1
|
Li J, Arnold J, Sima M, Al Faruque H, Galang J, Hu-Lieskovan S, Kopeček J, Yang J. Combination of multivalent DR5 receptor clustering agonists and histone deacetylase inhibitors for treatment of colon cancer. J Control Release 2024; 376:1014-1024. [PMID: 39489464 DOI: 10.1016/j.jconrel.2024.10.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/19/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
Death Receptor 5 (DR5) targeted therapies offer significant promise due to their pivotal role in mediating the extrinsic pathway of apoptosis. Despite DR5 overexpression in various malignancies and the potential of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), clinical applications of anti-DR5 monoclonal antibodies (mAbs) have been hampered by suboptimal outcomes potentially due to lack of receptor clustering. To address the limitation, we developed N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer-based conjugates integrating multiple copies of DR5-targeting peptide (cyclic WDCLDNRIGRRQCVKL; cDR5) to enhance receptor clustering and apoptosis. Three conjugates with variable number of cDR5 were prepared and denoted as PH-cDR5 (high valence), PM-cDR5 (medium valence) and PL-cDR5 (low valence). Our studies in TRAIL-sensitive and resistant cancer cell lines demonstrated that the HPMA copolymer-peptide conjugates (P-cDR5) significantly improved DR5 receptor clustering and induced apoptosis effectively. In TRAIL-sensitive colon cancer cells (COLO205, HCT-116), P-cDR5 showed efficacy comparable to anti-DR5 mAb Drozitumab (DRO), but P-cDR5 outperformed DRO in TRAIL-resistant cells (HT-29), highlighting the importance of efficient receptor clustering. In COLO205 cells PM-cDR5 exhibited an IC50 of 94 pM, while PH-cDR5 had an even lower IC50 of 15 pM (based on cDR5 equivalent concentration), indicating enhanced potency of the multivalent HPMA copolymer-based system with a flexible polymer backbone in comparison with the IC50 for TRAIL at 0.12 nM. Combining P-cDR5 with valproic acid, a histone deacetylase inhibitor, resulted in further enhancement of apoptosis inducing efficacy, along with destabilizing mitochondrial membranes and increased sensitivity of TRAIL-resistant cells. These findings suggest that attaching multiple cDR5 peptides to a flexible water-soluble polymer carrier not only overcomes the limitations of previous designs but also offers a promising avenue for treating resistant cancers, pointing toward the need for further preclinical exploration and validation of this innovative strategy.
Collapse
Affiliation(s)
- Jiahui Li
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jaden Arnold
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA
| | - Monika Sima
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Hasan Al Faruque
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jacob Galang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Sophia Hu-Lieskovan
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
2
|
Cunha-Garcia D, Monteiro-Fernandes D, Correia JS, Neves-Carvalho A, Vilaça-Ferreira AC, Guerra-Gomes S, Viana JF, Oliveira JF, Teixeira-Castro A, Maciel P, Duarte-Silva S. Genetic Ablation of Inositol 1,4,5-Trisphosphate Receptor Type 2 (IP 3R2) Fails to Modify Disease Progression in a Mouse Model of Spinocerebellar Ataxia Type 3. Int J Mol Sci 2023; 24:10606. [PMID: 37445783 PMCID: PMC10341520 DOI: 10.3390/ijms241310606] [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: 05/15/2023] [Revised: 06/07/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is a rare neurodegenerative disease caused by an abnormal polyglutamine expansion within the ataxin-3 protein (ATXN3). This leads to neurodegeneration of specific brain and spinal cord regions, resulting in a progressive loss of motor function. Despite neuronal death, non-neuronal cells, including astrocytes, are also involved in SCA3 pathogenesis. Astrogliosis is a common pathological feature in SCA3 patients and animal models of the disease. However, the contribution of astrocytes to SCA3 is not clearly defined. Inositol 1,4,5-trisphosphate receptor type 2 (IP3R2) is the predominant IP3R in mediating astrocyte somatic calcium signals, and genetically ablation of IP3R2 has been widely used to study astrocyte function. Here, we aimed to investigate the relevance of IP3R2 in the onset and progression of SCA3. For this, we tested whether IP3R2 depletion and the consecutive suppression of global astrocytic calcium signalling would lead to marked changes in the behavioral phenotype of a SCA3 mouse model, the CMVMJD135 transgenic line. This was achieved by crossing IP3R2 null mice with the CMVMJD135 mouse model and performing a longitudinal behavioral characterization of these mice using well-established motor-related function tests. Our results demonstrate that IP3R2 deletion in astrocytes does not modify SCA3 progression.
Collapse
Affiliation(s)
- Daniela Cunha-Garcia
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Daniela Monteiro-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Joana Sofia Correia
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Andreia Neves-Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Ana Catarina Vilaça-Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Sónia Guerra-Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - João Filipe Viana
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - João Filipe Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
- IPCA-EST-2Ai, Polytechnic Institute of Cávado and Ave, Applied Artificial Intelligence Laboratory, Campus of IPCA, 4750-810 Barcelos, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (D.C.-G.); (D.M.-F.); (J.S.C.); (A.N.-C.); (A.C.V.-F.); (S.G.-G.); (J.F.V.); (J.F.O.); (A.T.-C.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/4805-017 Guimarães, Portugal
| |
Collapse
|
3
|
Mayoral-Palarz K, Neves-Carvalho A, Duarte-Silva S, Monteiro-Fernandes D, Maciel P, Khodakhah K. Cerebellar neuronal dysfunction accompanies early motor symptoms in spinocerebellar ataxia type 3. Dis Model Mech 2022; 15:275597. [PMID: 35660856 PMCID: PMC9367011 DOI: 10.1242/dmm.049514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is an adult-onset, progressive ataxia. SCA3 presents with ataxia before any gross neuropathology. A feature of many cerebellar ataxias is aberrant cerebellar output that contributes to motor dysfunction. We examined whether abnormal cerebellar output was present in the CMVMJD135 SCA3 mouse model and, if so, whether it correlated with the disease onset and progression. In vivo recordings showed that the activity of deep cerebellar nuclei neurons, the main output of the cerebellum, was altered. The aberrant activity correlated with the onset of ataxia. However, although the severity of ataxia increased with age, the severity of the aberrant cerebellar output was not progressive. The abnormal cerebellar output, however, was accompanied by non-progressive abnormal activity of their upstream synaptic inputs, the Purkinje cells. In vitro recordings indicated that alterations in intrinsic Purkinje cell pacemaking and in their synaptic inputs contributed to abnormal Purkinje cell activity. These findings implicate abnormal cerebellar physiology as an early, consistent contributor to pathophysiology in SCA3, and suggest that the aberrant cerebellar output could be an appropriate therapeutic target in SCA3. Summary: In a mouse model of spinocerebellar ataxia type 3 (SCA3), aberrant cerebellar physiology is apparent early in disease, prior to cerebellar neuronal pathology. Aberrant cerebellar output could be a therapeutic target in SCA3.
Collapse
Affiliation(s)
- Kristin Mayoral-Palarz
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andreia Neves-Carvalho
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Daniela Monteiro-Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Kamran Khodakhah
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
4
|
Costa MD, Maciel P. Modifier pathways in polyglutamine (PolyQ) diseases: from genetic screens to drug targets. Cell Mol Life Sci 2022; 79:274. [PMID: 35503478 PMCID: PMC11071829 DOI: 10.1007/s00018-022-04280-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/14/2022] [Accepted: 03/30/2022] [Indexed: 12/17/2022]
Abstract
Polyglutamine (PolyQ) diseases include a group of inherited neurodegenerative disorders caused by unstable expansions of CAG trinucleotide repeats in the coding region of specific genes. Such genetic alterations produce abnormal proteins containing an unusually long PolyQ tract that renders them more prone to aggregate and cause toxicity. Although research in the field in the last years has contributed significantly to the knowledge of the biological mechanisms implicated in these diseases, effective treatments are still lacking. In this review, we revisit work performed in models of PolyQ diseases, namely the yeast Saccharomyces cerevisiae, the nematode worm Caenorhabditis elegans and the fruit fly Drosophila melanogaster, and provide a critical overview of the high-throughput unbiased genetic screens that have been performed using these systems to identify novel genetic modifiers of PolyQ diseases. These approaches have revealed a wide variety of cellular processes that modulate the toxicity and aggregation of mutant PolyQ proteins, reflecting the complexity of these disorders and demonstrating how challenging the development of therapeutic strategies can be. In addition to the unbiased large-scale genetic screenings in non-vertebrate models, complementary studies in mammalian systems, closer to humans, have contributed with novel genetic modifiers of PolyQ diseases, revealing neuronal function and inflammation as key disease modulators. A pathway enrichment analysis, using the human orthologues of genetic modifiers of PolyQ diseases clustered modifier genes into major themes translatable to the human disease context, such as protein folding and transport as well as transcription regulation. Innovative genetic strategies of genetic manipulation, together with significant advances in genomics and bioinformatics, are taking modifier genetic studies to more realistic disease contexts. The characterization of PolyQ disease modifier pathways is of extreme relevance to reveal novel therapeutic possibilities to delay disease onset and progression in patients.
Collapse
Affiliation(s)
- Marta Daniela Costa
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, 4710-057, Braga, Portugal
- ICVS/3Bs-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Maciel
- School of Medicine, Life and Health Sciences Research Institute (ICVS), University of Minho, 4710-057, Braga, Portugal.
- ICVS/3Bs-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
5
|
Watchon M, Luu L, Robinson KJ, Yuan KC, De Luca A, Suddull HJ, Tym MC, Guillemin GJ, Cole NJ, Nicholson GA, Chung RS, Lee A, Laird AS. Sodium valproate increases activity of the sirtuin pathway resulting in beneficial effects for spinocerebellar ataxia-3 in vivo. Mol Brain 2021; 14:128. [PMID: 34416891 PMCID: PMC8377983 DOI: 10.1186/s13041-021-00839-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Machado-Joseph disease (MJD, also known as spinocerebellar ataxia type 3) is a fatal neurodegenerative disease that impairs control and coordination of movement. Here we tested whether treatment with the histone deacetylase inhibitor sodium valproate (valproate) prevented a movement phenotype that develops in larvae of a transgenic zebrafish model of the disease. We found that treatment with valproate improved the swimming of the MJD zebrafish, affected levels of acetylated histones 3 and 4, but also increased expression of polyglutamine expanded human ataxin-3. Proteomic analysis of protein lysates generated from the treated and untreated MJD zebrafish also predicted that valproate treatment had activated the sirtuin longevity signaling pathway and this was confirmed by findings of increased SIRT1 protein levels and sirtuin activity in valproate treated MJD zebrafish and HEK293 cells expressing ataxin-3 84Q, respectively. Treatment with resveratrol (another compound known to activate the sirtuin pathway), also improved swimming in the MJD zebrafish. Co-treatment with valproate alongside EX527, a SIRT1 activity inhibitor, prevented induction of autophagy by valproate and the beneficial effects of valproate on the movement in the MJD zebrafish, supporting that they were both dependent on sirtuin activity. These findings provide the first evidence of sodium valproate inducing activation of the sirtuin pathway. Further, they indicate that drugs that target the sirtuin pathway, including sodium valproate and resveratrol, warrant further investigation for the treatment of MJD and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Maxinne Watchon
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Luan Luu
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Katherine J Robinson
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Kristy C Yuan
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Alana De Luca
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Hannah J Suddull
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Madelaine C Tym
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Gilles J Guillemin
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Nicholas J Cole
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Garth A Nicholson
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia.,ANZAC Research Institute, Concord Repatriation Hospital, Concord, NSW, Australia
| | - Roger S Chung
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Albert Lee
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia
| | - Angela S Laird
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 2 Technology Place, Sydney, NSW 2109, Australia.
| |
Collapse
|
6
|
Raj K, Akundi RS. Mutant Ataxin-3-Containing Aggregates (MATAGGs) in Spinocerebellar Ataxia Type 3: Dynamics of the Disorder. Mol Neurobiol 2021; 58:3095-3118. [PMID: 33629274 DOI: 10.1007/s12035-021-02314-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/25/2021] [Indexed: 11/25/2022]
Abstract
Spinocerebellar ataxia type 3 (SCA3) is the most common type of SCA worldwide caused by abnormal polyglutamine expansion in the coding region of the ataxin-3 gene. Ataxin-3 is a multi-faceted protein involved in various cellular processes such as deubiquitination, cytoskeletal organisation, and transcriptional regulation. The presence of an expanded poly(Q) stretch leads to altered processing and misfolding of the protein culminating in the production of insoluble protein aggregates in the cell. Various post-translational modifications affect ataxin-3 fibrillation and aggregation. This review provides an exhaustive assessment of the various pathogenic mechanisms undertaken by the mutant ataxin-3-containing aggregates (MATAGGs) for disease induction and neurodegeneration. This includes in-depth discussion on MATAGG dynamics including their formation, role in neuronal pathogenesis, and the debate over the toxic v/s protective nature of the MATAGGs in disease progression. Additionally, the currently available therapeutic strategies against SCA3 have been reviewed. The shift in the focus of such strategies, from targeting the steps that lead to or reduce aggregate formation to targeting the expression of mutant ataxin-3 itself via RNA-based therapeutics, has also been presented. We also discuss the intriguing promise that various growth and neurotrophic factors, especially the insulin pathway, hold in the modulation of SCA3 progression. These emerging areas show the newer directions through which SCA3 can be targeted including various preclinical and clinical trials. All these advances made in the last three decades since the discovery of the ataxin-3 gene have been critically reviewed here.
Collapse
Affiliation(s)
- Kritika Raj
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, Chanakyapuri, New Delhi, 110021, India
| | - Ravi Shankar Akundi
- Neuroinflammation Research Lab, Faculty of Life Sciences and Biotechnology, South Asian University, Chanakyapuri, New Delhi, 110021, India.
| |
Collapse
|
7
|
Barros I, Marcelo A, Silva TP, Barata J, Rufino-Ramos D, Pereira de Almeida L, Miranda CO. Mesenchymal Stromal Cells' Therapy for Polyglutamine Disorders: Where Do We Stand and Where Should We Go? Front Cell Neurosci 2020; 14:584277. [PMID: 33132851 PMCID: PMC7573388 DOI: 10.3389/fncel.2020.584277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/03/2020] [Indexed: 12/16/2022] Open
Abstract
Polyglutamine (polyQ) diseases are a group of inherited neurodegenerative disorders caused by the expansion of the cytosine-adenine-guanine (CAG) repeat. This mutation encodes extended glutamine (Q) tract in the disease protein, resulting in the alteration of its conformation/physiological role and in the formation of toxic fragments/aggregates of the protein. This group of heterogeneous disorders shares common molecular mechanisms, which opens the possibility to develop a pan therapeutic approach. Vast efforts have been made to develop strategies to alleviate disease symptoms. Nonetheless, there is still no therapy that can cure or effectively delay disease progression of any of these disorders. Mesenchymal stromal cells (MSC) are promising tools for the treatment of polyQ disorders, promoting protection, tissue regeneration, and/or modulation of the immune system in animal models. Accordingly, data collected from clinical trials have so far demonstrated that transplantation of MSC is safe and delays the progression of some polyQ disorders for some time. However, to achieve sustained phenotypic amelioration in clinics, several treatments may be necessary. Therefore, efforts to develop new strategies to improve MSC's therapeutic outcomes have been emerging. In this review article, we discuss the current treatments and strategies used to reduce polyQ symptoms and major pre-clinical and clinical achievements obtained with MSC transplantation as well as remaining flaws that need to be overcome. The requirement to cross the blood-brain-barrier (BBB), together with a short rate of cell engraftment in the lesioned area and low survival of MSC in a pathophysiological context upon transplantation may contribute to the transient therapeutic effects. We also review methods like pre-conditioning or genetic engineering of MSC that can be used to increase MSC survival in vivo, cellular-free approaches-i.e., MSC-conditioned medium (CM) or MSC-derived extracellular vesicles (EVs) as a way of possibly replacing the use of MSC and methods required to standardize the potential of MSC/MSC-derived products. These are fundamental questions that need to be addressed to obtain maximum MSC performance in polyQ diseases and therefore increase clinical benefits.
Collapse
Affiliation(s)
- Inês Barros
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,III-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Adriana Marcelo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Teresa P Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - João Barata
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - David Rufino-Ramos
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,Viravector-Viral Vector for Gene Transfer Core Facility, University of Coimbra, Coimbra, Portugal
| | - Catarina O Miranda
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,III-Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
8
|
Cho DS, Schmitt RE, Dasgupta A, Ducharme AM, Doles JD. Single-cell deconstruction of post-sepsis skeletal muscle and adipose tissue microenvironments. J Cachexia Sarcopenia Muscle 2020; 11:1351-1363. [PMID: 32643301 PMCID: PMC7567136 DOI: 10.1002/jcsm.12596] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 04/22/2020] [Accepted: 05/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Persistent loss of skeletal muscle mass and function as well as altered fat metabolism are frequently observed in severe sepsis survivors. Studies examining sepsis-associated tissue dysfunction from the perspective of the tissue microenvironment are scarce. In this study, we comprehensively assessed transcriptional changes in muscle and fat at single-cell resolution following experimental sepsis induction. METHODS Skeletal muscle and visceral white adipose tissue from control mice or mice 1 day or 1 month following faecal slurry-induced sepsis were used. Single cells were mechanically and enzymatically prepared from whole tissue, and viable cells were further isolated by fluorescence activated cell sorting. Droplet-based single-cell RNA-sequencing (scRNA-seq; 10× Genomics) was used to generate single-cell gene expression profiles of thousands of muscle and fat-resident cells. Bioinformatics analyses were performed to identify and compare individual cell populations in both tissues. RESULTS In skeletal muscle, scRNA-seq analysis classified 1438 single cells into myocytes, endothelial cells, fibroblasts, mesenchymal stem cells, macrophages, neutrophils, T-cells, B-cells, and dendritic cells. In adipose tissue, scRNA-seq analysis classified 2281 single cells into adipose stem cells, preadipocytes, endothelial cells, fibroblasts, macrophages, dendritic cells, B-cells, T-cells, NK cells, and gamma delta T-cells. One day post-sepsis, the proportion of most non-immune cell populations was decreased, while immune cell populations, particularly neutrophils and macrophages, were highly enriched. Proportional changes of endothelial cells, neutrophils, and macrophages were validated using faecal slurry and cecal ligation and puncture models. At 1 month post-sepsis, we observed persistent enrichment/depletion of cell populations and further uncovered a cell-type and tissue-specific ability to return to a baseline transcriptomic state. Differential gene expression analyses revealed key genes and pathways altered in post-sepsis muscle and fat and highlighted the engagement of infection/inflammation and tissue damage signalling. Finally, regulator analysis identified gonadotropin-releasing hormone and Bay 11-7082 as targets/compounds that we show can reduce sepsis-associated loss of lean or fat mass. CONCLUSIONS These data demonstrate persistent post-sepsis muscle and adipose tissue disruption at the single-cell level and highlight opportunities to combat long-term post-sepsis tissue wasting using bioinformatics-guided therapeutic interventions.
Collapse
Affiliation(s)
- Dong Seong Cho
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMNUSA
| | - Rebecca E. Schmitt
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMNUSA
| | - Aneesha Dasgupta
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMNUSA
| | | | - Jason D. Doles
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMNUSA
| |
Collapse
|
9
|
Niewiadomska-Cimicka A, Hache A, Trottier Y. Gene Deregulation and Underlying Mechanisms in Spinocerebellar Ataxias With Polyglutamine Expansion. Front Neurosci 2020; 14:571. [PMID: 32581696 PMCID: PMC7296114 DOI: 10.3389/fnins.2020.00571] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/11/2020] [Indexed: 12/14/2022] Open
Abstract
Polyglutamine spinocerebellar ataxias (polyQ SCAs) include SCA1, SCA2, SCA3, SCA6, SCA7, and SCA17 and constitute a group of adult onset neurodegenerative disorders caused by the expansion of a CAG repeat sequence located within the coding region of specific genes, which translates into polyglutamine tract in the corresponding proteins. PolyQ SCAs are characterized by degeneration of the cerebellum and its associated structures and lead to progressive ataxia and other diverse symptoms. In recent years, gene and epigenetic deregulations have been shown to play a critical role in the pathogenesis of polyQ SCAs. Here, we provide an overview of the functions of wild type and pathogenic polyQ SCA proteins in gene regulation, describe the extent and nature of gene expression changes and their pathological consequences in diseases, and discuss potential avenues to further investigate converging and distinct disease pathways and to develop therapeutic strategies.
Collapse
Affiliation(s)
- Anna Niewiadomska-Cimicka
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, Strasbourg, France
| | - Antoine Hache
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, Strasbourg, France
| | - Yvon Trottier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Université de Strasbourg, Strasbourg, France
| |
Collapse
|
10
|
Da Silva JD, Teixeira-Castro A, Maciel P. From Pathogenesis to Novel Therapeutics for Spinocerebellar Ataxia Type 3: Evading Potholes on the Way to Translation. Neurotherapeutics 2019; 16:1009-1031. [PMID: 31691128 PMCID: PMC6985322 DOI: 10.1007/s13311-019-00798-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD), is a neurodegenerative disorder caused by a polyglutamine expansion in the ATXN3 gene. In spite of the identification of a clear monogenic cause 25 years ago, the pathological process still puzzles researchers, impairing prospects for an effective therapy. Here, we propose the disruption of protein homeostasis as the hub of SCA3 pathogenesis, being the molecular mechanisms and cellular pathways that are deregulated in SCA3 downstream consequences of the misfolding and aggregation of ATXN3. Moreover, we attempt to provide a realistic perspective on how the translational/clinical research in SCA3 should evolve. This was based on molecular findings, clinical and epidemiological characteristics, studies of proposed treatments in other conditions, and how that information is essential for their (re-)application in SCA3. This review thus aims i) to critically evaluate the current state of research on SCA3, from fundamental to translational and clinical perspectives; ii) to bring up the current key questions that remain unanswered in this disorder; and iii) to provide a frame on how those answers should be pursued.
Collapse
Affiliation(s)
- Jorge Diogo Da Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
11
|
Buijsen RAM, Toonen LJA, Gardiner SL, van Roon-Mom WMC. Genetics, Mechanisms, and Therapeutic Progress in Polyglutamine Spinocerebellar Ataxias. Neurotherapeutics 2019; 16:263-286. [PMID: 30607747 PMCID: PMC6554265 DOI: 10.1007/s13311-018-00696-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Autosomal dominant cerebellar ataxias (ADCAs) are a group of neurodegenerative disorders characterized by degeneration of the cerebellum and its connections. All ADCAs have progressive ataxia as their main clinical feature, frequently accompanied by dysarthria and oculomotor deficits. The most common spinocerebellar ataxias (SCAs) are 6 polyglutamine (polyQ) SCAs. These diseases are all caused by a CAG repeat expansion in the coding region of a gene. Currently, no curative treatment is available for any of the polyQ SCAs, but increasing knowledge on the genetics and the pathological mechanisms of these polyQ SCAs has provided promising therapeutic targets to potentially slow disease progression. Potential treatments can be divided into pharmacological and gene therapies that target the toxic downstream effects, gene therapies that target the polyQ SCA genes, and stem cell replacement therapies. Here, we will provide a review on the genetics, mechanisms, and therapeutic progress in polyglutamine spinocerebellar ataxias.
Collapse
Affiliation(s)
- Ronald A M Buijsen
- Department of Human Genetics, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
| | - Lodewijk J A Toonen
- Department of Human Genetics, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Sarah L Gardiner
- Department of Human Genetics, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
- Department of Neurology, LUMC, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | | |
Collapse
|
12
|
Matos CA, de Almeida LP, Nóbrega C. Machado-Joseph disease/spinocerebellar ataxia type 3: lessons from disease pathogenesis and clues into therapy. J Neurochem 2018; 148:8-28. [PMID: 29959858 DOI: 10.1111/jnc.14541] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/05/2018] [Accepted: 06/27/2018] [Indexed: 12/25/2022]
Abstract
Machado-Joseph disease (MJD), also known as spinocerebellar ataxia type 3 (SCA3), is an incurable disorder, widely regarded as the most common form of spinocerebellar ataxia in the world. MJD/SCA3 arises from mutation of the ATXN3 gene, but this simple monogenic cause contrasts with the complexity of the pathogenic mechanisms that are currently admitted to underlie neuronal dysfunction and death. The aberrantly expanded protein product - ataxin-3 - is known to aggregate and generate toxic species that disrupt several cell systems, including autophagy, proteostasis, transcription, mitochondrial function and signalling. Over the years, research into putative therapeutic approaches has often been devoted to the development of strategies that counteract disease at different stages of cellular pathogenesis. Silencing the pathogenic protein, blocking aggregation, inhibiting toxic proteolytic processing and counteracting dysfunctions of the cellular systems affected have yielded promising ameliorating results in studies with cellular and animal models. The current review analyses the available studies dedicated to the investigation of MJD/SCA3 pathogenesis and the exploration of possible therapeutic strategies, focusing primarily on gene therapy and pharmacological approaches rooted on the molecular and cellular mechanisms of disease.
Collapse
Affiliation(s)
- Carlos A Matos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Clévio Nóbrega
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Department of Biomedical Sciences and Medicine, University of Algarve, Coimbra, Portugal.,Centre for Biomedical Research (CBMR), University of Algarve, Coimbra, Portugal.,Algarve Biomedical Center (ABC), University of Algarve, Faro, Portugal
| |
Collapse
|
13
|
Duarte-Silva S, Neves-Carvalho A, Soares-Cunha C, Silva JM, Teixeira-Castro A, Vieira R, Silva-Fernandes A, Maciel P. Neuroprotective Effects of Creatine in the CMVMJD135 Mouse Model of Spinocerebellar Ataxia Type 3. Mov Disord 2018; 33:815-826. [PMID: 29570846 DOI: 10.1002/mds.27292] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 11/27/2017] [Accepted: 12/18/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Mitochondrial dysfunction has been implicated in several neurodegenerative diseases. Creatine administration increases concentration of the energy buffer phosphocreatine, exerting protective effects in the brain. We evaluate whether a creatine-enriched diet would be beneficial for a mouse model of spinocerebellar ataxia type 3, a genetically defined neurodegenerative disease for which no treatment is available. METHODS We performed 2 independent preclinical trials using the CMVMJD135 mouse model (treating 2 groups of animals with different disease severity) and wild-type mice, to which 2% creatine was provided for 19 (preclinical trial 1) or 29 (preclinical trial 2) weeks, starting at a presymptomatic age. Motor behavior was evaluated at several time points from 5 to 34 weeks of age, and neuropathological studies were performed at the end of each trial. RESULTS Creatine supplementation led to an overall improvement in the motor phenotype of CMVMJD135 mice in both trials, rescuing motor balance and coordination and also restored brain weight, mitigated astrogliosis, and preserved Calbindin-positive cells in the cerebellum. Moreover, a reduction of mutant ataxin-3 aggregates occurred despite maintained steady-state levels of the protein and the absence of autophagy activation. Creatine treatment also restored the expression of the mitochondrial mass marker Porin and reduced the expression of antioxidant enzymes Heme oxygenase 1 (HO1) and NAD(P)H Quinone Dehydrogenase 1 (NQO1), suggesting a beneficial effect at the level of mitochondria and oxidative stress. CONCLUSIONS Creatine slows disease progression and improves motor dysfunction as well as ameliorates neuropathology of the CMVMJD135 animals, supporting this as a useful strategy to slow the progression of spinocerebellar ataxia type 3. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Sara Duarte-Silva
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andreia Neves-Carvalho
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana M Silva
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rita Vieira
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Anabela Silva-Fernandes
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
14
|
Wang ZJ, Hanet A, Weishäupl D, Martins IM, Sowa AS, Riess O, Schmidt T. Divalproex sodium modulates nuclear localization of ataxin-3 and prevents cellular toxicity caused by expanded ataxin-3. CNS Neurosci Ther 2018; 24:404-411. [PMID: 29318784 DOI: 10.1111/cns.12795] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 12/10/2017] [Accepted: 12/10/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND & AIMS Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD), is an autosomal dominantly inherited neurodegenerative disorder and the most common form of SCA worldwide. It is caused by the expansion of a polyglutamine (polyQ) tract in the ataxin-3 protein. Nuclear localization of the affected protein is a key event in the pathology of SCA3 via affecting nuclear organization, transcriptional dysfunction, and seeding aggregations, finally causing neurodegeneration and cell death. So far, there is no effective therapy to prevent or slow the progression of SCA3. METHODS In this study, we explored the effect of divalproex sodium as an HDACi in SCA3 cell models and explored how divalproex sodium interferes with pathogenetic processes causing SCA3. RESULTS We found that divalproex sodium rescues the hypoacetylation levels of histone H3 and attenuates cellular cytotoxicity induced by expanded ataxin-3 partly via preventing nuclear transport of ataxin-3 (particularly heat shock-dependent). CONCLUSION Our study provides novel insights into the mechanisms of action of divalproex sodium as a possible treatment for SCA3, beyond the known regulation of transcription.
Collapse
Affiliation(s)
- Zi-Jian Wang
- Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an, Shaanxi, China.,Institute of Medical Genetics & Applied Genomics, University of Tuebingen, Tuebingen, Germany.,Center for Rare Diseases (ZSE), University Hospital Tuebingen, Tuebingen, Germany.,Graduate Training Centre of Neuroscience, University of Tuebingen, Tuebingen, Germany
| | - Aoife Hanet
- Institute of Medical Genetics & Applied Genomics, University of Tuebingen, Tuebingen, Germany.,Center for Rare Diseases (ZSE), University Hospital Tuebingen, Tuebingen, Germany.,Department of Biochemistry, Max Planck Institute for Developmental Biology, Tuebingen, Germany
| | - Daniel Weishäupl
- Institute of Medical Genetics & Applied Genomics, University of Tuebingen, Tuebingen, Germany.,Center for Rare Diseases (ZSE), University Hospital Tuebingen, Tuebingen, Germany.,Graduate Training Centre of Neuroscience, University of Tuebingen, Tuebingen, Germany
| | - Inês M Martins
- Institute of Medical Genetics & Applied Genomics, University of Tuebingen, Tuebingen, Germany.,Center for Rare Diseases (ZSE), University Hospital Tuebingen, Tuebingen, Germany
| | - Anna S Sowa
- Institute of Medical Genetics & Applied Genomics, University of Tuebingen, Tuebingen, Germany.,Center for Rare Diseases (ZSE), University Hospital Tuebingen, Tuebingen, Germany.,Graduate Training Centre of Neuroscience, University of Tuebingen, Tuebingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics & Applied Genomics, University of Tuebingen, Tuebingen, Germany.,Center for Rare Diseases (ZSE), University Hospital Tuebingen, Tuebingen, Germany
| | - Thorsten Schmidt
- Institute of Medical Genetics & Applied Genomics, University of Tuebingen, Tuebingen, Germany.,Center for Rare Diseases (ZSE), University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
15
|
Duarte-Silva S, Maciel P. Pharmacological Therapies for Machado-Joseph Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1049:369-394. [PMID: 29427114 DOI: 10.1007/978-3-319-71779-1_19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Machado-Joseph disease (MJD), also known as Spinocerebellar Ataxia type 3 (SCA3), is the most common autosomal dominant ataxia worldwide. MJD integrates a large group of disorders known as polyglutamine diseases (polyQ). To date, no effective treatment exists for MJD and other polyQ diseases. Nevertheless, researchers are making efforts to find treatment possibilities that modify the disease course or alleviate disease symptoms. Since neuroimaging studies in mutation carrying individuals suggest that in nervous system dysfunction begins many years before the onset of any detectable symptoms, the development of therapeutic interventions becomes of great importance, not only to slow progression of manifest disease but also to delay, or ideally prevent, its onset. Potential therapeutic targets for MJD and polyQ diseases can be divided into (i) those that are aimed at the polyQ proteins themselves, namely gene silencing, attempts to enhance mutant protein degradation or inhibition/prevention of aggregation; and (ii) those that intercept the toxic downstream effects of the polyQ proteins, such as mitochondrial dysfunction and oxidative stress, transcriptional abnormalities, UPS impairment, excitotoxicity, or activation of cell death. The existence of relevant animal models and the recent contributions towards the identification of putative molecular mechanisms underlying MJD are impacting on the development of new drugs. To date only a few preclinical trials were conducted, nevertheless some had very promising results and some candidate drugs are close to being tested in humans. Clinical trials for MJD are also very few to date and their results not very promising, mostly due to trial design constraints. Here, we provide an overview of the pharmacological therapeutic strategies for MJD studied in animal models and patients, and of their possible translation into the clinical practice.
Collapse
Affiliation(s)
- Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
16
|
Wang Z. Experimental and Clinical Strategies for Treating Spinocerebellar Ataxia Type 3. Neuroscience 2017; 371:138-154. [PMID: 29229556 DOI: 10.1016/j.neuroscience.2017.11.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 01/02/2023]
Abstract
Spinocerebellar ataxia type 3 (SCA3), or Machado-Joseph disease (MJD), is an autosomal dominant neurodegenerative disorder caused by the expansion of a polyglutamine (polyQ) tract in the ataxin-3 protein. To date, there is no effective therapy available to prevent progression of this disease. However, clinical strategies for alleviating various symptoms are imperative to promote a better quality of life for SCA3/MJD patients. Furthermore, experimental therapeutic strategies, including gene silencing or mutant protein clearance, mutant polyQ protein modification, stabilizing the native protein conformation, rescue of cellular dysfunction and neuromodulation to slow the progression of SCA3/MJD, have been developed. In this study, based on the current knowledge, I detail the clinical and experimental therapeutic strategies for treating SCA3/MJD, paying particular attention to drug discovery.
Collapse
Affiliation(s)
- Zijian Wang
- Genetic Engineering Laboratory, College of Biological and Environmental Engineering, Xi'an University, Xi'an, Shaanxi 710065, China.
| |
Collapse
|
17
|
Li Z, Wu F, Zhang X, Chai Y, Chen D, Yang Y, Xu K, Yin J, Li R, Shi H, Wang Z, Li X, Xiao J, Zhang H. Valproate Attenuates Endoplasmic Reticulum Stress-Induced Apoptosis in SH-SY5Y Cells via the AKT/GSK3β Signaling Pathway. Int J Mol Sci 2017; 18:ijms18020315. [PMID: 28208696 PMCID: PMC5343851 DOI: 10.3390/ijms18020315] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 01/12/2017] [Accepted: 01/27/2017] [Indexed: 12/22/2022] Open
Abstract
Endoplasmic reticulum (ER) stress-induced apoptosis plays an important role in a range of neurological disorders, such as neurodegenerative diseases, spinal cord injury, and diabetic neuropathy. Valproate (VPA), a typical antiepileptic drug, is commonly used in the treatment of bipolar disorder and epilepsy. Recently, VPA has been reported to exert neurotrophic effects and promote neurite outgrowth, but its molecular mechanism is still unclear. In the present study, we investigated whether VPA inhibited ER stress and promoted neuroprotection and neuronal restoration in SH-SY5Y cells and in primary rat cortical neurons, respectively, upon exposure to thapsigargin (TG). In SH-SY5Y cells, cell viability was detected by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, and the expression of ER stress-related apoptotic proteins such as glucose‑regulated protein (GRP78), C/EBP homologous protein (CHOP), and cleaved caspase-12/-3 were analyzed with Western blot analyses and immunofluorescence assays. To explore the pathway involved in VPA-induced cell proliferation, we also examined p-AKT, GSK3β, p-JNK and MMP-9. Moreover, to detect the effect of VPA in primary cortical neurons, immunofluorescence staining of β-III tubulin and Anti-NeuN was analyzed in primary cultured neurons exposed to TG. Our results demonstrated that VPA administration improved cell viability in cells exposed to TG. In addition, VPA increased the levels of GRP78 and p-AKT and decreased the levels of ATF6, XBP-1, GSK3β, p-JNK and MMP-9. Furthermore, the levels of the ER stress-induced apoptosis response proteins CHOP, cleaved caspase-12 and cleaved caspase-3 were inhibited by VPA treatment. Meanwhile, VPA administration also increased the ratio of Bcl-2/Bax. Moreover, VPA can maintain neurite outgrowth of primary cortical neurons. Collectively, the neurotrophic effect of VPA is related to the inhibition of ER stress-induced apoptosis in SH-SY5Y cells and the maintenance of neuronal growth. Collectively, our results suggested a new approach for the therapeutic function of VPA in neurological disorders and neuroprotection.
Collapse
Affiliation(s)
- Zhengmao Li
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Fenzan Wu
- Science and Education division, Cixi People's Hospital, Wenzhou Medical University, Ningbo 315300, China.
| | - Xie Zhang
- Ningbo Medical Treatment Center, Li Huili Hospital, Ningbo 315000, China.
| | - Yi Chai
- Department of neurosurgery, The second Affiliated Hospital, Nanchang University, Nanchang 330006, China.
| | - Daqing Chen
- Emergency Department, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yuetao Yang
- Emergency Department, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China.
| | - Kebin Xu
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Jiayu Yin
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Rui Li
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Hongxue Shi
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Zhouguang Wang
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Xiaokun Li
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
- Institute of Life Sciences, Wenzhou University, Wenzhou 325035, China.
| | - Jian Xiao
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Hongyu Zhang
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|
18
|
Esteves S, Duarte-Silva S, Maciel P. Discovery of Therapeutic Approaches for Polyglutamine Diseases: A Summary of Recent Efforts. Med Res Rev 2016; 37:860-906. [PMID: 27870126 DOI: 10.1002/med.21425] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/01/2016] [Accepted: 10/05/2016] [Indexed: 12/19/2022]
Abstract
Polyglutamine (PolyQ) diseases are a group of neurodegenerative disorders caused by the expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the coding region of specific genes. This leads to the production of pathogenic proteins containing critically expanded tracts of glutamines. Although polyQ diseases are individually rare, the fact that these nine diseases are irreversibly progressive over 10 to 30 years, severely impairing and ultimately fatal, usually implicating the full-time patient support by a caregiver for long time periods, makes their economic and social impact quite significant. This has led several researchers worldwide to investigate the pathogenic mechanism(s) and therapeutic strategies for polyQ diseases. Although research in the field has grown notably in the last decades, we are still far from having an effective treatment to offer patients, and the decision of which compounds should be translated to the clinics may be very challenging. In this review, we provide a comprehensive and critical overview of the most recent drug discovery efforts in the field of polyQ diseases, including the most relevant findings emerging from two different types of approaches-hypothesis-based candidate molecule testing and hypothesis-free unbiased drug screenings. We hereby summarize and reflect on the preclinical studies as well as all the clinical trials performed to date, aiming to provide a useful framework for increasingly successful future drug discovery and development efforts.
Collapse
Affiliation(s)
- Sofia Esteves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| | - Sara Duarte-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, University of Minho, Guimarães, Braga, Portugal
| |
Collapse
|
19
|
Sun R, Zhang S, Hu W, Lu X, Lou N, Yang Z, Chen S, Zhang X, Yang H. Valproic acid attenuates skeletal muscle wasting by inhibiting C/EBPβ-regulated atrogin1 expression in cancer cachexia. Am J Physiol Cell Physiol 2016; 311:C101-15. [DOI: 10.1152/ajpcell.00344.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/13/2016] [Indexed: 12/15/2022]
Abstract
Muscle wasting is the hallmark of cancer cachexia and is associated with poor quality of life and increased mortality. Valproic acid (VPA), a histone deacetylase (HDAC) inhibitor, has important biological effects in the treatment of muscular dystrophy. To verify whether VPA could ameliorate muscle wasting induced by cancer cachexia, we explored the role of VPA in two cancer cachectic mouse models [induced by colon-26 (C26) adenocarcinoma or Lewis lung carcinoma (LLC)] and atrophied C2C12 myotubes [induced by C26 cell conditioned medium (CCM) or LLC cell conditioned medium (LCM)]. Our data demonstrated that treatment with VPA increased the mass and cross-sectional area of skeletal muscles in tumor-bearing mice. Furthermore, treatment with VPA also increased the diameter of myotubes cultured in conditioned medium. The skeletal muscles in cachectic mice or atrophied myotubes treated with VPA exhibited reduced levels of CCAAT/enhancer binding protein beta (C/EBPβ), resulting in atrogin1 downregulation and the eventual alleviation of muscle wasting and myotube atrophy. Moreover, atrogin1 promoter activity in myotubes was stimulated by CCM via activating the C/EBPβ-responsive cis-element and subsequently inhibited by VPA. In contrast to the effect of VPA on the levels of C/EBPβ, the levels of inactivating forkhead box O3 (FoxO3a) were unaffected. In summary, VPA attenuated muscle wasting and myotube atrophy and reduced C/EBPβ binding to atrogin1 promoter locus in the myotubes. Our discoveries indicate that HDAC inhibition by VPA might be a promising new approach for the preservation of skeletal muscle in cancer cachexia.
Collapse
Affiliation(s)
- Rulin Sun
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Santao Zhang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Wenjun Hu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Xing Lu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Ning Lou
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China; and
| | - Zhende Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| | - Shaoyong Chen
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China; and
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei Province, China
| |
Collapse
|