. We found that HSP90 inhibition resulted in clearance of cholesterol from LE/Ly, and this was associated with enhanced delivery of the mutant NPC1I1061T protein to LE/Ly. We also observed that inhibition of HSP90 increased the expression of HSP70, and overexpression of HSP70 also reduced cholesterol storage in NPC1I1061T fibroblasts. However, we did not see correction of cholesterol storage by arimoclomol, a drug that is reported to increase HSP70 expression, at doses up to 0.5 mM. The increase in other chaperones as a consequence of HSP90 improves folding of NPC1 protein and relieves cholesterol accumulation in NPC1 mutant fibroblasts.
Collapse
Affiliation(s)
- Nina H Pipalia
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, USA
| | - Syed Z Saad
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, USA
| | - Kanagaraj Subramanian
- Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Abigail Cross
- Natural Sciences Department, Fordham University, New York, NY, USA
| | - Aisha Al-Motawa
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, USA
| | - Kunal Garg
- Department of Biochemistry, Weill Cornell Medical College, New York, NY, USA
| | - Brian S J Blagg
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN, USA
| | - Len Neckers
- Urologic Oncology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Paul Helquist
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN, USA
| | - Olaf Wiest
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN, USA
| | - Daniel S Ory
- Department of Internal Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | |
Collapse
|
6
|
Orth M, Albrecht V, Seidl K, Kinzel L, Unger K, Hess J, Kreutzer L, Sun N, Stegen B, Nieto A, Maas J, Winssinger N, Friedl AA, Walch AK, Belka C, Zitzelsberger H, Niyazi M, Lauber K. Inhibition of HSP90 as a Strategy to Radiosensitize Glioblastoma: Targeting the DNA Damage Response and Beyond. Front Oncol 2021; 11:612354. [PMID: 33816244 PMCID: PMC8011354 DOI: 10.3389/fonc.2021.612354] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/25/2021] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy is an essential component of multi-modality treatment of glioblastoma (GBM). However, treatment failure and recurrence are frequent and give rise to the dismal prognosis of this aggressive type of primary brain tumor. A high level of inherent treatment resistance is considered to be the major underlying reason, stemming from constantly activated DNA damage response (DDR) mechanisms as a consequence of oncogene overexpression, persistent replicative stress, and other so far unknown reasons. The molecular chaperone heat shock protein 90 (HSP90) plays an important role in the establishment and maintenance of treatment resistance, since it crucially assists the folding and stabilization of various DDR regulators. Accordingly, inhibition of HSP90 represents a multi-target strategy to interfere with DDR function and to sensitize cancer cells to radiotherapy. Using NW457, a pochoxime-based HSP90 inhibitor with favorable brain pharmacokinetic profile, we show here that HSP90 inhibition at low concentrations with per se limited cytotoxicity leads to downregulation of various DNA damage response factors on the protein level, distinct transcriptomic alterations, impaired DNA damage repair, and reduced clonogenic survival in response to ionizing irradiation in glioblastoma cells in vitro. In vivo, HSP90 inhibition by NW457 improved the therapeutic outcome of fractionated CBCT-based irradiation in an orthotopic, syngeneic GBM mouse model, both in terms of tumor progression and survival. Nevertheless, in view of the promising in vitro results the in vivo efficacy was not as strong as expected, although apart from the radiosensitizing effects HSP90 inhibition also reduced irradiation-induced GBM cell migration and tumor invasiveness. Hence, our findings identify the combination of HSP90 inhibition and radiotherapy in principle as a promising strategy for GBM treatment whose performance needs to be further optimized by improved inhibitor substances, better formulations and/or administration routes, and fine-tuned treatment sequences.
Collapse
Affiliation(s)
- Michael Orth
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Valerie Albrecht
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Karin Seidl
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Linda Kinzel
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Julia Hess
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Lisa Kreutzer
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Na Sun
- Research Unit Analytical Pathology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Benjamin Stegen
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium, Munich, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Alexander Nieto
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Jessica Maas
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Nicolas Winssinger
- Department of Organic Chemistry, NCCR Chemical Biology, University of Geneva, Geneva, Switzerland
| | - Anna A Friedl
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Axel K Walch
- Research Unit Analytical Pathology, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium, Munich, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Center Munich, Neuherberg, Germany
| | - Horst Zitzelsberger
- Research Unit Radiation Cytogenetics, Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Center Munich, Neuherberg, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium, Munich, Germany.,Clinical Cooperation Group Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Center Munich, Neuherberg, Germany
| |
Collapse
|
7
|
Klingl YE, Pakravan D, Van Den Bosch L. Opportunities for histone deacetylase inhibition in amyotrophic lateral sclerosis. Br J Pharmacol 2021; 178:1353-1372. [PMID: 32726472 PMCID: PMC9327724 DOI: 10.1111/bph.15217] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease. ALS patients suffer from a progressive loss of motor neurons, leading to respiratory failure within 3 to 5 years after diagnosis. Available therapies only slow down the disease progression moderately or extend the lifespan by a few months. Epigenetic hallmarks have been linked to the disease, creating an avenue for potential therapeutic approaches. Interference with one class of epigenetic enzymes, histone deacetylases, has been shown to affect neurodegeneration in many preclinical models. Consequently, it is crucial to improve our understanding about histone deacetylases and their inhibitors in (pre)clinical models of ALS. We conclude that selective inhibitors with high tolerability and safety and sufficient blood-brain barrier permeability will be needed to interfere with both epigenetic and non-epigenetic targets of these enzymes. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.6/issuetoc.
Collapse
Affiliation(s)
- Yvonne E. Klingl
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| | - Donya Pakravan
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI)KU Leuven‐University of LeuvenLeuvenBelgium
- Laboratory of NeurobiologyVIB, Center for Brain & Disease ResearchLeuvenBelgium
| |
Collapse
|
8
|
Chen H, Gong Y, Ma Y, Thompson RC, Wang J, Cheng Z, Xue L. A Brain-Penetrating Hsp90 Inhibitor NXD30001 Inhibits Glioblastoma as a Monotherapy or in Combination With Radiation. Front Pharmacol 2020; 11:974. [PMID: 32695001 PMCID: PMC7338553 DOI: 10.3389/fphar.2020.00974] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/15/2020] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a highly heterogeneous disease, which is initiated and sustained by various molecular alterations in an array of signal transduction pathways. Heat-shock protein 90 (Hsp90) is a molecular chaperone and is critically implicated in folding and activation of a diverse group of client proteins, many of which are key regulators for glioblastoma biology. We here assessed the anti-neoplastic efficacy of a novel brain-penetrating Hsp90 inhibitor NXD30001 as a monotherapy and combined with radiation in vitro and in vivo. Our results demonstrated that NXD30001 potently inhibited neurosphere formation, growth, and survival of CD133+ GBM cells with the half maximal inhibitory concentration at low nanomolar range, but CD133- GBM cells were less sensitive to NXD30001. NXD30001 also increased radio-sensitivity in glioblastoma stem cells (GSCs) at suboptimal concentrations. Moreover, NXD30001 dose-dependently decreased phosphorylation levels of multiple Hsp90 client proteins which play key roles in GBM, such as EGFR, Akt, c-Myc, and Notch1. In addition, NXD30001 could impair DNA damage response and endoplasmic reticulum stress response after radiotherapy by alteration of the related proteins expression. In a murine orthotopic model of human glioblastoma, NXD30001 marvelously induced tumor regression and extended median survival of tumor-bearing mice by approximately 20% when compared with the vehicle group (37 d vs 31 d, P<0.05). Radiotherapy solely increased median survival of tumor-bearing mice from 31 d to 38 d (P<0.05), while NXD30001 combined with radiation further extended survival to 43 d (P<0.05). We concluded that GSCs are more sensitive to NXD30001 than non-stem GBM cells, and NXD30001 in combination with radiation exerts better inhibitive effect in GBM progression than monotherapy.
Collapse
Affiliation(s)
- Hao Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Yuanying Gong
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Yufang Ma
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Reid C. Thompson
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jialiang Wang
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Zhixiang Cheng
- Department of Pain Management, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
- Department of Pain Management, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Lixia Xue
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
9
|
Kuta R, Larochelle N, Fernandez M, Pal A, Minotti S, Tibshirani M, St Louis K, Gentil BJ, Nalbantoglu JN, Hermann A, Durham HD. Depending on the stress, histone deacetylase inhibitors act as heat shock protein co-inducers in motor neurons and potentiate arimoclomol, exerting neuroprotection through multiple mechanisms in ALS models. Cell Stress Chaperones 2020; 25:173-191. [PMID: 31900865 PMCID: PMC6985055 DOI: 10.1007/s12192-019-01064-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 12/12/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
Upregulation of heat shock proteins (HSPs) is an approach to treatment of neurodegenerative disorders with impaired proteostasis. Many neurons, including motor neurons affected in amyotrophic lateral sclerosis (ALS), are relatively resistant to stress-induced upregulation of HSPs. This study demonstrated that histone deacetylase (HDAC) inhibitors enable the heat shock response in cultured spinal motor neurons, in a stress-dependent manner, and can improve the efficacy of HSP-inducing drugs in murine spinal cord cultures subjected to thermal or proteotoxic stress. The effect of particular HDAC inhibitors differed with the stress paradigm. The HDAC6 (class IIb) inhibitor, tubastatin A, acted as a co-inducer of Hsp70 (HSPA1A) expression with heat shock, but not with proteotoxic stress induced by expression of mutant SOD1 linked to familial ALS. Certain HDAC class I inhibitors (the pan inhibitor, SAHA, or the HDAC1/3 inhibitor, RGFP109) were HSP co-inducers comparable to the hydroxyamine arimoclomol in response to proteotoxic stress, but not thermal stress. Regardless, stress-induced Hsp70 expression could be enhanced by combining an HDAC inhibitor with either arimoclomol or with an HSP90 inhibitor that constitutively induced HSPs. HDAC inhibition failed to induce Hsp70 in motor neurons expressing ALS-linked mutant FUS, in which the heat shock response was suppressed; yet SAHA, RGFP109, and arimoclomol did reduce loss of nuclear FUS, a disease hallmark, and HDAC inhibition rescued the DNA repair response in iPSC-derived motor neurons carrying the FUSP525Lmutation, pointing to multiple mechanisms of neuroprotection by both HDAC inhibiting drugs and arimoclomol.
Collapse
Affiliation(s)
- Rachel Kuta
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Nancy Larochelle
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Mario Fernandez
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Arun Pal
- Department Neurology, Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Sandra Minotti
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Michael Tibshirani
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Kyle St Louis
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Benoit J Gentil
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Josephine N Nalbantoglu
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada
| | - Andreas Hermann
- Translational Neurodegeneration Section "Albrecht-Kossel", Department of Neurology and Center for Transdisciplinary Neuroscience (CTNR), University Medical Center Rostock, University of Rostock, Rostock, Germany and German Center for Neurodegenerative Diseases (DZNE) Rostock, Rostock, Germany
| | - Heather D Durham
- Department of Neurology and Neurosurgery and Montreal Neurological Institute, McGill University, 3801 University St., Montreal, QC, H3A 2B4, Canada.
| |
Collapse
|
10
|
Tau Protein Squired by Molecular Chaperones During Alzheimer’s Disease. J Mol Neurosci 2018; 66:356-368. [DOI: 10.1007/s12031-018-1174-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 09/14/2018] [Indexed: 01/19/2023]
|
11
|
San Gil R, Ooi L, Yerbury JJ, Ecroyd H. The heat shock response in neurons and astroglia and its role in neurodegenerative diseases. Mol Neurodegener 2017; 12:65. [PMID: 28923065 PMCID: PMC5604514 DOI: 10.1186/s13024-017-0208-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 09/01/2017] [Indexed: 12/13/2022] Open
Abstract
Protein inclusions are a predominant molecular pathology found in numerous neurodegenerative diseases, including amyotrophic lateral sclerosis and Huntington's disease. Protein inclusions form in discrete areas of the brain characteristic to the type of neurodegenerative disease, and coincide with the death of neurons in that region (e.g. spinal cord motor neurons in amyotrophic lateral sclerosis). This suggests that the process of protein misfolding leading to inclusion formation is neurotoxic, and that cell-autonomous and non-cell autonomous mechanisms that maintain protein homeostasis (proteostasis) can, at times, be insufficient to prevent protein inclusion formation in the central nervous system. The heat shock response is a pro-survival pathway induced under conditions of cellular stress that acts to maintain proteostasis through the up-regulation of heat shock proteins, a superfamily of molecular chaperones, other co-chaperones and mitotic regulators. The kinetics and magnitude of the heat shock response varies in a stress- and cell-type dependent manner. It remains to be determined if and/or how the heat shock response is activated in the different cell-types that comprise the central nervous system (e.g. neurons and astroglia) in response to protein misfolding events that precede cellular dysfunctions in neurodegenerative diseases. This is particularly relevant considering emerging evidence demonstrating the non-cell autonomous nature of amyotrophic lateral sclerosis and Huntington's disease (and other neurodegenerative diseases) and the destructive role of astroglia in disease progression. This review highlights the complexity of heat shock response activation and addresses whether neurons and glia sense and respond to protein misfolding and aggregation associated with neurodegenerative diseases, in particular Huntington's disease and amyotrophic lateral sclerosis, by inducing a pro-survival heat shock response.
Collapse
Affiliation(s)
- Rebecca San Gil
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Northfields Ave, Wollongong, 2522 Australia
| | - Lezanne Ooi
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Northfields Ave, Wollongong, 2522 Australia
| | - Justin J. Yerbury
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Northfields Ave, Wollongong, 2522 Australia
| | - Heath Ecroyd
- Illawarra Health and Medical Research Institute and School of Biological Sciences, University of Wollongong, Northfields Ave, Wollongong, 2522 Australia
| |
Collapse
|
12
|
Wang P, Wander CM, Yuan CX, Bereman MS, Cohen TJ. Acetylation-induced TDP-43 pathology is suppressed by an HSF1-dependent chaperone program. Nat Commun 2017; 8:82. [PMID: 28724966 PMCID: PMC5517419 DOI: 10.1038/s41467-017-00088-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 05/31/2017] [Indexed: 12/12/2022] Open
Abstract
TDP-43 pathology marks a spectrum of multisystem proteinopathies including amyotrophic lateral sclerosis, frontotemporal lobar degeneration, and sporadic inclusion body myositis. Surprisingly, it has been challenging to recapitulate this pathology, highlighting an incomplete understanding of TDP-43 regulatory mechanisms. Here we provide evidence supporting TDP-43 acetylation as a trigger for disease pathology. Using cultured cells and mouse skeletal muscle, we show that TDP-43 acetylation-mimics promote TDP-43 phosphorylation and ubiquitination, perturb mitochondria, and initiate degenerative inflammatory responses that resemble sporadic inclusion body myositis pathology. Analysis of functionally linked amyotrophic lateral sclerosis proteins revealed recruitment of p62, ubiquilin-2, and optineurin to TDP-43 aggregates. We demonstrate that TDP-43 acetylation-mimic pathology is potently suppressed by an HSF1-dependent mechanism that disaggregates TDP-43. Our study illustrates bidirectional TDP-43 processing in which TDP-43 aggregation is targeted by a coordinated chaperone response. Thus, activation or restoration of refolding mechanisms may alleviate TDP-43 aggregation in tissues that are uniquely susceptible to TDP-43 proteinopathies.TDP-43 aggregation is linked to various diseases including amyotrophic lateral sclerosis. Here the authors show that acetylation of the protein triggers TDP-43 pathology in cultured cells and mouse skeletal muscle, which can be cleared through an HSF1-dependent chaperone mechanism that disaggregates the protein.
Collapse
Affiliation(s)
- Ping Wang
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Connor M Wander
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| | | | - Michael S Bereman
- Department of Biological Sciences and Department of Chemistry, Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, 27695, USA
| | - Todd J Cohen
- Department of Neurology, UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
13
|
Wang B, Liu Y, Huang L, Chen J, Li JJ, Wang R, Kim E, Justicia C, Sakata K, Chen H, Planas A, Ostrom RS, Li W, Yang G, McDonald MP, Chen R, Heck D, Liao FF, Liao FF. A CNS-permeable Hsp90 inhibitor rescues synaptic dysfunction and memory loss in APP-overexpressing Alzheimer's mouse model via an HSF1-mediated mechanism. Mol Psychiatry 2017; 22:990-1001. [PMID: 27457810 PMCID: PMC5323357 DOI: 10.1038/mp.2016.104] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 03/27/2016] [Accepted: 04/20/2016] [Indexed: 11/09/2022]
Abstract
Induction of neuroprotective heat-shock proteins via pharmacological Hsp90 inhibitors is currently being investigated as a potential treatment for neurodegenerative diseases. Two major hurdles for therapeutic use of Hsp90 inhibitors are systemic toxicity and limited central nervous system permeability. We demonstrate here that chronic treatment with a proprietary Hsp90 inhibitor compound (OS47720) not only elicits a heat-shock-like response but also offers synaptic protection in symptomatic Tg2576 mice, a model of Alzheimer's disease, without noticeable systemic toxicity. Despite a short half-life of OS47720 in mouse brain, a single intraperitoneal injection induces rapid and long-lasting (>3 days) nuclear activation of the heat-shock factor, HSF1. Mechanistic study indicates that the remedial effects of OS47720 depend upon HSF1 activation and the subsequent HSF1-mediated transcriptional events on synaptic genes. Taken together, this work reveals a novel role of HSF1 in synaptic function and memory, which likely occurs through modulation of the synaptic transcriptome.
Collapse
Affiliation(s)
- Bin Wang
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Yu Liu
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Lianyan Huang
- Department of Anesthesiology, New York University School of Medicine, New York, NY 10016
| | - Jianjun Chen
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Jing jing Li
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Ruishan Wang
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Eunhee Kim
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Carles Justicia
- Department of Brain Ischemia and Neurodegeneration, Institute for Biomedical Research (IIBB-CSIC), Rossello 161, planta 6, 08036-Barcelona, Spain
| | - Kazuko Sakata
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Hao Chen
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Anna Planas
- Department of Brain Ischemia and Neurodegeneration, Institute for Biomedical Research (IIBB-CSIC), Rossello 161, planta 6, 08036-Barcelona, Spain
| | - Rennolds S Ostrom
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Guang Yang
- Department of Anesthesiology, New York University School of Medicine, New York, NY 10016
| | - Michael P. McDonald
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163,Department of Neurology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Ruihong Chen
- Oncosynergy, Inc; 409 Illinois St., San Francisco, CA, 94158
| | - Detlef Heck
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | - Francesca-Fang Liao
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163,Correspondence should be addressed to Francesca-Fang Liao, Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163.
| | - F-F Liao
- Department of Pharmacology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| |
Collapse
|
14
|
Geldanamycin-inspired compounds induce direct trans-differentiation of human mesenchymal stem cells to neurons. Eur J Med Chem 2017; 135:110-116. [DOI: 10.1016/j.ejmech.2017.03.082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/16/2017] [Accepted: 03/31/2017] [Indexed: 01/01/2023]
|
15
|
Ding Y, Adachi H, Katsuno M, Sahashi K, Kondo N, Iida M, Tohnai G, Nakatsuji H, Sobue G. BIIB021, a synthetic Hsp90 inhibitor, induces mutant ataxin-1 degradation through the activation of heat shock factor 1. Neuroscience 2016; 327:20-31. [PMID: 27058144 DOI: 10.1016/j.neuroscience.2016.03.064] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/14/2016] [Accepted: 03/30/2016] [Indexed: 12/20/2022]
Abstract
Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disease caused by the expansion of a polyglutamine (polyQ) tract in ataxin-1 (ATXN1). The pathological hallmarks of SCA1 are the loss of cerebellar Purkinje cells and neurons in the brainstem and the presence of nuclear aggregates containing the polyQ-expanded ATXN1 protein. Heat shock protein 90 (Hsp90) inhibitors have been shown to reduce polyQ-induced toxicity. This study was designed to examine the therapeutic effects of BIIB021, a purine-scaffold Hsp90 inhibitor, on the protein homeostasis of polyQ-expanded mutant ATXN1 in a cell culture model of SCA1. Our results demonstrated that BIIB021 activated heat shock factor 1 (HSF1) and suppressed the abnormal accumulation of ATXN1 and its toxicity. The pharmacological degradation of mutant ATXN1 via activated HSF1 was dependent on both the proteasome and autophagy systems. These findings indicate that HSF1 is a key molecule in the regulation of the protein homeostasis of the polyQ-expanded mutant ATXN1 and that Hsp90 has potential as a novel therapeutic target in patients with SCA1.
Collapse
Affiliation(s)
- Ying Ding
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hiroaki Adachi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Department of Neurology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807-8555, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Naohide Kondo
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Madoka Iida
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Genki Tohnai
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Hideaki Nakatsuji
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
16
|
Thirstrup K, Sotty F, Montezinho LCP, Badolo L, Thougaard A, Kristjánsson M, Jensen T, Watson S, Nielsen SM. Linking HSP90 target occupancy to HSP70 induction and efficacy in mouse brain. Pharmacol Res 2016; 104:197-205. [DOI: 10.1016/j.phrs.2015.12.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 12/22/2015] [Accepted: 12/22/2015] [Indexed: 12/31/2022]
|
17
|
Shabbir A, Bianchetti E, Cargonja R, Petrovic A, Mladinic M, Pilipović K, Nistri A. Role of HSP70 in motoneuron survival after excitotoxic stress in a rat spinal cord injury modelin vitro. Eur J Neurosci 2015; 42:3054-65. [DOI: 10.1111/ejn.13108] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 01/06/2023]
Affiliation(s)
- Ayisha Shabbir
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Elena Bianchetti
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Renato Cargonja
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Antonela Petrovic
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Miranda Mladinic
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
- Department of Biotechnology; University of Rijeka; Rijeka Croatia
| | - Kristina Pilipović
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| | - Andrea Nistri
- Neuroscience Department; Scuola Internazionale Superiore di Studi Avanzati (SISSA); Via Bonomea 265 34136 Trieste Italy
| |
Collapse
|
18
|
Sun X, Crawford R, Liu C, Luo T, Hu B. Development-dependent regulation of molecular chaperones after hypoxia-ischemia. Neurobiol Dis 2015; 82:123-131. [PMID: 26070787 DOI: 10.1016/j.nbd.2015.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/05/2015] [Accepted: 06/03/2015] [Indexed: 02/08/2023] Open
Abstract
Cellular stress response after hypoxia-Ischemia (HI) may be substantially different between immature and mature brains. To study this phenomenon, postnatal day 7 (P7) and P26 rats were subjected to HI followed by different periods of recovery. Nuclear accumulation of heat-shock transcription factor-1 (HSF1) and expression of molecular chaperone proteins and mRNAs were analyzed by in situ hybridization, Western blotting and confocal microscopy. Nuclear accumulation of HSF1 protein and induction of hsp70 mRNA occurred dramatically in P26 neurons, but minimally in P7 neurons and moderately in microglial cells after HI. Consistently, the level of HSF1 was significantly higher in P26 brain samples, compared with that in P7 brain. Translation of hsp70 mRNA into proteins in P26 mature neurons was seen at 4h and peaked at 24h, when some neurons had already died after HI. Induction of ER glucose-regulated protein-78 (grp78) and mitochondrial hsp60 mRNAs and proteins was moderate and occurred also only in P26 mature brain after HI. These results suggest that the cellular stress response after HI is development-dependent, being pronounced in mature but virtually negligible in neonatal neurons. Therefore, the effectiveness of therapeutic strategies targeting the stress pathway against HI may be significantly different between immature and mature brains. The delayed induction of molecular chaperones in mature brain may be somewhat late for protecting HI neurons from acute HI injury.
Collapse
Affiliation(s)
- Xin Sun
- Shock Trauma and Anesthesiology Research Center, University of MD School of Medicine, USA; Department of Neurology, The First Teaching Hospital, Jilin University, China
| | - Robert Crawford
- Shock Trauma and Anesthesiology Research Center, University of MD School of Medicine, USA
| | - Chunli Liu
- Shock Trauma and Anesthesiology Research Center, University of MD School of Medicine, USA
| | - Tianfei Luo
- Shock Trauma and Anesthesiology Research Center, University of MD School of Medicine, USA
| | - Bingren Hu
- Shock Trauma and Anesthesiology Research Center, University of MD School of Medicine, USA.
| |
Collapse
|
19
|
Smith HL, Li W, Cheetham ME. Molecular chaperones and neuronal proteostasis. Semin Cell Dev Biol 2015; 40:142-52. [PMID: 25770416 PMCID: PMC4471145 DOI: 10.1016/j.semcdb.2015.03.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 02/27/2015] [Accepted: 03/04/2015] [Indexed: 12/13/2022]
Abstract
Protein homeostasis (proteostasis) is essential for maintaining the functionality of the proteome. The disruption of proteostasis, due to genetic mutations or an age-related decline, leads to aberrantly folded proteins that typically lose their function. The accumulation of misfolded and aggregated protein is also cytotoxic and has been implicated in the pathogenesis of neurodegenerative diseases. Neurons have developed an intrinsic protein quality control network, of which molecular chaperones are an essential component. Molecular chaperones function to promote efficient folding and target misfolded proteins for refolding or degradation. Increasing molecular chaperone expression can suppress protein aggregation and toxicity in numerous models of neurodegenerative disease; therefore, molecular chaperones are considered exciting therapeutic targets. Furthermore, mutations in several chaperones cause inherited neurodegenerative diseases. In this review, we focus on the importance of molecular chaperones in neurodegenerative diseases, and discuss the advances in understanding their protective mechanisms.
Collapse
Affiliation(s)
- Heather L Smith
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Wenwen Li
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | |
Collapse
|
20
|
Smith DA, Carland CR, Guo Y, Bernstein SI. Getting folded: chaperone proteins in muscle development, maintenance and disease. Anat Rec (Hoboken) 2014; 297:1637-1649. [PMID: 25125177 PMCID: PMC4135391 DOI: 10.1002/ar.22980] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/11/2014] [Accepted: 04/12/2014] [Indexed: 09/26/2024]
Abstract
Chaperone proteins are critical for protein folding and stability, and hence are necessary for normal cellular organization and function. Recent studies have begun to interrogate the role of this specialized class of proteins in muscle biology. During development, chaperone-mediated folding of client proteins enables their integration into nascent functional sarcomeres. In addition to assisting with muscle differentiation, chaperones play a key role in the maintenance of muscle tissues. Furthermore, disruption of the chaperone network can result in neuromuscular disease. In this review, we discuss how chaperones are involved in myofibrillogenesis, sarcomere maintenance, and muscle disorders. We also consider the possibilities of therapeutically targeting chaperones to treat muscle disease.
Collapse
Affiliation(s)
- Daniel A. Smith
- Department of Biology and the Molecular Biology Institute, San Diego State
University, San Diego, CA 92182, USA
| | - Carmen R. Carland
- Department of Biology and the Molecular Biology Institute, San Diego State
University, San Diego, CA 92182, USA
| | - Yiming Guo
- Department of Biology and the Molecular Biology Institute, San Diego State
University, San Diego, CA 92182, USA
| | - Sanford I. Bernstein
- Department of Biology and the Molecular Biology Institute, San Diego State
University, San Diego, CA 92182, USA
| |
Collapse
|
21
|
Bhat R, Tummalapalli SR, Rotella DP. Progress in the discovery and development of heat shock protein 90 (Hsp90) inhibitors. J Med Chem 2014; 57:8718-28. [PMID: 25141341 DOI: 10.1021/jm500823a] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The discovery and clinical development of heat shock protein 90 (Hsp90) inhibitors continue to progress. A number of Hsp90 inhibitors are in clinical trials, and preclinical discoveries of new chemotypes that bind to distinct regions in the protein as well as isoform selective compounds are active areas of research. This review will highlight progress in the field since 2010.
Collapse
Affiliation(s)
- Rohit Bhat
- Department of Chemistry and Biochemistry, Sokol Institute for Pharmaceutical Life Sciences, Montclair State University , Montclair, New Jersey 07043, United States
| | | | | |
Collapse
|