1
|
Guerra San Juan I, Brunner JW, Eggan K, Toonen RF, Verhage M. KIF5A regulates axonal repair and time-dependent axonal transport of SFPQ granules and mitochondria in human motor neurons. Neurobiol Dis 2024; 204:106759. [PMID: 39644980 DOI: 10.1016/j.nbd.2024.106759] [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: 09/20/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024] Open
Abstract
Mutations in the microtubule-binding motor protein kinesin 5 A (KIF5A) are implicated in several adult-onset motor neuron diseases, including Amyotrophic Lateral Sclerosis, Spastic Paraplegia Type 10 and Charcot-Marie-Tooth Disease Type 2. While KIF5 family members transport a variety of cargos along axons, the specific cargos affected by KIF5A mutations remain poorly understood. Here, we generated KIF5Anull mutant human motor neurons and analyzed the impact on axonal transport and motor neuron outgrowth and regeneration in vitro. KIF5A deficiency caused reduced neurite complexity in young neurons (DIV14) and defects in axonal regeneration. KIF5A deficiency did not affect neurofilament transport but impaired mitochondrial motility and anterograde speed at DIV42. Notably, KIF5A deficiency strongly reduced anterograde transport of splicing factor proline/glutamine-rich (SFPQ)-associated RNA granules in DIV42 axons. Hence, KIF5A plays a critical role in promoting axonal regrowth after injury and in driving the anterograde transport of mitochondria and especially SFPQ-associated RNA granules in mature neurons.
Collapse
Affiliation(s)
- Irune Guerra San Juan
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, the Netherlands; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands.
| | - Jessie W Brunner
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, the Netherlands; Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ruud F Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, the Netherlands; Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, the Netherlands; Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
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
|
3
|
Guerra San Juan I, Brunner J, Eggan K, Toonen RF, Verhage M. KIF5A regulates axonal repair and time-dependent axonal transport of SFPQ granules and mitochondria in human motor neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611684. [PMID: 39314491 PMCID: PMC11418931 DOI: 10.1101/2024.09.06.611684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Mutations in the microtubule binding motor protein, kinesin family member 5A (KIF5A), cause the fatal motor neuron disease, Amyotrophic Lateral Sclerosis. While KIF5 family members transport a variety of cargos along axons, it is still unclear which cargos are affected by KIF5A mutations. We generated KIF5A null mutant human motor neurons to investigate the impact of KIF5A loss on the transport of various cargoes and its effect on motor neuron function at two different timepoints in vitro. The absence of KIF5A resulted in reduced neurite complexity in young motor neurons (DIV14) and significant defects in axonal regeneration capacity at all developmental stages. KIF5A loss did not affect neurofilament transport but resulted in decreased mitochondria motility and anterograde speed at DIV42. More prominently, KIF5A depletion strongly reduced anterograde transport of SFPQ-associated RNA granules in DIV42 motor neuron axons. We conclude that KIF5A most prominently functions in human motor neurons to promote axonal regrowth after injury as well as to anterogradely transport mitochondria and, to a larger extent, SFPQ-associated RNA granules in a time-dependent manner.
Collapse
Affiliation(s)
- Irune Guerra San Juan
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, The Netherlands
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Jessie Brunner
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Kevin Eggan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ruud F. Toonen
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Matthijs Verhage
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam and VU Medical Center, Amsterdam, The Netherlands
- Department of Human Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Bartak M, Bąska P, Chodkowski M, Tymińska B, Bańbura MW, Cymerys J. Neurons cytoskeletal architecture remodeling during the replication cycle of mouse coronavirus MHV-JHM: a morphological in vitro study. BMC Vet Res 2024; 20:18. [PMID: 38195523 PMCID: PMC10775625 DOI: 10.1186/s12917-023-03813-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 11/10/2023] [Indexed: 01/11/2024] Open
Abstract
Nowadays, the population is still struggling with a post-COVID19 syndrome known as long COVID, including a broad spectrum of neurological problems. There is an urgent need for a better understanding and exploration of the mechanisms of coronavirus neurotropism. For this purpose, the neurotropic strain of mouse hepatitis virus (MHV-JHM) originating from the beta-coronavirus genus, the same as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been used. The role of the cytoskeleton during virus replication in neurons in vitro was determined to understand the mechanisms of MHV-JHM neuroinfection. We have described for the first time the changes of actin filaments during MHV-JHM infection. We also observed productive replication of MHV-JHM in neurons during 168 h p.i. and syncytial cytopathic effect. We discovered that the MHV-JHM strain modulated neuronal cytoskeleton during infection, which were manifested by: (i) condensation of actin filaments in the cortical layer of the cytoplasm, (ii) formation of microtubule cisternae structures containing viral antigen targeting viral replication site (iii) formation of tunneling nanotubes used by MHV-JHM for intercellular transport. Additionally, we demonstrated that the use of cytoskeletal inhibitors have reduced virus replication in neurons, especially noscapine and nocodazole, the microtubule shortening factors.
Collapse
Affiliation(s)
- Michalina Bartak
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8 St., Warsaw, 02-786, Poland.
| | - Piotr Bąska
- Division of Pharmacology and Toxicology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-SGGW, Ciszewskiego 8 St., Warsaw, 02-786, Poland
| | - Marcin Chodkowski
- Laboratory of Nanobiology and Biomaterials, Military Institute of Hygiene and Epidemiology, Kozielska 4 St., Warsaw, 01-063, Poland
| | - Beata Tymińska
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8 St., Warsaw, 02-786, Poland
| | - Marcin W Bańbura
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8 St., Warsaw, 02-786, Poland
| | - Joanna Cymerys
- Division of Microbiology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Ciszewskiego 8 St., Warsaw, 02-786, Poland.
| |
Collapse
|
5
|
Li H, Li J, Wang P, Yuan F, Zhang S. Improvement of actin dynamics and cognitive impairment in diabetes through troxerutin-mediated downregulation of TRPM7/CaN/cofilin. Neuropeptides 2023; 102:102381. [PMID: 37837806 DOI: 10.1016/j.npep.2023.102381] [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: 06/29/2023] [Revised: 09/08/2023] [Accepted: 09/08/2023] [Indexed: 10/16/2023]
Abstract
Diabetic cognitive impairment is a central nervous complication of diabetes mellitus. Its specific pathogenesis is unknown, and no effective treatment strategy is currently available. An imbalance in actin dynamics is an important mechanism underlying cognitive impairment. Transient receptor potential channel 7 (TRPM7) mediates actin dynamics imbalance through calcineurin (CaN) and cofilin cascades involved in various neurodegenerative diseases. We previously demonstrated that TRPM7 expression is increased in diabetic cognitive impairment, and troxerutin has been shown to ameliorate diabetic cognitive impairment. However, the relationship between troxerutin and TRPM7 remains unclear. In this study, we hypothesize that troxerutin may improve diabetic cognitive impairment by enhancing actin dynamics through downregulation of the TRPM7/CaN/cofilin pathway. To test this hypothesis, we divided db/m and db/db mice into the following groups: normal control group (NC), normal + troxerutin group (NT), diabetic group (DM), diabetic + troxerutin group (DT) and diabetic + troxerutin + bradykinin group (DTB). The results showed that diabetic mice exhibited cognitive impairment at 17 weeks of age, TRPM7, CaN, cofilin and G-actin were highly expressed in the CA1 region of hippocampus, while p-cofilin and F-actin expression decreased. Furthermore, hippocampal neuronal cellsshowed varying degrees of damage. The length of synaptic active zone, the width of synaptic cleft, and the number of synapses per high-power field were decreased. Troxerutin intervention alleviated these manifestations in the DT group; however, the effect of troxerutin was weakened in the DTB group. In conclusion, our findings suggest that diabetes leads to cognitive impairment, activation of the TRPM7/CaN/cofilin pathway, actin dynamics imbalance, and destruction of hippocampal neuronal cells and synapses. Troxerutin can downregulate TRPM7/CaN/cofilin, improve actin dynamics imbalance, and ameliorate cognitive impairment in diabetic mice. This study provides a new avenue for exploring and treating cognitive impairment in diabetes.
Collapse
Affiliation(s)
- Hongyan Li
- Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China; Department of Endocrinology, Shijiazhuang people's hospital, Shijiazhuang, Hebei, China
| | - Jie Li
- Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Pin Wang
- Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Fang Yuan
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Songyun Zhang
- Department of Endocrinology, Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
6
|
Kuang J, Liu M, Yu Q, Cheng Y, Huang J, Han S, Shi J, Huang L, Li P. Antiviral Effect and Mechanism of Edaravone against Grouper Iridovirus Infection. Viruses 2023; 15:2237. [PMID: 38005914 PMCID: PMC10674758 DOI: 10.3390/v15112237] [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: 10/10/2023] [Revised: 11/01/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Singapore grouper iridovirus (SGIV) is a virus with high fatality rate in the grouper culture industry. The outbreak of SGIV is often accompanied by a large number of grouper deaths, which has a great impact on the economy. Therefore, it is of great significance to find effective drugs against SGIV. It has been reported that edaravone is a broad-spectrum antiviral drug, most widely used clinically in recent years, but no report has been found exploring the effect of edaravone on SGIV infections. In this study, we evaluated the antiviral effect of edaravone against SGIV, and the anti-SGIV mechanism of edaravone was also explored. It was found that the safe concentration of edaravone on grouper spleen (GS) cells was 50 µg/mL, and it possessed antiviral activity against SGIV infection in a dose-dependent manner. Furthermore, edaravone could significantly disrupt SGIV particles and interference with SGIV binding to host cells, as well as SGIV replication in host cells. However, edaravone was not effective during the SGIV invasion into host cells. This study was the first time that it was determined that edaravone could exert antiviral effects in response to SGIV infection by directly interfering with the processes of SGIV infecting cells, aiming to provide a theoretical basis for the control of grouper virus disease.
Collapse
Affiliation(s)
- Jihui Kuang
- School of Resources, Environment and Materials, Guangxi University, Nanning 537100, China;
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
| | - Mingzhu Liu
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Qing Yu
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Yuan Cheng
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Jing Huang
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Shuyu Han
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- Guangxi Fisheries Technology Extension Station, Nanning 530022, China
| | - Jingu Shi
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- Guangxi Fisheries Technology Extension Station, Nanning 530022, China
| | - Lin Huang
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| | - Pengfei Li
- School of Resources, Environment and Materials, Guangxi University, Nanning 537100, China;
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Marine Sciences, Guangxi Academy of Sciences, Nanning 530022, China; (M.L.); (Q.Y.); (Y.C.); (J.H.); (S.H.); (J.S.)
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Marine Sciences, Nanning 530022, China
| |
Collapse
|
7
|
Xue S, Zhou X, Yang ZH, Si XK, Sun X. Stroke-induced damage on the blood-brain barrier. Front Neurol 2023; 14:1248970. [PMID: 37840921 PMCID: PMC10569696 DOI: 10.3389/fneur.2023.1248970] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/08/2023] [Indexed: 10/17/2023] Open
Abstract
The blood-brain barrier (BBB) is a functional phenotype exhibited by the neurovascular unit (NVU). It is maintained and regulated by the interaction between cellular and non-cellular matrix components of the NVU. The BBB plays a vital role in maintaining the dynamic stability of the intracerebral microenvironment as a barrier layer at the critical interface between the blood and neural tissues. The large contact area (approximately 20 m2/1.3 kg brain) and short diffusion distance between neurons and capillaries allow endothelial cells to dominate the regulatory role. The NVU is a structural component of the BBB. Individual cells and components of the NVU work together to maintain BBB stability. One of the hallmarks of acute ischemic stroke is the disruption of the BBB, including impaired function of the tight junction and other molecules, as well as increased BBB permeability, leading to brain edema and a range of clinical symptoms. This review summarizes the cellular composition of the BBB and describes the protein composition of the barrier functional junction complex and the mechanisms regulating acute ischemic stroke-induced BBB disruption.
Collapse
Affiliation(s)
| | | | | | | | - Xin Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
8
|
Villablanca C, Vidal R, Gonzalez-Billault C. Are cytoskeleton changes observed in astrocytes functionally linked to aging? Brain Res Bull 2023; 196:59-67. [PMID: 36935053 DOI: 10.1016/j.brainresbull.2023.03.007] [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: 12/02/2022] [Revised: 02/22/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
Astrocytes are active participants in the performance of the Central Nervous System (CNS) in both health and disease. During aging, astrocytes are susceptible to reactive astrogliosis, a molecular state characterized by functional changes in response to pathological situations, and cellular senescence, characterized by loss of cell division, apoptosis resistance, and gain of proinflammatory functions. This results in two different states of astrocytes, which can produce proinflammatory phenotypes with harmful consequences in chronic conditions. Reactive astrocytes and senescent astrocytes share morpho-functional features that are dependent on the organization of the cytoskeleton. However, such changes in the cytoskeleton have yet to receive the necessary attention to explain their role in the alterations of astrocytes that are associated with aging and pathologies. In this review, we summarize all the available findings that connect changes in the cytoskeleton of the astrocytes with aging. In addition, we discuss future avenues that we believe will guide such a novel topic.
Collapse
Affiliation(s)
- Cristopher Villablanca
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - René Vidal
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Institute for Nutrition and Food Technologies, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
9
|
Li M, Peng L, Wang Z, Liu L, Cao M, Cui J, Wu F, Yang J. Roles of the cytoskeleton in human diseases. Mol Biol Rep 2023; 50:2847-2856. [PMID: 36609753 DOI: 10.1007/s11033-022-08025-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 10/12/2022] [Indexed: 01/08/2023]
Abstract
Recently, researches have revealed the key roles of the cytoskeleton in the occurrence and development of multiple diseases, suggesting that targeting the cytoskeleton is a viable approach for treating numerous refractory diseases. The cytoskeleton is a highly structured and complex network composed of actin filaments, microtubules, and intermediate filaments. In normal cells, these three cytoskeleton components are highly integrated and coordinated. However, the cytoskeleton undergoes drastic remodeling in cytoskeleton-related diseases, causing changes in cell polarity, affecting the cell cycle, leading to senescent diseases, and influencing cell migration to accelerate cancer metastasis. Additionally, mutations or abnormalities in cytoskeletal proteins and their related proteins are closely associated with several congenital diseases. Therefore, this review summarizes the roles of the cytoskeleton in cytoskeleton-related diseases as well as its potential roles in disease treatment to provide insights regarding the physiological functions and pathological roles of the cytoskeleton.
Collapse
Affiliation(s)
- Mengxin Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Li Peng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, 610065, Chengdu, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Lijia Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Mengjiao Cao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Jingyao Cui
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Fanzi Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China.
| |
Collapse
|
10
|
Circular RNAs in Parkinson's Disease: Reliable Biological Markers and Targets for Rehabilitation. Mol Neurobiol 2023; 60:3261-3276. [PMID: 36840847 DOI: 10.1007/s12035-023-03268-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 02/14/2023] [Indexed: 02/26/2023]
Abstract
In clinical practice, the underlying pathogenesis of Parkinson's disease (PD) remains unknown. Circular RNAs (circRNAs) have good biological properties and can be used as biological marker. Rehabilitation as a third treatment alongside drug and surgery has been shown to be clinically effective, but biomarkers of rehabilitation efficiency at genetic level is still lacking. In this study, we identified differentially expressed circRNAs in peripheral blood exosomes between PD patients and health controls (HCs) and determined whether these circRNAs changed after rehabilitation, to explore the competing RNA networks and epigenetic mechanisms affected. We found that there were 558 upregulated and 609 downregulated circRNAs in PD patients compared to HCs, 3398 upregulated and 479 downregulated circRNAs in PD patients after rehabilitation compared to them before rehabilitation, along with 3721 upregulated and 635 downregulated circRNAs in PD patients after rehabilitation compared to HCs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that differentially expressed circRNAs may affect the stability of the cellular actin backbone and synaptic structure by influencing the aggregation of α-synuclein (a-syn). We selected two circRNAs overexpressed in PD patients for validation (hsa_circ_0001535 and hsa_circ_0000437); the results revealed that their expression levels were all reduced to varying degrees (p < 0.05) after rehabilitation. After network analysis, we believe that hsa_circ_0001535 may be related to the aggregation of a-syn, while hsa_circ_0000437 may act on hsa-let-7b-5p or hsa-let-7c-5p through sponge effect to cause inflammatory response. Our findings suggest that rehabilitation can mitigate the pathological process of PD by epigenetic means.
Collapse
|
11
|
Macedo GC, Kreifeldt M, Goulding SP, Okhuarobo A, Sidhu H, Contet C. Chronic MAP4343 reverses escalated alcohol drinking in a mouse model of alcohol use disorder. Neuropsychopharmacology 2023; 48:821-830. [PMID: 36670228 PMCID: PMC10066354 DOI: 10.1038/s41386-023-01529-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 12/28/2022] [Accepted: 12/31/2022] [Indexed: 01/22/2023]
Abstract
Alcohol use disorders can be driven by negative reinforcement. Alterations of the microtubule cytoskeleton have been associated with mood regulation in the context of depression. Notably, MAP4343, a pregnenolone derivative known to promote tubulin assembly, has antidepressant properties. In the present study, we tested the hypothesis that MAP4343 may reduce excessive alcohol drinking in a mouse model of alcohol dependence by normalizing affect during withdrawal. Adult male C57BL/6J mice were given limited access to voluntary alcohol drinking and ethanol intake escalation was induced by chronic intermittent ethanol (CIE) vapor inhalation. Chronic, but not acute, administration of MAP4343 reduced ethanol intake and this effect was more pronounced in CIE-exposed mice. There was a complex interaction between the effects of MAP4343 and alcohol on affective behaviors. In the elevated plus maze, chronic MAP4343 tended to increase open-arm exploration in alcohol-naive mice but reduced it in alcohol-withdrawn mice. In the tail suspension test, chronic MAP4343 reduced immobility selectively in Air-exposed alcohol-drinking mice. Finally, chronic MAP4343 countered the plasma corticosterone reduction induced by CIE. Parallel analysis of tubulin post-translational modifications revealed lower α-tubulin acetylation in the medial prefrontal cortex of CIE-withdrawn mice. Altogether, these data support the relevance of microtubules as a therapeutic target for the treatment of AUD.
Collapse
Affiliation(s)
- Giovana C Macedo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Max Kreifeldt
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Scott P Goulding
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Agbonlahor Okhuarobo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.,Faculty of Pharmacy, Department of Pharmacology & Toxicology, University of Benin, Benin City, Nigeria
| | - Harpreet Sidhu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Candice Contet
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
12
|
Cheng Y, Liu M, Yu Q, Huang S, Han S, Shi J, Wei H, Zou J, Li P. Effect of EGCG Extracted from Green Tea against Largemouth Bass Virus Infection. Viruses 2023; 15:151. [PMID: 36680191 PMCID: PMC9864265 DOI: 10.3390/v15010151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023] Open
Abstract
(1) Background: Largemouth bass virus (LMBV) is a major viral pathogen in largemouth bass (Micropterus salmoides) aquaculture that often causes high mortality and heavy economic losses, thus developing treatments to combat this pathogen is of great commercial importance. Green tea is a well-known medicinal plant that contains active ingredients with antiviral, antibacterial, and other biological activities. The goals of this study were to explore the effect and mechanism of green tea source compounds on LMBV and provide data to serve as the basis for the screening of targeted drugs in the future. In this study, we evaluated the effects of the main component of green tea, epigallocatechin-3-gallate (EGCG), against LMBV infection. (2) Methods: The safe working concentration of EGCG was identified by cell viability detection and light microscopy. The antiviral activity and mechanism of action of EGCG against LMBV infection were evaluated with light microscopy, an aptamer 6-carboxy-fluorescein-based fluorescent molecular probe, and reverse transcription quantitative PCR. (3) Results: The safe working concentration of EGCG was ≤10 μg/mL. EGCG showed significant anti-LMBV infection activity in a concentration-dependent manner, and it also destroyed the structure of virus particles. EGCG impacted the binding of virus particles to cell receptors and virus invasion into the host cells. Inhibitory effects of EGCG on LMBV particles, LMBV binding to the host-cell membrane, and LMBV invasion were 84.89%, 98.99%, and 95.23%, respectively. Meanwhile, the effects of EGCG subsequently were verified in vivo. The fatality rate of the LMBV + EGCG group was significantly lower than that of the LMBV group. (4) Conclusions: Our results suggest that EGCG has effective antiviral properties against LMBV and may be a candidate for the effective treatment and control of LMBV infections in largemouth bass aquaculture.
Collapse
Affiliation(s)
- Yuan Cheng
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Sciences, Nanning 530000, China
| | - Mingzhu Liu
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Sciences, Nanning 530000, China
| | - Qing Yu
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Sciences, Nanning 530000, China
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Nanning 530000, China
| | - Shuaishuai Huang
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Sciences, Nanning 530000, China
| | - Shuyu Han
- Guangxi Fisheries Technology Extension Station, Nanning 530000, China
| | - Jingu Shi
- Beihai Fisheries Technology Extension Station, Beihai 536001, China
| | - Hongling Wei
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Sciences, Nanning 530000, China
| | - Jianwei Zou
- Beihai Fisheries Technology Extension Station, Beihai 536001, China
| | - Pengfei Li
- Guangxi Key Laboratory of Aquatic Biotechnology and Modern Ecological Aquaculture, Guangxi Engineering Research Center for Fishery Major Diseases Control and Efficient Healthy Breeding Industrial Technology (GERCFT), Guangxi Academy of Sciences, Nanning 530000, China
- China-ASEAN Modern Fishery Industry Technology Transfer Demonstration Center, Nanning 530000, China
| |
Collapse
|
13
|
López-Cerdán A, Andreu Z, Hidalgo MR, Grillo-Risco R, Català-Senent JF, Soler-Sáez I, Neva-Alejo A, Gordillo F, de la Iglesia-Vayá M, García-García F. Unveiling sex-based differences in Parkinson's disease: a comprehensive meta-analysis of transcriptomic studies. Biol Sex Differ 2022; 13:68. [PMID: 36414996 PMCID: PMC9682715 DOI: 10.1186/s13293-022-00477-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND In recent decades, increasing longevity (among other factors) has fostered a rise in Parkinson's disease incidence. Although not exhaustively studied in this devastating disease, the impact of sex represents a critical variable in Parkinson's disease as epidemiological and clinical features differ between males and females. METHODS To study sex bias in Parkinson's disease, we conducted a systematic review to select sex-labeled transcriptomic data from three relevant brain tissues: the frontal cortex, the striatum, and the substantia nigra. We performed differential expression analysis on each study chosen. Then we summarized the individual differential expression results with three tissue-specific meta-analyses and a global all-tissues meta-analysis. Finally, results from the meta-analysis were functionally characterized using different functional profiling approaches. RESULTS The tissue-specific meta-analyses linked Parkinson's disease to the enhanced expression of MED31 in the female frontal cortex and the dysregulation of 237 genes in the substantia nigra. The global meta-analysis detected 15 genes with sex-differential patterns in Parkinson's disease, which participate in mitochondrial function, oxidative stress, neuronal degeneration, and cell death. Furthermore, functional analyses identified pathways, protein-protein interaction networks, and transcription factors that differed by sex. While male patients exhibited changes in oxidative stress based on metal ions, inflammation, and angiogenesis, female patients exhibited dysfunctions in mitochondrial and lysosomal activity, antigen processing and presentation functions, and glutamic and purine metabolism. All results generated during this study are readily available by accessing an open web resource ( http://bioinfo.cipf.es/metafun-pd/ ) for consultation and reuse in further studies. CONCLUSIONS Our in silico approach has highlighted sex-based differential mechanisms in typical Parkinson Disease hallmarks (inflammation, mitochondrial dysfunction, and oxidative stress). Additionally, we have identified specific genes and transcription factors for male and female Parkinson Disease patients that represent potential candidates as biomarkers to diagnosis.
Collapse
Affiliation(s)
- Adolfo López-Cerdán
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
- Biomedical Imaging Unit FISABIO-CIPF, Fundación Para El Fomento de La Investigación Sanitaria Y Biomédica de La Comunidad Valenciana, 46012, Valencia, Spain
| | - Zoraida Andreu
- Foundation Valencian Institute of Oncology (FIVO), 46009, Valencia, Spain
| | - Marta R Hidalgo
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Rubén Grillo-Risco
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | | | - Irene Soler-Sáez
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Almudena Neva-Alejo
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - Fernando Gordillo
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain
| | - María de la Iglesia-Vayá
- Biomedical Imaging Unit FISABIO-CIPF, Fundación Para El Fomento de La Investigación Sanitaria Y Biomédica de La Comunidad Valenciana, 46012, Valencia, Spain
| | - Francisco García-García
- Bioinformatics and Biostatistics Unit, Principe Felipe Research Center (CIPF), 46012, Valencia, Spain.
| |
Collapse
|
14
|
Solomon T, Rajendran M, Rostovtseva T, Hool L. How cytoskeletal proteins regulate mitochondrial energetics in cell physiology and diseases. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210324. [PMID: 36189806 PMCID: PMC9527905 DOI: 10.1098/rstb.2021.0324] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Mitochondria are ubiquitous organelles that play a pivotal role in the supply of energy through the production of adenosine triphosphate in all eukaryotic cells. The importance of mitochondria in cells is demonstrated in the poor survival outcomes observed in patients with defects in mitochondrial gene or RNA expression. Studies have identified that mitochondria are influenced by the cell's cytoskeletal environment. This is evident in pathological conditions such as cardiomyopathy where the cytoskeleton is in disarray and leads to alterations in mitochondrial oxygen consumption and electron transport. In cancer, reorganization of the actin cytoskeleton is critical for trans-differentiation of epithelial-like cells into motile mesenchymal-like cells that promotes cancer progression. The cytoskeleton is critical to the shape and elongation of neurons, facilitating communication during development and nerve signalling. Although it is recognized that cytoskeletal proteins physically tether mitochondria, it is not well understood how cytoskeletal proteins alter mitochondrial function. Since end-stage disease frequently involves poor energy production, understanding the role of the cytoskeleton in the progression of chronic pathology may enable the development of therapeutics to improve energy production and consumption and slow disease progression. This article is part of the theme issue ‘The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease’.
Collapse
Affiliation(s)
- Tanya Solomon
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Megha Rajendran
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Tatiana Rostovtseva
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Livia Hool
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Lafanechère L. The microtubule cytoskeleton: An old validated target for novel therapeutic drugs. Front Pharmacol 2022; 13:969183. [PMID: 36188585 PMCID: PMC9521402 DOI: 10.3389/fphar.2022.969183] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Compounds targeting microtubules are widely used in cancer therapy with a proven efficacy. However, because they also target non-cancerous cells, their administration leads to numerous adverse effects. With the advancement of knowledge on the structure of tubulin, the regulation of microtubule dynamics and their deregulation in pathological processes, new therapeutic strategies are emerging, both for the treatment of cancer and for other diseases, such as neuronal or even heart diseases and parasite infections. In addition, a better understanding of the mechanism of action of well-known drugs such as colchicine or certain kinase inhibitors contributes to the development of these new therapeutic approaches. Nowadays, chemists and biologists are working jointly to select drugs which target the microtubule cytoskeleton and have improved properties. On the basis of a few examples this review attempts to depict the panorama of these recent advances.
Collapse
|
16
|
Grignard J, Lamamy V, Vermersch E, Delagrange P, Stephan JP, Dorval T, Fages F. Mathematical modeling of the microtubule detyrosination/tyrosination cycle for cell-based drug screening design. PLoS Comput Biol 2022; 18:e1010236. [PMID: 35759459 PMCID: PMC9236252 DOI: 10.1371/journal.pcbi.1010236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 05/20/2022] [Indexed: 11/18/2022] Open
Abstract
Microtubules and their post-translational modifications are involved in major cellular processes. In severe diseases such as neurodegenerative disorders, tyrosinated tubulin and tyrosinated microtubules are in lower concentration. We present here a mechanistic mathematical model of the microtubule tyrosination cycle combining computational modeling and high-content image analyses to understand the key kinetic parameters governing the tyrosination status in different cellular models. That mathematical model is parameterized, firstly, for neuronal cells using kinetic values taken from the literature, and, secondly, for proliferative cells, by a change of two parameter values obtained, and shown minimal, by a continuous optimization procedure based on temporal logic constraints to formalize experimental high-content imaging data. In both cases, the mathematical models explain the inability to increase the tyrosination status by activating the Tubulin Tyrosine Ligase enzyme. The tyrosinated tubulin is indeed the product of a chain of two reactions in the cycle: the detyrosinated microtubule depolymerization followed by its tyrosination. The tyrosination status at equilibrium is thus limited by both reaction rates and activating the tyrosination reaction alone is not effective. Our computational model also predicts the effect of inhibiting the Tubulin Carboxy Peptidase enzyme which we have experimentally validated in MEF cellular model. Furthermore, the model predicts that the activation of two particular kinetic parameters, the tyrosination and detyrosinated microtubule depolymerization rate constants, in synergy, should suffice to enable an increase of the tyrosination status in living cells.
Collapse
Affiliation(s)
- Jeremy Grignard
- Pole of Activity Data Sciences and Data Management, Institut de Recherches Servier (IdRS), Croissy-sur-Seine, France
- * E-mail: (JG); (TD); (FF)
| | - Véronique Lamamy
- Pole of Activity Cellular Sciences, Institut de Recherches Servier (IdRS), Croissy-sur-Seine, France
| | - Eva Vermersch
- Pole of Activity Cellular Sciences, Institut de Recherches Servier (IdRS), Croissy-sur-Seine, France
| | - Philippe Delagrange
- Therapeutic Area Neuropsychiatry and Immunoinflammation, Institut de Recherches Servier (IdRS), Croissy-sur-Seine, France
| | - Jean-Philippe Stephan
- In Vitro Pharmacology Unit, Institut de Recherches Servier (IdRS), Croissy-sur-Seine, France
| | - Thierry Dorval
- Pole of Activity Data Sciences and Data Management, Institut de Recherches Servier (IdRS), Croissy-sur-Seine, France
- * E-mail: (JG); (TD); (FF)
| | - François Fages
- Team Project Lifeware, Institut National de Recherche en Informatique et Automatique, Inria Saclay, Palaiseau, France
- * E-mail: (JG); (TD); (FF)
| |
Collapse
|
17
|
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] [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
|
18
|
Ying L, Zhao J, Ye Y, Liu Y, Xiao B, Xue T, Zhu H, Wu Y, He J, Qin S, Jiang Y, Guo F, Zhang L, Liu N, Zhang L. Regulation of Cdc42 signaling by the dopamine D2 receptor in a mouse model of Parkinson's disease. Aging Cell 2022; 21:e13588. [PMID: 35415964 PMCID: PMC9124300 DOI: 10.1111/acel.13588] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/29/2022] [Accepted: 02/26/2022] [Indexed: 12/02/2022] Open
Abstract
Substantial spine loss in striatal medium spiny neurons (MSNs) and abnormal behaviors are common features of Parkinson's disease (PD). The caudate putamen (CPu) mainly contains MSNs expressing dopamine D1 receptor (dMSNs) and dopamine D2 receptor (iMSNs) exerting critical effects on motor and cognition behavior. However, the molecular mechanisms contributing to spine loss and abnormal behaviors in dMSNs and iMSNs under parkinsonian state remain unknown. In the present study, we revealed that Cell division control protein 42 (Cdc42) signaling was significantly decreased in the caudate putamen (CPu) in parkinsonian mice. In addition, overexpression of constitutively active Cdc42 in the CPu reversed spine abnormalities and improved the behavior deficits in parkinsonian mice. Utilizing conditional dopamine D1 receptor (D1R) or D2 receptor (D2R) knockout mice, we found that such a decrease under parkinsonian state was further reduced by conditional knockout of the D2R but not D1R. Moreover, the thin spine loss in iMSNs and deficits in motor coordination and cognition induced by conditional knockout of D2R were reversed by overexpression of constitutively active Cdc42 in the CPu. Additionally, conditional knockout of Cdc42 from D2R‐positive neurons in the CPu was sufficient to induce spine and behavior deficits similar to those observed in parkinsonian mice. Overall, our results indicate that impaired Cdc42 signaling regulated by D2R plays an important role in spine loss and behavioral deficits in PD.
Collapse
Affiliation(s)
- Li Ying
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Jinlan Zhao
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Yingshan Ye
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Yutong Liu
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Bin Xiao
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Tao Xue
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Hangfei Zhu
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Yue Wu
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Jing He
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Sifei Qin
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Yong Jiang
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology Children's Hospital Research Foundation Cincinnati Ohio USA
| | - Lin Zhang
- Department of Histology and Embryology NMPA Key Laboratory for Safety Evaluation of Cosmetics Key Laboratory of Construction and Detection in Tissue Engineering of Guangdong Province School of Basic Medical Sciences Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Nuyun Liu
- Laboratory Animal Center Elderly Health Services Research Center Southern Medical University Guangzhou China
| | - Lu Zhang
- Key Laboratory of Functional Proteomics of Guangdong Province Key Laboratory of Mental Health of the Ministry of Education School of Basic Medical Sciences Pediatric Center of Zhujiang Hospital Center for Orthopaedic Surgery of the Third Affiliated Hospital Southern Medical University Guangzhou China
| |
Collapse
|
19
|
García-Padilla C, Muñoz-Gallardo MDM, Lozano-Velasco E, Castillo-Casas JM, Caño-Carrillo S, García-López V, Aránega A, Franco D, García-Martínez V, López-Sánchez C. New Insights into the Roles of lncRNAs as Modulators of Cytoskeleton Architecture and Their Implications in Cellular Homeostasis and in Tumorigenesis. Noncoding RNA 2022; 8:ncrna8020028. [PMID: 35447891 PMCID: PMC9033079 DOI: 10.3390/ncrna8020028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/31/2022] [Accepted: 04/09/2022] [Indexed: 11/20/2022] Open
Abstract
The importance of the cytoskeleton not only in cell architecture but also as a pivotal element in the transduction of signals that mediate multiple biological processes has recently been highlighted. Broadly, the cytoskeleton consists of three types of structural proteins: (1) actin filaments, involved in establishing and maintaining cell shape and movement; (2) microtubules, necessary to support the different organelles and distribution of chromosomes during cell cycle; and (3) intermediate filaments, which have a mainly structural function showing specificity for the cell type where they are expressed. Interaction between these protein structures is essential for the cytoskeletal mesh to be functional. Furthermore, the cytoskeleton is subject to intense spatio-temporal regulation mediated by the assembly and disassembly of its components. Loss of cytoskeleton homeostasis and integrity of cell focal adhesion are hallmarks of several cancer types. Recently, many reports have pointed out that lncRNAs could be critical mediators in cellular homeostasis controlling dynamic structure and stability of the network formed by cytoskeletal structures, specifically in different types of carcinomas. In this review, we summarize current information available about the roles of lncRNAs as modulators of actin dependent cytoskeleton and their impact on cancer pathogenesis. Finally, we explore other examples of cytoskeletal lncRNAs currently unrelated to tumorigenesis, to illustrate knowledge about them.
Collapse
Affiliation(s)
- Carlos García-Padilla
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (E.L.-V.); (V.G.-L.); (V.G.-M.)
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (M.d.M.M.-G.); (J.M.C.-C.); (S.C.-C.); (A.A.); (D.F.)
- Correspondence: (C.G.-P.); (C.L.-S.)
| | - María del Mar Muñoz-Gallardo
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (M.d.M.M.-G.); (J.M.C.-C.); (S.C.-C.); (A.A.); (D.F.)
| | - Estefanía Lozano-Velasco
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (E.L.-V.); (V.G.-L.); (V.G.-M.)
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (M.d.M.M.-G.); (J.M.C.-C.); (S.C.-C.); (A.A.); (D.F.)
- Fundación Medina, 18016 Granada, Spain
| | - Juan Manuel Castillo-Casas
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (M.d.M.M.-G.); (J.M.C.-C.); (S.C.-C.); (A.A.); (D.F.)
| | - Sheila Caño-Carrillo
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (M.d.M.M.-G.); (J.M.C.-C.); (S.C.-C.); (A.A.); (D.F.)
| | - Virginio García-López
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (E.L.-V.); (V.G.-L.); (V.G.-M.)
| | - Amelia Aránega
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (M.d.M.M.-G.); (J.M.C.-C.); (S.C.-C.); (A.A.); (D.F.)
- Fundación Medina, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain; (M.d.M.M.-G.); (J.M.C.-C.); (S.C.-C.); (A.A.); (D.F.)
- Fundación Medina, 18016 Granada, Spain
| | - Virginio García-Martínez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (E.L.-V.); (V.G.-L.); (V.G.-M.)
| | - Carmen López-Sánchez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain; (E.L.-V.); (V.G.-L.); (V.G.-M.)
- Correspondence: (C.G.-P.); (C.L.-S.)
| |
Collapse
|
20
|
Baron DM, Fenton AR, Saez-Atienzar S, Giampetruzzi A, Sreeram A, Shankaracharya, Keagle PJ, Doocy VR, Smith NJ, Danielson EW, Andresano M, McCormack MC, Garcia J, Bercier V, Van Den Bosch L, Brent JR, Fallini C, Traynor BJ, Holzbaur ELF, Landers JE. ALS-associated KIF5A mutations abolish autoinhibition resulting in a toxic gain of function. Cell Rep 2022; 39:110598. [PMID: 35385738 PMCID: PMC9134378 DOI: 10.1016/j.celrep.2022.110598] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/02/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Understanding the pathogenic mechanisms of disease mutations is critical to advancing treatments. ALS-associated mutations in the gene encoding the microtubule motor KIF5A result in skipping of exon 27 (KIF5AΔExon27) and the encoding of a protein with a novel 39 amino acid residue C-terminal sequence. Here, we report that expression of ALS-linked mutant KIF5A results in dysregulated motor activity, cellular mislocalization, altered axonal transport, and decreased neuronal survival. Single-molecule analysis revealed that the altered C terminus of mutant KIF5A results in a constitutively active state. Furthermore, mutant KIF5A possesses altered protein and RNA interactions and its expression results in altered gene expression/splicing. Taken together, our data support the hypothesis that causative ALS mutations result in a toxic gain of function in the intracellular motor KIF5A that disrupts intracellular trafficking and neuronal homeostasis.
Collapse
Affiliation(s)
- Desiree M Baron
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Adam R Fenton
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sara Saez-Atienzar
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA
| | - Anthony Giampetruzzi
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Aparna Sreeram
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Shankaracharya
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Pamela J Keagle
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Victoria R Doocy
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Nathan J Smith
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Eric W Danielson
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Megan Andresano
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Mary C McCormack
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jaqueline Garcia
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Valérie Bercier
- KU Leuven-University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven-University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Jonathan R Brent
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Claudia Fallini
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA; Department of Cell and Molecular Biology, University of Rhode Island, Kingston, RI 02881, USA; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 20892, USA; Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA; Therapeutic Development Branch, National Center for Advancing Translational Sciences, NIH, Rockville, MD 20850, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - John E Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
21
|
Dias MS, Luo X, Ribas VT, Petrs-Silva H, Koch JC. The Role of Axonal Transport in Glaucoma. Int J Mol Sci 2022; 23:ijms23073935. [PMID: 35409291 PMCID: PMC8999615 DOI: 10.3390/ijms23073935] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 11/16/2022] Open
Abstract
Glaucoma is a neurodegenerative disease that affects the retinal ganglion cells (RGCs) and leads to progressive vision loss. The first pathological signs can be seen at the optic nerve head (ONH), the structure where RGC axons leave the retina to compose the optic nerve. Besides damage of the axonal cytoskeleton, axonal transport deficits at the ONH have been described as an important feature of glaucoma. Axonal transport is essential for proper neuronal function, including transport of organelles, synaptic components, vesicles, and neurotrophic factors. Impairment of axonal transport has been related to several neurodegenerative conditions. Studies on axonal transport in glaucoma include analysis in different animal models and in humans, and indicate that its failure happens mainly in the ONH and early in disease progression, preceding axonal and somal degeneration. Thus, a better understanding of the role of axonal transport in glaucoma is not only pivotal to decipher disease mechanisms but could also enable early therapies that might prevent irreversible neuronal damage at an early time point. In this review we present the current evidence of axonal transport impairment in glaucomatous neurodegeneration and summarize the methods employed to evaluate transport in this disease.
Collapse
Affiliation(s)
- Mariana Santana Dias
- Intermediate Laboratory of Gene Therapy and Viral Vectors, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.S.D.); (H.P.-S.)
| | - Xiaoyue Luo
- Department of Neurology, University Medical Center Göttingen, 37077 Göttingen, Germany;
| | - Vinicius Toledo Ribas
- Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil;
| | - Hilda Petrs-Silva
- Intermediate Laboratory of Gene Therapy and Viral Vectors, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (M.S.D.); (H.P.-S.)
| | - Jan Christoph Koch
- Department of Neurology, University Medical Center Göttingen, 37077 Göttingen, Germany;
- Correspondence:
| |
Collapse
|
22
|
Yaremenko LM, Grabovoy AN, Shepelev SE. Expression of Cytoskeletal Proteins in Neurons of the Rat Sensorimotor Cortex upon Hypoperfusion of the Brain and Sensitization by Cerebral Antigen. NEUROPHYSIOLOGY+ 2022. [DOI: 10.1007/s11062-022-09917-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
23
|
Arlt A, Kohlschmidt N, Hentschel A, Bartels E, Groß C, Töpf A, Edem P, Szabo N, Sickmann A, Meyer N, Schara-Schmidt U, Lau J, Lochmüller H, Horvath R, Oktay Y, Roos A, Hiz S. Novel insights into PORCN mutations, associated phenotypes and pathophysiological aspects. Orphanet J Rare Dis 2022; 17:29. [PMID: 35101074 PMCID: PMC8802438 DOI: 10.1186/s13023-021-02068-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 09/30/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Goltz syndrome (GS) is a X-linked disorder defined by defects of mesodermal- and ectodermal-derived structures and caused by PORCN mutations. Features include striated skin-pigmentation, ocular and skeletal malformations and supernumerary or hypoplastic nipples. Generally, GS is associated with in utero lethality in males and most of the reported male patients show mosaicism (only three non-mosaic surviving males have been described so far). Also, precise descriptions of neurological deficits in GS are rare and less severe phenotypes might not only be caused by mosaicism but also by less pathogenic mutations suggesting the need of a molecular genetics and functional work-up of these rare variants. RESULTS We report two cases: one girl suffering from typical skin and skeletal abnormalities, developmental delay, microcephaly, thin corpus callosum, periventricular gliosis and drug-resistant epilepsy caused by a PORCN nonsense-mutation (c.283C > T, p.Arg95Ter). Presence of these combined neurological features indicates that CNS-vulnerability might be a guiding symptom in the diagnosis of GS patients. The other patient is a boy with a supernumerary nipple and skeletal anomalies but also, developmental delay, microcephaly, cerebral atrophy with delayed myelination and drug-resistant epilepsy as predominant features. Skin abnormalities were not observed. Genotyping revealed a novel PORCN missense-mutation (c.847G > C, p.Asp283His) absent in the Genome Aggregation Database (gnomAD) but also identified in his asymptomatic mother. Given that non-random X-chromosome inactivation was excluded in the mother, fibroblasts of the index had been analyzed for PORCN protein-abundance and -distribution, vulnerability against additional ER-stress burden as well as for protein secretion revealing changes. CONCLUSIONS Our combined findings may suggest incomplete penetrance for the p.Asp283His variant and provide novel insights into the molecular etiology of GS by adding impaired ER-function and altered protein secretion to the list of pathophysiological processes resulting in the clinical manifestation of GS.
Collapse
Affiliation(s)
- Annabelle Arlt
- Institute of Clinical Genetics and Tumor Genetics, Bonn, Germany
| | | | | | - Enrika Bartels
- Institute of Clinical Genetics and Tumor Genetics, Bonn, Germany
| | - Claudia Groß
- Institute of Clinical Genetics and Tumor Genetics, Bonn, Germany
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Pınar Edem
- Department of Medical Biology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Nora Szabo
- Department of Clinical Neurosciences, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Albert Sickmann
- Leibniz Institute for Analytical Sciences (ISAS), Dortmund, Germany
| | - Nancy Meyer
- Pediatric Neurology, Faculty of Medicine, University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Ulrike Schara-Schmidt
- Pediatric Neurology, Faculty of Medicine, University of Duisburg-Essen, University Hospital, Essen, Germany
| | - Jarred Lau
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada
- Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Catalonia, Spain
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Yavuz Oktay
- Department of Medical Biology, School of Medicine, Dokuz Eylul University, Izmir, Turkey
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Andreas Roos
- Department of Clinical Neurosciences, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada.
| | - Semra Hiz
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| |
Collapse
|
24
|
Oláh J, Szénási T, Lehotzky A, Norris V, Ovádi J. Challenges in Discovering Drugs That Target the Protein-Protein Interactions of Disordered Proteins. Int J Mol Sci 2022; 23:ijms23031550. [PMID: 35163473 PMCID: PMC8835748 DOI: 10.3390/ijms23031550] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022] Open
Abstract
Protein–protein interactions (PPIs) outnumber proteins and are crucial to many fundamental processes; in consequence, PPIs are associated with several pathological conditions including neurodegeneration and modulating them by drugs constitutes a potentially major class of therapy. Classically, however, the discovery of small molecules for use as drugs entails targeting individual proteins rather than targeting PPIs. This is largely because discovering small molecules to modulate PPIs has been seen as extremely challenging. Here, we review the difficulties and limitations of strategies to discover drugs that target PPIs directly or indirectly, taking as examples the disordered proteins involved in neurodegenerative diseases.
Collapse
Affiliation(s)
- Judit Oláh
- Institute of Enzymology, Research Centre for Natural Sciences, ELKH, 1117 Budapest, Hungary; (J.O.); (T.S.); (A.L.)
| | - Tibor Szénási
- Institute of Enzymology, Research Centre for Natural Sciences, ELKH, 1117 Budapest, Hungary; (J.O.); (T.S.); (A.L.)
| | - Attila Lehotzky
- Institute of Enzymology, Research Centre for Natural Sciences, ELKH, 1117 Budapest, Hungary; (J.O.); (T.S.); (A.L.)
| | - Victor Norris
- Laboratory of Microbiology Signals and Microenvironment, University of Rouen, 76821 Mont Saint Aignan, France;
| | - Judit Ovádi
- Institute of Enzymology, Research Centre for Natural Sciences, ELKH, 1117 Budapest, Hungary; (J.O.); (T.S.); (A.L.)
- Correspondence:
| |
Collapse
|
25
|
Piermarini E, Akarsu S, Connors T, Kneussel M, Lane MA, Morfini G, Karabay A, Baas PW, Qiang L. Modeling gain-of-function and loss-of-function components of SPAST-based hereditary spastic paraplegia using transgenic mice. Hum Mol Genet 2021; 31:1844-1859. [PMID: 34935948 PMCID: PMC9169457 DOI: 10.1093/hmg/ddab367] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Hereditary spastic paraplegia (HSP) is a disease in which dieback degeneration of corticospinal tracts, accompanied by axonal swellings, leads to gait deficiencies. SPG4-HSP, the most common form of the disease, results from mutations of human spastin gene (SPAST), which is the gene that encodes spastin, a microtubule-severing protein. The lack of a vertebrate model that recapitulates both the etiology and symptoms of SPG4-HSP has stymied the development of effective therapies for the disease. hSPAST-C448Y mice, which express human mutant spastin at the ROSA26 locus, display corticospinal dieback and gait deficiencies but not axonal swellings. On the other hand, mouse spastin gene (Spast)-knockout (KO) mice display axonal swellings but not corticospinal dieback or gait deficiencies. One possibility is that reduced spastin function, resulting in axonal swellings, is not the cause of the disease but exacerbates the toxic effects of the mutant protein. To explore this idea, Spast-KO and hSPAST-C448Y mice were crossbred, and the offspring were compared with the parental lines via histological and behavioral analyses. The crossbred animals displayed axonal swellings as well as earlier onset, worsened gait deficiencies and corticospinal dieback compared with the hSPAST-C448Y mouse. These results, together with observations on changes in histone deacetylases 6 and tubulin modifications in the axon, indicate that each of these three transgenic mouse lines is valuable for investigating a different component of the disease pathology. Moreover, the crossbred mice are the best vertebrate model to date for testing potential therapies for SPG4-HSP.
Collapse
Affiliation(s)
- Emanuela Piermarini
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Seyma Akarsu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA,Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul 34469, Turkey
| | - Theresa Connors
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Matthias Kneussel
- Department of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Gerardo Morfini
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Arzu Karabay
- Department of Molecular Biology and Genetics, Istanbul Technical University, Istanbul 34469, Turkey
| | - Peter W Baas
- To whom correspondence should be addressed at: Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA. Tel: +1 2159918311; Fax: +1 2158439082; ; Tel: +1 2159918298;
| | - Liang Qiang
- To whom correspondence should be addressed at: Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA. Tel: +1 2159918311; Fax: +1 2158439082; ; Tel: +1 2159918298;
| |
Collapse
|
26
|
Sanyal C, Pietsch N, Ramirez Rios S, Peris L, Carrier L, Moutin MJ. The detyrosination/re-tyrosination cycle of tubulin and its role and dysfunction in neurons and cardiomyocytes. Semin Cell Dev Biol 2021; 137:46-62. [PMID: 34924330 DOI: 10.1016/j.semcdb.2021.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/28/2022]
Abstract
Among the variety of post-translational modifications to which microtubules are subjected, the detyrosination/re-tyrosination cycle is specific to tubulin. It is conserved by evolution and characterized by the enzymatic removal and re-addition of a gene-encoded tyrosine residue at the C-terminus of α-tubulin. Detyrosinated tubulin can be further converted to Δ2-tubulin by the removal of an additional C-terminal glutamate residue. Detyrosinated and Δ2-tubulin are carried by stable microtubules whereas tyrosinated microtubules are present on dynamic polymers. The cycle regulates trafficking of many cargo transporting molecular motors and is linked to the microtubule dynamics via regulation of microtubule interactions with specific cellular effectors such as kinesin-13. Here, we give an historical overview of the general features discovered for the cycle. We highlight the recent progress toward structure and functioning of the enzymes that keep the levels of tyrosinated and detyrosinated tubulin in cells, the long-known tubulin tyrosine ligase and the recently discovered vasohibin-SVBP complexes. We further describe how the cycle controls microtubule functions in healthy neurons and cardiomyocytes and how deregulations of the cycle are involved in dysfunctions of these highly differentiated cells, leading to neurodegeneration and heart failure in humans.
Collapse
Affiliation(s)
- Chadni Sanyal
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Niels Pietsch
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Sacnicte Ramirez Rios
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Leticia Peris
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Marie-Jo Moutin
- Univ. Grenoble Alpes, Inserm, U1216, CNRS, Grenoble Institut Neurosciences, 38000 Grenoble, France.
| |
Collapse
|
27
|
Raposo M, Bettencourt C, Melo ARV, Ferreira AF, Alonso I, Silva P, Vasconcelos J, Kay T, Saraiva-Pereira ML, Costa MD, Vilasboas-Campos D, Bettencourt BF, Bruges-Armas J, Houlden H, Heutink P, Jardim LB, Sequeiros J, Maciel P, Lima M. Novel Machado-Joseph disease-modifying genes and pathways identified by whole-exome sequencing. Neurobiol Dis 2021; 162:105578. [PMID: 34871736 DOI: 10.1016/j.nbd.2021.105578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/08/2021] [Accepted: 12/02/2021] [Indexed: 11/17/2022] Open
Abstract
Machado-Joseph disease (MJD/SCA3) is a neurodegenerative polyglutamine disorder exhibiting a wide spectrum of phenotypes. The abnormal size of the (CAG)n at ATXN3 explains ~55% of the age at onset variance, suggesting the involvement of other factors, namely genetic modifiers, whose identification remains limited. Our aim was to find novel genetic modifiers, analyse their epistatic effects and identify disease-modifying pathways contributing to MJD variable expressivity. We performed whole-exome sequencing in a discovery sample of four age at onset concordant and four discordant first-degree relative pairs of Azorean patients, to identify candidate variants which genotypes differed for each discordant pair but were shared in each concordant pair. Variants identified by this approach were then tested in an independent multi-origin cohort of 282 MJD patients. Whole-exome sequencing identified 233 candidate variants, from which 82 variants in 53 genes were prioritized for downstream analysis. Eighteen disease-modifying pathways were identified; two of the most enriched pathways were relevant for the nervous system, namely the neuregulin signaling and the agrin interactions at neuromuscular junction. Variants at PARD3, NFKB1, CHD5, ACTG1, CFAP57, DLGAP2, ITGB1, DIDO1 and CERS4 modulate age at onset in MJD, with those identified in CFAP57, ACTG1 and DIDO1 showing consistent effects across cohorts of different geographical origins. Network analyses of the nine novel MJD modifiers highlighted several important molecular interactions, including genes/proteins previously related with MJD pathogenesis, namely between ACTG1/APOE and VCP/ITGB1. We describe novel pathways, modifiers, and their interaction partners, providing a broad molecular portrait of age at onset modulation to be further exploited as new disease-modifying targets for MJD and related diseases.
Collapse
Affiliation(s)
- Mafalda Raposo
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal; Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal.
| | - Conceição Bettencourt
- Department of Neurodegenerative Disease and Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, London, UK.
| | - Ana Rosa Vieira Melo
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal; Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal
| | - Ana F Ferreira
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal; Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal.
| | - Isabel Alonso
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Paulo Silva
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.
| | - João Vasconcelos
- Departamento de Neurologia, Hospital do Divino Espírito Santo, Ponta Delgada, Portugal
| | - Teresa Kay
- Departamento de Genética Clínica, Hospital D. Estefânia, Lisboa, Portugal
| | - Maria Luiza Saraiva-Pereira
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Brazil; Serviço de Genética Médica/Centro de Pesquisa Clínica e Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| | - Marta D Costa
- Instituto de Investigação em Ciências da Vida e Saúde (ICVS), Escola de Medicina, Universidade do Minho, Braga, Portugal; ICVS/3B's - Laboratório Associado, Braga/Guimarães, Portugal.
| | - Daniela Vilasboas-Campos
- Instituto de Investigação em Ciências da Vida e Saúde (ICVS), Escola de Medicina, Universidade do Minho, Braga, Portugal; ICVS/3B's - Laboratório Associado, Braga/Guimarães, Portugal
| | - Bruno Filipe Bettencourt
- Serviço Especializado de Epidemiologia e Biologia Molecular (SEEBMO), Hospital de Santo Espírito da Ilha Terceira (HSEIT), Angra do Heroísmo, Azores, Portugal
| | - Jácome Bruges-Armas
- Serviço Especializado de Epidemiologia e Biologia Molecular (SEEBMO), Hospital de Santo Espírito da Ilha Terceira (HSEIT), Angra do Heroísmo, Azores, Portugal; CHRC - Comprehensive Health Research Centre, Faculdade de Ciências Médicas & CEDOC - Chronic Diseases Research Center, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Henry Houlden
- Department of Molecular Neuroscience, Institute of Neurology, University College London and Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, University College London Hospitals, London, United Kingdom, London.
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany.
| | - Laura Bannach Jardim
- Departamento de Medicina Interna, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Serviço de Genética Médica/Centro de Pesquisa Clínica e Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| | - Jorge Sequeiros
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.
| | - Patrícia Maciel
- Instituto de Investigação em Ciências da Vida e Saúde (ICVS), Escola de Medicina, Universidade do Minho, Braga, Portugal; ICVS/3B's - Laboratório Associado, Braga/Guimarães, Portugal.
| | - Manuela Lima
- Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal; Faculdade de Ciências e Tecnologia, Universidade dos Açores, Ponta Delgada, Portugal.
| |
Collapse
|
28
|
Serratos IN, Hernández-Pérez E, Campos C, Aschner M, Santamaría A. An Update on the Critical Role of α-Synuclein in Parkinson's Disease and Other Synucleinopathies: from Tissue to Cellular and Molecular Levels. Mol Neurobiol 2021; 59:620-642. [PMID: 34750787 DOI: 10.1007/s12035-021-02596-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022]
Abstract
The aggregation of alpha-synuclein (α-Syn) plays a critical role in the development of Parkinson's disease (PD) and other synucleinopathies. α-Syn, which is encoded by the SNCA gene, is a lysine-rich soluble amphipathic protein normally expressed in neurons. Located in the cytosolic domain, this protein has the ability to remodel itself in plasma membranes, where it assumes an alpha-helix conformation. However, the protein can also adopt another conformation rich in cross-beta sheets, undergoing mutations and post-translational modifications, then leading the protein to an unusual aggregation in the form of Lewy bodies (LB), which are cytoplasmic inclusions constituted predominantly by α-Syn. Pathogenic mechanisms affecting the structural and functional stability of α-Syn - such as endoplasmic reticulum stress, Golgi complex fragmentation, disfunctional protein degradation systems, aberrant interactions with mitochondrial membranes and nuclear DNA, altered cytoskeleton dynamics, disrupted neuronal plasmatic membrane, dysfunctional vesicular transport, and formation of extracellular toxic aggregates - contribute all to the pathogenic progression of PD and synucleinopathies. In this review, we describe the collective knowledge on this topic and provide an update on the critical role of α-Syn aggregates, both at the cellular and molecular levels, in the deregulation of organelles affecting the cellular homeostasis and leading to neuronal cell death in PD and other synucleinopathies.
Collapse
Affiliation(s)
- Iris N Serratos
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Mexico City, Mexico
| | - Elizabeth Hernández-Pérez
- Departamento de Ciencias de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Mexico City, Mexico
| | - Carolina Campos
- Departamento de Ciencias de La Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, Mexico City, Mexico.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores/Laboratorio de Neurofarmacología Molecular y Nanotecnología, Instituto Nacional de Neurología y Neurocirugía, SSA, 14269, Mexico City, Mexico.
| |
Collapse
|
29
|
Santiago-Mujika E, Luthi-Carter R, Giorgini F, Kalaria RN, Mukaetova-Ladinska EB. Tubulin and Tubulin Posttranslational Modifications in Alzheimer's Disease and Vascular Dementia. Front Aging Neurosci 2021; 13:730107. [PMID: 34776926 PMCID: PMC8586541 DOI: 10.3389/fnagi.2021.730107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 10/04/2021] [Indexed: 01/26/2023] Open
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) are the two most common forms of dementia in older people. Although these two dementia types differ in their etiology, they share many pathophysiological and morphological features, including neuronal loss, which is associated with the microtubule (MT) destabilization. Stabilization of MTs is achieved in different ways: through interactions with MT binding proteins (MTBP) or by posttranslational modifications (PTMs) of tubulin. Polyglutamylation and tyrosination are two foremost PTMs that regulate the interaction between MTs and MTBPs, and play, therefore, a role in neurodegeneration. In this review, we summarize key information on tubulin PTMs in relation to AD and VaD and address the importance of studying further the tubulin code to reveal sites of potential intervention in development of novel and effective dementia therapy.
Collapse
Affiliation(s)
- Estibaliz Santiago-Mujika
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
| | - Ruth Luthi-Carter
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
| | - Flaviano Giorgini
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Raj N. Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Elizabeta B. Mukaetova-Ladinska
- Department of Neuroscience, Behavior and Psychology, University of Leicester, Leicester, United Kingdom
- Evington Centre, Leicester General Hospital, Leicester, United Kingdom
| |
Collapse
|
30
|
Dehghan F, Zamani S, Barreiro C, Jami MS. Irisin injection mimics exercise effects on the brain proteome. Eur J Neurosci 2021; 54:7422-7441. [PMID: 34655501 DOI: 10.1111/ejn.15493] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 12/13/2022]
Abstract
Physical inactivity can endanger human health and increase the incidence of neurodegenerative disease. Exercise has tremendous beneficial effects on brain health and cognitive function, especially in older adults. It also improves brain-related outcomes in depression, epilepsy and neurodegenerative disorders, such as Parkinson's disease and Alzheimer's disease. Irisin is a mediator of the beneficial effects of exercise. This study aimed to assess the proteome alterations in adult male National Maritime Research Institute (NMRI) mice brain tissue upon three different conditions including endurance exercise, resistance exercise and irisin injection. Quantification of irisin levels in blood was performed using irisin-ELISA Kit. Quantification and identification of proteins via two-dimensional gel electrophoresis and matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry (MS)/MS showed the alteration of at least 21 proteins due to different treatments. Cellular pathway analysis revealed common beneficial effects of sole irisin treatment and different exercise procedures suggesting the capability of irisin injection to substitute the exercise when physical activity is not possible.
Collapse
Affiliation(s)
- Fariba Dehghan
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Saeed Zamani
- Department of Anatomical Sciences, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Department of Anatomical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Carlos Barreiro
- INBIOTEC (Instituto de Biotecnología de León), León, Spain.,Biochemistry and Molecular Biology Area, Department of Molecular Biology, University of León, Vegazana Campus, León, Spain
| | - Mohammad-Saeid Jami
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.,QIANBIOTEC, Research and Development Center for Biotechnology, Isfahan, Iran.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
31
|
Molecular Pathways Involved in Frontotemporal Lobar Degeneration with TDP-43 Proteinopathy: What Can We Learn from Proteomics? Int J Mol Sci 2021; 22:ijms221910298. [PMID: 34638637 PMCID: PMC8508653 DOI: 10.3390/ijms221910298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022] Open
Abstract
Frontotemporal lobar degeneration (FTLD) is a neurodegenerative disorder clinically characterized by behavioral, language, and motor symptoms, with major impact on the lives of patients and their families. TDP-43 proteinopathy is the underlying neuropathological substrate in the majority of cases, referred to as FTLD-TDP. Several genetic causes have been identified, which have revealed some components of its pathophysiology. However, the exact mechanisms driving FTLD-TDP remain largely unknown, forestalling the development of therapies. Proteomic approaches, in particular high-throughput mass spectrometry, hold promise to help elucidate the pathogenic molecular and cellular alterations. In this review, we describe the main findings of the proteomic profiling studies performed on human FTLD-TDP brain tissue. Subsequently, we address the major biological pathways implicated in FTLD-TDP, by reviewing these data together with knowledge derived from genomic and transcriptomic literature. We illustrate that an integrated perspective, encompassing both proteomic, genetic, and transcriptomic discoveries, is vital to unravel core disease processes, and to enable the identification of disease biomarkers and therapeutic targets for this devastating disorder.
Collapse
|
32
|
Pérez-Martín E, Muñoz-Castañeda R, Moutin MJ, Ávila-Zarza CA, Muñoz-Castañeda JM, Del Pilar C, Alonso JR, Andrieux A, Díaz D, Weruaga E. Oleoylethanolamide Delays the Dysfunction and Death of Purkinje Cells and Ameliorates Behavioral Defects in a Mouse Model of Cerebellar Neurodegeneration. Neurotherapeutics 2021; 18:1748-1767. [PMID: 33829414 PMCID: PMC8609004 DOI: 10.1007/s13311-021-01044-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2021] [Indexed: 02/04/2023] Open
Abstract
Oleoylethanolamide (OEA) is an endocannabinoid that has been proposed to prevent neuronal damage and neuroinflammation. In this study, we evaluated the effects of OEA on the disruption of both cerebellar structure and physiology and on the behavior of Purkinje cell degeneration (PCD) mutant mice. These mice exhibit cerebellar degeneration, displaying microtubule alterations that trigger the selective loss of Purkinje cells and consequent behavioral impairments. The effects of different doses (1, 5, and 10 mg/kg, i.p.) and administration schedules (chronic and acute) of OEA were assessed at the behavioral, histological, cellular, and molecular levels to determine the most effective OEA treatment regimen. Our in vivo results demonstrated that OEA treatment prior to the onset of the preneurodegenerative phase prevented morphological alterations in Purkinje neurons (the somata and dendritic arbors) and decreased Purkinje cell death. This effect followed an inverted U-shaped time-response curve, with acute administration on postnatal day 12 (10 mg/kg, i.p.) being the most effective treatment regimen tested. Indeed, PCD mice that received this specific OEA treatment regimen showed improvements in motor, cognitive and social functions, which were impaired in these mice. Moreover, these in vivo neuroprotective effects of OEA were mediated by the PPARα receptor, as pretreatment with the PPARα antagonist GW6471 (2.5 mg/kg, i.p.) abolished them. Finally, our in vitro results suggested that the molecular effect of OEA was related to microtubule stability and structure since OEA administration normalized some alterations in microtubule features in PCD-like cells. These findings provide strong evidence supporting the use of OEA as a pharmacological agent to limit severe cerebellar neurodegenerative processes.
Collapse
Affiliation(s)
- Ester Pérez-Martín
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Rodrigo Muñoz-Castañeda
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Marie-Jo Moutin
- GIN, Univ. Grenoble Alpes, CNRS, CEA, Grenoble Institute Neurosciences, Inserm, U121638000, Grenoble, France
| | - Carmelo A Ávila-Zarza
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
- Department of Statistics, University of Salamanca, 37007, Salamanca, Spain
| | - José M Muñoz-Castañeda
- Department of Theoretical, Atomic and Optical Physics, University of Valladolid, 47071, Valladolid, Spain
| | - Carlos Del Pilar
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - José R Alonso
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
- Universidad de Tarapacá, Arica, Chile
| | - Annie Andrieux
- GIN, Univ. Grenoble Alpes, CNRS, CEA, Grenoble Institute Neurosciences, Inserm, U121638000, Grenoble, France
| | - David Díaz
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
| | - Eduardo Weruaga
- Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neurosciences of Castile and Leon (INCyL), University of Salamanca, 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
33
|
Huang L, Fang L, Liu Q, Torshizi AD, Wang K. Integrated analysis on transcriptome and behaviors defines HTT repeat-dependent network modules in Huntington's disease. Genes Dis 2021; 9:479-493. [PMID: 35224162 PMCID: PMC8843892 DOI: 10.1016/j.gendis.2021.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/13/2021] [Accepted: 05/12/2021] [Indexed: 01/02/2023] Open
Abstract
Huntington's disease (HD) is caused by a CAG repeat expansion in the huntingtin (HTT) gene. Knock-in mice carrying a CAG repeat-expanded Htt will develop HD phenotypes. Previous studies suggested dysregulated molecular networks in a CAG length genotype- and the age-dependent manner in brain tissues from knock-in mice carrying expanded Htt CAG repeats. Furthermore, a large-scale phenome analysis defined a behavioral signature for HD genotype in knock-in mice carrying expanded Htt CAG repeats. However, an integrated analysis correlating phenotype features with genotypes (CAG repeat expansions) was not conducted previously. In this study, we revealed the landscape of the behavioral features and gene expression correlations based on 445 mRNA samples and 445 microRNA samples, together with behavioral features (396 PhenoCube behaviors and 111 NeuroCube behaviors) in Htt CAG-knock-in mice. We identified 37 behavioral features that were significantly associated with CAG repeat length including the number of steps and hind limb stand duration. The behavioral features were associated with several gene coexpression groups involved in neuronal dysfunctions, which were also supported by the single-cell RNA sequencing data in the striatum and the spatial gene expression in the brain. We also identified 15 chemicals with significant responses for genes with enriched behavioral features, most of them are agonist or antagonist for dopamine receptors and serotonin receptors used for neurology/psychiatry. Our study provides further evidence that abnormal neuronal signal transduction in the striatum plays an important role in causing HD-related phenotypic behaviors and provided rich information for the further pharmacotherapeutic intervention possibility for HD.
Collapse
Affiliation(s)
- Lulin Huang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Clinical Laboratory, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, PR China
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Corresponding author. The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Clinical Laboratory, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610054, PR China.
| | - Li Fang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Qian Liu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Abolfazl Doostparast Torshizi
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Kai Wang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Corresponding author.
| |
Collapse
|
34
|
Effect of ethanol and cocaine on [ 11C]MPC-6827 uptake in SH-SY5Y cells. Mol Biol Rep 2021; 48:3871-3876. [PMID: 33880672 PMCID: PMC8172511 DOI: 10.1007/s11033-021-06336-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/02/2021] [Indexed: 10/26/2022]
Abstract
Microtubules (MTs) are structural units in the cytoskeleton. In brain cells they are responsible for axonal transport, information processing, and signaling mechanisms. Proper function of these processes is critical for healthy brain functions. Alcohol and substance use disorders (AUD/SUDs) affects the function and organization of MTs in the brain, making them a potential neuroimaging marker to study the resulting impairment of overall neurobehavioral and cognitive processes. Our lab reported the first brain-penetrant MT-tracking Positron Emission Tomography (PET) ligand [11C]MPC-6827 and demonstrated its in vivo utility in rodents and non-human primates. To further explore the in vivo imaging potential of [11C]MPC-6827, we need to investigate its mechanism of action. Here, we report preliminary in vitro binding results in SH-SY5Y neuroblastoma cells exposed to ethanol (EtOH) or cocaine in combination with multiple agents that alter MT stability. EtOH and cocaine treatments increased MT stability and decreased free tubulin monomers. Our initial cell-binding assay demonstrated that [11C]MPC-6827 may have high affinity to free/unbound tubulin units. Consistent with this mechanism of action, we observed lower [11C]MPC-6827 uptake in SH-SY5Y cells after EtOH and cocaine treatments (e.g., fewer free tubulin units). We are currently performing in vivo PET imaging and ex vivo biodistribution studies in rodent and nonhuman primate models of AUD and SUDs and Alzheimer's disease.
Collapse
|
35
|
Efimova N, Yang C, Chia JX, Li N, Lengner CJ, Neufeld KL, Svitkina TM. Branched actin networks are assembled on microtubules by adenomatous polyposis coli for targeted membrane protrusion. J Cell Biol 2021; 219:151902. [PMID: 32597939 PMCID: PMC7480092 DOI: 10.1083/jcb.202003091] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/26/2022] Open
Abstract
Cell migration is driven by pushing and pulling activities of the actin cytoskeleton, but migration directionality is largely controlled by microtubules. This function of microtubules is especially critical for neuron navigation. However, the underlying mechanisms are poorly understood. Here we show that branched actin filament networks, the main pushing machinery in cells, grow directly from microtubule tips toward the leading edge in growth cones of hippocampal neurons. Adenomatous polyposis coli (APC), a protein with both tumor suppressor and cytoskeletal functions, concentrates at the microtubule-branched network interface, whereas APC knockdown nearly eliminates branched actin in growth cones and prevents growth cone recovery after repellent-induced collapse. Conversely, encounters of dynamic APC-positive microtubule tips with the cell edge induce local actin-rich protrusions. Together, we reveal a novel mechanism of cell navigation involving APC-dependent assembly of branched actin networks on microtubule tips.
Collapse
Affiliation(s)
- Nadia Efimova
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - Changsong Yang
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - Jonathan X Chia
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| | - Ning Li
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA
| | - Christopher J Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA.,Department of Cell and Developmental Biology, Perelman School of Medicine and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA
| | - Kristi L Neufeld
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS
| | - Tatyana M Svitkina
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
36
|
Shafiq M, Zafar S, Younas N, Noor A, Puig B, Altmeppen HC, Schmitz M, Matschke J, Ferrer I, Glatzel M, Zerr I. Prion protein oligomers cause neuronal cytoskeletal damage in rapidly progressive Alzheimer's disease. Mol Neurodegener 2021; 16:11. [PMID: 33618749 PMCID: PMC7898440 DOI: 10.1186/s13024-021-00422-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 01/02/2021] [Indexed: 12/27/2022] Open
Abstract
Background High-density oligomers of the prion protein (HDPs) have previously been identified in brain tissues of patients with rapidly progressive Alzheimer’s disease (rpAD). The current investigation aims at identifying interacting partners of HDPs in the rpAD brains to unravel the pathological involvement of HDPs in the rapid progression. Methods HDPs from the frontal cortex tissues of rpAD brains were isolated using sucrose density gradient centrifugation. Proteins interacting with HDPs were identified by co-immunoprecipitation coupled with mass spectrometry. Further verifications were carried out using proteomic tools, immunoblotting, and confocal laser scanning microscopy. Results We identified rpAD-specific HDP-interactors, including the growth arrest specific 2-like 2 protein (G2L2). Intriguingly, rpAD-specific disturbances were found in the localization of G2L2 and its associated proteins i.e., the end binding protein 1, α-tubulin, and β-actin. Discussion The results show the involvement of HDPs in the destabilization of the neuronal actin/tubulin infrastructure. We consider this disturbance to be a contributing factor for the rapid progression in rpAD. Supplementary Information The online version contains supplementary material available at 10.1186/s13024-021-00422-x.
Collapse
Affiliation(s)
- Mohsin Shafiq
- Department of Neurology, University Medicine Goettingen and German Center for Neurodegenerative Diseases (DZNE), 37075, Goettingen, Germany.,Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), 20246, Hamburg, Germany
| | - Saima Zafar
- Department of Neurology, University Medicine Goettingen and German Center for Neurodegenerative Diseases (DZNE), 37075, Goettingen, Germany. .,Biomedical Engineering and Sciences Department, School of Mechanical and Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Islamabad, Pakistan.
| | - Neelam Younas
- Department of Neurology, University Medicine Goettingen and German Center for Neurodegenerative Diseases (DZNE), 37075, Goettingen, Germany
| | - Aneeqa Noor
- Department of Neurology, University Medicine Goettingen and German Center for Neurodegenerative Diseases (DZNE), 37075, Goettingen, Germany
| | - Berta Puig
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), 20246, Hamburg, Germany.,Department of Neurology, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Hermann Clemens Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), 20246, Hamburg, Germany
| | - Matthias Schmitz
- Department of Neurology, University Medicine Goettingen and German Center for Neurodegenerative Diseases (DZNE), 37075, Goettingen, Germany
| | - Jakob Matschke
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), 20246, Hamburg, Germany
| | - Isidre Ferrer
- Institut de Neuropatologica, Servei Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, Carrer Feixa LLarga sn, 08907, Hospitalet de LLobregat, CIBERNED, Barcelona, Spain
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), 20246, Hamburg, Germany
| | - Inga Zerr
- Department of Neurology, University Medicine Goettingen and German Center for Neurodegenerative Diseases (DZNE), 37075, Goettingen, Germany
| |
Collapse
|
37
|
Hao R, Sun B, Yang L, Ma C, Li S. RVG29-modified microRNA-loaded nanoparticles improve ischemic brain injury by nasal delivery. Drug Deliv 2021; 27:772-781. [PMID: 32400219 PMCID: PMC7269067 DOI: 10.1080/10717544.2020.1760960] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Effective nose-to-brain delivery needs to be developed to treat neurodegenerative diseases. Regulating miR-124 can effectively improve the symptoms of ischemic brain injury and provide a certain protective effect from brain damage after cerebral ischemia. We used rat models of middle cerebral artery occlusion (t-MCAO) with ischemic brain injury, and we delivered RVG29-NPs-miR124 intranasally to treat neurological damage after cerebral ischemia. Rhoa and neurological scores in rats treated by intranasal administration of RVG29-PEG-PLGA/miRNA-124 were significantly lower than those in PEG-PLGA/miRNA-124 nasal administration and RVG29-PLGA/miRNA-124 nasal administration group treated rats. These results indicate that the nose-to-brain delivery of PLGA/miRNA-124 conjugated with PEG and RVG29 alleviated the symptoms of cerebral ischemia-reperfusion injury. Thus, nasal delivery of RVG29-PEG-PLGA/miRNA-124 could be a new method for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Rubin Hao
- Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, ChangChun, Jilin, China
| | - Bixi Sun
- School of Pharmaceutical Sciences, Jilin University, ChangChun, Jilin, China
| | - Lihua Yang
- Affiliated Hospital of Changchun University of Chinese medicine, ChangChun, Jilin, China
| | - Chun Ma
- Affiliated Hospital of Changchun University of Chinese medicine, ChangChun, Jilin, China
| | - Shuling Li
- Affiliated Hospital of Changchun University of Chinese medicine, ChangChun, Jilin, China
| |
Collapse
|
38
|
Mechanisms and Therapeutic Implications of GSK-3 in Treating Neurodegeneration. Cells 2021; 10:cells10020262. [PMID: 33572709 PMCID: PMC7911291 DOI: 10.3390/cells10020262] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders are spreading worldwide and are one of the greatest threats to public health. There is currently no adequate therapy for these disorders, and therefore there is an urgent need to accelerate the discovery and development of effective treatments. Although neurodegenerative disorders are broad ranging and highly complex, they may share overlapping mechanisms, and thus potentially manifest common targets for therapeutic interventions. Glycogen synthase kinase-3 (GSK-3) is now acknowledged to be a central player in regulating mood behavior, cognitive functions, and neuron viability. Indeed, many targets controlled by GSK-3 are critically involved in progressing neuron deterioration and disease pathogenesis. In this review, we focus on three pathways that represent prominent mechanisms linking GSK-3 with neurodegenerative disorders: cytoskeleton organization, the mammalian target of rapamycin (mTOR)/autophagy axis, and mitochondria. We also consider the challenges and opportunities in the development of GSK-3 inhibitors for treating neurodegeneration.
Collapse
|
39
|
Xu W, Gao C, Wu J. CD151 Alleviates Early Blood-Brain Barrier Dysfunction After Experimental Focal Brain Ischemia in Rats. Cell Mol Neurobiol 2021; 41:151-162. [PMID: 32285246 DOI: 10.1007/s10571-020-00842-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/04/2020] [Indexed: 12/12/2022]
Abstract
Preservation of the blood-brain barrier (BBB) function is a potential protective strategy against cerebral ischemic injuries. CD151 has a beneficial effect in maintaining vascular stability and plays a role in pro-angiogenesis. Both vascular stability and angiogenesis can affect BBB function. Therefore, we aimed to examine the action of CD151 in regulating BBB permeability after cerebral ischemic injury in the present study. Using a transient focal cerebral ischemia (tFCI) rat model, we established that CD151 overexpression in the brain mitigated the leakage of endogenous IgG at 6-24 h after tFCI in vivo. Moreover, we found that CD151 can decrease the diffusion of macromolecules through monolayer brain microvessel endothelial cells (BMVECs) after glucose and oxygen deprivation (OGD)-reoxygenation in vitro. Furthermore, overexpression of CD151 in BMVECs suppressed OGD-reoxygenation-induced F-actin formation and RhoA activity. However, while preserving BBB integrity after tFCI, CD151 overexpression did not affect the post-stroke outcomes. Taken together, the present study demonstrated that CD151 overexpression in the brain protects BBB permeability at early phase after tFCI. CD151 may be a potential target for early BBB protection in ischemic stroke.
Collapse
Affiliation(s)
- Wendeng Xu
- Department of Neurology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Ceshu Gao
- Department of Neurology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Jian Wu
- Department of Neurology, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China.
| |
Collapse
|
40
|
Leite SC, Pinto-Costa R, Sousa MM. Actin dynamics in the growth cone: a key player in axon regeneration. Curr Opin Neurobiol 2020; 69:11-18. [PMID: 33359956 DOI: 10.1016/j.conb.2020.11.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023]
Abstract
Neuronal development, maintenance and function depends on the tight regulation of cytoskeleton organization and dynamics. Following injury, adult central nervous system neurons have a limited ability to regenerate and to recapitulate their robust developmental axon growth. This decreased regenerative capacity is set by their inability to establish regeneration-competent growth cones. Growth cones are actin-enriched structures that regulate axon extension rate and direction. During neuronal development, increasing actin dynamics in the growth cone through the regulation of the activity of specific actin-binding proteins leads to increased axon elongation. Here, we will focus on recent findings showing that enhanced axon regeneration in the adult nervous system can be achieved by promoting actin dynamics, or by decreasing actomyosin contraction in the growth cone. These discoveries underscore the importance of actin organization in the growth cone as a key factor to promote axon (re)growth that should be further explored in the future.
Collapse
Affiliation(s)
- Sérgio Carvalho Leite
- Institute of Psychiatry and Neurosciences of Paris, INSERM U1266, Université de Paris, 75014 Paris, France
| | - Rita Pinto-Costa
- Nerve Regeneration Group, i3S- Instituto de Investigação e Inovação em Saúde and IBMC- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal
| | - Monica Mendes Sousa
- Nerve Regeneration Group, i3S- Instituto de Investigação e Inovação em Saúde and IBMC- Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
41
|
Ruffini N, Klingenberg S, Schweiger S, Gerber S. Common Factors in Neurodegeneration: A Meta-Study Revealing Shared Patterns on a Multi-Omics Scale. Cells 2020; 9:E2642. [PMID: 33302607 PMCID: PMC7764447 DOI: 10.3390/cells9122642] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/24/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are heterogeneous, progressive diseases with frequently overlapping symptoms characterized by a loss of neurons. Studies have suggested relations between neurodegenerative diseases for many years (e.g., regarding the aggregation of toxic proteins or triggering endogenous cell death pathways). We gathered publicly available genomic, transcriptomic, and proteomic data from 177 studies and more than one million patients to detect shared genetic patterns between the neurodegenerative diseases on three analyzed omics-layers. The results show a remarkably high number of shared differentially expressed genes between the transcriptomic and proteomic levels for all conditions, while showing a significant relation between genomic and proteomic data between AD and PD and AD and ALS. We identified a set of 139 genes being differentially expressed in several transcriptomic experiments of all four diseases. These 139 genes showed overrepresented gene ontology (GO) Terms involved in the development of neurodegeneration, such as response to heat and hypoxia, positive regulation of cytokines and angiogenesis, and RNA catabolic process. Furthermore, the four analyzed neurodegenerative diseases (NDDs) were clustered by their mean direction of regulation throughout all transcriptomic studies for this set of 139 genes, with the closest relation regarding this common gene set seen between AD and HD. GO-Term and pathway analysis of the proteomic overlap led to biological processes (BPs), related to protein folding and humoral immune response. Taken together, we could confirm the existence of many relations between Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis on transcriptomic and proteomic levels by analyzing the pathways and GO-Terms arising in these intersections. The significance of the connection and the striking relation of the results to processes leading to neurodegeneration between the transcriptomic and proteomic data for all four analyzed neurodegenerative diseases showed that exploring many studies simultaneously, including multiple omics-layers of different neurodegenerative diseases simultaneously, holds new relevant insights that do not emerge from analyzing these data separately. Furthermore, the results shed light on processes like the humoral immune response that have previously been described only for certain diseases. Our data therefore suggest human patients with neurodegenerative diseases should be addressed as complex biological systems by integrating multiple underlying data sources.
Collapse
Affiliation(s)
- Nicolas Ruffini
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
- Leibniz Institute for Resilience Research, Leibniz Association, Wallstraße 7, 55122 Mainz, Germany
| | - Susanne Klingenberg
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susann Schweiger
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| | - Susanne Gerber
- Institute for Human Genetics, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany; (N.R.); (S.K.); (S.S.)
| |
Collapse
|
42
|
Di Giaimo R, Penna E, Pizzella A, Cirillo R, Perrone-Capano C, Crispino M. Cross Talk at the Cytoskeleton-Plasma Membrane Interface: Impact on Neuronal Morphology and Functions. Int J Mol Sci 2020; 21:ijms21239133. [PMID: 33266269 PMCID: PMC7730950 DOI: 10.3390/ijms21239133] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/18/2020] [Accepted: 11/29/2020] [Indexed: 12/13/2022] Open
Abstract
The cytoskeleton and its associated proteins present at the plasma membrane not only determine the cell shape but also modulate important aspects of cell physiology such as intracellular transport including secretory and endocytic pathways. Continuous remodeling of the cell structure and intense communication with extracellular environment heavily depend on interactions between cytoskeletal elements and plasma membrane. This review focuses on the plasma membrane-cytoskeleton interface in neurons, with a special emphasis on the axon and nerve endings. We discuss the interaction between the cytoskeleton and membrane mainly in two emerging topics of neurobiology: (i) production and release of extracellular vesicles and (ii) local synthesis of new proteins at the synapses upon signaling cues. Both of these events contribute to synaptic plasticity. Our review provides new insights into the physiological and pathological significance of the cytoskeleton-membrane interface in the nervous system.
Collapse
Affiliation(s)
- Rossella Di Giaimo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
- Correspondence: (R.D.G.); (M.C.)
| | - Eduardo Penna
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Raffaella Cirillo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
| | - Carla Perrone-Capano
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy;
- Institute of Genetics and Biophysics “Adriano Buzzati Traverso”, National Research Council (CNR), 80131 Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy; (E.P.); (A.P.); (R.C.)
- Correspondence: (R.D.G.); (M.C.)
| |
Collapse
|
43
|
Lin CH, Lin HY, Fang JM, Chen CC. A dual inhibitor targeting HMG-CoA reductase and histone deacetylase mitigates neurite degeneration in LRRK2-G2019S parkinsonism. Aging (Albany NY) 2020; 12:25581-25598. [PMID: 33231564 PMCID: PMC7803522 DOI: 10.18632/aging.104165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/27/2020] [Indexed: 06/11/2023]
Abstract
Parkinson's disease (PD) is among the most common neurodegenerative disorders, and its etiology involves both genetic and environmental factors. The leucine-rich repeat kinase (LRRK2) G2019S mutation is the most common genetic cause of familial and sporadic PD. Current treatment is limited to dopaminergic supplementation, as no disease-modifying therapy is available yet. Recent evidence reveals that HMG-CoA reductase (HMGR) inhibitors (statins) exert neuroprotection through anti-neuroinflammatory effects, and histone deacetylase (HDAC) inhibitors mitigate neurodegeneration by promoting the transcription of neuronal survival factors. We designed and synthesized a dual inhibitor, statin hydroxamate JMF3086, that simultaneously inhibits HMGR and HDAC, and examined its neuroprotective effects on LRRK2-G2019S parkinsonism. JMF3086 restored dopaminergic neuron loss in aged LRRK2-G2019S flies and rescued neurite degeneration in primary hippocampal and dopaminergic neurons isolated from transgenic LRRK2-G2019S mice. The molecular mechanisms included downregulation of ERK1/2 phosphorylation, increased anti-apoptotic Akt phosphorylation, and inhibition of GSK3β activity to maintain cytoskeletal stability in stably transfected LRRK2-G2019S SH-SY5Y human dopaminergic cells. JMF3086 also promoted a-tubulin acetylation and kinesin-1 expression, facilitating antegrade mitochondrial transport in axons. Our findings demonstrate that JMF3086 exerted beneficial effects on restoring LRRK2-G2019S neurite degeneration by maintaining microtubule stability. This dual-target compound may be a promising mechanism-based therapy for PD.
Collapse
Affiliation(s)
- Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Han-Yi Lin
- Department of Neurology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Jim-Min Fang
- Department of Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Ching-Chow Chen
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| |
Collapse
|
44
|
Liu M, Yu Q, Xiao H, Li M, Huang Y, Zhang Q, Li P. The Inhibitory Activities and Antiviral Mechanism of Medicinal Plant Ingredient Quercetin Against Grouper Iridovirus Infection. Front Microbiol 2020; 11:586331. [PMID: 33178170 PMCID: PMC7596301 DOI: 10.3389/fmicb.2020.586331] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/14/2020] [Indexed: 11/13/2022] Open
Abstract
Singapore grouper iridovirus (SGIV) causes high mortality rates in mariculture, and effective treatments against SGIV infection are urgently required. Illicium verum Hook. f. (I. verum) is a well-known medicinal plant with a variety of biological activities. The natural ingredient quercetin isolated from I. verum could effectively inhibit SGIV infection in a dose-dependent manner. The possible antiviral mechanism of quercetin was further analyzed in this study. It showed that quercetin did obvious damages to SGIV particles. Furthermore, quercetin could interfere with SGIV binding to targets on host cells (by 76.14%), disturb SGIV invading into host cells (by 56.03%), and effect SGIV replication in host cells (by 52.73%), respectively. Quercetin had the best antiviral effects during the SGIV life cycle of binding to the receptors on host cells' membranes. Overall, the results suggest that quercetin has direct and host-mediated antiviral effects against SGIV and holds great potential for developing effective drugs to control SGIV infection in aquaculture.
Collapse
Affiliation(s)
- Mingzhu Liu
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Sciences, Nanning, China
| | - Qing Yu
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Sciences, Nanning, China
| | - Hehe Xiao
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Sciences, Nanning, China
| | - Mengmeng Li
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Sciences, Nanning, China.,College of Life Science, Henan Normal University, Xinxiang, China
| | - Yaming Huang
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Colleges and Universities Key Laboratory of Utilization of Microbial and Botanical Resources, School of Marine Science and Biotechnology, Guangxi University for Nationalities, Nanning, China
| | - Qin Zhang
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Colleges and Universities Key Laboratory of Utilization of Microbial and Botanical Resources, School of Marine Science and Biotechnology, Guangxi University for Nationalities, Nanning, China
| | - Pengfei Li
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Beibu Gulf Marine Industrial Research Institute, Guangxi Academy of Sciences, Nanning, China.,Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, College of Marine Sciences, Beibu Gulf University, Qinzhou, China
| |
Collapse
|
45
|
Gerasymchuk D, Hubiernatorova A, Domanskyi A. MicroRNAs Regulating Cytoskeleton Dynamics, Endocytosis, and Cell Motility-A Link Between Neurodegeneration and Cancer? Front Neurol 2020; 11:549006. [PMID: 33240194 PMCID: PMC7680873 DOI: 10.3389/fneur.2020.549006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
The cytoskeleton is one of the most mobile and complex cell structures. It is involved in cellular transport, cell division, cell shape formation and adaptation in response to extra- and intracellular stimuli, endo- and exocytosis, migration, and invasion. These processes are crucial for normal cellular physiology and are affected in several pathological processes, including neurodegenerative diseases, and cancer. Some proteins, participating in clathrin-mediated endocytosis (CME), play an important role in actin cytoskeleton reorganization, and formation of invadopodia in cancer cells and are also deregulated in neurodegenerative disorders. However, there is still limited information about the factors contributing to the regulation of their expression. MicroRNAs are potent negative regulators of gene expression mediating crosstalk between different cellular pathways in cellular homeostasis and stress responses. These molecules regulate numerous genes involved in neuronal differentiation, plasticity, and degeneration. Growing evidence suggests the role of microRNAs in the regulation of endocytosis, cell motility, and invasiveness. By modulating the levels of such microRNAs, it may be possible to interfere with CME or other processes to normalize their function. In malignancy, the role of microRNAs is undoubtful, and therefore changing their levels can attenuate the carcinogenic process. Here we review the current advances in our understanding of microRNAs regulating actin cytoskeleton dynamics, CME and cell motility with a special focus on neurodegenerative diseases, and cancer. We investigate whether current literature provides an evidence that microRNA-mediated regulation of essential cellular processes, such as CME and cell motility, is conserved in neurons, and cancer cells. We argue that more research effort should be addressed to study the neuron-specific functions on microRNAs. Disease-associated microRNAs affecting essential cellular processes deserve special attention both from the view of fundamental science and as future neurorestorative or anti-cancer therapies.
Collapse
Affiliation(s)
- Dmytro Gerasymchuk
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Institute of Molecular Biology and Genetics, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | | | - Andrii Domanskyi
- Institute of Biotechnology, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
46
|
Pathak S, Parkar H, Tripathi S, Kale A. Ofloxacin as a Disruptor of Actin Aggresome "Hirano Bodies": A Potential Repurposed Drug for the Treatment of Neurodegenerative Diseases. Front Aging Neurosci 2020; 12:591579. [PMID: 33132905 PMCID: PMC7573105 DOI: 10.3389/fnagi.2020.591579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/09/2020] [Indexed: 01/27/2023] Open
Abstract
There is a growing number of aging populations that are more prone to the prevalence of neuropathological disorders. Two major diseases that show a late onset of the symptoms include Alzheimer’s disorder (AD) and Parkinson’s disorder (PD), which are causing an unexpected social and economic impact on the families. A large number of researches in the last decade have focused upon the role of amyloid precursor protein, Aβ-plaque, and intraneuronal neurofibrillary tangles (tau-proteins). However, there is very few understanding of actin-associated paracrystalline structures formed in the hippocampus region of the brain and are called Hirano bodies. These actin-rich inclusion bodies are known to modulate the synaptic plasticity and employ conspicuous effects on long-term potentiation and paired-pulse paradigms. Since the currently known drugs have very little effect in controlling the progression of these diseases, there is a need to develop therapeutic agents, which can have improved efficacy and bioavailability, and can transport across the blood–brain barrier. Moreover, finding novel targets involving compound screening is both laborious and is an expensive process in itself followed by equally tedious Food and Drug Administration (FDA) approval exercise. Finding alternative functions to the already existing FDA-approved molecules for reversing the progression of age-related proteinopathies is of utmost importance. In the current study, we decipher the role of a broad-spectrum general antibiotic (Ofloxacin) on actin polymerization dynamics using various biophysical techniques like right-angle light scattering, dynamic light scattering, circular dichroism spectrometry, isothermal titration calorimetry, scanning electron microscopy, etc. We have also performed in silico docking studies to deduce a plausible mechanism of the drug binding to the actin. We report that actin gets disrupted upon binding to Ofloxacin in a concentration-dependent manner. We have inferred that Ofloxacin, when attached to a drug delivery system, can act as a good candidate for the treatment of neuropathological diseases.
Collapse
Affiliation(s)
- Samridhi Pathak
- School of Chemical Sciences, University of Mumbai - Department of Atomic Energy Center for Excellence in Basic Sciences, University of Mumbai, Vidyanagari Campus, Mumbai, India
| | - Haifa Parkar
- School of Chemical Sciences, University of Mumbai - Department of Atomic Energy Center for Excellence in Basic Sciences, University of Mumbai, Vidyanagari Campus, Mumbai, India
| | - Sarita Tripathi
- School of Chemical Sciences, University of Mumbai - Department of Atomic Energy Center for Excellence in Basic Sciences, University of Mumbai, Vidyanagari Campus, Mumbai, India
| | - Avinash Kale
- School of Chemical Sciences, University of Mumbai - Department of Atomic Energy Center for Excellence in Basic Sciences, University of Mumbai, Vidyanagari Campus, Mumbai, India
| |
Collapse
|
47
|
Benn CL, Dawson LA. Clinically Precedented Protein Kinases: Rationale for Their Use in Neurodegenerative Disease. Front Aging Neurosci 2020; 12:242. [PMID: 33117143 PMCID: PMC7494159 DOI: 10.3389/fnagi.2020.00242] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Kinases are an intensively studied drug target class in current pharmacological research as evidenced by the large number of kinase inhibitors being assessed in clinical trials. Kinase-targeted therapies have potential for treatment of a broad array of indications including central nervous system (CNS) disorders. In addition to the many variables which contribute to identification of a successful therapeutic molecule, drug discovery for CNS-related disorders also requires significant consideration of access to the target organ and specifically crossing the blood-brain barrier (BBB). To date, only a small number of kinase inhibitors have been reported that are specifically designed to be BBB permeable, which nonetheless demonstrates the potential for success. This review considers the potential for kinase inhibitors in the context of unmet medical need for neurodegenerative disease. A subset of kinases that have been the focus of clinical investigations over a 10-year period have been identified and discussed individually. For each kinase target, the data underpinning the validity of each in the context of neurodegenerative disease is critically evaluated. Selected molecules for each kinase are identified with information on modality, binding site and CNS penetrance, if known. Current clinical development in neurodegenerative disease are summarized. Collectively, the review indicates that kinase targets with sufficient rationale warrant careful design approaches with an emphasis on improving brain penetrance and selectivity.
Collapse
|
48
|
Oliveira da Silva MI, Liz MA. Linking Alpha-Synuclein to the Actin Cytoskeleton: Consequences to Neuronal Function. Front Cell Dev Biol 2020; 8:787. [PMID: 32903460 PMCID: PMC7434970 DOI: 10.3389/fcell.2020.00787] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/27/2020] [Indexed: 12/20/2022] Open
Abstract
Alpha-Synuclein (αSyn), a protein highly enriched in neurons where it preferentially localizes at the pre-synapse, has been in the spotlight because its intraneuronal aggregation is a central phenomenon in Parkinson's disease. However, the consequences of αSyn accumulation to neuronal function are not fully understood. Considering the crucial role of actin on synaptic function and the fact that dysregulation of this cytoskeleton component is emerging in neurodegenerative disorders, the impact of αSyn on actin is a critical point to be addressed. In this review we explore the link between αSyn and actin and its significance for physiology and pathology. We discuss the relevance of αSyn-actin interaction for synaptic function and highlight the actin-depolymerizing protein cofilin-1 as a key player on αSyn-induced actin dysfunction in Parkinson's disease.
Collapse
Affiliation(s)
- Marina I Oliveira da Silva
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Neurodegeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Nerve Regeneration Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Márcia A Liz
- Neurodegeneration Group, Instituto de Biologia Molecular e Celular (IBMC) and Nerve Regeneration Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| |
Collapse
|
49
|
Transthyretin interacts with actin regulators in a Drosophila model of familial amyloid polyneuropathy. Sci Rep 2020; 10:13596. [PMID: 32788615 PMCID: PMC7423984 DOI: 10.1038/s41598-020-70377-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 07/23/2020] [Indexed: 11/23/2022] Open
Abstract
Familial amyloid polyneuropathy (FAP) is a neurodegenerative disorder whose major hallmark is the deposition of mutated transthyretin (TTR) in the form of amyloid fibrils in the peripheral nervous system (PNS). The exposure of PNS axons to extracellular TTR deposits leads to an axonopathy that culminates in neuronal death. However, the molecular mechanisms underlying TTR-induced neurodegeneration are still unclear, despite the extensive studies in vertebrate models. In this work we used a Drosophila FAP model, based on the expression of the amyloidogenic TTR (V30M) in the fly retina, to uncover genetic interactions with cytoskeleton regulators. We show that TTR interacts with actin regulators and induces cytoskeleton alterations, leading to axonal defects. Moreover, our study pinpoints an interaction between TTRV30M and members of Rho GTPase signaling pathways, the major actin regulators. Based on these findings we propose that actin cytoskeleton alterations may mediate the axonopathy observed in FAP patients, and highlight a molecular pathway, mediated by Rho GTPases, underlying TTR-induced neurodegeneration. We expect this work to prompt novel studies and approaches towards FAP therapy.
Collapse
|
50
|
Dubey T, Gorantla NV, Chandrashekara KT, Chinnathambi S. Photodynamic exposure of Rose-Bengal inhibits Tau aggregation and modulates cytoskeletal network in neuronal cells. Sci Rep 2020; 10:12380. [PMID: 32704015 PMCID: PMC7378248 DOI: 10.1038/s41598-020-69403-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023] Open
Abstract
The intracellular Tau aggregates are known to be associated with Alzheimer’s disease. The inhibition of Tau aggregation is an important strategy for screening of therapeutic molecules in Alzheimer's disease. Several classes of dyes possess a unique property of photo-excitation, which is applied as a therapeutic measure against numerous neurological dysfunctions. Rose Bengal is a Xanthene dye, which has been widely used as a photosensitizer in photodynamic therapy. The aim of this work was to study the protective role of Rose Bengal against Tau aggregation and cytoskeleton modulations. The aggregation inhibition and disaggregation potency of Rose Bengal and photo-excited Rose Bengal were observed by in-vitro fluorescence, circular dichroism, and electron microscopy. Rose Bengal and photo-excited Rose Bengal induce minimal cytotoxicity in neuronal cells. In our studies, we observed that Rose Bengal and photo-excited Rose Bengal modulate the cytoskeleton network of actin and tubulin. The immunofluorescence studies showed the increased filopodia structures after photo-excited Rose Bengal treatment. Furthermore, Rose Bengal treatment increases the connections between the cells. Rose Bengal and photo-excited Rose Bengal treatment-induced actin-rich podosome-like structures associated with cell membranes. The in-vivo studies on UAS E-14 Tau mutant Drosophila suggested that exposure to Rose Bengal and photo-excited Rose Bengal efficiency rescues the behavioural and memory deficit in flies. Thus, the overall results suggest that Rose Bengal could have a therapeutic potency against Tau aggregation.
Collapse
Affiliation(s)
- Tushar Dubey
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India
| | - Nalini Vijay Gorantla
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India
| | | | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India. .,Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India.
| |
Collapse
|