1
|
Szabó A, Borkúti P, Kovács Z, Kristó I, Vilmos P. Recent advances in nuclear actin research. Nucleus 2025; 16:2498643. [PMID: 40320716 PMCID: PMC12054378 DOI: 10.1080/19491034.2025.2498643] [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: 02/14/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
Actin was first observed in the nucleus more than sixty years ago but research on nuclear actin did not receive significant attention for the next forty years. It only started to accelerate around the year 2000, when the first convincing experimental data emerged indicating that actin participates in essential nuclear processes. Today, we know that actin is involved in transcription, replication, DNA repair, chromatin remodeling, and participates in the determination of nuclear shape and size. In this paper we review the results of the last five years of increasingly intensive research on nuclear actin, because on one hand, the field has expanded with several new directions during this time, and on the other hand, the enrichment of our picture of nuclear actin will certainly provide a more solid foundation and new impetus for its future investigation.
Collapse
Affiliation(s)
- Anikó Szabó
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Péter Borkúti
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Zoltán Kovács
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Ildikó Kristó
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| | - Péter Vilmos
- HUN-REN Biological Research Centre, Institute of Genetics, Szeged, Hungary
| |
Collapse
|
2
|
Zheng H, Wang H, Zhang D, Gan Y, Wu Y, Xiang W, Fu P. Identification of therapeutic targets and immune landscape in glioblastoma through crosstalk with glioma-associated mesenchymal stem cells. Int Immunopharmacol 2025; 150:114228. [PMID: 39946771 DOI: 10.1016/j.intimp.2025.114228] [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: 11/16/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Glioma-associated mesenchymal stem cells (GA-MSCs) are one of the key factors limiting the effectiveness of glioblastoma (GBM) treatment and contributing to poor patient prognosis, making them a potential therapeutic target for GBM. In-depth research into the complex crosstalk between GA-MSCs and GBM cells not only aids in understanding the mechanisms of GBM progression but also provides valuable insights for developing new potential drugs. METHODS We conducted a comprehensive bioinformatics analysis aimed at identifying shared dysregulated genes between GBM and GA-MSCs. Through hub gene enrichment and immune infiltration analyses, we explored key molecular pathways and the immune landscape. Additionally, Cox regression analysis was employed to identify key factors influencing overall survival in GBM. The expression patterns and functional roles of hub genes were validated across various cancer types and datasets. Finally, dynamic simulations were used to assess the binding affinity of potential drugs to the targets, further supporting their potential as therapeutic candidates. RESULTS We identified 32 candidate genes primarily involved in the 1-kappa-B kinase/NF-kappa-B and MAPK signaling pathways, both of which played critical roles in tumor survival, proliferation, and invasion. Notable hub genes included DUSP1, FYN, FLNC, FN1, G3BP1, MYO1B, and WLS, each contributing uniquely to GBM progression. Among them, FLNC was highlighted as a key regulatory factor in GBM progression. Molecular dynamics simulations further revealed its potential as a therapeutic target, particularly demonstrating a high binding affinity with staurosporine. Additionally, a high proportion of dendritic cells contributed to the formation of the GBM immune microenvironment. CONCLUSIONS This study revealed the co-expression patterns and metabolic pathways between GA-MSCs and GBM, providing new insights into the molecular mechanisms of GBM progression. Targeting FLNC with staurosporine presents a promising therapeutic strategy for GBM. Aditionally, targeting the shared pathways of both may offer a valuable approach for treating malignant brain tumors.
Collapse
Affiliation(s)
- Haoyang Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Duo Zhang
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 China
| | - Yong Gan
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030 China
| | - Yuyi Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China.
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China.
| |
Collapse
|
3
|
Cullell N, Caruana G, Elias-Mas A, Delgado-Sanchez A, Artero C, Buongiorno MT, Almería M, Ray NJ, Correa SAL, Krupinski J. Glymphatic system clearance and Alzheimer's disease risk: a CSF proteome-wide study. Alzheimers Res Ther 2025; 17:31. [PMID: 39891246 PMCID: PMC11786353 DOI: 10.1186/s13195-024-01612-7] [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: 08/29/2024] [Accepted: 10/28/2024] [Indexed: 02/03/2025]
Abstract
BACKGROUND The emerging evidence of the role of the glymphatic system (GS) in Alzheimer's disease (AD) provides new opportunities for intervention from the earliest stages of the disease. The aim of the study is to evaluate the efficacy of GS in AD to identify new disease biomarkers. METHODS We performed a two-stage proteomic study to evaluate the GS health using intravenous gadolinium-based contrast agent (GBCA) with serial T1 3T magnetic resonance imaging (MRI) in individuals with amnestic mild cognitive impairment (aMCI). In Stage 1 (evaluated in the Cohort 1 of aMCI participants (n = 11)), we correlated the levels of 7K cerebrospinal fluid (CSF) proteins (estimated by SOMAscan) with GS health in 78 Freesurfer-segmented brain regions of interest (ROIs). RESULTS A total of seven different proteins were significantly associated with GS health (p-value < 6.4 × 10-4). The stronger correlations were identified for NSUN6, GRAAK, OLFML3, ACTN2, RUXF, SHPS1 and TIM-4. A pathway enrichment analysis revealed that the proteins associated with GS health were mainly implicated in neurodegenerative processes, immunity and inflammation. In Stage 2, we validated these proteomic results in a new cohort of aMCI participants (with and without evidence of AD pathology in CSF (aMCI(-) and aMCI/AD( +); n = 22 and 7, respectively) and healthy controls (n = 10). Proteomic prediction models were generated in each ROI. These were compared with demographic-only models for identifying participants with aMCI(-) and aMCI/AD( +) vs controls. This analysis was repeated to determine if the models could identify those with aMCI/AD( +) from both aMCI(-) and controls. The proteomic models were found to outperform the demographic-only models. CONCLUSIONS Our study identifies proteins linked with GS health and involved the immune system in aMCI participants.
Collapse
Affiliation(s)
- Natalia Cullell
- Fundació per a Docència I Recerca, MútuaTerrassa, Terrassa, Barcelona, Spain.
- Department of Neurology, F.Ass. MútuaTerrassa, Terrassa, Barcelona, Spain.
| | - Giovanni Caruana
- Department of Radiology, F.Ass. MútuaTerrassa, Terrassa, Barcelona, Spain
| | - Andrea Elias-Mas
- Department of Radiology, F.Ass. MútuaTerrassa, Terrassa, Barcelona, Spain
- Institute for Research and Innovation Parc Taulí (I3PT), Sabadell, Spain
- Genetics Doctorate Program, Universitat de Barcelona (UB), Barcelona, Spain
| | - Ariane Delgado-Sanchez
- Department of Psychology, Brooks Building, Faculty of Science and Education, Manchester Metropolitan University, Manchester, UK
| | - Cristina Artero
- Department of Neurology, F.Ass. MútuaTerrassa, Terrassa, Barcelona, Spain
| | | | - Marta Almería
- Department of Neurology, F.Ass. MútuaTerrassa, Terrassa, Barcelona, Spain
| | - Nicola J Ray
- Department of Psychology, Brooks Building, Faculty of Science and Education, Manchester Metropolitan University, Manchester, UK
| | - Sonia A L Correa
- Department of Life Sciences John Dalton Building, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Jerzy Krupinski
- Fundació per a Docència I Recerca, MútuaTerrassa, Terrassa, Barcelona, Spain.
- Department of Life Sciences John Dalton Building, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK.
| |
Collapse
|
4
|
P A H, Basavaraju N, Gupta A, Kommaddi RP. Actin Cytoskeleton at the Synapse: An Alzheimer's Disease Perspective. Cytoskeleton (Hoboken) 2025. [PMID: 39840749 DOI: 10.1002/cm.21993] [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: 02/28/2024] [Revised: 12/18/2024] [Accepted: 01/05/2025] [Indexed: 01/23/2025]
Abstract
Actin, a ubiquitous and highly conserved cytoskeletal protein, plays a pivotal role in various cellular functions such as structural support, facilitating cell motility, and contributing to the dynamic processes of synaptic function. Apart from its established role in inducing morphological changes, recent developments in the field indicate an active involvement of actin in modulating both the structure and function of pre- and postsynaptic terminals. Within the presynapse, it is involved in the organization and trafficking of synaptic vesicles, contributing to neurotransmitter release. In the postsynapse, actin dynamically modulates dendritic spines, influencing the postsynaptic density organization and anchoring of neurotransmitter receptors. In addition, the dynamic interplay of actin at the synapse underscores its essential role in regulating neural communication. This review strives to offer a comprehensive overview of the recent advancements in understanding the multifaceted role of the actin cytoskeleton in synaptic functions. By emphasizing its aberrant regulation, we aim to provide valuable insights into the underlying mechanisms of Alzheimer's disease pathophysiology.
Collapse
Affiliation(s)
- Haseena P A
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Nimisha Basavaraju
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | - Anant Gupta
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
- Manipal Academy of Higher Education, Manipal, India
| | | |
Collapse
|
5
|
Biel N, Rashid F, Natua S, Wang TY, Chou TF, Nguyen TVP, Golding I, Kalsotra A, Sokac AM. Reducing Cofilin dosage makes embryos resilient to heat stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.02.631102. [PMID: 39803506 PMCID: PMC11722379 DOI: 10.1101/2025.01.02.631102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
In addition to regulating the actin cytoskeleton, Cofilin also senses and responds to environmental stress. Cofilin can promote cell survival or death depending on context. Yet, many aspects of Cofilin's role in survival need clarification. Here, we show that exposing early Drosophila embryos to mild heat stress (32°C) induces a Cofilin-mediated Actin Stress Response and upregulation of heat- and ER- stress response genes. However, these responses do not alleviate the negative impacts of heat exposure. Instead, heat stressed embryos show downregulation of hundreds of developmental genes, including determinants of the embryonic body plan, and are less likely to hatch as larvae and adults. Remarkably, reducing Cofilin dosage blunts induction of all stress response pathways, mitigates downregulation of developmental genes, and completely rescues survival. Thus, Cofilin intersects with multiple stress response pathways, and modulates the transcriptomic response to heat stress. Strikingly, Cofilin knockdown emerges as a potent pro-survival manipulation for embryos.
Collapse
Affiliation(s)
- Natalie Biel
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX, 77030 USA
- Department of Cell and Developmental Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Faizan Rashid
- Department of Cell and Developmental Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- These authors contributed equally
| | - Subhashis Natua
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- These authors contributed equally
| | - Ting-Yu Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Tsui-Fen Chou
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Thu Vu Phuc Nguyen
- Department of Physics, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Present address: Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Ido Golding
- Department of Physics, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Anna Marie Sokac
- Department of Cell and Developmental Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
- Lead contact
| |
Collapse
|
6
|
Sun ZG, Murrell M. Cofilin-Mediated Filament Softening and Crosslinking Counterbalance to Enhance Actin Network Flexibility. PHYSICAL REVIEW LETTERS 2024; 133:218402. [PMID: 39642486 DOI: 10.1103/physrevlett.133.218402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/30/2024] [Indexed: 12/09/2024]
Abstract
Filamentous-actin (F-actin) crosslinking within the cell cytoskeleton mediates the transmission of mechanical forces, enabling changes in cell shape, as occurs during cell division and cell migration. Crosslinking by actin binding proteins (ABPs) generally increases the connectivity of the F-actin network, but also increases network rigidity. As a result, there is a narrow range in the concentration of crosslinker protein at which F-actin networks are both connected and labile. Another ABP, cofilin, severs F-actin filaments at high pH through increasing their bending flexibility and concentrating mechanical stress, inducing fragmentation. By contrast, at lower pH, cofilin increases filament flexibility yet does not sever. Instead, it forms disulfide bonds, which crosslink F-actin into bundles, and bundles into networks. Here, we combine light microscopy and rheology to determine the impact of two potentially opposing effects on the mechanics of F-actin networks-increased flexibility at the filament level, and increased connectivity at the network level. Indeed, by linear rheology, we find that these mechanisms are counterbalanced, such that cofilactin network moduli are only weakly dependent on cofilin concentration over a broad range, in contrast to the dramatic stiffening that occurs with F-actin crosslinking protein. Further, by nonlinear rheology, the network stiffens at a higher stress than crosslinking protein, indicative of a broader range in which the material remains flexible. These results may enable F-actin networks to increase connectivity without heavy penalties to rigidity, and thus provide a new route to modulating active polymer mechanics unseen using traditional F-actin accessory proteins.
Collapse
Affiliation(s)
- Zachary Gao Sun
- Department of Physics, Yale University, 217 Prospect Street, New Haven, Connecticut 06511, USA
- Systems Biology Institute, Yale University, West Haven, Connecticut 06516, USA
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, Connecticut 06520, USA
| | - Michael Murrell
- Department of Physics, Yale University, 217 Prospect Street, New Haven, Connecticut 06511, USA
- Systems Biology Institute, Yale University, West Haven, Connecticut 06516, USA
- Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, Connecticut 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
7
|
Schmid ET, Schinaman JM, Liu-Abramowicz N, Williams KS, Walker DW. Accumulation of F-actin drives brain aging and limits healthspan in Drosophila. Nat Commun 2024; 15:9238. [PMID: 39455560 PMCID: PMC11512044 DOI: 10.1038/s41467-024-53389-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The actin cytoskeleton is a key determinant of cell structure and homeostasis. However, possible tissue-specific changes to actin dynamics during aging, notably brain aging, are not understood. Here, we show that there is an age-related increase in filamentous actin (F-actin) in Drosophila brains, which is counteracted by prolongevity interventions. Critically, decreasing F-actin levels in aging neurons prevents age-onset cognitive decline and extends organismal healthspan. Mechanistically, we show that autophagy, a recycling process required for neuronal homeostasis, is disabled upon actin dysregulation in the aged brain. Remarkably, disrupting actin polymerization in aged animals with cytoskeletal drugs restores brain autophagy to youthful levels and reverses cellular hallmarks of brain aging. Finally, reducing F-actin levels in aging neurons slows brain aging and promotes healthspan in an autophagy-dependent manner. Our data identify excess actin polymerization as a hallmark of brain aging, which can be targeted to reverse brain aging phenotypes and prolong healthspan.
Collapse
Affiliation(s)
- Edward T Schmid
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Joseph M Schinaman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Naomi Liu-Abramowicz
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kylie S Williams
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - David W Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
8
|
Prange SE, Bhakta IN, Sysoeva D, Jean GE, Madisetti A, Le HHN, Duong LU, Hwu PT, Melton JG, Thompson-Peer KL. Dendrite injury triggers neuroprotection in Drosophila models of neurodegenerative disease. Sci Rep 2024; 14:24766. [PMID: 39433621 PMCID: PMC11494097 DOI: 10.1038/s41598-024-74670-4] [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: 05/09/2024] [Accepted: 09/26/2024] [Indexed: 10/23/2024] Open
Abstract
Dendrite defects and loss are early cellular alterations observed across neurodegenerative diseases that play a role in early disease pathogenesis. Dendrite degeneration can be modeled by expressing pathogenic polyglutamine disease transgenes in Drosophila neurons in vivo. Here, we show that we can protect against dendrite loss in neurons modeling neurodegenerative polyglutamine diseases through injury to a single primary dendrite branch. We find that this neuroprotection is specific to injury-induced activation of dendrite regeneration: neither injury to the axon nor injury just to surrounding tissues induces this response. We show that the mechanism of this regenerative response is stabilization of the actin (but not microtubule) cytoskeleton. We also demonstrate that this regenerative response may extend to other neurodegenerative diseases. Together, we provide evidence that activating dendrite regeneration pathways has the potential to slow-or even reverse-dendrite loss in neurodegenerative disease.
Collapse
Affiliation(s)
- Sydney E Prange
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, Irvine, CA, USA
| | - Isha N Bhakta
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Daria Sysoeva
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Grace E Jean
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Anjali Madisetti
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Hieu H N Le
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Ly U Duong
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Patrick T Hwu
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA
| | - Jaela G Melton
- Center for the Neurobiology of Learning and Memory, Irvine, CA, USA
| | - Katherine L Thompson-Peer
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA.
- Center for the Neurobiology of Learning and Memory, Irvine, CA, USA.
- Sue and Bill Gross Stem Cell Research Center, Irvine, CA, USA.
- Reeve-Irvine Research Center, Irvine, CA, USA.
| |
Collapse
|
9
|
Mann N, Hill J, Wang K, Hughes RM. OptoProfilin: A Single Component Biosensor of Applied Cellular Stress. Chembiochem 2024; 25:e202400007. [PMID: 38457348 PMCID: PMC11218921 DOI: 10.1002/cbic.202400007] [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: 01/03/2024] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/10/2024]
Abstract
The actin cytoskeleton is a biosensor of cellular stress and a potential prognosticator of human disease. In particular, aberrant cytoskeletal structures such as stress granules formed in response to energetic and oxidative stress are closely linked to ageing, cancer, cardiovascular disease, and viral infection. Whether these cytoskeletal phenomena can be harnessed for the development of biosensors for cytoskeletal dysfunction and, by extension, disease progression, remains an open question. In this work, we describe the design and development of an optogenetic iteration of profilin, an actin monomer binding protein with critical functions in cytoskeletal dynamics. We demonstrate that this optically activated profilin ('OptoProfilin') can act as an optically triggered biosensor of applied cellular stress in select immortalized cell lines. Notably, OptoProfilin is a single component biosensor, likely increasing its utility for experimentalists. While a large body of preexisting work closely links profilin activity with cellular stress and neurodegenerative disease, this, to our knowledge, is the first example of profilin as an optogenetic biosensor of stress-induced changes in the cytoskeleton.
Collapse
Affiliation(s)
- Noah Mann
- Department of Chemistry, East Carolina University, Greenville, North Carolina, United States
| | - Jahiem Hill
- Department of Chemistry, East Carolina University, Greenville, North Carolina, United States
| | - Kenneth Wang
- Department of Chemistry, Davidson College, Davidson, North Carolina, United States
| | - Robert M Hughes
- Department of Chemistry, East Carolina University, Greenville, North Carolina, United States
| |
Collapse
|
10
|
Oliveira da Silva MI, Santejo M, Babcock IW, Magalhães A, Minamide LS, Won SJ, Castillo E, Gerhardt E, Fahlbusch C, Swanson RA, Outeiro TF, Taipa R, Ruff M, Bamburg JR, Liz MA. α-Synuclein triggers cofilin pathology and dendritic spine impairment via a PrP C-CCR5 dependent pathway. Cell Death Dis 2024; 15:264. [PMID: 38615035 PMCID: PMC11016063 DOI: 10.1038/s41419-024-06630-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/15/2024]
Abstract
Cognitive dysfunction and dementia are critical symptoms of Lewy Body dementias (LBD). Specifically, alpha-synuclein (αSyn) accumulation in the hippocampus leading to synaptic dysfunction is linked to cognitive deficits in LBD. Here, we investigated the pathological impact of αSyn on hippocampal neurons. We report that either αSyn overexpression or αSyn pre-formed fibrils (PFFs) treatment triggers the formation of cofilin-actin rods, synapse disruptors, in cultured hippocampal neurons and in the hippocampus of synucleinopathy mouse models and of LBD patients. In vivo, cofilin pathology is present concomitantly with synaptic impairment and cognitive dysfunction. Rods generation prompted by αSyn involves the co-action of the cellular prion protein (PrPC) and the chemokine receptor 5 (CCR5). Importantly, we show that CCR5 inhibition, with a clinically relevant peptide antagonist, reverts dendritic spine impairment promoted by αSyn. Collectively, we detail the cellular and molecular mechanism through which αSyn disrupts hippocampal synaptic structure and we identify CCR5 as a novel therapeutic target to prevent synaptic impairment and cognitive dysfunction in LBD.
Collapse
Grants
- R01 AG049668 NIA NIH HHS
- R01 NS105774 NINDS NIH HHS
- R43 AG071064 NIA NIH HHS
- S10 OD025127 NIH HHS
- Applicable Funding Source FEDER - Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020 – Operacional Programme for Competitiveness and Internationalisation (POCI), Portugal 2020, and by Portuguese funds through FCT - Fundação para a Ciência e a Tecnologia/Ministério da Ciência, Tecnologia e Ensino Superior in the framework of the project POCI-01-0145-FEDER-028336 (PTDC/MED-NEU/28336/2017); National Funds through FCT – Fundação para a Ciência e a Tecnologia under the project IF/00902/2015; R&D@PhD from Luso-American Development Foundation (FLAD); FLAD Healthcare 2020; and Programme for Cooperation in Science between Portugal and Germany 2018/2019 (FCT/DAAD). Márcia A Liz is supported by CEECINST/00091/2018.
- FEDER - Fundo Europeu de Desenvolvimento Regional funds through the COMPETE 2020 – Operacional Programme for Competitiveness and Internationalisation (POCI), Portugal 2020, and by Portuguese funds through FCT - Fundação para a Ciência e a Tecnologia/Ministério da Ciência, Tecnologia e Ensino Superior in the framework of the project POCI-01-0145-FEDER-028336 (PTDC/MED-NEU/28336/2017); National Funds through FCT – Fundação para a Ciência e a Tecnologia under the project IF/00902/2015; R&D@PhD from Luso-American Development Foundation (FLAD); FLAD Healthcare 2020; and Programme for Cooperation in Science between Portugal and Germany 2018/2019 (FCT/DAAD).
- Generous gifts to the Colorado State University Development Fund (J.R.B) and by the National Institutes on Aging of the National Institutes of Health under award numbers R01AG049668, 1S10OD025127 (J.R.B), and R43AG071064 (J.R.B).
- National Institutes on Aging of the National Institutes of Health under award number RO1NS105774 (R.A.S).
- Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany’s Excellence Strategy - EXC 2067/1- 390729940) and SFB1286 (Project B8)
- Generous gifts to the Colorado State University Development Fund (J.R.B) and by the National Institutes on Aging of the National Institutes of Health under award numbers R01AG049668, 1S10OD025127 (J.R.B), R43AG071064 (J.R.B)
Collapse
Affiliation(s)
- Marina I Oliveira da Silva
- Neurodegeneration Team, Nerve Regeneration Group, IBMC -Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135, Porto, Portugal
| | - Miguel Santejo
- Neurodegeneration Team, Nerve Regeneration Group, IBMC -Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135, Porto, Portugal
| | - Isaac W Babcock
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Ana Magalhães
- Addiction Biology Group, IBMC -Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135, Porto, Portugal
| | - Laurie S Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Seok-Joon Won
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Erika Castillo
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Ellen Gerhardt
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Christiane Fahlbusch
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073, Göttingen, Germany
| | - Raymond A Swanson
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, 37073, Göttingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077, Göttingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
- Scientific employee with an honorary contract at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 37075, Göttingen, Germany
| | - Ricardo Taipa
- Neuropathology Unit, Centro Hospitalar Universitário de Santo António, 4099-001, Porto, Portugal
- Autoimmune and Neuroscience Research Group, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, 4050-313, Porto, Portugal
- ITR - Laboratory for Integrative and Translational Research in Population Health, 4050-600, Porto, Portugal
| | - Michael Ruff
- Creative Bio-Peptides, Rockville, MD, 20854, USA
| | - James R Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Márcia A Liz
- Neurodegeneration Team, Nerve Regeneration Group, IBMC -Instituto de Biologia Molecular e Celular and i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135, Porto, Portugal.
| |
Collapse
|
11
|
Sun ZG, Yadav V, Amiri S, Cao W, De La Cruz EM, Murrell M. Cofilin-mediated actin filament network flexibility facilitates 2D to 3D actomyosin shape change. Eur J Cell Biol 2024; 103:151379. [PMID: 38168598 DOI: 10.1016/j.ejcb.2023.151379] [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: 08/11/2023] [Revised: 12/06/2023] [Accepted: 12/16/2023] [Indexed: 01/05/2024] Open
Abstract
The organization of actin filaments (F-actin) into crosslinked networks determines the transmission of mechanical stresses within the cytoskeleton and subsequent changes in cell and tissue shape. Principally mediated by proteins such as α-actinin, F-actin crosslinking increases both network connectivity and rigidity, thereby facilitating stress transmission at low crosslinking yet attenuating transmission at high crosslinker concentration. Here, we engineer a two-dimensional model of the actomyosin cytoskeleton, in which myosin-induced mechanical stresses are controlled by light. We alter the extent of F-actin crosslinking by the introduction of oligomerized cofilin. At pH 6.5, F-actin severing by cofilin is weak, but cofilin bundles and crosslinks filaments. Given its effect of lowering the F-actin bending stiffness, cofilin- crosslinked networks are significantly more flexible and softer in bending than networks crosslinked by α-actinin. Thus, upon local activation of myosin-induced contractile stress, the network bends out-of-plane in contrast to the in-plane compression as observed with networks crosslinked by α-actinin. Here, we demonstrate that local effects on filament mechanics by cofilin introduces novel large-scale network material properties that enable the sculpting of complex shapes in the cell cytoskeleton.
Collapse
Affiliation(s)
- Zachary Gao Sun
- Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Physics, Yale University, 217 Prospect Street, New Haven, CT 06511, USA; Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, CT 06520, USA
| | - Vikrant Yadav
- Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Sorosh Amiri
- Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Mechanical Engineering and Material Science, Yale University, New Haven, CT 06511, USA
| | - Wenxiang Cao
- Department of Molecular Biology & Biophysics, Yale University, New Haven, CT 06511, USA
| | - Enrique M De La Cruz
- Department of Molecular Biology & Biophysics, Yale University, New Haven, CT 06511, USA
| | - Michael Murrell
- Systems Biology Institute, Yale University, West Haven, CT 06516, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA; Department of Physics, Yale University, 217 Prospect Street, New Haven, CT 06511, USA; Integrated Graduate Program in Physical and Engineering Biology, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
12
|
Sousa-Squiavinato ACM, Morgado-Díaz JA. A glimpse into cofilin-1 role in cancer therapy: A potential target to improve clinical outcomes? Biochim Biophys Acta Rev Cancer 2024; 1879:189087. [PMID: 38395237 DOI: 10.1016/j.bbcan.2024.189087] [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: 10/13/2023] [Revised: 12/22/2023] [Accepted: 02/18/2024] [Indexed: 02/25/2024]
Abstract
Cofilin-1 (CFL1) modulates dynamic actin networks by severing and enhancing depolymerization. The upregulation of cofilin-1 expression in several cancer types is associated with tumor progression and metastasis. However, recent discoveries indicated relevant cofilin-1 functions under oxidative stress conditions, interplaying with mitochondrial dynamics, and apoptosis networks. In this scenario, these emerging roles might impact the response to clinical therapy and could be used to enhance treatment efficacy. Here, we highlight new perspectives of cofilin-1 in the therapy resistance context and discussed how cofilin-1 is involved in these events, exploring aspects of its contribution to therapeutic resistance. We also provide an analysis of CFL1 expression in several tumors predicting survival. Therefore, understanding how exactly coflin-1 plays, particularly in therapy resistance, may pave the way to the development of treatment strategies and improvement of patient survival.
Collapse
Affiliation(s)
| | - Jose Andrés Morgado-Díaz
- Cellular and Molecular Oncobiology Program, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil.
| |
Collapse
|
13
|
Liu J, Mouradian MM. Pathogenetic Contributions and Therapeutic Implications of Transglutaminase 2 in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:2364. [PMID: 38397040 PMCID: PMC10888553 DOI: 10.3390/ijms25042364] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
Neurodegenerative diseases encompass a heterogeneous group of disorders that afflict millions of people worldwide. Characteristic protein aggregates are histopathological hallmark features of these disorders, including Amyloid β (Aβ)-containing plaques and tau-containing neurofibrillary tangles in Alzheimer's disease, α-Synuclein (α-Syn)-containing Lewy bodies and Lewy neurites in Parkinson's disease and dementia with Lewy bodies, and mutant huntingtin (mHTT) in nuclear inclusions in Huntington's disease. These various aggregates are found in specific brain regions that are impacted by neurodegeneration and associated with clinical manifestations. Transglutaminase (TG2) (also known as tissue transglutaminase) is the most ubiquitously expressed member of the transglutaminase family with protein crosslinking activity. To date, Aβ, tau, α-Syn, and mHTT have been determined to be substrates of TG2, leading to their aggregation and implicating the involvement of TG2 in several pathophysiological events in neurodegenerative disorders. In this review, we summarize the biochemistry and physiologic functions of TG2 and describe recent advances in the pathogenetic role of TG2 in these diseases. We also review TG2 inhibitors tested in clinical trials and discuss recent TG2-targeting approaches, which offer new perspectives for the design of future highly potent and selective drugs with improved brain delivery as a disease-modifying treatment for neurodegenerative disorders.
Collapse
Affiliation(s)
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics and Department of Neurology, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| |
Collapse
|
14
|
Shehjar F, Almarghalani DA, Mahajan R, Hasan SAM, Shah ZA. The Multifaceted Role of Cofilin in Neurodegeneration and Stroke: Insights into Pathogenesis and Targeting as a Therapy. Cells 2024; 13:188. [PMID: 38247879 PMCID: PMC10814918 DOI: 10.3390/cells13020188] [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: 11/21/2023] [Revised: 01/03/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
This comprehensive review explores the complex role of cofilin, an actin-binding protein, across various neurodegenerative diseases (Alzheimer's, Parkinson's, schizophrenia, amyotrophic lateral sclerosis (ALS), Huntington's) and stroke. Cofilin is an essential protein in cytoskeletal dynamics, and any dysregulation could lead to potentially serious complications. Cofilin's involvement is underscored by its impact on pathological hallmarks like Aβ plaques and α-synuclein aggregates, triggering synaptic dysfunction, dendritic spine loss, and impaired neuronal plasticity, leading to cognitive decline. In Parkinson's disease, cofilin collaborates with α-synuclein, exacerbating neurotoxicity and impairing mitochondrial and axonal function. ALS and frontotemporal dementia showcase cofilin's association with genetic factors like C9ORF72, affecting actin dynamics and contributing to neurotoxicity. Huntington's disease brings cofilin into focus by impairing microglial migration and influencing synaptic plasticity through AMPA receptor regulation. Alzheimer's, Parkinson's, and schizophrenia exhibit 14-3-3 proteins in cofilin dysregulation as a shared pathological mechanism. In the case of stroke, cofilin takes center stage, mediating neurotoxicity and neuronal cell death. Notably, there is a potential overlap in the pathologies and involvement of cofilin in various diseases. In this context, referencing cofilin dysfunction could provide valuable insights into the common pathologies associated with the aforementioned conditions. Moreover, this review explores promising therapeutic interventions, including cofilin inhibitors and gene therapy, demonstrating efficacy in preclinical models. Challenges in inhibitor development, brain delivery, tissue/cell specificity, and long-term safety are acknowledged, emphasizing the need for precision drug therapy. The call to action involves collaborative research, biomarker identification, and advancing translational efforts. Cofilin emerges as a pivotal player, offering potential as a therapeutic target. However, unraveling its complexities requires concerted multidisciplinary efforts for nuanced and effective interventions across the intricate landscape of neurodegenerative diseases and stroke, presenting a hopeful avenue for improved patient care.
Collapse
Affiliation(s)
- Faheem Shehjar
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (F.S.); (R.M.)
| | - Daniyah A. Almarghalani
- Stroke Research Unit, Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Reetika Mahajan
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (F.S.); (R.M.)
| | - Syed A.-M. Hasan
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA;
| | - Zahoor A. Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, Toledo, OH 43614, USA; (F.S.); (R.M.)
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA;
| |
Collapse
|
15
|
Almarghalani DA, Bahader GA, Ali M, Tillekeratne LMV, Shah ZA. Cofilin Inhibitor Improves Neurological and Cognitive Functions after Intracerebral Hemorrhage by Suppressing Endoplasmic Reticulum Stress Related-Neuroinflammation. Pharmaceuticals (Basel) 2024; 17:114. [PMID: 38256947 PMCID: PMC10818666 DOI: 10.3390/ph17010114] [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: 11/27/2023] [Revised: 01/05/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Neuroinflammation after intracerebral hemorrhage (ICH) is a crucial factor that determines the extent of the injury. Cofilin is a cytoskeleton-associated protein that drives neuroinflammation and microglia activation. A novel cofilin inhibitor (CI) synthesized and developed in our lab has turned out to be a potential therapeutic agent for targeting cofilin-mediated neuroinflammation in an in vitro model of ICH and traumatic brain injury. The current study aims to examine the therapeutic potential of CI in a mouse collagenase model of ICH and examine the neurobehavioral outcomes and its mechanism of action. Male mice were subjected to intrastriatal collagenase injection to induce ICH, and sham mice received needle insertion. Various concentrations (25, 50, and 100 mg/kg) of CI were administered to different cohorts of the animals as a single intravenous injection 3 h following ICH and intraperitoneally every 12 h for 3 days. The animals were tested for neurobehavioral parameters for up to 7 days and sacrificed to collect brains for hematoma volume measurement, Western blotting, and immunohistochemistry. Blood was collected for cofilin, TNF-α, and IL-1β assessments. The results indicated that 50 mg/kg CI improved neurological outcomes, reversed post-stroke cognitive impairment, accelerated hematoma resolution, mitigated cofilin rods/aggregates, and reduced microglial and astrocyte activation in mice with ICH. Microglia morphological analysis demonstrated that CI restored the homeostasis ramification pattern of microglia in mice treated with CI. CI suppressed endoplasmic reticulum stress-related neuroinflammation by inhibiting inflammasomes and cell death signaling pathways. We also showed that CI prevented synaptic loss by reviving the pre- and post-synaptic markers. Our results unveil a novel therapeutic approach to treating ICH and open a window for using CI in clinical practice.
Collapse
Affiliation(s)
- Daniyah A. Almarghalani
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Ghaith A. Bahader
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Mohammad Ali
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - L. M. Viranga Tillekeratne
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A. Shah
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
16
|
Lee W. The Cytoskeleton and Its Binding Proteins as Mechanosensors, Transducers, and Functional Regulators of Cells. Int J Mol Sci 2023; 25:172. [PMID: 38203343 PMCID: PMC10779244 DOI: 10.3390/ijms25010172] [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: 12/01/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
Due to its complement of diverse proteins, such as actin filaments, intermediate filaments, and microtubules, the cytoskeleton is essential not only for structural stability but also for regulating cellular signaling, intracellular transportation, and cell division [...].
Collapse
Affiliation(s)
- Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; ; Tel.: +82-54-770-2409; Fax: +82-54-770-2447
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
17
|
Mann N, Hill J, Wang K, Hughes RM. OptoProfilin: A Single Component Biosensor of Applied Cellular Stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.04.560945. [PMID: 37873064 PMCID: PMC10592976 DOI: 10.1101/2023.10.04.560945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
The actin cytoskeleton is a biosensor of cellular stress and a potential prognosticator of human disease. In particular, aberrant cytoskeletal structures such as cofilin-actin rods and stress granules formed in response to energetic and oxidative stress are closely linked to neurodegenerative diseases such as Alzheimer's, Parkinson's, and ALS. Whether these cytoskeletal phenomena can be harnessed for the development of biosensors for cytoskeletal dysfunction and, by extension, neurodegenerative disease progression, remains an open question. In this work, we describe the design and development of an optogenetic iteration of profilin, an actin monomer binding protein with critical functions in cytoskeletal dynamics. We demonstrate that this optically activated profilin ('OptoProfilin') can act as an optically triggered biosensor of applied cellular stress in select immortalized cell lines. Notably, OptoProfilin is a single component biosensor, likely increasing its utility for experimentalists. While a large body of preexisting work closely links profilin activity with cellular stress and neurodegenerative disease, this, to our knowledge, is the first example of profilin as an optogenetic biosensor of stress-induced changes in the cytoskeleton.
Collapse
Affiliation(s)
- Noah Mann
- Department of Chemistry, East Carolina University, Greenville, North Carolina, United States
| | - Jahiem Hill
- Department of Chemistry, East Carolina University, Greenville, North Carolina, United States
| | - Kenneth Wang
- Department of Chemistry, Davidson College, Davidson, North Carolina, United States
| | - Robert M. Hughes
- Department of Chemistry, East Carolina University, Greenville, North Carolina, United States
| |
Collapse
|
18
|
Sattler R, Traynor BJ, Robertson J, Van Den Bosch L, Barmada SJ, Svendsen CN, Disney MD, Gendron TF, Wong PC, Turner MR, Boxer A, Babu S, Benatar M, Kurnellas M, Rohrer JD, Donnelly CJ, Bustos LM, Van Keuren-Jensen K, Dacks PA, Sabbagh MN. Roadmap for C9ORF72 in Frontotemporal Dementia and Amyotrophic Lateral Sclerosis: Report on the C9ORF72 FTD/ALS Summit. Neurol Ther 2023; 12:1821-1843. [PMID: 37847372 PMCID: PMC10630271 DOI: 10.1007/s40120-023-00548-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/14/2023] [Indexed: 10/18/2023] Open
Abstract
A summit held March 2023 in Scottsdale, Arizona (USA) focused on the intronic hexanucleotide expansion in the C9ORF72 gene and its relevance in frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS; C9ORF72-FTD/ALS). The goal of this summit was to connect basic scientists, clinical researchers, drug developers, and individuals affected by C9ORF72-FTD/ALS to evaluate how collaborative efforts across the FTD-ALS disease spectrum might break down existing disease silos. Presentations and discussions covered recent discoveries in C9ORF72-FTD/ALS disease mechanisms, availability of disease biomarkers and recent advances in therapeutic development, and clinical trial design for prevention and treatment for individuals affected by C9ORF72-FTD/ALS and asymptomatic pathological expansion carriers. The C9ORF72-associated hexanucleotide repeat expansion is an important locus for both ALS and FTD. C9ORF72-FTD/ALS may be characterized by loss of function of the C9ORF72 protein and toxic gain of functions caused by both dipeptide repeat (DPR) proteins and hexanucleotide repeat RNA. C9ORF72-FTD/ALS therapeutic strategies discussed at the summit included the use of antisense oligonucleotides, adeno-associated virus (AAV)-mediated gene silencing and gene delivery, and engineered small molecules targeting RNA structures associated with the C9ORF72 expansion. Neurofilament light chain, DPR proteins, and transactive response (TAR) DNA-binding protein 43 (TDP-43)-associated molecular changes were presented as biomarker candidates. Similarly, brain imaging modalities (i.e., magnetic resonance imaging [MRI] and positron emission tomography [PET]) measuring structural, functional, and metabolic changes were discussed as important tools to monitor individuals affected with C9ORF72-FTD/ALS, at both pre-symptomatic and symptomatic disease stages. Finally, summit attendees evaluated current clinical trial designs available for FTD or ALS patients and concluded that therapeutics relevant to FTD/ALS patients, such as those specifically targeting C9ORF72, may need to be tested with composite endpoints covering clinical symptoms of both FTD and ALS. The latter will require novel clinical trial designs to be inclusive of all patient subgroups spanning the FTD/ALS spectrum.
Collapse
Affiliation(s)
- Rita Sattler
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Janice Robertson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Ludo Van Den Bosch
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology and KU Leuven, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), University of Leuven, Leuven, Belgium
| | - Sami J Barmada
- Department of Neurology, Neuroscience Program, University of Michigan, Ann Arbor, MI, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Matthew D Disney
- Department of Chemistry, The Herbert Wertheim UF-Scripps Institute for Biomedical Research and Innovation, The Scripps Research Institute, Jupiter, FL, USA
| | - Tania F Gendron
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Philip C Wong
- Departments of Pathology and Neuroscience, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Martin R Turner
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Adam Boxer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of San Francisco, San Francisco, CA, USA
| | - Suma Babu
- Sean M. Healey and AMG Center for ALS and the Neurological Clinical Research Institute, Massachusetts General Hospital-Harvard Medical School, Boston, MA, USA
| | - Michael Benatar
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, 33129, USA
| | | | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Christopher J Donnelly
- LiveLikeLou Center for ALS Research, Brain Institute, University of Pittsburgh, Pittsburgh, USA
- Department of Neurobiology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lynette M Bustos
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA
| | | | - Penny A Dacks
- The Association for Frontotemporal Degeneration and FTD Disorders Registry, King of Prussia, PA, USA
| | - Marwan N Sabbagh
- Barrow Neurological Institute, 2910 N Third Ave, Phoenix, AZ, 85013, USA.
| |
Collapse
|
19
|
Tahtamouni LH, Alderfer SA, Kuhn TB, Minamide LS, Chanda S, Ruff MR, Bamburg JR. Characterization of a Human Neuronal Culture System for the Study of Cofilin-Actin Rod Pathology. Biomedicines 2023; 11:2942. [PMID: 38001943 PMCID: PMC10669520 DOI: 10.3390/biomedicines11112942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Cofilactin rod pathology, which can initiate synapse loss, has been extensively studied in rodent neurons, hippocampal slices, and in vivo mouse models of human neurodegenerative diseases such as Alzheimer's disease (AD). In these systems, rod formation induced by disease-associated factors, such as soluble oligomers of Amyloid-β (Aβ) in AD, utilizes a pathway requiring cellular prion protein (PrPC), NADPH oxidase (NOX), and cytokine/chemokine receptors (CCR5 and/or CXCR4). However, rod pathways have not been systematically assessed in a human neuronal model. Here, we characterize glutamatergic neurons differentiated from human-induced pluripotent stem cells (iPSCs) for the formation of rods in response to activators of the PrPC-dependent pathway. Optimization of substratum, cell density, and use of glial-conditioned medium yielded a robust system for studying the development of Aβ-induced rods in the absence of glia, suggesting a cell-autonomous pathway. Rod induction in younger neurons requires ectopic expression of PrPC, but this dependency disappears by Day 55. The quantification of proteins within the rod-inducing pathway suggests that increased PrPC and CXCR4 expression may be factors in the doubling of the rod response to Aβ between Days 35 and 55. FDA-approved antagonists to CXCR4 and CCR5 inhibit the rod response. Rods were predominantly observed in dendrites, although severe cytoskeletal disruptions prevented the assignment of over 40% of the rods to either an axon or dendrite. In the absence of glia, a condition in which rods are more readily observed, neurons mature and fire action potentials but do not form functional synapses. However, PSD95-containing dendritic spines associate with axonal regions of pre-synaptic vesicles containing the glutamate transporter, VGLUT1. Thus, our results identified stem cell-derived neurons as a robust model for studying cofilactin rod formation in a human cellular environment and for developing effective therapeutic strategies for the treatment of dementias arising from multiple proteinopathies with different rod initiators.
Collapse
Affiliation(s)
- Lubna H. Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa 13133, Jordan;
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| | - Sydney A. Alderfer
- Department of Chemical and Biological Engineering and School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA;
| | - Thomas B. Kuhn
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| | - Laurie S. Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| | - Soham Chanda
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| | - Michael R. Ruff
- Creative Bio-Peptides, Inc., 10319 Glen Road, Suite 100, Potomac, MD 20854, USA;
| | - James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (T.B.K.); (L.S.M.); (S.C.)
| |
Collapse
|
20
|
McGoldrick P, Robertson J. Unraveling the impact of disrupted nucleocytoplasmic transport systems in C9orf72-associated ALS. Front Cell Neurosci 2023; 17:1247297. [PMID: 37720544 PMCID: PMC10501458 DOI: 10.3389/fncel.2023.1247297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/08/2023] [Indexed: 09/19/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two adult-onset neurodegenerative diseases that are part of a common disease spectrum due to clinical, genetic, and pathological overlap. A prominent genetic factor contributing to both diseases is a hexanucleotide repeat expansion in a non-coding region of the C9orf72 gene. This mutation in C9orf72 leads to nuclear depletion and cytoplasmic aggregation of Tar DNA-RNA binding protein 43 (TDP-43). TDP-43 pathology is characteristic of the majority of ALS cases, irrespective of disease causation, and is present in ~50% of FTD cases. Defects in nucleocytoplasmic transport involving the nuclear pore complex, the Ran-GTPase cycle, and nuclear transport factors have been linked with the mislocalization of TDP-43. Here, we will explore and discuss the implications of these system abnormalities of nucleocytoplasmic transport in C9orf72-ALS/FTD, as well as in other forms of familial and sporadic ALS.
Collapse
Affiliation(s)
- Philip McGoldrick
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Janice Robertson
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
21
|
Schmid ET, Schinaman JM, Williams KS, Walker DW. Accumulation of F-actin drives brain aging and limits healthspan in Drosophila. RESEARCH SQUARE 2023:rs.3.rs-3158290. [PMID: 37577708 PMCID: PMC10418561 DOI: 10.21203/rs.3.rs-3158290/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The actin cytoskeleton is a key determinant of cell and tissue homeostasis. However, tissue-specific roles for actin dynamics in aging, notably brain aging, are not understood. Here, we show that there is an age-related increase in filamentous actin (F-actin) in Drosophila brains, which is counteracted by prolongevity interventions. Critically, modulating F-actin levels in aging neurons prevents age-onset cognitive decline and extends organismal healthspan. Mechanistically, we show that autophagy, a recycling process required for neuronal homeostasis, is disabled upon actin dysregulation in the aged brain. Remarkably, disrupting actin polymerization in aged animals with cytoskeletal drugs restores brain autophagy to youthful levels and reverses cellular hallmarks of brain aging. Finally, reducing F-actin levels in aging neurons slows brain aging and promotes healthspan in an autophagy-dependent manner. Our data identify excess actin polymerization as a hallmark of brain aging, which can be targeted to reverse brain aging phenotypes and prolong healthspan.
Collapse
Affiliation(s)
- Edward T. Schmid
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Joseph M. Schinaman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Kylie S. Williams
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - David W. Walker
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|