1
|
Ho AK, Jeganathan F, Bictash M, Chen HJ. Identification of novel small molecule chaperone activators for neurodegenerative disease treatment. Biomed Pharmacother 2025; 187:118049. [PMID: 40239269 PMCID: PMC12086176 DOI: 10.1016/j.biopha.2025.118049] [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: 01/10/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
A pathological hallmark of neurodegenerative disease is the accumulation of aberrant protein aggregates which contribute to the cytotoxicity and are therefore a target for therapy development. One key mechanism to manage cellular protein homeostasis is heat shock proteins (HSPs), protein chaperones which are known to target aberrant protein accumulation. Activation of HSPs target aberrant TDP-43, tau and amyloid to rescue neurodegenerative disease. As an attempt to target HSP activation for neurodegeneration therapy, we here develop a drug screening assay to identify compounds that will activate the master regulator of HSPs, the transcription factor heat shock factor 1 (HSF1). As HSF1 is bound by HSP90 which prevents its activation, we developed a NanoBRET assay, which allows us to monitor and quantify the HSF1-HSP90 interaction in living cells to screen for compounds disrupting this interaction and thereby releasing HSF1 for activation. After the optimisation and validation of the assay, a two thousand compound library was screened which produced 10 hits including two known HSP90 inhibitors. Follow-up functional study showed that one of the hits oxyphenbutazone (OPB) significantly reduces the accumulation of insoluble TDP-43 in a cell model, eliciting no signs of stress or toxicity. Overall, this study demonstrates a viable strategy for new drug discovery in targeting aberrant proteins and identifies potential candidates for translation into neurodegenerative disease treatment.
Collapse
Affiliation(s)
- Anita K Ho
- York Biomedical Research Institute, Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - Fiona Jeganathan
- Alzheimer's Research UK UCL Drug Discovery Institute, University College London, The Cruciform Building, Gower Street, London WC1E 6BT, London WC1E 6BT, UK
| | - Magda Bictash
- Alzheimer's Research UK UCL Drug Discovery Institute, University College London, The Cruciform Building, Gower Street, London WC1E 6BT, London WC1E 6BT, UK
| | - Han-Jou Chen
- York Biomedical Research Institute, Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK.
| |
Collapse
|
2
|
Freisem D, Hoenigsperger H, Catanese A, Sparrer KMJ. Inborn errors of canonical autophagy in neurodegenerative diseases. Hum Mol Genet 2025:ddae179. [PMID: 40304712 DOI: 10.1093/hmg/ddae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 05/02/2025] Open
Abstract
Neurodegenerative disorders (NDDs), characterized by a progressive loss of neurons and cognitive function, are a severe burden to human health and mental fitness worldwide. A hallmark of NDDs such as Alzheimer's disease, Huntington's disease, Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS) and prion diseases is disturbed cellular proteostasis, resulting in pathogenic deposition of aggregated protein species. Autophagy is a major cellular process maintaining proteostasis and integral to innate immune defenses that mediates lysosomal protein turnover. Defects in autophagy are thus frequently associated with NDDs. In this review, we discuss the interplay between NDDs associated proteins and autophagy and provide an overview over recent discoveries in inborn errors in canonical autophagy proteins that are associated with NDDs. While mutations in autophagy receptors seems to be associated mainly with the development of ALS, errors in mitophagy are mainly found to promote PD. Finally, we argue whether autophagy may impact progress and onset of the disease, as well as the potential of targeting autophagy as a therapeutic approach. Concludingly, understanding disorders due to inborn errors in autophagy-"autophagopathies"-will help to unravel underlying NDD pathomechanisms and provide unique insights into the neuroprotective role of autophagy, thus potentially paving the way for novel therapeutic interventions.
Collapse
Affiliation(s)
- Dennis Freisem
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, Baden-Wuerttemberg, Ulm 89081, Germany
| | - Helene Hoenigsperger
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, Baden-Wuerttemberg, Ulm 89081, Germany
| | - Alberto Catanese
- German Center for Neurodegenerative Diseases, Albert-Einstein-Allee 11, Baden-Wuerttemberg, Ulm 89081, Germany
- Institute of Anatomy and Cell Biology, Ulm University Medical Center, Albert-Einstein-Allee 11, Baden-Wuerttemberg, Ulm 89081, Germany
| | - Konstantin M J Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Meyerhofstr. 1, Baden-Wuerttemberg, Ulm 89081, Germany
| |
Collapse
|
3
|
Faller KME, Chaytow H, Gillingwater TH. Targeting common disease pathomechanisms to treat amyotrophic lateral sclerosis. Nat Rev Neurol 2025; 21:86-102. [PMID: 39743546 DOI: 10.1038/s41582-024-01049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
The motor neuron disease amyotrophic lateral sclerosis (ALS) is a devastating condition with limited treatment options. The past few years have witnessed a ramping up of translational ALS research, offering the prospect of disease-modifying therapies. Although breakthroughs using gene-targeted approaches have shown potential to treat patients with specific disease-causing mutations, the applicability of such therapies remains restricted to a minority of individuals. Therapies targeting more general mechanisms that underlie motor neuron pathology in ALS are therefore of considerable interest. ALS pathology is associated with disruption to a complex array of key cellular pathways, including RNA processing, proteostasis, metabolism and inflammation. This Review details attempts to restore cellular homeostasis by targeting these pathways in order to develop effective, broadly-applicable ALS therapeutics.
Collapse
Affiliation(s)
- Kiterie M E Faller
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Helena Chaytow
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
4
|
Tedeschi V, Nele V, Valsecchi V, Anzilotti S, Vinciguerra A, Zucaro L, Sisalli MJ, Cassiano C, De Iesu N, Pignataro G, Canzoniero LMT, Pannaccione A, De Rosa G, Secondo A. Nanoparticles encapsulating phosphatidylinositol derivatives promote neuroprotection and functional improvement in preclinical models of ALS via a long-lasting activation of TRPML1 lysosomal channel. Pharmacol Res 2024; 210:107491. [PMID: 39491634 DOI: 10.1016/j.phrs.2024.107491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease currently incurable, in which motor neuron degeneration leads to voluntary skeletal muscle atrophy. Molecularly, ALS is characterized by protein aggregation, synaptic and organellar dysfunction, and Ca2+ dyshomeostasis. Of interest, autophagy dysfunction is emerging as one of the main putative targets of ALS therapy. A tune regulation of this cleansing process is affordable by a proper stimulation of TRPML1, one of the main lysosomal channels. However, TRPML1 activation by PI(3,5)P2 has low open probability to remain in an active conformation. To overcome this drawback we developed a lipid-based formulation of PI(3,5)P2 whose putative therapeutic potential has been tested in in vitro and in vivo ALS models. Pharmacodynamic properties of PI(3,5)P2 lipid-based formulations (F1 and F2) on TRPML1 activity have been characterized by means of patch-clamp electrophysiology and Fura-2AM video-imaging in motor neuronal cells. Once selected for the ability to stabilize TRPML1 activity, the most effective preparation F1 was studied in vivo to measure neuromuscular function and survival of SOD1G93A ALS mice, thereby establishing its therapeutic profile. F1, but not PI(3,5)P2 alone, stabilized the open state of the lysosomal channel TRPML1 and increased the persistence of intracellular calcium concentration ([Ca2+]i). Then, F1 was effective in delaying motor neuron loss, improving innervated endplants and muscle performance in SOD1G93A mice, extending overall lifespan by an average of 10 days. Of note F1 prevented gliosis and autophagy dysfunction in ALS mice by restoring PI(3,5)P2 level. Our novel self-assembling lipidic formulation for PI(3,5)P2 delivery exerts a neuroprotective effect in preclinical models of ALS mainly regulating dysfunctional autophagy through TRPML1 activity stabilization.
Collapse
Affiliation(s)
- Valentina Tedeschi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | - Valeria Nele
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy
| | - Valeria Valsecchi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | - Serenella Anzilotti
- Department of Science and Technology-DST, University of Sannio, Via Port'Arsa 11, Benevento 82100, Italy
| | - Antonio Vinciguerra
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica Delle Marche", Via Tronto 10/A, Ancona 60126, Italy
| | - Laura Zucaro
- Biogem Scarl, Istituto di Ricerche Genetiche, Ariano Irpino, AV, Italy; Department of Translational Medical Sciences, Università degli Studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Josè Sisalli
- Department of Translational Medical Sciences, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | - Chiara Cassiano
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy
| | | | - Giuseppe Pignataro
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | | | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, School of Medicine, University of Naples "Federico II", Via S. Pansini 5, Naples 80131, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, Naples 80131, Italy.
| | - Agnese Secondo
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica Delle Marche", Via Tronto 10/A, Ancona 60126, Italy.
| |
Collapse
|
5
|
Uneri A, McArdle CJ, Deng Z, Barth SH, Keene D, Craft S, Raab-Graham KF. DJ-1-mediated repression of the RNA-binding protein FMRP is predicted to impact known Alzheimer's disease-related protein networks. J Alzheimers Dis 2024; 102:763-777. [PMID: 39610285 DOI: 10.1177/13872877241291175] [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] [Indexed: 11/30/2024]
Abstract
BACKGROUND RNA-binding proteins (RBPs) modulate the synaptic proteome and are instrumental in maintaining synaptic homeostasis. Moreover, aberrant expression of an RBP in a disease state would have deleterious downstream effects on synaptic function. While many underlying mechanisms of synaptic dysfunction in Alzheimer's disease (AD) have been proposed, the contribution of RBPs has been relatively unexplored. OBJECTIVE To investigate alterations in RBP-messenger RNA (mRNA) interactions in AD, and its overall impact on the disease-related proteome. METHODS We first utilized RNA-immunoprecipitation to investigate interactions between RBP, DJ-1 (Parkinson's Disease protein 7) and target mRNAs in controls and AD. Surface Sensing of Translation - Proximity Ligation Assay (SUnSET-PLA) and western blotting additionally quantified alterations in mRNA translation and protein expression of DJ-1 targets. Finally, we utilized an unbiased bioinformatic approach that connects AD-related pathways to two RBPs, DJ-1 and FMRP (Fragile X messenger ribonucleoprotein 1). RESULTS We find that oligomeric DJ-1 in AD donor synapses were less dynamic in their ability to bind and unbind mRNA compared to synapses from cognitively unimpaired, neuropathologically-verified controls. Furthermore, we find that DJ-1 associates with the mRNA coding for FMRP, Fmr1, leading to its reduced synaptic expression in AD. Through the construction of protein-protein interaction networks, aberrant expression of DJ-1 and FMRP are predicted to lead to the upregulation of key AD-related pathways, such as thyroid hormone stimulating pathway, autophagy, and ubiquitin mediated proteolysis. CONCLUSIONS DJ-1 and FMRP are novel targets that may restore established neurobiological mechanisms underlying AD.
Collapse
Affiliation(s)
- Ayse Uneri
- Department of Translational Neuroscience, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Colin J McArdle
- Department of Translational Neuroscience, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Zhiyong Deng
- Department of Translational Neuroscience, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Samuel H Barth
- Department of Translational Neuroscience, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
| | - Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Gerontology and Geriatric Medicine, Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA
| | - Suzanne Craft
- Department of Gerontology and Geriatric Medicine, Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA
| | - Kimberly F Raab-Graham
- Department of Translational Neuroscience, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, NC, USA
- Department of Gerontology and Geriatric Medicine, Wake Forest Alzheimer's Disease Research Center, Winston-Salem, NC, USA
| |
Collapse
|
6
|
Casiraghi V, Sorce MN, Santangelo S, Invernizzi S, Bossolasco P, Lattuada C, Battaglia C, Venturin M, Silani V, Colombrita C, Ratti A. Modeling of TDP-43 proteinopathy by chronic oxidative stress identifies rapamycin as beneficial in ALS patient-derived 2D and 3D iPSC models. Exp Neurol 2024; 383:115057. [PMID: 39536963 DOI: 10.1016/j.expneurol.2024.115057] [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: 05/29/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disorder characterized neuropathologically by TDP-43 proteinopathy with loss of TDP-43 nuclear splicing activity and formation of cytoplasmic TDP-43 aggregates. The lack of suitable experimental models of TDP-43 proteinopathy has hampered the discovery of effective therapies. We already showed that chronic and mild oxidative insult by sodium arsenite (ARS) triggered TDP-43 cytoplasmic aggregation and stress granules (SGs) formation in ALS patient-derived fibroblasts and motor neurons differentiated from induced pluripotent stem cells (iPSC-MNs). However, whether this insult induces a reduction of TDP-43 splicing activity in the nucleus, thus recapitulating both gain and loss of function pathomechanisms, still remains to be determined. In this study we first showed that chronic ARS in human neuroblastoma cells triggered TDP-43 cytoplasmic mislocalization, SGs formation and defective splicing of TDP-43 target genes UNC13A and POLDIP3 as functional readouts of TDP-43 proteinopathy. Additionally, a dysregulation of autophagy and senescence markers was observed in this condition. In a preliminary drug screening approach with autophagy-promoting drugs, namely rapamycin, lithium carbonate and metformin, only rapamycin prevented ARS-induced loss of TDP-43 splicing activity. We then demonstrated that, in addition to TDP-43 cytoplasmic aggregation, chronic ARS triggered TDP-43 loss of splicing activity also in ALS patient-derived primary fibroblasts and iPSC-MNs and that rapamycin was beneficial to reduce these TDP-43 pathological features. By switching to a neuro-glial 3D in vitro model, we observed that treatment of ALS iPSC-brain organoids with chronic ARS also induced a defective TDP-43 splicing activity which was prevented by rapamycin. Collectively, we established different human cell models of TDP-43 proteinopathy which recapitulate TDP-43 gain and loss of function, prevented by rapamycin administration. Human neuroblastoma cells and patient-derived fibroblasts and 2D- and 3D-iPSC models exposed to chronic oxidative stress represent therefore suitable in vitro platforms for future drug screening approaches in ALS.
Collapse
Affiliation(s)
- Valeria Casiraghi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Marta Nice Sorce
- Department of Neuroscience - Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149 Milan, Italy
| | - Serena Santangelo
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Sabrina Invernizzi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Patrizia Bossolasco
- Department of Neuroscience - Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149 Milan, Italy
| | - Chiara Lattuada
- Department of Neuroscience - Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149 Milan, Italy
| | - Cristina Battaglia
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Marco Venturin
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy
| | - Vincenzo Silani
- Department of Neuroscience - Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149 Milan, Italy; "Dino Ferrari" Center, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via Francesco Sforza 35, 20122 Milan, Italy
| | - Claudia Colombrita
- Department of Neuroscience - Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149 Milan, Italy
| | - Antonia Ratti
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Via Fratelli Cervi 93, 20090 Segrate, Milan, Italy; Department of Neuroscience - Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149 Milan, Italy.
| |
Collapse
|
7
|
Lescouzères L, Patten SA. Promising animal models for amyotrophic lateral sclerosis drug discovery: a comprehensive update. Expert Opin Drug Discov 2024; 19:1213-1233. [PMID: 39115327 DOI: 10.1080/17460441.2024.2387791] [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: 03/08/2024] [Accepted: 07/30/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. Several animal models have been generated to understand ALS pathogenesis. They have provided valuable insight into disease mechanisms and the development of therapeutic strategies. AREAS COVERED In this review, the authors provide a concise overview of simple genetic model organisms, including C. elegans, Drosophila, zebrafish, and mouse genetic models that have been generated to study ALS. They emphasize the benefits of each model and their application in translational research for discovering new chemicals, gene therapy approaches, and antibody-based strategies for treating ALS. EXPERT OPINION Significant progress is being made in identifying new therapeutic targets for ALS. This progress is being enabled by promising animal models of the disease using increasingly effective genetic and pharmacological strategies. There are still challenges to be overcome in order to achieve improved success rates for translating drugs from animal models to clinics for treating ALS. Several promising future directions include the establishment of novel preclinical protocol standards, as well as the combination of animal models with human induced pluripotent stem cells (iPSCs).
Collapse
Affiliation(s)
- Léa Lescouzères
- INRS - Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, Canada
| | - Shunmoogum A Patten
- INRS - Centre Armand Frappier Santé Biotechnologie, Laval, QC, Canada
- Departement de Neurosciences, Université de Montréal, Montreal, Canada
| |
Collapse
|
8
|
Ho PC, Hsieh TC, Tsai KJ. TDP-43 proteinopathy in frontotemporal lobar degeneration and amyotrophic lateral sclerosis: From pathomechanisms to therapeutic strategies. Ageing Res Rev 2024; 100:102441. [PMID: 39069095 DOI: 10.1016/j.arr.2024.102441] [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: 05/31/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Proteostasis failure is a common pathological characteristic in neurodegenerative diseases. Revitalizing clearance systems could effectively mitigate these diseases. The transactivation response (TAR) DNA-binding protein 43 (TDP-43) plays a critical role as an RNA/DNA-binding protein in RNA metabolism and synaptic function. Accumulation of TDP-43 aggregates in the central nervous system is a hallmark of frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Autophagy, a major and highly conserved degradation pathway, holds the potential for degrading aggregated TDP-43 and alleviating FTLD/ALS. This review explores the causes of TDP-43 aggregation, FTLD/ALS-related genes, key autophagy factors, and autophagy-based therapeutic strategies targeting TDP-43 proteinopathy. Understanding the underlying pathological mechanisms of TDP-43 proteinopathy can facilitate therapeutic interventions.
Collapse
Affiliation(s)
- Pei-Chuan Ho
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Chi Hsieh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
9
|
Sophronea T, Agrawal S, Kumari N, Mishra J, Walecha V, Luthra PM. A 2AR antagonists triggered the AMPK/m-TOR autophagic pathway to reverse the calcium-dependent cell damage in 6-OHDA induced model of PD. Neurochem Int 2024; 178:105793. [PMID: 38880232 DOI: 10.1016/j.neuint.2024.105793] [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: 02/14/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
Calcium dyshomeostasis, oxidative stress, autophagy and apoptosis are the pathogenesis of selective dopaminergic neuronal loss in Parkinson's disease (PD). Earlier, we reported that A2A R modulates IP3-dependent intracellular Ca2+ signalling via PKA. Moreover, A2A R antagonist has been reported to reduce oxidative stress and apoptosis in PD models, however intracellular Ca2+ ([Ca2+]i) dependent autophagy regulation in the 6-OHDA model of PD has not been explored. In the present study, we investigated the A2A R antagonists mediated neuroprotective effects in 6-OHDA-induced primary midbrain neuronal (PMN) cells and unilateral lesioned rat model of PD. 6-OHDA-induced oxidative stress (ROS and superoxide) and [Ca2+]i was measured using Fluo4AM, DCFDA and DHE dye respectively. Furthermore, autophagy was assessed by Western blot of p-m-TOR/mTOR, p-AMPK/AMPK, LC3I/II, Beclin and β-actin. Apoptosis was measured by Annexin V-APC-PI detection and Western blot of Bcl2, Bax, caspase3 and β-actin. Dopamine levels were measured by Dopamine ELISA kit and Western blot of tyrosine hydroxylase. Our results suggest that 6-OHDA-induced PMN cell death occurred due to the interruption of [Ca2+]i homeostasis, accompanied by activation of autophagy and apoptosis. A2A R antagonists prevented 6-OHDA-induced neuronal cell death by decreasing [Ca2+]i overload and oxidative stress. In addition, we found that A2A R antagonists upregulated mTOR phosphorylation and downregulated AMPK phosphorylation thereby reducing autophagy and apoptosis both in 6-OHDA induced PMN cells and 6-OHDA unilateral lesioned rat model. In conclusion, A2A R antagonists alleviated 6-OHDA toxicity by modulating [Ca2+]i signalling to inhibit autophagy mediated by the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Tuithung Sophronea
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Saurabh Agrawal
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Namrata Kumari
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Jyoti Mishra
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Vaishali Walecha
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India
| | - Pratibha Mehta Luthra
- Neuropharmaceutical Chemistry Laboratory, Dr. B. R. Ambedkar Centre for Biomedical Research, North Campus, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
10
|
Pi J, Chen D, Wang J, Yang E, Yang J, Liu Y, Yu J, Xia J, Huang X, Chen L, Ruan Y, Xu JF, Yang F, Shen L. Macrophage targeted graphene oxide nanosystem synergize antibiotic killing and host immune defense for Tuberculosis Therapy. Pharmacol Res 2024; 208:107379. [PMID: 39218421 DOI: 10.1016/j.phrs.2024.107379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
Tuberculosis (TB), a deadly disease caused by Mycobacterium tuberculosis (Mtb) infection, remains one of the top killers among infectious diseases worldwide. How to increase targeting effects of current anti-TB chemotherapeutics and enhance anti-TB immunological responses remains a big challenge in TB and drug-resistant TB treatment. Here, mannose functionalized and polyetherimide protected graphene oxide system (GO-PEI-MAN) was designed for macrophage-targeted antibiotic (rifampicin) and autophagy inducer (carbamazepine) delivery to achieve more effective Mtb killings by combining targeted drug killing and host immunological clearance. GO-PEI-MAN system demonstrated selective uptake by in vitro macrophages and ex vivo macrophages from macaques. The endocytosed GO-PEI-MAN system would be transported into lysosomes, where the drug loaded Rif@Car@GO-PEI-MAN system would undergo accelerated drug release in acidic lysosomal conditions. Rif@Car@GO-PEI-MAN could significantly promote autophagy and apoptosis in Mtb infected macrophages, as well as induce anti-bacterial M1 polarization of Mtb infected macrophages to increase anti-bacterial IFN-γ and nitric oxide production. Collectively, Rif@Car@GO-PEI-MAN demonstrated effectively enhanced intracellular Mtb killing effects than rifampicin, carbamazepine or GO-PEI-MAN alone in Mtb infected macrophages, and could significantly reduce mycobacterial burdens in the lung of infected mice with alleviated pathology and inflammation without systemic toxicity. This macrophage targeted nanosystem synergizing increased drug killing efficiency and enhanced host immunological defense may be served as more effective therapeutics against TB and drug-resistant TB.
Collapse
Affiliation(s)
- Jiang Pi
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Dongsheng Chen
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China
| | - Jiajun Wang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China
| | - Enzhuo Yang
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA; Clinic and Research Center of Tuberculosis, Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiayi Yang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yilin Liu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jiaqi Yu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Jiaojiao Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Xueqin Huang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Lingming Chen
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Yongdui Ruan
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China
| | - Jun-Fa Xu
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China
| | - Fen Yang
- Research Center of Nano Technology and Application Engineering, The First Dongguan Affiliated Hospital, Dongguan Innovation Institute, Guangdong Medical University, China; Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong, China.
| | - Ling Shen
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
11
|
Wenzhi Y, Xiangyi L, Dongsheng F. The prion-like effect and prion-like protein targeting strategy in amyotrophic lateral sclerosis. Heliyon 2024; 10:e34963. [PMID: 39170125 PMCID: PMC11336370 DOI: 10.1016/j.heliyon.2024.e34963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Pathological proteins in amyotrophic lateral sclerosis (ALS), such as superoxide dismutase 1, TAR DNA-binding protein 43, and fused in sarcoma, exhibit a prion-like pattern. All these proteins have a low-complexity domain and seeding activity in cells. In this review, we summarize the studies on the prion-like effect of these proteins and list six prion-like protein targeting strategies that we believe have potential for ALS therapy, including antisense oligonucleotides, antibody-based technology, peptide, protein chaperone, autophagy enhancement, and heteromultivalent compounds. Considering the pathological complexity and heterogeneity of ALS, we believe that the final solution to ALS therapy is most likely to be an individualized cocktail therapy, including clearance of toxicity, blockage of pathological progress, and protection of neurons.
Collapse
Affiliation(s)
- Yang Wenzhi
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Liu Xiangyi
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| | - Fan Dongsheng
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
| |
Collapse
|
12
|
Wang H, Zeng R. Aberrant protein aggregation in amyotrophic lateral sclerosis. J Neurol 2024; 271:4826-4851. [PMID: 38869826 DOI: 10.1007/s00415-024-12485-z] [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: 03/12/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/14/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease. As its pathological mechanisms are not well understood, there are no efficient therapeutics for it at present. While it is highly heterogenous both etiologically and clinically, it has a common salient hallmark, i.e., aberrant protein aggregation (APA). The upstream pathogenesis and the downstream effects of APA in ALS are sophisticated and the investigation of this pathology would be of consequence for understanding ALS. In this paper, the pathomechanism of APA in ALS and the candidate treatment strategies for it are discussed.
Collapse
Affiliation(s)
- Huaixiu Wang
- Department Neurology, Shanxi Provincial Peoples Hospital: Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China.
- Beijing Ai-Si-Kang Medical Technology Co. Ltd., No. 18 11th St Economical & Technological Development Zone, Beijing, 100176, China.
| | - Rong Zeng
- Department Neurology, Shanxi Provincial Peoples Hospital: Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| |
Collapse
|
13
|
Li X, Bedlack R. Evaluating emerging drugs in phase II & III for the treatment of amyotrophic lateral sclerosis. Expert Opin Emerg Drugs 2024; 29:93-102. [PMID: 38516735 DOI: 10.1080/14728214.2024.2333420] [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: 11/19/2023] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Amyotrophic Lateral Sclerosis is a rapidly progressive motor neuron disorder causing severe disability and premature death. Owing to the advances in uncovering ALS pathophysiology, efficient clinical trial design and research advocacy program, several disease-modifying drugs have been approved for treating ALS. Despite this progress, ALS remains a rapidly disabling and life shortening condition. There is a critical need for more effective therapies. AREAS COVERED Here, we reviewed the emerging ALS therapeutics undergoing phase II & III clinical trials. To identify the investigational drugs, we searched ALS and phase II/III trials that are active and recruiting or not yet recruiting on clinicaltrials.gov and Pharmaprojects database. EXPERT OPINION The current pipeline is larger and more diverse than ever, with drugs targeting potential genetic and retroviral causes of ALS and drugs targeting a wide array of downstream pathways, including RNA metabolism, protein aggregation, integrated stress response and neuroinflammation.We remain most excited about those that target direct causes of ALS, e.g. antisense oligonucleotides targeting causative genes. Drugs that eliminate abnormal protein aggregates are also up-and-coming. Eventually, because of the heterogeneity of ALS pathophysiology, biomarkers that determine which biological events are most important for an individual ALS patient are needed.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Neurology, Duke University, Durham, NC, USA
| | | |
Collapse
|
14
|
Xu D, Dai J, Tang L, Pan J, Zhang H. Nontargeted metabolomics reveals sequential changes in amino acid and ferroptosis-related metabolism in Parkinson's disease. Biomed Chromatogr 2024; 38:e5834. [PMID: 38308389 DOI: 10.1002/bmc.5834] [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: 10/25/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 02/04/2024]
Abstract
Parkinson's disease (PD) is inseparable from metabolic disorders but lacks assessment of specific metabolite alteration. To explore the sequential metabolic changes in PD progression, we evenly divided 78 C57BL/6 mice (10 weeks) into six groups (one control group and five experimental groups) and collected the hippocampus tissue of mice after treating with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, and probenecid (twice a week) at five periods (1, 2, 3, 4, and 5 weeks) for metabolome analysis. Our study identified 567 differentially abundant metabolites (DAMs) (total 4348 metabolites). Compared with controls, 145, 146, 171, 208, and 213 DAMs were obtained from the five experimental groups, respectively. Notably, 40 shared DAMs were present in five experimental groups, of which 22 shared DAMs formed a new metabolic network based on amino acid metabolism. Compared with group W3, 84 DAMs were identified in group W5, including 12 unique DAMs. DAMs in different stages of PD were significantly enriched in amino acid metabolism pathway, lipid metabolism pathway, and ferroptosis pathway. l-Glutamine, spermidine, and l-tryptophan were the key hubs in the whole metabolic process of PD. N-Formyl-l-methionine gradually increased in abundance with PD progression, whereas 5-methylcytosine gradually decreased. The study emphasized the sequential changes in DAMs in PD progression, stimulating subsequent studies.
Collapse
Affiliation(s)
- Delai Xu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jing Dai
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liuxing Tang
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Pan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua Zhang
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
15
|
Gonzalo-Consuegra C, Santos-García I, García-Toscano L, Martín-Baquero R, Rodríguez-Cueto C, Wittwer MB, Dzygiel P, Grether U, de Lago E, Fernández-Ruiz J. Involvement of CB 1 and CB 2 receptors in neuroprotective effects of cannabinoids in experimental TDP-43 related frontotemporal dementia using male mice. Biomed Pharmacother 2024; 174:116473. [PMID: 38522237 DOI: 10.1016/j.biopha.2024.116473] [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: 10/31/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUND The elevation of endocannabinoid levels through inhibiting their degradation afforded neuroprotection in CaMKIIα-TDP-43 mice, a conditional transgenic model of frontotemporal dementia. However, which cannabinoid receptors are mediating these benefits is still pending to be elucidated. METHODS We have investigated the involvement of the CB1 and the CB2 receptor using chronic treatments with selective ligands in CaMKIIα-TDP-43 mice, analysis of their cognitive deterioration with the Novel Object Recognition test, and immunostaining for neuronal and glial markers in two areas of interest in frontotemporal dementia. RESULTS Our results confirmed the therapeutic value of activating either the CB1 or the CB2 receptor, with improvements in the animal performance in the Novel Object Recognition test, preservation of pyramidal neurons, in particular in the medial prefrontal cortex, and attenuation of glial reactivity, in particular in the hippocampus. In addition, the activation of both CB1 and CB2 receptors reduced the elevated levels of TDP-43 in the medial prefrontal cortex of CaMKIIα-TDP-43 mice, an effect exerted by mechanisms that are currently under investigation. CONCLUSIONS These data reinforce the notion that the activation of CB1 and CB2 receptors may represent a promising therapy against TDP-43-induced neuropathology in frontotemporal dementia. Future studies will have to confirm these benefits, in particular with one of the selective CB2 agonists used here, which has been thoroughly characterized for clinical development.
Collapse
MESH Headings
- Animals
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/metabolism
- Male
- Neuroprotective Agents/pharmacology
- Mice, Transgenic
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB1/agonists
- Frontotemporal Dementia/drug therapy
- Frontotemporal Dementia/metabolism
- Frontotemporal Dementia/pathology
- Mice
- Cannabinoids/pharmacology
- Disease Models, Animal
- Prefrontal Cortex/drug effects
- Prefrontal Cortex/metabolism
- Prefrontal Cortex/pathology
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- DNA-Binding Proteins/metabolism
- Mice, Inbred C57BL
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hippocampus/pathology
Collapse
Affiliation(s)
- Claudia Gonzalo-Consuegra
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Irene Santos-García
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Laura García-Toscano
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Raquel Martín-Baquero
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carmen Rodríguez-Cueto
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Matthias B Wittwer
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Pawel Dzygiel
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Uwe Grether
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Eva de Lago
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| | - Javier Fernández-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| |
Collapse
|
16
|
Jin SW, Seong Y, Yoon D, Kwon YS, Song H. Dissolution of ribonucleoprotein condensates by the embryonic stem cell protein L1TD1. Nucleic Acids Res 2024; 52:3310-3326. [PMID: 38165001 PMCID: PMC11014241 DOI: 10.1093/nar/gkad1244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 11/22/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
L1TD1 is a cytoplasmic RNA-binding protein specifically expressed in pluripotent stem cells and, unlike its mouse ortholog, is essential for the maintenance of stemness in human cells. Although L1TD1 is the only known protein-coding gene domesticated from a LINE-1 (L1) retroelement, the functional legacy of its ancestral protein, ORF1p of L1, and how it is manifested in L1TD1 are still unknown. Here, we determined RNAs associated with L1TD1 and found that, like ORF1p, L1TD1 binds L1 RNAs and localizes to high-density ribonucleoprotein (RNP) condensates. Unexpectedly, L1TD1 enhanced the translation of a subset of mRNAs enriched in the condensates. L1TD1 depletion promoted the formation of stress granules in embryonic stem cells. In HeLa cells, ectopically expressed L1TD1 facilitated the dissolution of stress granules and granules formed by pathological mutations of TDP-43 and FUS. The glutamate-rich domain and the ORF1-homology domain of L1TD1 facilitated dispersal of the RNPs and induced autophagy, respectively. These results provide insights into how L1TD1 regulates gene expression in pluripotent stem cells. We propose that the ability of L1TD1 to dissolve stress granules may provide novel opportunities for treatment of neurodegenerative diseases caused by disturbed stress granule dynamics.
Collapse
Affiliation(s)
- Sang Woo Jin
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Youngmo Seong
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Dayoung Yoon
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Young-Soo Kwon
- Department of Integrative Bioscience & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Hoseok Song
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
17
|
Alayoubi AM, Iqbal M, Aman H, Hashmi JA, Alayadhi L, Al-Regaiey K, Basit S. Loss-of-function variant in spermidine/spermine N1-acetyl transferase like 1 (SATL1) gene as an underlying cause of autism spectrum disorder. Sci Rep 2024; 14:5765. [PMID: 38459140 PMCID: PMC10923806 DOI: 10.1038/s41598-024-56253-5] [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: 10/11/2023] [Accepted: 03/04/2024] [Indexed: 03/10/2024] Open
Abstract
Autism spectrum disorder (ASD) is a complicated, lifelong neurodevelopmental disorder affecting verbal and non-verbal communication and social interactions. ASD signs and symptoms appear early in development before the age of 3 years. It is unlikely for a person to acquire autism after a period of normal development. However, we encountered an 8-year-old child who developed ASD later in life although his developmental milestones were normal at the beginning of life. Sequencing the complete coding part of the genome identified a hemizygous nonsense mutation (NM_001367857.2):c.1803C>G; (p.Tyr601Ter) in the gene (SATL1) encoding spermidine/spermine N1-acetyl transferase like 1. Screening an ASD cohort of 28 isolated patients for the SATL1 gene identified another patient with the same variant. Although SATL1 mutations have not been associated with any human diseases, our data suggests that a mutation in SATL1 is the underlying cause of ASD in our cases. In mammals, mutations in spermine synthase (SMS), an enzyme needed for the synthesis of spermidine polyamine, have been reported in a syndromic form of the X-linked mental retardation. Moreover, SATL1 gene expression studies showed a relatively higher expression of SATL1 transcripts in ASD related parts of the brain including the cerebellum, amygdala and frontal cortex. Additionally, spermidine has been characterized in the context of learning and memory and supplementations with spermidine increase neuroprotective effects and decrease age-induced memory impairment. Furthermore, spermidine biosynthesis is required for spontaneous axonal regeneration and prevents α-synuclein neurotoxicity in invertebrate models. Thus, we report, for the first time, that a mutation in the SATL1 gene could be a contributing factor in the development of autistic symptoms in our patients.
Collapse
Affiliation(s)
- Abdulfatah M Alayoubi
- Department of Basic Medical Sciences, Taibah University Medina, Almadinah Almunawwarah, Saudi Arabia
| | - Muhammad Iqbal
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hassan Aman
- Al-Amal Psychiatry Hospital Medina, Almadinah Almunawwarrah, Saudi Arabia
| | - Jamil A Hashmi
- Center for Genetics and Inherited Diseases, Taibah University Medina, Almadinah Almunawwarrah, Saudi Arabia
| | - Laila Alayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
- Autism Research and Treatment Center, Riyadh, Saudi Arabia
| | - Khalid Al-Regaiey
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sulman Basit
- Department of Basic Medical Sciences, Taibah University Medina, Almadinah Almunawwarah, Saudi Arabia.
- Center for Genetics and Inherited Diseases, Taibah University Medina, Almadinah Almunawwarrah, Saudi Arabia.
- Department of Basic Medical Sciences, Taibah University Medina, Almadinah Almunawwarrah, Saudi Arabia.
| |
Collapse
|
18
|
Watchon M, Wright AL, Ahel HI, Robinson KJ, Plenderleith SK, Kuriakose A, Yuan KC, Laird AS. Spermidine treatment: induction of autophagy but also apoptosis? Mol Brain 2024; 17:15. [PMID: 38443995 PMCID: PMC10916058 DOI: 10.1186/s13041-024-01085-7] [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: 10/24/2023] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
Machado-Joseph disease (MJD), also known as spinocerebellar ataxia type 3, is a fatal neurodegenerative disease that causes loss of balance and motor co-ordination, eventually leading to paralysis. It is caused by the autosomal dominant inheritance of a long CAG trinucleotide repeat sequence within the ATXN3 gene, encoding for an expanded polyglutamine (polyQ) repeat sequence within the ataxin-3 protein. Ataxin-3 containing an expanded polyQ repeat is known to be highly prone to intraneuronal aggregation, and previous studies have demonstrated that protein quality control pathways, such as autophagy, are impaired in MJD patients and animal models of the disease. In this study, we tested the therapeutic potential of spermidine on zebrafish and rodent models of MJD to determine its capacity to induce autophagy and improve functional output. Spermidine treatment of transgenic MJD zebrafish induced autophagy and resulted in increased distances swum by the MJD zebrafish. Interestingly, treatment of the CMVMJD135 mouse model of MJD with spermidine added to drinking water did not produce any improvement in motor behaviour assays, neurological testing or neuropathology. In fact, wild type mice treated with spermidine were found to have decreased rotarod performance when compared to control animals. Immunoblot analysis of protein lysates extracted from mouse cerebellar tissue found little differences between the groups, except for an increased level of phospho-ULK1 in spermidine treated animals, suggesting that autophagy was indeed induced. As we detected decreased motor performance in wild type mice following treatment with spermidine, we conducted follow up studies into the effects of spermidine treatment in zebrafish. Interestingly, we found that in addition to inducing autophagy, spermidine treatment also induced apoptosis, particularly in wild type zebrafish. These findings suggest that spermidine treatment may not be therapeutically beneficial for the treatment of MJD, and in fact warrants caution due to the potential negative side effects caused by induction of apoptosis.
Collapse
Affiliation(s)
- Maxinne Watchon
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Rd, 2109, Sydney, NSW, Australia
| | - Amanda L Wright
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Rd, 2109, Sydney, NSW, Australia
| | - Holly I Ahel
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Rd, 2109, Sydney, NSW, Australia
| | - Katherine J Robinson
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Rd, 2109, Sydney, NSW, Australia
| | - Stuart K Plenderleith
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Rd, 2109, Sydney, NSW, Australia
| | - Andrea Kuriakose
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Rd, 2109, Sydney, NSW, Australia
| | - Kristy C Yuan
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Rd, 2109, Sydney, NSW, Australia
| | - Angela S Laird
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Level 1, 75 Talavera Rd, 2109, Sydney, NSW, Australia.
| |
Collapse
|
19
|
Jagaraj CJ, Shadfar S, Kashani SA, Saravanabavan S, Farzana F, Atkin JD. Molecular hallmarks of ageing in amyotrophic lateral sclerosis. Cell Mol Life Sci 2024; 81:111. [PMID: 38430277 PMCID: PMC10908642 DOI: 10.1007/s00018-024-05164-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/21/2024] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal, severely debilitating and rapidly progressing disorder affecting motor neurons in the brain, brainstem, and spinal cord. Unfortunately, there are few effective treatments, thus there remains a critical need to find novel interventions that can mitigate against its effects. Whilst the aetiology of ALS remains unclear, ageing is the major risk factor. Ageing is a slowly progressive process marked by functional decline of an organism over its lifespan. However, it remains unclear how ageing promotes the risk of ALS. At the molecular and cellular level there are specific hallmarks characteristic of normal ageing. These hallmarks are highly inter-related and overlap significantly with each other. Moreover, whilst ageing is a normal process, there are striking similarities at the molecular level between these factors and neurodegeneration in ALS. Nine ageing hallmarks were originally proposed: genomic instability, loss of telomeres, senescence, epigenetic modifications, dysregulated nutrient sensing, loss of proteostasis, mitochondrial dysfunction, stem cell exhaustion, and altered inter-cellular communication. However, these were recently (2023) expanded to include dysregulation of autophagy, inflammation and dysbiosis. Hence, given the latest updates to these hallmarks, and their close association to disease processes in ALS, a new examination of their relationship to pathophysiology is warranted. In this review, we describe possible mechanisms by which normal ageing impacts on neurodegenerative mechanisms implicated in ALS, and new therapeutic interventions that may arise from this.
Collapse
Affiliation(s)
- Cyril Jones Jagaraj
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sina Shadfar
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sara Assar Kashani
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Sayanthooran Saravanabavan
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Fabiha Farzana
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia
| | - Julie D Atkin
- MND Research Centre, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 75 Talavera Road, Sydney, NSW, 2109, Australia.
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
20
|
Potenza RL, Armida M, Popoli P. Can Some Anticancer Drugs Be Repurposed to Treat Amyotrophic Lateral Sclerosis? A Brief Narrative Review. Int J Mol Sci 2024; 25:1751. [PMID: 38339026 PMCID: PMC10855887 DOI: 10.3390/ijms25031751] [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: 12/14/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare progressive motor neuron disease that, due to its high complexity, still lacks effective treatments. Development of a new drug is a highly costly and time-consuming process, and the repositioning of approved drugs can represent an efficient strategy to provide therapeutic opportunities. This is particularly true for rare diseases, which are characterised by small patient populations and therefore attract little commercial interest. Based on the overlap between the biological background of cancer and neurodegeneration, the repurposing of antineoplastic drugs for ALS has been suggested. The objective of this narrative review was to summarise the current experimental evidence on the use of approved anticancer drugs in ALS. Specifically, anticancer drugs belonging to different classes were found to act on mechanisms involved in the ALS pathogenesis, and some of them proved to exert beneficial effects in ALS models. However, additional studies are necessary to confirm the real therapeutic potential of anticancer drugs for repositioning in ALS treatment.
Collapse
Affiliation(s)
- Rosa Luisa Potenza
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy; (M.A.); (P.P.)
| | | | | |
Collapse
|
21
|
Cunha-Oliveira T, Montezinho L, Simões RF, Carvalho M, Ferreiro E, Silva FSG. Mitochondria: A Promising Convergent Target for the Treatment of Amyotrophic Lateral Sclerosis. Cells 2024; 13:248. [PMID: 38334639 PMCID: PMC10854804 DOI: 10.3390/cells13030248] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/10/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by the progressive loss of motor neurons, for which current treatment options are limited. Recent studies have shed light on the role of mitochondria in ALS pathogenesis, making them an attractive therapeutic intervention target. This review contains a very comprehensive critical description of the involvement of mitochondria and mitochondria-mediated mechanisms in ALS. The review covers several key areas related to mitochondria in ALS, including impaired mitochondrial function, mitochondrial bioenergetics, reactive oxygen species, metabolic processes and energy metabolism, mitochondrial dynamics, turnover, autophagy and mitophagy, impaired mitochondrial transport, and apoptosis. This review also highlights preclinical and clinical studies that have investigated various mitochondria-targeted therapies for ALS treatment. These include strategies to improve mitochondrial function, such as the use of dichloroacetate, ketogenic and high-fat diets, acetyl-carnitine, and mitochondria-targeted antioxidants. Additionally, antiapoptotic agents, like the mPTP-targeting agents minocycline and rasagiline, are discussed. The paper aims to contribute to the identification of effective mitochondria-targeted therapies for ALS treatment by synthesizing the current understanding of the role of mitochondria in ALS pathogenesis and reviewing potential convergent therapeutic interventions. The complex interplay between mitochondria and the pathogenic mechanisms of ALS holds promise for the development of novel treatment strategies to combat this devastating disease.
Collapse
Affiliation(s)
- Teresa Cunha-Oliveira
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Liliana Montezinho
- Center for Investigation Vasco da Gama (CIVG), Escola Universitária Vasco da Gama, 3020-210 Coimbra, Portugal;
| | - Rui F. Simões
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Marcelo Carvalho
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Elisabete Ferreiro
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Filomena S. G. Silva
- CNC—Center for Neuroscience and Cell Biology, CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Mitotag Lda, Biocant Park, 3060-197 Cantanhede, Portugal
| |
Collapse
|
22
|
Ma H, Huo J, Xin C, Yang J, Liu Q, Dong H, Li R, Liu Y. RABGGTB plays a critical role in ALS pathogenesis. Brain Res Bull 2024; 206:110833. [PMID: 38042502 DOI: 10.1016/j.brainresbull.2023.110833] [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: 07/11/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/04/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease with unknown causes, which mainly affects motor neurons in the anterior horn of the spinal cord, brain stem, and cerebral cortex, also known as motor neuron disease. An important pathological feature of ALS is the formation of aggregates of mutant SOD1 protein, CTF25 of TDP-43, or other abnormal proteins in motor neurons, which require autophagy for degradation. Protein prenylation is known to participate in membrane association and proper localization of proteins. RABGGTB is the β subunit of GGTase II (one of the prenyltransferases) that can regulate autophagy via Rab7 geranylgeranylation. In this study, we overexpressed RABGGTB via lentiviral transfection in NSC34-hSOD1G93A and TDP-43 cells. Overexpression of RABGGTB improved ALS cell proliferation by facilitating autophagosome-lysosome fusion. Furthermore, the abnormal aggregation of SOD1 protein was reduced. This indicates that protein prenylation is important for the proliferation and autophagy of cells autophagy. Enhanced autophagy has been observed in two of the most widely used ALS cell models. These findings indicate the widespread applicability of prenylation in ALS. In summary, overexpression of RABGGTB improved the geranylgeranylation of the Rab7 protein and had a positive effect on cells. These findings provide insights into the development of a novel therapeutic strategy for ALS.
Collapse
Affiliation(s)
- Haiyang Ma
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Jia Huo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Cheng Xin
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Jing Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Qi Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Hui Dong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Rui Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China.
| | - Yaling Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China; The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China; Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China.
| |
Collapse
|
23
|
Zhang Y, Bai J, Cui Z, Li Y, Gao Q, Miao Y, Xiong B. Polyamine metabolite spermidine rejuvenates oocyte quality by enhancing mitophagy during female reproductive aging. NATURE AGING 2023; 3:1372-1386. [PMID: 37845508 DOI: 10.1038/s43587-023-00498-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 08/30/2023] [Indexed: 10/18/2023]
Abstract
Advanced age is a primary risk factor for female infertility due to reduced ovarian reserve and declining oocyte quality. However, as an important contributing factor, the role of metabolic regulation during reproductive aging is poorly understood. Here, we applied untargeted metabolomics to identify spermidine as a critical metabolite in ovaries to protect oocytes against aging. In particular, we found that the spermidine level was reduced in ovaries of aged mice and that supplementation with spermidine promoted follicle development, oocyte maturation, early embryonic development and female fertility of aged mice. By microtranscriptomic analysis, we further discovered that spermidine-induced recovery of oocyte quality was mediated by enhancement of mitophagy activity and mitochondrial function in aged mice, and this mechanism of action was conserved in porcine oocytes under oxidative stress. Altogether, our findings suggest that spermidine supplementation could represent a therapeutic strategy to ameliorate oocyte quality and reproductive outcome in cis-gender women and other persons trying to conceive at an advanced age. Future work is needed to test whether this approach can be safely and effectively translated to humans.
Collapse
Affiliation(s)
- Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Jie Bai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhaokang Cui
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Qian Gao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yilong Miao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Bo Xiong
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
24
|
Maragakis NJ, de Carvalho M, Weiss MD. Therapeutic targeting of ALS pathways: Refocusing an incomplete picture. Ann Clin Transl Neurol 2023; 10:1948-1971. [PMID: 37641443 PMCID: PMC10647018 DOI: 10.1002/acn3.51887] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023] Open
Abstract
Numerous potential amyotrophic lateral sclerosis (ALS)-relevant pathways have been hypothesized and studied preclinically, with subsequent translation to clinical trial. However, few successes have been observed with only modest effects. Along with an improved but incomplete understanding of ALS as a neurodegenerative disease is the evolution of more sophisticated and diverse in vitro and in vivo preclinical modeling platforms, as well as clinical trial designs. We highlight proposed pathological pathways that have been major therapeutic targets for investigational compounds. It is likely that the failures of so many of these therapeutic compounds may not have occurred because of lack of efficacy but rather because of a lack of preclinical modeling that would help define an appropriate disease pathway, as well as a failure to establish target engagement. These challenges are compounded by shortcomings in clinical trial design, including lack of biomarkers that could predict clinical success and studies that are underpowered. Although research investments have provided abundant insights into new ALS-relevant pathways, most have not yet been developed more fully to result in clinical study. In this review, we detail some of the important, well-established pathways, the therapeutics targeting them, and the subsequent clinical design. With an understanding of some of the shortcomings in translational efforts over the last three decades of ALS investigation, we propose that scientists and clinicians may choose to revisit some of these therapeutic pathways reviewed here with an eye toward improving preclinical modeling, biomarker development, and the investment in more sophisticated clinical trial designs.
Collapse
Affiliation(s)
| | - Mamede de Carvalho
- Faculdade de MedicinaInsqatituto de Medicina Molecular João Lobo Antunes, Centro Académico de Medicina de Lisboa, Universidade de LisboaLisbonPortugal
| | - Michael D. Weiss
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
25
|
Duranti E, Villa C. Muscle Involvement in Amyotrophic Lateral Sclerosis: Understanding the Pathogenesis and Advancing Therapeutics. Biomolecules 2023; 13:1582. [PMID: 38002264 PMCID: PMC10669302 DOI: 10.3390/biom13111582] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal condition characterized by the selective loss of motor neurons in the motor cortex, brainstem, and spinal cord. Muscle involvement, muscle atrophy, and subsequent paralysis are among the main features of this disease, which is defined as a neuromuscular disorder. ALS is a persistently progressive disease, and as motor neurons continue to degenerate, individuals with ALS experience a gradual decline in their ability to perform daily activities. Ultimately, muscle function loss may result in paralysis, presenting significant challenges in mobility, communication, and self-care. While the majority of ALS research has traditionally focused on pathogenic pathways in the central nervous system, there has been a great interest in muscle research. These studies were carried out on patients and animal models in order to better understand the molecular mechanisms involved and to develop therapies aimed at improving muscle function. This review summarizes the features of ALS and discusses the role of muscle, as well as examines recent studies in the development of treatments.
Collapse
Affiliation(s)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| |
Collapse
|
26
|
Richardson PJ, Smith DP, de Giorgio A, Snetkov X, Almond-Thynne J, Cronin S, Mead RJ, McDermott CJ, Shaw PJ. Janus kinase inhibitors are potential therapeutics for amyotrophic lateral sclerosis. Transl Neurodegener 2023; 12:47. [PMID: 37828541 PMCID: PMC10568794 DOI: 10.1186/s40035-023-00380-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a poorly treated multifactorial neurodegenerative disease associated with multiple cell types and subcellular organelles. As with other multifactorial diseases, it is likely that drugs will need to target multiple disease processes and cell types to be effective. We review here the role of Janus kinase (JAK)/Signal transducer and activator of transcription (STAT) signalling in ALS, confirm the association of this signalling with fundamental ALS disease processes using the BenevolentAI Knowledge Graph, and demonstrate that inhibitors of this pathway could reduce the ALS pathophysiology in neurons, glia, muscle fibres, and blood cells. Specifically, we suggest that inhibition of the JAK enzymes by approved inhibitors known as Jakinibs could reduce STAT3 activation and modify the progress of this disease. Analysis of the Jakinibs highlights baricitinib as a suitable candidate due to its ability to penetrate the central nervous system and exert beneficial effects on the immune system. Therefore, we recommend that this drug be tested in appropriately designed clinical trials for ALS.
Collapse
Affiliation(s)
| | | | | | | | | | - Sara Cronin
- BenevolentAI, 15 MetroTech Centre, 8th FL, Brooklyn, NY, 11201, USA
| | - Richard J Mead
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
| | - Christopher J McDermott
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, University of Sheffield and Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK
- NIHR Sheffield Biomedical Research Centre, University of Sheffield and Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
27
|
Nag S, Schneider JA. Limbic-predominant age-related TDP43 encephalopathy (LATE) neuropathological change in neurodegenerative diseases. Nat Rev Neurol 2023; 19:525-541. [PMID: 37563264 PMCID: PMC10964248 DOI: 10.1038/s41582-023-00846-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2023] [Indexed: 08/12/2023]
Abstract
TAR DNA-binding protein 43 (TDP43) is a focus of research in late-onset dementias. TDP43 pathology in the brain was initially identified in amyotrophic lateral sclerosis and frontotemporal lobar degeneration, and later in Alzheimer disease (AD), other neurodegenerative diseases and ageing. Limbic-predominant age-related TDP43 encephalopathy (LATE), recognized as a clinical entity in 2019, is characterized by amnestic dementia resembling AD dementia and occurring most commonly in adults over 80 years of age. Neuropathological findings in LATE, referred to as LATE neuropathological change (LATE-NC), consist of neuronal and glial cytoplasmic TDP43 localized predominantly in limbic areas with or without coexisting hippocampal sclerosis and/or AD neuropathological change and without frontotemporal lobar degeneration or amyotrophic lateral sclerosis pathology. LATE-NC is frequently associated with one or more coexisting pathologies, mainly AD neuropathological change. The focus of this Review is the pathology, genetic risk factors and nature of the cognitive impairments and dementia in pure LATE-NC and in LATE-NC associated with coexisting pathologies. As the clinical and cognitive profile of LATE is currently not easily distinguishable from AD dementia, it is important to develop biomarkers to aid in the diagnosis of this condition in the clinic. The pathogenesis of LATE-NC should be a focus of future research to form the basis for the development of preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Sukriti Nag
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Department of Pathology (Neuropathology), Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
28
|
Mandrioli J, D'Amico R, Zucchi E, De Biasi S, Banchelli F, Martinelli I, Simonini C, Lo Tartaro D, Vicini R, Fini N, Gianferrari G, Pinti M, Lunetta C, Gerardi F, Tarlarini C, Mazzini L, De Marchi F, Scognamiglio A, Sorarù G, Fortuna A, Lauria G, Bella ED, Caponnetto C, Meo G, Chio A, Calvo A, Cossarizza A. Randomized, double-blind, placebo-controlled trial of rapamycin in amyotrophic lateral sclerosis. Nat Commun 2023; 14:4970. [PMID: 37591957 PMCID: PMC10435464 DOI: 10.1038/s41467-023-40734-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
In preclinical studies rapamycin was found to target neuroinflammation, by expanding regulatory T cells, and affecting autophagy, two pillars of amyotrophic lateral sclerosis (ALS) pathogenesis. Herein we report a multicenter, randomized, double-blind trial, in 63 ALS patients who were randomly assigned in a 1:1:1 ratio to receive rapamycin 2 mg/m2/day,1 mg/m2/day or placebo (EUDRACT 2016-002399-28; NCT03359538). The primary outcome, the number of patients exhibiting an increase >30% in regulatory T cells from baseline to treatment end, was not attained. Secondary outcomes were changes from baseline of T, B, NK cell subpopulations, inflammasome mRNA expression and activation status, S6-ribosomal protein phosphorylation, neurofilaments; clinical outcome measures of disease progression; survival; safety and quality of life. Of the secondary outcomes, rapamycin decreased mRNA relative expression of the pro-inflammatory cytokine IL-18, reduced plasmatic IL-18 protein, and increased the percentage of classical monocytes and memory switched B cells, although no corrections were applied for multiple tests. In conclusion, we show that rapamycin treatment is well tolerated and provides reassuring safety findings in ALS patients, but further trials are necessary to understand the biological and clinical effects of this drug in ALS.
Collapse
Affiliation(s)
- Jessica Mandrioli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy.
| | - Roberto D'Amico
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Zucchi
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Neurosciences PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Ilaria Martinelli
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Cecilia Simonini
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Vicini
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Nicola Fini
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giulia Gianferrari
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Christian Lunetta
- NEuroMuscular Omnicenter, Serena Onlus Foundation, Milan, Italy
- Istituto Maugeri IRCCS Milano, Milan, Italy
| | | | | | - Letizia Mazzini
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Fabiola De Marchi
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Ada Scognamiglio
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padua, Padua, Italy
- Centro Regionale Specializzato Malattie del Motoneurone, Azienda Ospedale Università di Padova, Padua, Italy
| | - Andrea Fortuna
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Giuseppe Lauria
- 3rd Neurology Unit and ALS Centre, IRCCS 'Carlo Besta' Neurological Institute, Milan, Italy
| | - Eleonora Dalla Bella
- 3rd Neurology Unit and ALS Centre, IRCCS 'Carlo Besta' Neurological Institute, Milan, Italy
| | - Claudia Caponnetto
- Department of Neurosciences, Rehabilitatioņ Ophthalmology, Genetics, Mother and Child Disease, Ospedale Policlinico San Martino, Genova, Italy
| | - Giuseppe Meo
- Department of Neurosciences, Rehabilitatioņ Ophthalmology, Genetics, Mother and Child Disease, Ospedale Policlinico San Martino, Genova, Italy
| | - Adriano Chio
- 'Rita Levi Montalcini' Department of Neurosciences, ALS Centre, University of Turin and Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Andrea Calvo
- 'Rita Levi Montalcini' Department of Neurosciences, ALS Centre, University of Turin and Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
- National Institute for Cardiovascular Research, via Irnerio 48, 40126, Bologna, Italy
| |
Collapse
|
29
|
Tejwani L, Jung Y, Kokubu H, Sowmithra S, Ni L, Lee C, Sanders B, Lee PJ, Xiang Y, Luttik K, Soriano A, Yoon J, Park J, Ro HH, Ju H, Liao C, Tieze SM, Rigo F, Jafar-Nejad P, Lim J. Reduction of nemo-like kinase increases lysosome biogenesis and ameliorates TDP-43-related neurodegeneration. J Clin Invest 2023; 133:e138207. [PMID: 37384409 PMCID: PMC10425213 DOI: 10.1172/jci138207] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/28/2023] [Indexed: 07/01/2023] Open
Abstract
Protein aggregation is a hallmark of many neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS). Although mutations in TARDBP, encoding transactive response DNA-binding protein 43 kDa (TDP-43), account for less than 1% of all ALS cases, TDP-43-positive aggregates are present in nearly all ALS patients, including patients with sporadic ALS (sALS) or carrying other familial ALS-causing (fALS-causing) mutations. Interestingly, TDP-43 inclusions are also present in subsets of patients with frontotemporal dementia, Alzheimer's disease, and Parkinson's disease; therefore, methods of activating intracellular protein quality control machinery capable of clearing toxic cytoplasmic TDP-43 species may alleviate disease-related phenotypes. Here, we identify a function of nemo-like kinase (Nlk) as a negative regulator of lysosome biogenesis. Genetic or pharmacological reduction of Nlk increased lysosome formation and improved clearance of aggregated TDP-43. Furthermore, Nlk reduction ameliorated pathological, behavioral, and life span deficits in 2 distinct mouse models of TDP-43 proteinopathy. Because many toxic proteins can be cleared through the autophagy/lysosome pathway, targeted reduction of Nlk represents a potential approach to therapy development for multiple neurodegenerative disorders.
Collapse
Affiliation(s)
- Leon Tejwani
- Interdepartmental Neuroscience Program
- Department of Neuroscience, and
| | - Youngseob Jung
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hiroshi Kokubu
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sowmithra Sowmithra
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Luhan Ni
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Changwoo Lee
- Interdepartmental Neuroscience Program
- Department of Neuroscience, and
| | - Benjamin Sanders
- Interdepartmental Neuroscience Program
- Department of Neuroscience, and
| | - Paul J. Lee
- Interdepartmental Neuroscience Program
- Department of Neuroscience, and
| | - Yangfei Xiang
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kimberly Luttik
- Interdepartmental Neuroscience Program
- Department of Neuroscience, and
| | | | | | - Junhyun Park
- Interdepartmental Neuroscience Program
- Department of Neuroscience, and
| | | | - Hyoungseok Ju
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | | | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | - Janghoo Lim
- Interdepartmental Neuroscience Program
- Department of Neuroscience, and
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, and
- Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
30
|
Chu YP, Jin LW, Wang LC, Ho PC, Wei WY, Tsai KJ. Transthyretin attenuates TDP-43 proteinopathy by autophagy activation via ATF4 in FTLD-TDP. Brain 2023; 146:2089-2106. [PMID: 36355566 PMCID: PMC10411944 DOI: 10.1093/brain/awac412] [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: 05/06/2022] [Revised: 10/01/2022] [Accepted: 10/06/2022] [Indexed: 11/12/2022] Open
Abstract
TAR DNA-binding protein-43 (TDP-43) proteinopathies are accompanied by the pathological hallmark of cytoplasmic inclusions in the neurodegenerative diseases, including frontal temporal lobar degeneration-TDP and amyotrophic lateral sclerosis. We found that transthyretin accumulates with TDP-43 cytoplasmic inclusions in frontal temporal lobar degeneration-TDP human patients and transgenic mice, in which transthyretin exhibits dramatic expression decline in elderly mice. The upregulation of transthyretin expression was demonstrated to facilitate the clearance of cytoplasmic TDP-43 inclusions through autophagy, in which transthyretin induces autophagy upregulation via ATF4. Of interest, transthyretin upregulated ATF4 expression and promoted ATF4 nuclear import, presenting physical interaction. Neuronal expression of transthyretin in frontal temporal lobar degeneration-TDP mice restored autophagy function and facilitated early soluble TDP-43 aggregates for autophagosome targeting, ameliorating neuropathology and behavioural deficits. Thus, transthyretin conducted two-way regulations by either inducing autophagy activation or escorting TDP-43 aggregates targeted autophagosomes, suggesting that transthyretin is a potential modulator therapy for neurological disorders caused by TDP-43 proteinopathy.
Collapse
Affiliation(s)
- Yuan-Ping Chu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, CA, USA
| | - Liang-Chao Wang
- Division of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chuan Ho
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Yen Wei
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
31
|
Davis SE, Cook AK, Hall JA, Voskobiynyk Y, Carullo NV, Boyle NR, Hakim AR, Anderson KM, Hobdy KP, Pugh DA, Murchison CF, McMeekin LJ, Simmons M, Margolies KA, Cowell RM, Nana AL, Spina S, Grinberg LT, Miller BL, Seeley WW, Arrant AE. Patients with sporadic FTLD exhibit similar increases in lysosomal proteins and storage material as patients with FTD due to GRN mutations. Acta Neuropathol Commun 2023; 11:70. [PMID: 37118844 PMCID: PMC10148425 DOI: 10.1186/s40478-023-01571-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
Loss of function progranulin (GRN) mutations are a major autosomal dominant cause of frontotemporal dementia (FTD). Patients with FTD due to GRN mutations (FTD-GRN) develop frontotemporal lobar degeneration with TDP-43 pathology type A (FTLD-TDP type A) and exhibit elevated levels of lysosomal proteins and storage material in frontal cortex, perhaps indicating lysosomal dysfunction as a mechanism of disease. To investigate whether patients with sporadic FTLD exhibit similar signs of lysosomal dysfunction, we compared lysosomal protein levels, transcript levels, and storage material in patients with FTD-GRN or sporadic FTLD-TDP type A. We analyzed samples from frontal cortex, a degenerated brain region, and occipital cortex, a relatively spared brain region. In frontal cortex, patients with sporadic FTLD-TDP type A exhibited similar increases in lysosomal protein levels, transcript levels, and storage material as patients with FTD-GRN. In occipital cortex of both patient groups, most lysosomal measures did not differ from controls. Frontal cortex from a transgenic mouse model of TDP-opathy had similar increases in cathepsin D and lysosomal storage material, showing that TDP-opathy and neurodegeneration can drive these changes independently of progranulin. To investigate these changes in additional FTLD subtypes, we analyzed frontal cortical samples from patients with sporadic FTLD-TDP type C or Pick's disease, an FTLD-tau subtype. All sporadic FTLD groups had similar increases in cathepsin D activity, lysosomal membrane proteins, and storage material as FTD-GRN patients. However, patients with FTLD-TDP type C or Pick's disease did not have similar increases in lysosomal transcripts as patients with FTD-GRN or sporadic FTLD-TDP type A. Based on these data, accumulation of lysosomal proteins and storage material may be a common aspect of end-stage FTLD. However, the unique changes in gene expression in patients with FTD-GRN or sporadic FTLD-TDP type A may indicate distinct underlying lysosomal changes among FTLD subtypes.
Collapse
Affiliation(s)
- Skylar E Davis
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna K Cook
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Justin A Hall
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yuliya Voskobiynyk
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nancy V Carullo
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicholas R Boyle
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ahmad R Hakim
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kristian M Anderson
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kierra P Hobdy
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Derian A Pugh
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Charles F Murchison
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Laura J McMeekin
- Department of Neuroscience, Southern Research, Birmingham, AL, USA
| | - Micah Simmons
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Neuroscience, Southern Research, Birmingham, AL, USA
| | | | - Rita M Cowell
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Neuroscience, Southern Research, Birmingham, AL, USA
| | - Alissa L Nana
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, UCSF Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew E Arrant
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
32
|
Diab R, Pilotto F, Saxena S. Autophagy and neurodegeneration: Unraveling the role of C9ORF72 in the regulation of autophagy and its relationship to ALS-FTD pathology. Front Cell Neurosci 2023; 17:1086895. [PMID: 37006471 PMCID: PMC10060823 DOI: 10.3389/fncel.2023.1086895] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/01/2023] [Indexed: 03/18/2023] Open
Abstract
The proper functioning of the cell clearance machinery is critical for neuronal health within the central nervous system (CNS). In normal physiological conditions, the cell clearance machinery is actively involved in the elimination of misfolded and toxic proteins throughout the lifetime of an organism. The highly conserved and regulated pathway of autophagy is one of the important processes involved in preventing and neutralizing pathogenic buildup of toxic proteins that could eventually lead to the development of neurodegenerative diseases (NDs) such as Alzheimer’s disease or Amyotrophic lateral sclerosis (ALS). The most common genetic cause of ALS and frontotemporal dementia (FTD) is a hexanucleotide expansion consisting of GGGGCC (G4C2) repeats in the chromosome 9 open reading frame 72 gene (C9ORF72). These abnormally expanded repeats have been implicated in leading to three main modes of disease pathology: loss of function of the C9ORF72 protein, the generation of RNA foci, and the production of dipeptide repeat proteins (DPRs). In this review, we discuss the normal physiological role of C9ORF72 in the autophagy-lysosome pathway (ALP), and present recent research deciphering how dysfunction of the ALP synergizes with C9ORF72 haploinsufficiency, which together with the gain of toxic mechanisms involving hexanucleotide repeat expansions and DPRs, drive the disease process. This review delves further into the interactions of C9ORF72 with RAB proteins involved in endosomal/lysosomal trafficking, and their role in regulating various steps in autophagy and lysosomal pathways. Lastly, the review aims to provide a framework for further investigations of neuronal autophagy in C9ORF72-linked ALS-FTD as well as other neurodegenerative diseases.
Collapse
Affiliation(s)
- Rim Diab
- Department of Neurology, Center for Experimental Neurology, Inselspital University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Federica Pilotto
- Department of Neurology, Center for Experimental Neurology, Inselspital University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Smita Saxena
- Department of Neurology, Center for Experimental Neurology, Inselspital University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- *Correspondence: Smita Saxena,
| |
Collapse
|
33
|
Paul S, Dansithong W, Gandelman M, Figueroa KP, Zu T, Ranum LPW, Scoles DR, Pulst SM. Staufen Impairs Autophagy in Neurodegeneration. Ann Neurol 2023; 93:398-416. [PMID: 36151701 PMCID: PMC9892312 DOI: 10.1002/ana.26515] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The mechanistic target of rapamycin (mTOR) kinase is one of the master coordinators of cellular stress responses, regulating metabolism, autophagy, and apoptosis. We recently reported that staufen1 (STAU1), a stress granule (SG) protein, was overabundant in fibroblast cell lines from patients with spinocerebellar ataxia type 2 (SCA2), amyotrophic lateral sclerosis, frontotemporal degeneration, Huntington's, Alzheimer's, and Parkinson's diseases as well as animal models, and patient tissues. STAU1 overabundance is associated with mTOR hyperactivation and links SG formation with autophagy. Our objective was to determine the mechanism of mTOR regulation by STAU1. METHODS We determined STAU1 abundance with disease- and chemical-induced cellular stressors in patient cells and animal models. We also used RNA-binding assays to contextualize STAU1 interaction with MTOR mRNA. RESULTS STAU1 and mTOR were overabundant in bacterial artificial chromosome (BAC)-C9ORF72, ATXN2Q127 , and Thy1-TDP-43 transgenic mouse models. Reducing STAU1 levels in these mice normalized mTOR levels and activity and autophagy-related marker proteins. We also saw increased STAU1 levels in HEK293 cells transfected to express C9ORF72-relevant dipeptide repeats (DPRs). Conversely, DPR accumulations were not observed in cells treated by STAU1 RNA interference (RNAi). Overexpression of STAU1 in HEK293 cells increased mTOR levels through direct MTOR mRNA interaction, activating downstream targets and impairing autophagic flux. Targeting mTOR by rapamycin or RNAi normalized STAU1 abundance in an SCA2 cellular model. INTERPRETATION STAU1 interaction with mTOR drives its hyperactivation and inhibits autophagic flux in multiple models of neurodegeneration. Staufen, therefore, constitutes a novel target to modulate mTOR activity and autophagy, and for the treatment of neurodegenerative diseases. ANN NEUROL 2023;93:398-416.
Collapse
Affiliation(s)
- Sharan Paul
- Department of Neurology, University of Utah, Salt Lake City, UT
| | | | - Mandi Gandelman
- Department of Neurology, University of Utah, Salt Lake City, UT
| | | | - Tao Zu
- Center for NeuroGenetics and Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL
| | - Laura P W Ranum
- Center for NeuroGenetics and Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL
| | - Daniel R Scoles
- Department of Neurology, University of Utah, Salt Lake City, UT
| | - Stefan M Pulst
- Department of Neurology, University of Utah, Salt Lake City, UT
| |
Collapse
|
34
|
Sustained therapeutic benefits by transient reduction of TDP-43 using ENA-modified antisense oligonucleotides in ALS/FTD mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 31:353-366. [PMID: 36817728 PMCID: PMC9925842 DOI: 10.1016/j.omtn.2023.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 01/12/2023] [Indexed: 01/17/2023]
Abstract
The abnormal aggregation of TDP-43 into cytoplasmic inclusions in affected neurons is a pathological hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Although how TDP-43 forms cytoplasmic aggregates and causes neurodegeneration in patients with ALS/FTD remains unclear, reducing cellular TDP-43 levels is likely to prevent aggregation and to rescue neurons from TDP-43 toxicity. To address this issue, here we developed gapmer-type antisense oligonucleotides (ASOs) against human TDP-43 using 2'-O,4'-C-ethylene nucleic acids (ENAs), which are modified nucleic acids with high stability, and tested the therapeutic potential of lowering TDP-43 levels using ENA-modified ASOs. We demonstrated that intracerebroventricular administration of ENA-modified ASOs into a mouse model of ALS/FTD expressing human TDP-43 results in the efficient reduction of TDP-43 levels in the brain and spinal cord. Surprisingly, a single injection of ENA-modified ASOs into TDP-43 mice led to long-lasting improvement of behavioral abnormalities and the suppression of cytoplasmic TDP-43 aggregation, even after TDP-43 levels had returned to the initial levels. Our results demonstrate that transient reduction of TDP-43 using ENA-modified ASOs leads to sustained therapeutic benefits in vivo, indicating the possibility of a disease-modifying therapy by lowering TDP-43 levels for the treatment of the TDP-43 proteinopathies, including ALS/FTD.
Collapse
|
35
|
Lopez-Herdoiza MB, Bauché S, Wilmet B, Le Duigou C, Roussel D, Frah M, Béal J, Devely G, Boluda S, Frick P, Bouteiller D, Dussaud S, Guillabert P, Dalle C, Dumont M, Camuzat A, Saracino D, Barbier M, Bruneteau G, Ravassard P, Neumann M, Nicole S, Le Ber I, Brice A, Latouche M. C9ORF72 knockdown triggers FTD-like symptoms and cell pathology in mice. Front Cell Neurosci 2023; 17:1155929. [PMID: 37138765 PMCID: PMC10149765 DOI: 10.3389/fncel.2023.1155929] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/27/2023] [Indexed: 05/05/2023] Open
Abstract
The GGGGCC intronic repeat expansion within C9ORF72 is the most common genetic cause of ALS and FTD. This mutation results in toxic gain of function through accumulation of expanded RNA foci and aggregation of abnormally translated dipeptide repeat proteins, as well as loss of function due to impaired transcription of C9ORF72. A number of in vivo and in vitro models of gain and loss of function effects have suggested that both mechanisms synergize to cause the disease. However, the contribution of the loss of function mechanism remains poorly understood. We have generated C9ORF72 knockdown mice to mimic C9-FTD/ALS patients haploinsufficiency and investigate the role of this loss of function in the pathogenesis. We found that decreasing C9ORF72 leads to anomalies of the autophagy/lysosomal pathway, cytoplasmic accumulation of TDP-43 and decreased synaptic density in the cortex. Knockdown mice also developed FTD-like behavioral deficits and mild motor phenotypes at a later stage. These findings show that C9ORF72 partial loss of function contributes to the damaging events leading to C9-FTD/ALS.
Collapse
Affiliation(s)
| | - Stephanie Bauché
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Baptiste Wilmet
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Caroline Le Duigou
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Delphine Roussel
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Magali Frah
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Jonas Béal
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Gabin Devely
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Susana Boluda
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Petra Frick
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | | | - Sébastien Dussaud
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Pierre Guillabert
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Carine Dalle
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Magali Dumont
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Agnes Camuzat
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Dario Saracino
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Mathieu Barbier
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Gaelle Bruneteau
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | | | - Manuela Neumann
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Neuropathology, Tübingen University Hospital, Tübingen, Germany
| | - Sophie Nicole
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Isabelle Le Ber
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Alexis Brice
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
| | - Morwena Latouche
- Institut du Cerveau–Paris Brain Institute–ICM, Inserm, CNRS, Paris, France
- EPHE, Neurogenetics Team, PSL Research University, Paris, France
- *Correspondence: Morwena Latouche,
| |
Collapse
|
36
|
Therapeutic Effect of Rapamycin on TDP-43-Related Pathogenesis in Ischemic Stroke. Int J Mol Sci 2022; 24:ijms24010676. [PMID: 36614118 PMCID: PMC9820757 DOI: 10.3390/ijms24010676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
Stroke is a major cause of death and disability across the world, and its detrimental impact should not be underestimated. Therapies are available and effective for ischemic stroke (e.g., thrombolytic recanalization and mechanical thrombectomy); however, there are limitations to therapeutic interventions. Recanalization therapy has developed dramatically, while the use of adjunct neuroprotective agents as complementary therapies remains deficient. Pathological TAR DNA-binding protein (TDP-43) has been identified as a major component of insoluble aggregates in numerous neurodegenerative pathologies, including ALS, FTLD and Alzheimer's disease. Here, we show that increased pathological TDP-43 fractions accompanied by impaired mitochondrial function and increased gliosis were observed in an ischemic stroke rat model, suggesting a pathological role of TDP-43 in ischemic stroke. In ischemic rats administered rapamycin, the insoluble TDP-43 fraction was significantly decreased in the ischemic cortex region, accompanied by a recovery of mitochondrial function, the attenuation of cellular apoptosis, a reduction in infarct areas and improvements in motor defects. Accordingly, our results suggest that rapamycin provides neuroprotective benefits not only by ameliorating pathological TDP-43 levels, but also by reversing mitochondrial function and attenuating cell apoptosis in ischemic stroke.
Collapse
|
37
|
Choi SH, Yousefian-Jazi A, Hyeon SJ, Nguyen PTT, Chu J, Kim S, Kim S, Ryu HL, Kowall NW, Ryu H, Lee J. Modulation of histone H3K4 dimethylation by spermidine ameliorates motor neuron survival and neuropathology in a mouse model of ALS. J Biomed Sci 2022; 29:106. [PMID: 36536341 PMCID: PMC9764677 DOI: 10.1186/s12929-022-00890-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by progressive paralysis due to motor neuron degeneration. It has been proposed that epigenetic modification and transcriptional dysregulation may contribute to motor neuron death. In this study, we investigate the basis for therapeutic approaches to target lysine-specific histone demethylase 1 (LSD1) and elucidate the mechanistic role of LSD1-histone H3K4 signaling pathway in ALS pathogenesis. METHODS In order to examine the role of spermidine (SD), we administered SD to an animal model of ALS (G93A) and performed neuropathological analysis, body weight, and survival evaluation. RESULTS Herein, we found that LSD1 activity is increased while levels of H3K4me2, a substrate of LSD1, is decreased in cellular and animal models of ALS. SD administration modulated the LSD1 activity and restored H3K4me2 levels in ChAT-positive motor neurons in the lumbar spinal cord of ALS mice. SD prevented cellular damage by improving the number and size of motor neurons in ALS mice. SD administration also reduced GFAP-positive astrogliogenesis in the white and gray matter of the lumbar spinal cord, improving the neuropathology of ALS mice. Moreover, SD administration improved the rotarod performance and gait analysis of ALS mice. Finally, SD administration delayed disease onset and prolonged the lifespan of ALS (G93A) transgenic mice. CONCLUSION Together, modulating epigenetic targets such as LSD1 by small compounds may be a useful therapeutic strategy for treating ALS.
Collapse
Affiliation(s)
- Seung-Hye Choi
- grid.35541.360000000121053345K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 South Korea
| | - Ali Yousefian-Jazi
- grid.35541.360000000121053345K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 South Korea
| | - Seung Jae Hyeon
- grid.35541.360000000121053345K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 South Korea
| | - Phuong Thi Thanh Nguyen
- grid.35541.360000000121053345K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 South Korea ,grid.412786.e0000 0004 1791 8264KIST School, Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Seoul, 02792 South Korea
| | - Jiyeon Chu
- grid.35541.360000000121053345K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 South Korea ,grid.222754.40000 0001 0840 2678Integrated Biomedical and Life Science Department, Graduate School, Korea University, Seoul, 02841 South Korea
| | - Sojung Kim
- grid.35541.360000000121053345K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 South Korea
| | - Suhyun Kim
- grid.35541.360000000121053345K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 South Korea
| | - Hannah L. Ryu
- grid.189504.10000 0004 1936 7558Department of Neurology, Boston University Alzheimer’s Disease Research Center, Boston University School of Medicine, Boston, MA 02118 USA
| | - Neil W. Kowall
- grid.189504.10000 0004 1936 7558Department of Neurology, Boston University Alzheimer’s Disease Research Center, Boston University School of Medicine, Boston, MA 02118 USA ,grid.410370.10000 0004 4657 1992VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA 02130 USA
| | - Hoon Ryu
- grid.35541.360000000121053345K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792 South Korea ,grid.412786.e0000 0004 1791 8264KIST School, Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Seoul, 02792 South Korea
| | - Junghee Lee
- grid.189504.10000 0004 1936 7558Department of Neurology, Boston University Alzheimer’s Disease Research Center, Boston University School of Medicine, Boston, MA 02118 USA ,grid.410370.10000 0004 4657 1992VA Boston Healthcare System, 150 S. Huntington Avenue, Boston, MA 02130 USA
| |
Collapse
|
38
|
Riku Y, Yoshida M, Iwasaki Y, Sobue G, Katsuno M, Ishigaki S. TDP-43 Proteinopathy and Tauopathy: Do They Have Pathomechanistic Links? Int J Mol Sci 2022; 23:ijms232415755. [PMID: 36555399 PMCID: PMC9779029 DOI: 10.3390/ijms232415755] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Transactivation response DNA binding protein 43 kDa (TDP-43) and tau are major pathological proteins of neurodegenerative disorders, of which neuronal and glial aggregates are pathological hallmarks. Interestingly, accumulating evidence from neuropathological studies has shown that comorbid TDP-43 pathology is observed in a subset of patients with tauopathies, and vice versa. The concomitant pathology often spreads in a disease-specific manner and has morphological characteristics in each primary disorder. The findings from translational studies have suggested that comorbid TDP-43 or tau pathology has clinical impacts and that the comorbid pathology is not a bystander, but a part of the disease process. Shared genetic risk factors or molecular abnormalities between TDP-43 proteinopathies and tauopathies, and direct interactions between TDP-43 and tau aggregates, have been reported. Further investigations to clarify the pathogenetic factors that are shared by a broad spectrum of neurodegenerative disorders will establish key therapeutic targets.
Collapse
Affiliation(s)
- Yuichi Riku
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
- Correspondence: or
| | - Mari Yoshida
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
| | - Yasushi Iwasaki
- Institute for Medical Science of Aging, Aichi Medical University, Nagakute 480-1195, Japan
| | - Gen Sobue
- Graduate School of Medicine, Aichi Medical University, Nagakute 480-1195, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya 744-8550, Japan
| | - Shinsuke Ishigaki
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu 520-2192, Japan
| |
Collapse
|
39
|
Tedesco B, Ferrari V, Cozzi M, Chierichetti M, Casarotto E, Pramaggiore P, Mina F, Piccolella M, Cristofani R, Crippa V, Rusmini P, Galbiati M, Poletti A. The role of autophagy-lysosomal pathway in motor neuron diseases. Biochem Soc Trans 2022; 50:1489-1503. [PMID: 36111809 PMCID: PMC9704526 DOI: 10.1042/bst20220778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 10/22/2023]
Abstract
Motor neuron diseases (MNDs) include a broad group of diseases in which neurodegeneration mainly affects upper and/or lower motor neurons (MNs). Although the involvement of specific MNs, symptoms, age of onset, and progression differ in MNDs, the main pathogenic mechanism common to most MNDs is represented by proteostasis alteration and proteotoxicity. This pathomechanism may be directly related to mutations in genes encoding proteins involved in the protein quality control system, particularly the autophagy-lysosomal pathway (ALP). Alternatively, proteostasis alteration can be caused by aberrant proteins that tend to misfold and to aggregate, two related processes that, over time, cannot be properly handled by the ALP. Here, we summarize the main ALP features, focusing on different routes utilized to deliver substrates to the lysosome and how the various ALP pathways intersect with the intracellular trafficking of membranes and vesicles. Next, we provide an overview of the mutated genes that have been found associated with MNDs, how these gene products are involved in different steps of ALP and related processes. Finally, we discuss how autophagy can be considered a valid therapeutic target for MNDs treatment focusing on traditional autophagy modulators and on emerging approaches to overcome their limitations.
Collapse
Affiliation(s)
- Barbara Tedesco
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
- Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Veronica Ferrari
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Marta Cozzi
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Marta Chierichetti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Elena Casarotto
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Paola Pramaggiore
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Francesco Mina
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Margherita Piccolella
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Valeria Crippa
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Paola Rusmini
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Mariarita Galbiati
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
40
|
Park SK, Park S, Liebman SW. TDP-43 Toxicity in Yeast Is Associated with a Reduction in Autophagy, and Deletions of TIP41 and PBP1 Counteract These Effects. Viruses 2022; 14:2264. [PMID: 36298819 PMCID: PMC9607128 DOI: 10.3390/v14102264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/27/2022] Open
Abstract
When human TDP-43 is overexpressed in yeast it is toxic and forms cytoplasmic aggregates. The mechanism of this toxicity is unknown. Genetic screens for TDP-43 toxicity modifiers in the yeast system previously identified proteins, including PBP1, that enhance TDP-43 toxicity. The determination in yeast that deletion of PBP1 reduces TDP-43 toxicity while overexpression enhances toxicity, led to the discovery that its human homolog, ATXN2, is associated with ALS risk. Thus, the yeast system has relevance to human disease. We now show that deletion of a new yeast gene, tip41Δ, likewise suppresses TDP-43 toxicity. We also found that TDP-43 overexpression and toxicity is associated with reduced autophagy. This is consistent with findings in other systems that increasing autophagy reduces TDP-43 toxicity and is in contrast to a report of enhanced autophagy when TDP-43 was overexpressed in yeast. Interestingly, we found that deletions of PBP1 and TIP41, which reduced TDP-43 toxicity, eliminated TDP-43's inhibition of autophagy. This suggests that toxicity of TDP-43 expressed in yeast is in part due to its inhibition of autophagy and that deletions of PBP1 and TIP41 may reduce TDP-43 toxicity by preventing TDP-43 from inhibiting autophagy.
Collapse
Affiliation(s)
| | | | - Susan W. Liebman
- Department of Pharmacology, University of Nevada, Reno, NV 89557, USA
| |
Collapse
|
41
|
Wu H, Wang J, Jiang H, Liu X, Sun X, Chen Y, Hu C, Wang Z, Han T, Sun C, Wei W, Jiang W. The association of dietary spermidine with all-cause mortality and CVD mortality: The U.S. National Health and Nutrition Examination Survey, 2003 to 2014. Front Public Health 2022; 10:949170. [PMID: 36249217 PMCID: PMC9554131 DOI: 10.3389/fpubh.2022.949170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/25/2022] [Indexed: 01/21/2023] Open
Abstract
Background Current studies on the protective effects of dietary spermidine (SPD) on cardiovascular disease (CVD) are mainly limited to animal studies, and the relationship between dietary SPD and CVD mortality remains inconclusive. Objective This study aims to evaluate the association between dietary SPD intake and CVD and all-cause mortality. Methods A total of 23,894 people enrolled in the National Health and Nutrition Examination Survey (NHANES) from 2003 to 2014 were recruited for this study. The dietary intake of SPD from 11 specific food origins and total SPD was categorized into tertiles or quartiles. Cox proportional hazard regression models were developed to evaluate the association of SPD intake with CVD and all-cause mortalities. Results Among the 23,894 participants, 2,365 deaths, including 736 deaths due to CVD, were documented. After adjustment for potential confounders, compared with participants in the lowest quartile, participants in the highest quartile of total SPD had a significantly lower risk of CVD mortality (HR = 0.68, 95% CI: 0.51-0.91) and all-cause mortality (HR = 0.70, 95% CI: 0.60-0.82); participants in the highest tertiles or quartiles of vegetable-derived SPD, cereal-derived SPD, legume-derived SPD, nut-derived SPD, and cheese-derived SPD had a lower risk of CVD mortality (HR vegetable - derivedSPD = 0.68, 95% CI: 0.54-0.86; HR cereal - derivedSPD = 0.75, 95% CI: 0.57-0.97; HR legume - derivedSPD = 0.68, 95% CI: 0.52-0.88; HR nut - derivedSPD = 0.66, 95% CI: 0.53-0.80; HR cheese - derivedSPD = 0.68, 95% CI: 0.52-0.88) and all-cause mortality (HR vegetable - derivedSPD = 0.73, 95% CI: 0.64-0.84; HR cereal - derivedSPD = 0.80, 95% CI: 0.69-0.93; HR legume - derivedSPD = 0.70, 95% CI: 0.60-0.80;HR nut - derivedSPD = 0.72, 95% CI: 0.64-0.81; HR cheese - derivedSPD = 0.70, 95% CI: 0.61-0.81) than those in the lowest tertiles or quartiles. Moreover, subgroup analysis showed consistent associations among the people with hypertension and hyperlipidemia. Conclusion Higher intake of dietary SPD is associated with decreased risk of CVD and all-cause mortality, and among specific food origin SPD, SPD derived from vegetables, cereals, legumes, nuts, and cheese was associated with reduced CVD and all-cause mortality.
Collapse
Affiliation(s)
- Huanyu Wu
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Jianing Wang
- Department of Cerebrovascular Disease, The Fifth Afliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Hongyan Jiang
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Xin Liu
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Xinyi Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Yunyan Chen
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Cong Hu
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Zheng Wang
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Tianshu Han
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China,Changhao Sun
| | - Wei Wei
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China,Wei Wei
| | - Wenbo Jiang
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, China,*Correspondence: Wenbo Jiang
| |
Collapse
|
42
|
Pu M, Tai Y, Yuan L, Zhang Y, Guo H, Hao Z, Chen J, Qi X, Wang G, Tao Z, Ren J. The contribution of proteasomal impairment to autophagy activation by C9orf72 poly-GA aggregates. Cell Mol Life Sci 2022; 79:501. [PMID: 36036324 PMCID: PMC11803000 DOI: 10.1007/s00018-022-04518-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/18/2022] [Accepted: 08/08/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Poly-GA, a dipeptide repeat protein unconventionally translated from GGGGCC (G4C2) repeat expansions in C9orf72, is abundant in C9orf72-related amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) (C9orf72-ALS/FTD). Although the poly-GA aggregates have been identified in C9orf72-ALS/FTD neurons, the effects on UPS (ubiquitin-proteasome system) and autophagy and their exact molecular mechanisms have not been fully elucidated. RESULTS Herein, our in vivo experiments indicate that the mice expressing ploy-GA with 150 repeats instead of 30 repeats exhibit significant aggregates in cells. Mice expressing 150 repeats ploy-GA shows behavioral deficits and activates autophagy in the brain. In vitro findings suggest that the poly-GA aggregates influence proteasomal by directly binding proteasome subunit PSMD2. Subsequently, the poly-GA aggregates activate phosphorylation and ubiquitination of p62 to recruit autophagosomes. Ultimately, the poly-GA aggregates lead to compensatory activation of autophagy. In vivo studies further reveal that rapamycin (autophagy activator) treatment significantly improves the degenerative symptoms and alleviates neuronal injury in mice expressing 150 repeats poly-GA. Meanwhile, rapamycin administration to mice expressing 150 repeats poly-GA reduces neuroinflammation and aggregates in the brain. CONCLUSION In summary, we elucidate the relationship between poly-GA in the proteasome and autophagy: when poly-GA forms complexes with the proteasome, it recruits autophagosomes and affects proteasome function. Our study provides support for further promoting the comprehension of the pathogenesis of C9orf72, which may bring a hint for the exploration of rapamycin for the treatment of ALS/FTD.
Collapse
Affiliation(s)
- Mei Pu
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yusi Tai
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Luyang Yuan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Zhang
- Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Huijie Guo
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zongbing Hao
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Jing Chen
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
| | - Xinming Qi
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China
| | - Guanghui Wang
- Laboratory of Molecular Neuropathology, Jiangsu Key Laboratory of Neuropsychiatric Diseases and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| | - Zhouteng Tao
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China.
- State Key Laboratory of Transient Optics and Photonics, Xi'an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi'an, China.
| | - Jin Ren
- Center for Drug Safety Evaluation and Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
43
|
Hu L, Mao S, Lin L, Bai G, Liu B, Mao J. Stress granules in the spinal muscular atrophy and amyotrophic lateral sclerosis: The correlation and promising therapy. Neurobiol Dis 2022; 170:105749. [PMID: 35568100 DOI: 10.1016/j.nbd.2022.105749] [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: 01/22/2022] [Revised: 03/27/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022] Open
Abstract
Increasing genetic and biochemical evidence has broadened our view of the pathomechanisms that lead to Spinal muscular atrophy (SMA) and Amyotrophic lateral sclerosis (ALS), two fatal neurodegenerative diseases with similar symptoms and causes. Stress granules are dynamic cytosolic storage hubs for mRNAs in response to stress exposures, that are evolutionarily conserved cytoplasmic RNA granules in somatic cells. A lot of previous studies have shown that the impaired stress granules are crucial events in SMA/ALS pathogenesis. In this review, we described the key stress granules related RNA binding proteins (SMN, TDP-43, and FUS) involved in SMA/ALS, summarized the reported mutations in these RNA binding proteins involved in SMA/ALS pathogenesis, and discussed the mechanisms through which stress granules dynamics participate in the diseases. Meanwhile, we described the applications and limitation of current therapies targeting SMA/ALS. We futher proposed the promising targets on stress granules in the future therapeutic interventions of SMA/ALS.
Collapse
Affiliation(s)
- LiDan Hu
- the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China.
| | - Shanshan Mao
- the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Li Lin
- the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Guannan Bai
- the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Bingjie Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jianhua Mao
- the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China.
| |
Collapse
|
44
|
Amalyan S, Tamboli S, Lazarevich I, Topolnik D, Bouman LH, Topolnik L. Enhanced motor cortex output and disinhibition in asymptomatic female mice with C9orf72 genetic expansion. Cell Rep 2022; 40:111043. [PMID: 35793625 DOI: 10.1016/j.celrep.2022.111043] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 04/29/2022] [Accepted: 06/12/2022] [Indexed: 11/28/2022] Open
Abstract
Information and action coding by cortical circuits relies on a balanced dialogue between excitation and inhibition. Circuit hyperexcitability is considered a potential pathophysiological mechanism in various brain disorders, but the underlying deficits, especially at early disease stages, remain largely unknown. We report that asymptomatic female mice carrying the chromosome 9 open reading frame 72 (C9orf72) repeat expansion, which represents a high-prevalence genetic abnormality for human amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) spectrum disorder, exhibit abnormal motor cortex output. The number of primary motor cortex (M1) layer 5 pyramidal neurons is reduced in asymptomatic mice, with the surviving neurons receiving a decreased inhibitory drive that results in a higher M1 output, specifically during high-speed animal locomotion. Importantly, using deep-learning algorithms revealed that speed-dependent M1 output predicts the likelihood of C9orf72 genetic expansion. Our data link early circuit abnormalities with a gene mutation in asymptomatic ALS/FTLD carriers.
Collapse
Affiliation(s)
- Sona Amalyan
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada
| | - Suhel Tamboli
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada
| | - Ivan Lazarevich
- École Normale Supérieure, Laboratoire de Neurosciences Cognitives, Group for Neural Theory, Paris, France
| | - Dimitry Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada
| | - Leandra Harriet Bouman
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada
| | - Lisa Topolnik
- Department of Biochemistry, Microbiology and Bio-informatics, Laval University, Québec, QC, Canada; Neuroscience Axis, CHU de Québec Research Center (CHUL), Québec, QC, Canada.
| |
Collapse
|
45
|
Hayes LR, Kalab P. Emerging Therapies and Novel Targets for TDP-43 Proteinopathy in ALS/FTD. Neurotherapeutics 2022; 19:1061-1084. [PMID: 35790708 PMCID: PMC9587158 DOI: 10.1007/s13311-022-01260-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2022] [Indexed: 10/17/2022] Open
Abstract
Nuclear clearance and cytoplasmic mislocalization of the essential RNA binding protein, TDP-43, is a pathologic hallmark of amyotrophic lateral sclerosis, frontotemporal dementia, and related neurodegenerative disorders collectively termed "TDP-43 proteinopathies." TDP-43 mislocalization causes neurodegeneration through both loss and gain of function mechanisms. Loss of TDP-43 nuclear RNA processing function destabilizes the transcriptome by multiple mechanisms including disruption of pre-mRNA splicing, the failure of repression of cryptic exons, and retrotransposon activation. The accumulation of cytoplasmic TDP-43, which is prone to aberrant liquid-liquid phase separation and aggregation, traps TDP-43 in the cytoplasm and disrupts a host of downstream processes including the trafficking of RNA granules, local translation within axons, and mitochondrial function. In this review, we will discuss the TDP-43 therapy development pipeline, beginning with therapies in current and upcoming clinical trials, which are primarily focused on accelerating the clearance of TDP-43 aggregates. Then, we will look ahead to emerging strategies from preclinical studies, first from high-throughput genetic and pharmacologic screens, and finally from mechanistic studies focused on the upstream cause(s) of TDP-43 disruption in ALS/FTD. These include modulation of stress granule dynamics, TDP-43 nucleocytoplasmic shuttling, RNA metabolism, and correction of aberrant splicing events.
Collapse
Affiliation(s)
- Lindsey R Hayes
- Johns Hopkins School of Medicine, Dept. of Neurology, Baltimore, MD, USA.
| | - Petr Kalab
- Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
46
|
Chua JP, Bedi K, Paulsen MT, Ljungman M, Tank EMH, Kim ES, McBride JP, Colón-Mercado JM, Ward ME, Weisman LS, Barmada SJ. Myotubularin-related phosphatase 5 is a critical determinant of autophagy in neurons. Curr Biol 2022; 32:2581-2595.e6. [PMID: 35580604 PMCID: PMC9233098 DOI: 10.1016/j.cub.2022.04.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 02/18/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022]
Abstract
Autophagy is a conserved, multi-step process of capturing proteolytic cargo in autophagosomes for lysosome degradation. The capacity to remove toxic proteins that accumulate in neurodegenerative disorders attests to the disease-modifying potential of the autophagy pathway. However, neurons respond only marginally to conventional methods for inducing autophagy, limiting efforts to develop therapeutic autophagy modulators for neurodegenerative diseases. The determinants underlying poor autophagy induction in neurons and the degree to which neurons and other cell types are differentially sensitive to autophagy stimuli are incompletely defined. Accordingly, we sampled nascent transcript synthesis and stabilities in fibroblasts, induced pluripotent stem cells (iPSCs), and iPSC-derived neurons (iNeurons), thereby uncovering a neuron-specific stability of transcripts encoding myotubularin-related phosphatase 5 (MTMR5). MTMR5 is an autophagy suppressor that acts with its binding partner, MTMR2, to dephosphorylate phosphoinositides critical for autophagy initiation and autophagosome maturation. We found that MTMR5 is necessary and sufficient to suppress autophagy in iNeurons and undifferentiated iPSCs. Using optical pulse labeling to visualize the turnover of endogenously encoded proteins in live cells, we observed that knockdown of MTMR5 or MTMR2, but not the unrelated phosphatase MTMR9, significantly enhances neuronal degradation of TDP-43, an autophagy substrate implicated in several neurodegenerative diseases. Our findings thus establish a regulatory mechanism of autophagy intrinsic to neurons and targetable for clearing disease-related proteins in a cell-type-specific manner. In so doing, our results not only unravel novel aspects of neuronal biology and proteostasis but also elucidate a strategy for modulating neuronal autophagy that could be of high therapeutic potential for multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Jason P. Chua
- Department of Neurology, Johns Hopkins Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Lead contact
| | - Karan Bedi
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Michelle T. Paulsen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI, USA
| | - Mats Ljungman
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center and Center for RNA Biomedicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Erin S. Kim
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Jonathon P. McBride
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Michael E. Ward
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Lois S. Weisman
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Sami J. Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
47
|
Ojaimi YA, Dangoumau A, Alarcan H, Hergesheimer R, Vourc'h P, Corcia P, Lanznaster D, Blasco H. TAR DNA-binding protein of 43 kDa (TDP-43) and amyotrophic lateral sclerosis (ALS): a promising therapeutic target. Expert Opin Ther Targets 2022; 26:575-592. [PMID: 35652285 DOI: 10.1080/14728222.2022.2083958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease that lacks an effective treatment. Aggregates of the TAR DNA-binding protein-43 (TDP-43) are observed in 97% of all ALS cases, thus making this protein a major therapeutic target in ALS. . AREAS COVERED The authors describe the major cellular functions of TDP-43 and the features and consequences of TDP-43 proteinopathy. Drawing from fundamental and preclinical studies on cellular and animal TDP-43 models of ALS and selected clinical trials, the major pathways that have been targeted for the mitigation of TDP-43 pathology in ALS are discussed. The authors provide insights on the approaches targeting the tendency of TDP-43 for aggregation, defective nucleocytoplasmic transport, dysfunctional proteostasis, abnormal stress granule dynamics, and pathological post-translational modifications of TDP-43. EXPERT OPINION The complexity of ALS and TDP-43 proteinopathy generates challenges for the development of novel therapeutic approaches. However, the critical involvement of TDP-43 in the initiation and progression of ALS, makes it a promising therapeutic target. Further research should be centered on the development of precision strategies, consideration of patient subgroups, the prevention of the mislocalization of TDP-43 and restoration of the lost functions of TPD-43. .
Collapse
Affiliation(s)
| | | | - Hugo Alarcan
- UMR 1253 iBrain, Université de Tours, Tours, France.,Laboratoire de biochimie et biologie moléculaire, CHRU Bretonneau, Tours, France
| | | | - Patrick Vourc'h
- UMR 1253 iBrain, Université de Tours, Tours, France.,Laboratoire de biochimie et biologie moléculaire, CHRU Bretonneau, Tours, France
| | - Philippe Corcia
- Laboratoire de biochimie et biologie moléculaire, CHRU Bretonneau, Tours, France.,Service de neurologie, CHRU Bretonneau, Tours, France
| | | | - Hélène Blasco
- UMR 1253 iBrain, Université de Tours, Tours, France.,Laboratoire de biochimie et biologie moléculaire, CHRU Bretonneau, Tours, France
| |
Collapse
|
48
|
Houghton OH, Mizielinska S, Gomez-Suaga P. The Interplay Between Autophagy and RNA Homeostasis: Implications for Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Front Cell Dev Biol 2022; 10:838402. [PMID: 35573690 PMCID: PMC9096704 DOI: 10.3389/fcell.2022.838402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/14/2022] [Indexed: 01/18/2023] Open
Abstract
Amyotrophic lateral sclerosis and frontotemporal dementia are neurodegenerative disorders that lie on a disease spectrum, sharing genetic causes and pathology, and both without effective therapeutics. Two pathways that have been shown to play major roles in disease pathogenesis are autophagy and RNA homeostasis. Intriguingly, there is an increasing body of evidence suggesting a critical interplay between these pathways. Autophagy is a multi-stage process for bulk and selective clearance of malfunctional cellular components, with many layers of regulation. Although the majority of autophagy research focuses on protein degradation, it can also mediate RNA catabolism. ALS/FTD-associated proteins are involved in many stages of autophagy and autophagy-mediated RNA degradation, particularly converging on the clearance of persistent pathological stress granules. In this review, we will summarise the progress in understanding the autophagy-RNA homeostasis interplay and how that knowledge contributes to our understanding of the pathobiology of ALS/FTD.
Collapse
Affiliation(s)
- O H Houghton
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom.,UK Dementia Research Institute at King's College London, London, United Kingdom
| | - S Mizielinska
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom.,UK Dementia Research Institute at King's College London, London, United Kingdom
| | - P Gomez-Suaga
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom.,Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, Cáceres, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), Cáceres, Spain
| |
Collapse
|
49
|
Lumkwana D, Peddie C, Kriel J, Michie LL, Heathcote N, Collinson L, Kinnear C, Loos B. Investigating the Role of Spermidine in a Model System of Alzheimer’s Disease Using Correlative Microscopy and Super-resolution Techniques. Front Cell Dev Biol 2022; 10:819571. [PMID: 35656544 PMCID: PMC9152225 DOI: 10.3389/fcell.2022.819571] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 04/07/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Spermidine has recently received major attention for its potential therapeutic benefits in the context of neurodegeneration, cancer, and aging. However, it is unclear whether concentration dependencies of spermidine exist, to differentially enhance autophagic flux. Moreover, the relationship between low or high autophagy activity relative to basal neuronal autophagy flux and subsequent protein clearance as well as cellular toxicity has remained largely unclear. Methods: Here, we used high-resolution imaging and biochemical techniques to investigate the effects of a low and of a high concentration of spermidine on autophagic flux, neuronal toxicity, and protein clearance in in vitro models of paraquat (PQ) induced neuronal toxicity and amyloid precursor protein (APP) overexpression, as well as in an in vivo model of PQ-induced rodent brain injury. Results: Our results reveal that spermidine induces autophagic flux in a concentration-dependent manner, however the detectable change in the autophagy response critically depends on the specificity and sensitivity of the method employed. By using correlative imaging techniques through Super-Resolution Structured Illumination Microscopy (SR-SIM) and Focused Ion Beam Scanning Electron Microscopy (FIB-SEM), we demonstrate that spermidine at a low concentration induces autophagosome formation capable of large volume clearance. In addition, we provide evidence of distinct, context-dependent protective roles of spermidine in models of Alzheimer’s disease. In an in vitro environment, a low concentration of spermidine protected against PQ-induced toxicity, while both low and high concentrations provided protection against cytotoxicity induced by APP overexpression. In the in vivo scenario, we demonstrate brain region-specific susceptibility to PQ-induced neuronal toxicity, with the hippocampus being highly susceptible compared to the cortex. Regardless of this, spermidine administered at both low and high dosages protected against paraquat-induced toxicity. Conclusions: Taken together, our results demonstrate that firstly, administration of spermidine may present a favourable therapeutic strategy for the treatment of Alzheimer’s disease and secondly, that concentration and dosage-dependent precision autophagy flux screening may be more critical for optimal autophagy and cell death control than previously thought.
Collapse
Affiliation(s)
- D. Lumkwana
- Microscopy and Imaging Translational Technology Platform, Cancer Research UK, University College London, London, United Kingdom
- *Correspondence: D. Lumkwana,
| | - C. Peddie
- Science Technology Platform, Electron Microscopy, Francis Crick Institute, London, United Kingdom
| | - J. Kriel
- Central Analytical Facilities, Electron Microscopy Unit, Stellenbosch University, Stellenbosch, South Africa
| | - L. L. Michie
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - N. Heathcote
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - L. Collinson
- Science Technology Platform, Electron Microscopy, Francis Crick Institute, London, United Kingdom
| | - C. Kinnear
- DST/NRF Centre of Excellence in Biomedical Tuberculosis Research, SAMRC Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - B. Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
50
|
Wang C, Zheng C. Using Caenorhabditis elegans to Model Therapeutic Interventions of Neurodegenerative Diseases Targeting Microbe-Host Interactions. Front Pharmacol 2022; 13:875349. [PMID: 35571084 PMCID: PMC9096141 DOI: 10.3389/fphar.2022.875349] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/08/2022] [Indexed: 12/02/2022] Open
Abstract
Emerging evidence from both clinical studies and animal models indicates the importance of the interaction between the gut microbiome and the brain in the pathogenesis of neurodegenerative diseases (NDs). Although how microbes modulate neurodegeneration is still mostly unclear, recent studies have started to probe into the mechanisms for the communication between microbes and hosts in NDs. In this review, we highlight the advantages of using Caenorhabditis elegans (C. elegans) to disentangle the microbe-host interaction that regulates neurodegeneration. We summarize the microbial pro- and anti-neurodegenerative factors identified using the C. elegans ND models and the effects of many are confirmed in mouse models. Specifically, we focused on the role of bacterial amyloid proteins, such as curli, in promoting proteotoxicity and neurodegeneration by cross-seeding the aggregation of endogenous ND-related proteins, such as α-synuclein. Targeting bacterial amyloid production may serve as a novel therapeutic strategy for treating NDs, and several compounds, such as epigallocatechin-3-gallate (EGCG), were shown to suppress neurodegeneration at least partly by inhibiting curli production. Because bacterial amyloid fibrils contribute to biofilm formation, inhibition of amyloid production often leads to the disruption of biofilms. Interestingly, from a list of 59 compounds that showed neuroprotective effects in C. elegans and mouse ND models, we found that about half of them are known to inhibit bacterial growth or biofilm formation, suggesting a strong correlation between the neuroprotective and antibiofilm activities. Whether these potential therapeutics indeed protect neurons from proteotoxicity by inhibiting the cross-seeding between bacterial and human amyloid proteins awaits further investigations. Finally, we propose to screen the long list of antibiofilm agents, both FDA-approved drugs and novel compounds, for their neuroprotective effects and develop new pharmaceuticals that target the gut microbiome for the treatment of NDs. To this end, the C. elegans ND models can serve as a platform for fast, high-throughput, and low-cost drug screens that target the microbe-host interaction in NDs.
Collapse
Affiliation(s)
| | - Chaogu Zheng
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|