51
|
Slater PG, Domínguez-Romero ME, Villarreal M, Eisner V, Larraín J. Mitochondrial function in spinal cord injury and regeneration. Cell Mol Life Sci 2022; 79:239. [PMID: 35416520 PMCID: PMC11072423 DOI: 10.1007/s00018-022-04261-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 12/21/2022]
Abstract
Many people around the world suffer from some form of paralysis caused by spinal cord injury (SCI), which has an impact on quality and life expectancy. The spinal cord is part of the central nervous system (CNS), which in mammals is unable to regenerate, and to date, there is a lack of full functional recovery therapies for SCI. These injuries start with a rapid and mechanical insult, followed by a secondary phase leading progressively to greater damage. This secondary phase can be potentially modifiable through targeted therapies. The growing literature, derived from mammalian and regenerative model studies, supports a leading role for mitochondria in every cellular response after SCI: mitochondrial dysfunction is the common event of different triggers leading to cell death, cellular metabolism regulates the immune response, mitochondrial number and localization correlate with axon regenerative capacity, while mitochondrial abundance and substrate utilization regulate neural stem progenitor cells self-renewal and differentiation. Herein, we present a comprehensive review of the cellular responses during the secondary phase of SCI, the mitochondrial contribution to each of them, as well as evidence of mitochondrial involvement in spinal cord regeneration, suggesting that a more in-depth study of mitochondrial function and regulation is needed to identify potential targets for SCI therapeutic intervention.
Collapse
Affiliation(s)
- Paula G Slater
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile.
| | - Miguel E Domínguez-Romero
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Maximiliano Villarreal
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Verónica Eisner
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Juan Larraín
- Center for Aging and Regeneration, Departamento de Biología Celular Y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| |
Collapse
|
52
|
Xie L, Cen LP, Li Y, Gilbert HY, Strelko O, Berlinicke C, Stavarache MA, Ma M, Wang Y, Cui Q, Kaplitt MG, Zack DJ, Benowitz LI, Yin Y. Monocyte-derived SDF1 supports optic nerve regeneration and alters retinal ganglion cells' response to Pten deletion. Proc Natl Acad Sci U S A 2022; 119:e2113751119. [PMID: 35394873 PMCID: PMC9169637 DOI: 10.1073/pnas.2113751119] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 02/23/2022] [Indexed: 12/22/2022] Open
Abstract
Although mammalian retinal ganglion cells (RGCs) normally cannot regenerate axons nor survive after optic nerve injury, this failure is partially reversed by inducing sterile inflammation in the eye. Infiltrative myeloid cells express the axogenic protein oncomodulin (Ocm) but additional, as-yet-unidentified, factors are also required. We show here that infiltrative macrophages express stromal cell–derived factor 1 (SDF1, CXCL12), which plays a central role in this regard. Among many growth factors tested in culture, only SDF1 enhances Ocm activity, an effect mediated through intracellular cyclic AMP (cAMP) elevation and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) activation. SDF1 deficiency in myeloid cells (CXCL12flx/flxLysM-Cre−/+ mice) or deletion of the SDF1 receptor CXCR4 in RGCs (intraocular AAV2-Cre in CXCR4flx/flx mice) or SDF1 antagonist AMD3100 greatly suppresses inflammation-induced regeneration and decreases RGC survival to baseline levels. Conversely, SDF1 induces optic nerve regeneration and RGC survival, and, when combined with Ocm/cAMP, SDF1 increases axon regeneration to levels similar to those induced by intraocular inflammation. In contrast to deletion of phosphatase and tensin homolog (Pten), which promotes regeneration selectively from αRGCs, SDF1 promotes regeneration from non-αRGCs and enables the latter cells to respond robustly to Pten deletion; however, SDF1 surprisingly diminishes the response of αRGCs to Pten deletion. When combined with inflammation and Pten deletion, SDF1 enables many RGCs to regenerate axons the entire length of the optic nerve. Thus, SDF1 complements the effects of Ocm in mediating inflammation-induced regeneration and enables different RGC subtypes to respond to Pten deletion.
Collapse
Affiliation(s)
- Lili Xie
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Ling-Ping Cen
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Joint Shantou International Eye Center, Shantou University and The Chinese University of Hong Kong, Shantou 515000, China
| | - Yiqing Li
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510085, China
| | - Hui-Ya Gilbert
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Oleksandr Strelko
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Cynthia Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Mihaela A. Stavarache
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065
| | - Madeline Ma
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| | - Yongting Wang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qi Cui
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Joint Shantou International Eye Center, Shantou University and The Chinese University of Hong Kong, Shantou 515000, China
| | - Michael G. Kaplitt
- Laboratory of Molecular Neurosurgery, Department of Neurological Surgery, Weill Cornell Medical College, New York, NY 10065
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Larry I. Benowitz
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
- Department of Ophthalmology, Harvard Medical School, Boston, MA 02115
- Program in Neuroscience, Harvard Medical School, Boston, MA 02115
| | - Yuqin Yin
- Department of Neurosurgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115
- F. M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA 02115
| |
Collapse
|
53
|
Shah SH, Schiapparelli LM, Ma Y, Yokota S, Atkins M, Xia X, Cameron EG, Huang T, Saturday S, Sun CB, Knasel C, Blackshaw S, Yates Iii JR, Cline HT, Goldberg JL. Quantitative transportomics identifies Kif5a as a major regulator of neurodegeneration. eLife 2022; 11:68148. [PMID: 35259089 PMCID: PMC8947766 DOI: 10.7554/elife.68148] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 03/07/2022] [Indexed: 11/29/2022] Open
Abstract
Many neurons in the adult central nervous system, including retinal ganglion cells (RGCs), degenerate and die after injury. Early axon protein and organelle trafficking failure is a key component in many neurodegenerative disorders yet changes to axoplasmic transport in disease models have not been quantified. We analyzed early changes in the protein ‘transportome’ from RGC somas to their axons after optic nerve injury and identified transport failure of an anterograde motor protein Kif5a early in RGC degeneration. We demonstrated that manipulating Kif5a expression affects anterograde mitochondrial trafficking in RGCs and characterized axon transport in Kif5a knockout mice to identify proteins whose axon localization was Kif5a-dependent. Finally, we found that knockout of Kif5a in RGCs resulted in progressive RGC degeneration in the absence of injury. Together with expression data localizing Kif5a to human RGCs, these data identify Kif5a transport failure as a cause of RGC neurodegeneration and point to a mechanism for future therapeutics.
Collapse
Affiliation(s)
- Sahil H Shah
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | | | - Yuanhui Ma
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| | - Satoshi Yokota
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Melissa Atkins
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Xin Xia
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Evan G Cameron
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Thanh Huang
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Sarah Saturday
- Neuroscience Department, The Scripps Research Institute, La Jolla, United States
| | - Catalin B Sun
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Cara Knasel
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, United States
| | - John R Yates Iii
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| | - Hollis T Cline
- Neuroscience Department, The Scripps Research Institute, La Jolla, United States
| | - Jeffrey L Goldberg
- Byers Eye Institute and Spencer Center for Vision Research, Stanford University, Palo Alto, United States
| |
Collapse
|
54
|
Winter CC, He Z, Jacobi A. Axon Regeneration: A Subcellular Extension in Multiple Dimensions. Cold Spring Harb Perspect Biol 2022; 14:a040923. [PMID: 34518340 PMCID: PMC8886981 DOI: 10.1101/cshperspect.a040923] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Axons are a unique cellular structure that allows for the communication between neurons. Axon damage compromises neuronal communications and often leads to functional deficits. Thus, developing strategies that promote effective axon regeneration for functional restoration is highly desirable. One fruitful approach is to dissect the regenerative mechanisms used by some types of neurons in both mammalian and nonmammalian systems that exhibit spontaneous regenerative capacity. Additionally, numerous efforts have been devoted to deciphering the barriers that prevent successful axon regeneration in the most regeneration-refractory system-the adult mammalian central nervous system. As a result, several regeneration-promoting strategies have been developed, but significant limitations remain. This review is aimed to summarize historic progression and current understanding of this exciting yet incomplete endeavor.
Collapse
Affiliation(s)
- Carla C Winter
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
- PhD Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Anne Jacobi
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology and Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
55
|
Widodo N, Puspitarini S, Widyananda MH, Alamsyah A, Wicaksono ST, Masruri M, Jatmiko YD. Anticancer activity of Caesalpinia sappan by downregulating mitochondrial genes in A549 lung cancer cell line. F1000Res 2022; 11:169. [PMID: 36128561 PMCID: PMC9468624 DOI: 10.12688/f1000research.76187.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 09/05/2024] Open
Abstract
Background: The standardization and mechanism of action of Caesalpinia sappan as an anticancer agent are still lacking. This study aimed to understand the mechanism of action of C,sappan extract as an anticancer agent. Methods: This study was conducted using the A549 lung cancer cell line to understand the mechanism of action of C. sappan extract as an anticancer agent. The cytotoxicity activity, cell cycle progression, apoptosis, protein-related apoptosis (i.e., BCL-2and BAX protein) assays, and RNA sequencing were performed level were measured. Moreover, the antioxidant activity, total flavonoids, and phenolics of C.sappan were also assessed. Results: C.sappan has strong antioxidant activity (22.14 ± 0.93 ppm) total flavonoid content of (529.3 ± 4.56 mgQE/g), and phenolics content of (923.37 ± 5 mgGAE/g). The C.sappan ethanol extract inhibited cancer cell growth and arrested at G0/G1 phase of cell cycle, inducing apoptosis by increasing BAX/BCL-2 protein ratio in A549 lung cancer cell line. Furthermore, results from RNA sequencing analysis showed that C.sappan ethanol extract caused downregulation of genes acting on mitochondrial function including adenosine triphosphate (ATP) production and respiration. Conclusions: This study demonstrated that C.sappan has the ability to inhibit cancer cell growth by inducing apoptosis and mitochondrial dysfunction in A549 cells.
Collapse
Affiliation(s)
- Nashi Widodo
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Sapti Puspitarini
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | | | - Adzral Alamsyah
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Septian Tri Wicaksono
- Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Masruri Masruri
- Chemistry Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Yoga Dwi Jatmiko
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| |
Collapse
|
56
|
Widodo N, Puspitarini S, Widyananda MH, Alamsyah A, Wicaksono ST, Masruri M, Jatmiko YD. Anticancer activity of Caesalpinia sappan by downregulating mitochondrial genes in A549 lung cancer cell line. F1000Res 2022; 11:169. [PMID: 36128561 PMCID: PMC9468624 DOI: 10.12688/f1000research.76187.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 11/20/2022] Open
Abstract
Background: The standardization and mechanism of action of Caesalpinia sappan as an anticancer agent are still lacking. This study aimed to understand the mechanism of action of C,sappan extract as an anticancer agent. Methods: This study was conducted using the A549 lung cancer cell line to understand the mechanism of action of C. sappan extract as an anticancer agent. The cytotoxicity activity, cell cycle progression, apoptosis, protein-related apoptosis (i.e., BCL-2and BAX protein) assays, and RNA sequencing were performed level were measured. Moreover, the antioxidant activity, total flavonoids, and phenolics of C.sappan were also assessed. Results: C.sappan has strong antioxidant activity (22.14 ± 0.93 ppm) total flavonoid content of (529.3 ± 4.56 mgQE/g), and phenolics content of (923.37 ± 5 mgGAE/g). The C.sappan ethanol extract inhibited cancer cell growth and arrested at G0/G1 phase of cell cycle, inducing apoptosis by increasing BAX/BCL-2 protein ratio in A549 lung cancer cell line. Furthermore, results from RNA sequencing analysis showed that C.sappan ethanol extract caused downregulation of genes acting on mitochondrial function including adenosine triphosphate (ATP) production and respiration. Conclusions: This study demonstrated that C.sappan has the ability to inhibit cancer cell growth by inducing apoptosis and mitochondrial dysfunction in A549 cells.
Collapse
Affiliation(s)
- Nashi Widodo
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Sapti Puspitarini
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | | | - Adzral Alamsyah
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Septian Tri Wicaksono
- Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Masruri Masruri
- Chemistry Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Yoga Dwi Jatmiko
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| |
Collapse
|
57
|
Babetto E, Beirowski B. Stressed axons craving for glial sugar: links to regeneration? Neural Regen Res 2022; 17:304-306. [PMID: 34269193 PMCID: PMC8463966 DOI: 10.4103/1673-5374.317965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Elisabetta Babetto
- Hunter James Kelly Research Institute; Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Bogdan Beirowski
- Hunter James Kelly Research Institute; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
58
|
Lin L, He Z, Zhang T, Zuo Y, Chen X, Abdelrahman Z, Chen F, Wei Z, Si K, Gong W, Wang X, He S, Chen Z. A biocompatible two-photon absorbing fluorescent mitochondrial probe for deep in vivo bioimaging. J Mater Chem B 2022; 10:887-898. [DOI: 10.1039/d1tb02040d] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We reported a mitochondria-targeted two-photon fluorescent dye with an excellent two-photon absorption cross-section. With this dye, we reached an imaging depth of ca. 640 μm during mitochondrial imaging of cortical cells in live animals.
Collapse
Affiliation(s)
- Lingmin Lin
- Department of Neurobiology and Department of Orthopedics, Zhejiang University School of Medicine, 2nd Affiliated Hospital, Hangzhou, Zhejiang Province 310009, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
| | - Zewei He
- State Key Laboratory for Modern Optical Instrumentation, Centre for Optical and Electromagnetic Research, East Building No. 5, Zijingang Campus and Zhejiang University, Hangzhou 310058, China
| | - Tianfang Zhang
- Department of Rehabilitation Medicine, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
| | - Yanming Zuo
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
| | - Xiangfeng Chen
- Department of Neurobiology and Department of Orthopedics, Zhejiang University School of Medicine, 2nd Affiliated Hospital, Hangzhou, Zhejiang Province 310009, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
| | - Zeinab Abdelrahman
- Department of Neurobiology and Department of Orthopedics, Zhejiang University School of Medicine, 2nd Affiliated Hospital, Hangzhou, Zhejiang Province 310009, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
| | - Feihong Chen
- State Key Laboratory for Modern Optical Instrumentation, Centre for Optical and Electromagnetic Research, East Building No. 5, Zijingang Campus and Zhejiang University, Hangzhou 310058, China
| | - Zhongcao Wei
- Guangdong Provincial Key Laboratory of Nanophotonic Functional Materials and Devices, School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510006, China
| | - Ke Si
- State Key Laboratory for Modern Optical Instrumentation, Centre for Optical and Electromagnetic Research, East Building No. 5, Zijingang Campus and Zhejiang University, Hangzhou 310058, China
| | - Wei Gong
- Center for Neuroscience and Department of Neurobiology of the Second Affiliated Hospital, State Key Laboratory of Modern Optical Instrumentation, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xuhua Wang
- Department of Neurobiology and Department of Orthopedics, Zhejiang University School of Medicine, 2nd Affiliated Hospital, Hangzhou, Zhejiang Province 310009, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001 Jiangsu, P. R. China
| | - Sailing He
- State Key Laboratory for Modern Optical Instrumentation, Centre for Optical and Electromagnetic Research, East Building No. 5, Zijingang Campus and Zhejiang University, Hangzhou 310058, China
| | - Zuobing Chen
- Department of Rehabilitation Medicine, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, 310003, China
| |
Collapse
|
59
|
König T, Nolte H, Aaltonen MJ, Tatsuta T, Krols M, Stroh T, Langer T, McBride HM. MIROs and DRP1 drive mitochondrial-derived vesicle biogenesis and promote quality control. Nat Cell Biol 2021; 23:1271-1286. [PMID: 34873283 DOI: 10.1038/s41556-021-00798-4] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 10/19/2021] [Indexed: 12/11/2022]
Abstract
Mitochondrial-derived vesicles (MDVs) are implicated in diverse physiological processes-for example, mitochondrial quality control-and are linked to various neurodegenerative diseases. However, their specific cargo composition and complex molecular biogenesis are still unknown. Here we report the proteome and lipidome of steady-state TOMM20+ MDVs. We identified 107 high-confidence MDV cargoes, which include all β-barrel proteins and the TOM import complex. MDV cargoes are delivered as fully assembled complexes to lysosomes, thus representing a selective mitochondrial quality control mechanism for multi-subunit complexes, including the TOM machinery. Moreover, we define key biogenesis steps of phosphatidic acid-enriched MDVs starting with the MIRO1/2-dependent formation of thin membrane protrusions pulled along microtubule filaments, followed by MID49/MID51/MFF-dependent recruitment of the dynamin family GTPase DRP1 and finally DRP1-dependent scission. In summary, we define the function of MDVs in mitochondrial quality control and present a mechanistic model for global GTPase-driven MDV biogenesis.
Collapse
Affiliation(s)
- Tim König
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Hendrik Nolte
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Mari J Aaltonen
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Takashi Tatsuta
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Michiel Krols
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Thomas Stroh
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Heidi M McBride
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
60
|
Napoli E, Panoutsopoulos AA, Kysar P, Satriya N, Sterling K, Shibata B, Imai D, Ruskin DN, Zarbalis KS, Giulivi C. Wdfy3 regulates glycophagy, mitophagy, and synaptic plasticity. J Cereb Blood Flow Metab 2021; 41:3213-3231. [PMID: 34187232 PMCID: PMC8669292 DOI: 10.1177/0271678x211027384] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Autophagy is essential to cell function, as it enables the recycling of intracellular constituents during starvation and in addition functions as a quality control mechanism by eliminating spent organelles and proteins that could cause cellular damage if not properly removed. Recently, we reported on Wdfy3's role in mitophagy, a clinically relevant macroautophagic scaffold protein that is linked to intellectual disability, neurodevelopmental delay, and autism spectrum disorder. In this study, we confirm our previous report that Wdfy3 haploinsufficiency in mice results in decreased mitophagy with accumulation of mitochondria with altered morphology, but expanding on that observation, we also note decreased mitochondrial localization at synaptic terminals and decreased synaptic density, which may contribute to altered synaptic plasticity. These changes are accompanied by defective elimination of glycogen particles and a shift to increased glycogen synthesis over glycogenolysis and glycophagy. This imbalance leads to an age-dependent higher incidence of brain glycogen deposits with cerebellar hypoplasia. Our results support and further extend Wdfy3's role in modulating both brain bioenergetics and synaptic plasticity by including glycogen as a target of macroautophagic degradation.
Collapse
Affiliation(s)
- Eleonora Napoli
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Alexios A Panoutsopoulos
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA
| | - Patricia Kysar
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | - Nathaniel Satriya
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Kira Sterling
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Bradley Shibata
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, CA, USA
| | - Denise Imai
- Anatomic Pathology Service, Veterinary Medical Teaching Hospital, University of California, Davis, CA, USA
| | - David N Ruskin
- Department of Psychology and Neuroscience Program, Trinity College, Hartford, CT, USA
| | - Konstantinos S Zarbalis
- Department of Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA.,Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA, USA.,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, CA, USA
| | - Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA.,Medical Investigations of Neurodevelopmental Disorders (MIND) Institute, University of California Davis, CA, USA
| |
Collapse
|
61
|
Zinsmaier KE. Mitochondrial Miro GTPases coordinate mitochondrial and peroxisomal dynamics. Small GTPases 2021; 12:372-398. [PMID: 33183150 PMCID: PMC8583064 DOI: 10.1080/21541248.2020.1843957] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Mitochondria and peroxisomes are highly dynamic, multifunctional organelles. Both perform key roles for cellular physiology and homoeostasis by mediating bioenergetics, biosynthesis, and/or signalling. To support cellular function, they must be properly distributed, of proper size, and be able to interact with other organelles. Accumulating evidence suggests that the small atypical GTPase Miro provides a central signalling node to coordinate mitochondrial as well as peroxisomal dynamics. In this review, I summarize our current understanding of Miro-dependent functions and molecular mechanisms underlying the proper distribution, size and function of mitochondria and peroxisomes.
Collapse
Affiliation(s)
- Konrad E. Zinsmaier
- Departments of Neuroscience and Molecular and Cellular Biology, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
62
|
Cade BE, Lee J, Sofer T, Wang H, Zhang M, Chen H, Gharib SA, Gottlieb DJ, Guo X, Lane JM, Liang J, Lin X, Mei H, Patel SR, Purcell SM, Saxena R, Shah NA, Evans DS, Hanis CL, Hillman DR, Mukherjee S, Palmer LJ, Stone KL, Tranah GJ, Abecasis GR, Boerwinkle EA, Correa A, Cupples LA, Kaplan RC, Nickerson DA, North KE, Psaty BM, Rotter JI, Rich SS, Tracy RP, Vasan RS, Wilson JG, Zhu X, Redline S, TOPMed Sleep Working Group CadeBrianChenHanGharibSinaGoodmanMatthewGottliebDanielHaleLaurenKnutsonKristenLauderdaleDianeLaneJacquelineLeeJiwonLiangJingjingLinXihongLiuYaowuMeiHaoMitchellBraxtonNgoDebbyO’ConnellJeffOchs-BalcomHeatherPatelSanjayPurcellShaunRedlineSusanRhodesJessicaSaxenaRichaShahNeomiSoferTamarSulJae HoonSunyaevShamilWangHemingWilsonJamesZhangManZhouHufengZhuXiaofeng. Whole-genome association analyses of sleep-disordered breathing phenotypes in the NHLBI TOPMed program. Genome Med 2021; 13:136. [PMID: 34446064 PMCID: PMC8394596 DOI: 10.1186/s13073-021-00917-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/28/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Sleep-disordered breathing is a common disorder associated with significant morbidity. The genetic architecture of sleep-disordered breathing remains poorly understood. Through the NHLBI Trans-Omics for Precision Medicine (TOPMed) program, we performed the first whole-genome sequence analysis of sleep-disordered breathing. METHODS The study sample was comprised of 7988 individuals of diverse ancestry. Common-variant and pathway analyses included an additional 13,257 individuals. We examined five complementary traits describing different aspects of sleep-disordered breathing: the apnea-hypopnea index, average oxyhemoglobin desaturation per event, average and minimum oxyhemoglobin saturation across the sleep episode, and the percentage of sleep with oxyhemoglobin saturation < 90%. We adjusted for age, sex, BMI, study, and family structure using MMSKAT and EMMAX mixed linear model approaches. Additional bioinformatics analyses were performed with MetaXcan, GIGSEA, and ReMap. RESULTS We identified a multi-ethnic set-based rare-variant association (p = 3.48 × 10-8) on chromosome X with ARMCX3. Additional rare-variant associations include ARMCX3-AS1, MRPS33, and C16orf90. Novel common-variant loci were identified in the NRG1 and SLC45A2 regions, and previously associated loci in the IL18RAP and ATP2B4 regions were associated with novel phenotypes. Transcription factor binding site enrichment identified associations with genes implicated with respiratory and craniofacial traits. Additional analyses identified significantly associated pathways. CONCLUSIONS We have identified the first gene-based rare-variant associations with objectively measured sleep-disordered breathing traits. Our results increase the understanding of the genetic architecture of sleep-disordered breathing and highlight associations in genes that modulate lung development, inflammation, respiratory rhythmogenesis, and HIF1A-mediated hypoxic response.
Collapse
Affiliation(s)
- Brian E. Cade
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Sleep Medicine, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Jiwon Lee
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA
| | - Tamar Sofer
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Sleep Medicine, Harvard Medical School, Boston, MA 02115 USA
| | - Heming Wang
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Sleep Medicine, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Man Zhang
- grid.411024.20000 0001 2175 4264Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Han Chen
- grid.267308.80000 0000 9206 2401Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA ,grid.267308.80000 0000 9206 2401Center for Precision Health, School of Public Health and School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA
| | - Sina A. Gharib
- grid.34477.330000000122986657Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA 98195 USA
| | - Daniel J. Gottlieb
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Sleep Medicine, Harvard Medical School, Boston, MA 02115 USA ,grid.410370.10000 0004 4657 1992VA Boston Healthcare System, Boston, MA 02132 USA
| | - Xiuqing Guo
- grid.239844.00000 0001 0157 6501The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Jacqueline M. Lane
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Sleep Medicine, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA ,grid.32224.350000 0004 0386 9924Center for Genomic Medicine and Department of Anesthesia, Pain, and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Jingjing Liang
- grid.67105.350000 0001 2164 3847Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106 USA
| | - Xihong Lin
- grid.38142.3c000000041936754XDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA
| | - Hao Mei
- grid.410721.10000 0004 1937 0407Department of Data Science, University of Mississippi Medical Center, Jackson, MS 29216 USA
| | - Sanjay R. Patel
- grid.21925.3d0000 0004 1936 9000Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Shaun M. Purcell
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Sleep Medicine, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA
| | - Richa Saxena
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Sleep Medicine, Harvard Medical School, Boston, MA 02115 USA ,grid.66859.34Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142 USA ,grid.32224.350000 0004 0386 9924Center for Genomic Medicine and Department of Anesthesia, Pain, and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Neomi A. Shah
- grid.59734.3c0000 0001 0670 2351Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Daniel S. Evans
- grid.17866.3e0000000098234542California Pacific Medical Center Research Institute, San Francisco, CA 94107 USA
| | - Craig L. Hanis
- grid.267308.80000 0000 9206 2401Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA
| | - David R. Hillman
- grid.3521.50000 0004 0437 5942Department of Pulmonary Physiology and Sleep Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia 6009 Australia
| | - Sutapa Mukherjee
- Sleep Health Service, Respiratory and Sleep Services, Southern Adelaide Local Health Network, Adelaide, South Australia Australia ,grid.1014.40000 0004 0367 2697Adelaide Institute for Sleep Health, Flinders University, Adelaide, South Australia Australia
| | - Lyle J. Palmer
- grid.1010.00000 0004 1936 7304School of Public Health, University of Adelaide, Adelaide, South Australia 5000 Australia
| | - Katie L. Stone
- grid.17866.3e0000000098234542California Pacific Medical Center Research Institute, San Francisco, CA 94107 USA
| | - Gregory J. Tranah
- grid.17866.3e0000000098234542California Pacific Medical Center Research Institute, San Francisco, CA 94107 USA
| | | | - Gonçalo R. Abecasis
- grid.214458.e0000000086837370Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109 USA
| | - Eric A. Boerwinkle
- grid.267308.80000 0000 9206 2401Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030 USA ,grid.39382.330000 0001 2160 926XHuman Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030 USA
| | - Adolfo Correa
- grid.410721.10000 0004 1937 0407Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216 USA ,Jackson Heart Study, Jackson, MS 39216 USA
| | - L. Adrienne Cupples
- grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118 USA ,grid.510954.c0000 0004 0444 3861Framingham Heart Study, Framingham, MA 01702 USA
| | - Robert C. Kaplan
- grid.251993.50000000121791997Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, 10461 USA
| | - Deborah A. Nickerson
- grid.34477.330000000122986657Department of Genome Sciences, University of Washington, Seattle, WA 98195 USA ,grid.34477.330000000122986657Northwest Genomics Center, Seattle, WA 98105 USA
| | - Kari E. North
- grid.410711.20000 0001 1034 1720Department of Epidemiology and Carolina Center of Genome Sciences, University of North Carolina, Chapel Hill, NC 27514 USA
| | - Bruce M. Psaty
- grid.34477.330000000122986657Cardiovascular Health Study, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA 98101 USA ,grid.488833.c0000 0004 0615 7519Kaiser Permanente Washington Health Research Institute, Seattle, WA 98101 USA
| | - Jerome I. Rotter
- grid.239844.00000 0001 0157 6501The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Stephen S. Rich
- grid.27755.320000 0000 9136 933XCenter for Public Health Genomics, University of Virginia, Charlottesville, VA 22908 USA
| | - Russell P. Tracy
- grid.59062.380000 0004 1936 7689Department of Pathology, University of Vermont, Colchester, VT 05405 USA
| | - Ramachandran S. Vasan
- grid.510954.c0000 0004 0444 3861Framingham Heart Study, Framingham, MA 01702 USA ,grid.189504.10000 0004 1936 7558Sections of Preventive Medicine and Epidemiology and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA 02118 USA ,grid.189504.10000 0004 1936 7558Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118 USA
| | - James G. Wilson
- grid.410721.10000 0004 1937 0407Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216 USA
| | - Xiaofeng Zhu
- grid.67105.350000 0001 2164 3847Department of Population and Quantitative Health Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106 USA
| | - Susan Redline
- grid.38142.3c000000041936754XDivision of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDivision of Sleep Medicine, Harvard Medical School, Boston, MA 02115 USA ,grid.239395.70000 0000 9011 8547Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215 USA
| | | |
Collapse
|
63
|
Chemokine CCL5 promotes robust optic nerve regeneration and mediates many of the effects of CNTF gene therapy. Proc Natl Acad Sci U S A 2021; 118:2017282118. [PMID: 33627402 DOI: 10.1073/pnas.2017282118] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ciliary neurotrophic factor (CNTF) is a leading therapeutic candidate for several ocular diseases and induces optic nerve regeneration in animal models. Paradoxically, however, although CNTF gene therapy promotes extensive regeneration, recombinant CNTF (rCNTF) has little effect. Because intraocular viral vectors induce inflammation, and because CNTF is an immune modulator, we investigated whether CNTF gene therapy acts indirectly through other immune mediators. The beneficial effects of CNTF gene therapy remained unchanged after deleting CNTF receptor alpha (CNTFRα) in retinal ganglion cells (RGCs), the projection neurons of the retina, but were diminished by depleting neutrophils or by genetically suppressing monocyte infiltration. CNTF gene therapy increased expression of C-C motif chemokine ligand 5 (CCL5) in immune cells and retinal glia, and recombinant CCL5 induced extensive axon regeneration. Conversely, CRISPR-mediated knockdown of the cognate receptor (CCR5) in RGCs or treating wild-type mice with a CCR5 antagonist repressed the effects of CNTF gene therapy. Thus, CCL5 is a previously unrecognized, potent activator of optic nerve regeneration and mediates many of the effects of CNTF gene therapy.
Collapse
|
64
|
Abstract
The damage or loss of retinal ganglion cells (RGCs) and their axons accounts for the visual functional defects observed after traumatic injury, in degenerative diseases such as glaucoma, or in compressive optic neuropathies such as from optic glioma. By using optic nerve crush injury models, recent studies have revealed the cellular and molecular logic behind the regenerative failure of injured RGC axons in adult mammals and suggested several strategies with translational potential. This review summarizes these findings and discusses challenges for developing clinically applicable neural repair strategies.
Collapse
Affiliation(s)
- Philip R Williams
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Larry I Benowitz
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California 94303, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
65
|
Huang N, Li S, Xie Y, Han Q, Xu XM, Sheng ZH. Reprogramming an energetic AKT-PAK5 axis boosts axon energy supply and facilitates neuron survival and regeneration after injury and ischemia. Curr Biol 2021; 31:3098-3114.e7. [PMID: 34087103 PMCID: PMC8319057 DOI: 10.1016/j.cub.2021.04.079] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/29/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022]
Abstract
Mitochondria supply adenosine triphosphate (ATP) essential for neuronal survival and regeneration. Brain injury and ischemia trigger acute mitochondrial damage and a local energy crisis, leading to degeneration. Boosting local ATP supply in injured axons is thus critical to meet increased energy demand during nerve repair and regeneration in adult brains, where mitochondria remain largely stationary. Here, we elucidate an intrinsic energetic repair signaling axis that boosts axonal energy supply by reprogramming mitochondrial trafficking and anchoring in response to acute injury-ischemic stress in mature neurons and adult brains. P21-activated kinase 5 (PAK5) is a brain mitochondrial kinase with declined expression in mature neurons. PAK5 synthesis and signaling is spatiotemporally activated within axons in response to ischemic stress and axonal injury. PAK5 signaling remobilizes and replaces damaged mitochondria via the phosphorylation switch that turns off the axonal mitochondrial anchor syntaphilin. Injury-ischemic insults trigger AKT growth signaling that activates PAK5 and boosts local energy supply, thus protecting axon survival and facilitating regeneration in in vitro and in vivo models. Our study reveals an axonal mitochondrial signaling axis that responds to injury and ischemia by remobilizing damaged mitochondria for replacement, thereby maintaining local energy supply to support central nervous system (CNS) survival and regeneration.
Collapse
Affiliation(s)
- Ning Huang
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Sunan Li
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Yuxiang Xie
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN 46202, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, 320 W. 15th Street, Indianapolis, IN 46202, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA.
| |
Collapse
|
66
|
Muench NA, Patel S, Maes ME, Donahue RJ, Ikeda A, Nickells RW. The Influence of Mitochondrial Dynamics and Function on Retinal Ganglion Cell Susceptibility in Optic Nerve Disease. Cells 2021; 10:cells10071593. [PMID: 34201955 PMCID: PMC8306483 DOI: 10.3390/cells10071593] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/30/2022] Open
Abstract
The important roles of mitochondrial function and dysfunction in the process of neurodegeneration are widely acknowledged. Retinal ganglion cells (RGCs) appear to be a highly vulnerable neuronal cell type in the central nervous system with respect to mitochondrial dysfunction but the actual reasons for this are still incompletely understood. These cells have a unique circumstance where unmyelinated axons must bend nearly 90° to exit the eye and then cross a translaminar pressure gradient before becoming myelinated in the optic nerve. This region, the optic nerve head, contains some of the highest density of mitochondria present in these cells. Glaucoma represents a perfect storm of events occurring at this location, with a combination of changes in the translaminar pressure gradient and reassignment of the metabolic support functions of supporting glia, which appears to apply increased metabolic stress to the RGC axons leading to a failure of axonal transport mechanisms. However, RGCs themselves are also extremely sensitive to genetic mutations, particularly in genes affecting mitochondrial dynamics and mitochondrial clearance. These mutations, which systemically affect the mitochondria in every cell, often lead to an optic neuropathy as the sole pathologic defect in affected patients. This review summarizes knowledge of mitochondrial structure and function, the known energy demands of neurons in general, and places these in the context of normal and pathological characteristics of mitochondria attributed to RGCs.
Collapse
Affiliation(s)
- Nicole A. Muench
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
| | - Sonia Patel
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
| | - Margaret E. Maes
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria;
| | - Ryan J. Donahue
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
- Boston Children’s Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Robert W. Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
- Correspondence:
| |
Collapse
|
67
|
Nahacka Z, Zobalova R, Dubisova M, Rohlena J, Neuzil J. Miro proteins connect mitochondrial function and intercellular transport. Crit Rev Biochem Mol Biol 2021; 56:401-425. [PMID: 34139898 DOI: 10.1080/10409238.2021.1925216] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mitochondria are organelles present in most eukaryotic cells, where they play major and multifaceted roles. The classical notion of the main mitochondrial function as the powerhouse of the cell per se has been complemented by recent discoveries pointing to mitochondria as organelles affecting a number of other auxiliary processes. They go beyond the classical energy provision via acting as a relay point of many catabolic and anabolic processes, to signaling pathways critically affecting cell growth by their implication in de novo pyrimidine synthesis. These additional roles further underscore the importance of mitochondrial homeostasis in various tissues, where its deregulation promotes a number of pathologies. While it has long been known that mitochondria can move within a cell to sites where they are needed, recent research has uncovered that mitochondria can also move between cells. While this intriguing field of research is only emerging, it is clear that mobilization of mitochondria requires a complex apparatus that critically involves mitochondrial proteins of the Miro family, whose role goes beyond the mitochondrial transfer, as will be covered in this review.
Collapse
Affiliation(s)
- Zuzana Nahacka
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Renata Zobalova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Maria Dubisova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic.,School of Medical Science, Griffith University, Southport, Australia
| |
Collapse
|
68
|
Kedra J, Lin S, Pacheco A, Gallo G, Smith GM. Axotomy Induces Drp1-Dependent Fragmentation of Axonal Mitochondria. Front Mol Neurosci 2021; 14:668670. [PMID: 34149354 PMCID: PMC8209475 DOI: 10.3389/fnmol.2021.668670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/07/2021] [Indexed: 02/02/2023] Open
Abstract
It is well established that CNS axons fail to regenerate, undergo retrograde dieback, and form dystrophic growth cones due to both intrinsic and extrinsic factors. We sought to investigate the role of axonal mitochondria in the axonal response to injury. A viral vector (AAV) containing a mitochondrially targeted fluorescent protein (mitoDsRed) as well as fluorescently tagged LC3 (GFP-LC3), an autophagosomal marker, was injected into the primary motor cortex, to label the corticospinal tract (CST), of adult rats. The axons of the CST were then injured by dorsal column lesion at C4-C5. We found that mitochondria in injured CST axons near the injury site are fragmented and fragmentation of mitochondria persists for 2 weeks before returning to pre-injury lengths. Fragmented mitochondria have consistently been shown to be dysfunctional and detrimental to cellular health. Inhibition of Drp1, the GTPase responsible for mitochondrial fission, using a specific pharmacological inhibitor (mDivi-1) blocked fragmentation. Additionally, it was determined that there is increased mitophagy in CST axons following Spinal cord injury (SCI) based on increased colocalization of mitochondria and LC3. In vitro models revealed that mitochondrial divalent ion uptake is necessary for injury-induced mitochondrial fission, as inhibiting the mitochondrial calcium uniporter (MCU) using RU360 prevented injury-induced fission. This phenomenon was also observed in vivo. These studies indicate that following the injury, both in vivo and in vitro, axonal mitochondria undergo increased fission, which may contribute to the lack of regeneration seen in CNS neurons.
Collapse
Affiliation(s)
- Joseph Kedra
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Shen Lin
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Almudena Pacheco
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - George M Smith
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Neuroscience, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
69
|
Shil SK, Kagawa Y, Umaru BA, Nanto-Hara F, Miyazaki H, Yamamoto Y, Kobayashi S, Suzuki C, Abe T, Owada Y. Ndufs4 ablation decreases synaptophysin expression in hippocampus. Sci Rep 2021; 11:10969. [PMID: 34040028 PMCID: PMC8155116 DOI: 10.1038/s41598-021-90127-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 05/04/2021] [Indexed: 02/04/2023] Open
Abstract
Altered function of mitochondrial respiratory chain in brain cells is related to many neurodegenerative diseases. NADH Dehydrogenase (Ubiquinone) Fe-S protein 4 (Ndufs4) is one of the subunits of mitochondrial complex I and its mutation in human is associated with Leigh syndrome. However, the molecular biological role of Ndufs4 in neuronal function is poorly understood. In this study, upon Ndufs4 expression confirmation in NeuN-positive neurons, and GFAP-positive astrocytes in WT mouse hippocampus, we found significant decrease of mitochondrial respiration in Ndufs4-KO mouse hippocampus. Although there was no change in the number of NeuN positive neurons in Ndufs4-KO hippocampus, the expression of synaptophysin, a presynaptic protein, was significantly decreased. To investigate the detailed mechanism, we silenced Ndufs4 in Neuro-2a cells and we observed shorter neurite lengths with decreased expression of synaptophysin. Furthermore, western blot analysis for phosphorylated extracellular regulated kinase (pERK) revealed that Ndufs4 silencing decreases the activity of ERK signalling. These results suggest that Ndufs4-modulated mitochondrial activity may be involved in neuroplasticity via regulating synaptophysin expression.
Collapse
Affiliation(s)
- Subrata Kumar Shil
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yoshiteru Kagawa
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| | - Banlanjo Abdulaziz Umaru
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Fumika Nanto-Hara
- Division of Animal Metabolism and Nutrition, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, 305-0901, Japan
| | - Hirofumi Miyazaki
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Yui Yamamoto
- Department of Anatomy, Tohoku Medical and Pharmaceutical University, Sendai, 981-0905, Japan
| | - Shuhei Kobayashi
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan
| | - Chitose Suzuki
- Department of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Takaaki Abe
- Department of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Yuji Owada
- Department of Organ Anatomy, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
70
|
The Role of Lipids, Lipid Metabolism and Ectopic Lipid Accumulation in Axon Growth, Regeneration and Repair after CNS Injury and Disease. Cells 2021; 10:cells10051078. [PMID: 34062747 PMCID: PMC8147289 DOI: 10.3390/cells10051078] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/20/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
Axons in the adult mammalian nervous system can extend over formidable distances, up to one meter or more in humans. During development, axonal and dendritic growth requires continuous addition of new membrane. Of the three major kinds of membrane lipids, phospholipids are the most abundant in all cell membranes, including neurons. Not only immature axons, but also severed axons in the adult require large amounts of lipids for axon regeneration to occur. Lipids also serve as energy storage, signaling molecules and they contribute to tissue physiology, as demonstrated by a variety of metabolic disorders in which harmful amounts of lipids accumulate in various tissues through the body. Detrimental changes in lipid metabolism and excess accumulation of lipids contribute to a lack of axon regeneration, poor neurological outcome and complications after a variety of central nervous system (CNS) trauma including brain and spinal cord injury. Recent evidence indicates that rewiring lipid metabolism can be manipulated for therapeutic gain, as it favors conditions for axon regeneration and CNS repair. Here, we review the role of lipids, lipid metabolism and ectopic lipid accumulation in axon growth, regeneration and CNS repair. In addition, we outline molecular and pharmacological strategies to fine-tune lipid composition and energy metabolism in neurons and non-neuronal cells that can be exploited to improve neurological recovery after CNS trauma and disease.
Collapse
|
71
|
Advances in Regeneration of Retinal Ganglion Cells and Optic Nerves. Int J Mol Sci 2021; 22:ijms22094616. [PMID: 33924833 PMCID: PMC8125313 DOI: 10.3390/ijms22094616] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma, the second leading cause of blindness worldwide, is an incurable neurodegenerative disorder due to the dysfunction of retinal ganglion cells (RGCs). RGCs function as the only output neurons conveying the detected light information from the retina to the brain, which is a bottleneck of vision formation. RGCs in mammals cannot regenerate if injured, and RGC subtypes differ dramatically in their ability to survive and regenerate after injury. Recently, novel RGC subtypes and markers have been uncovered in succession. Meanwhile, apart from great advances in RGC axon regeneration, some degree of experimental RGC regeneration has been achieved by the in vitro differentiation of embryonic stem cells and induced pluripotent stem cells or in vivo somatic cell reprogramming, which provides insights into the future therapy of myriad neurodegenerative disorders. Further approaches to the combination of different factors will be necessary to develop efficacious future therapeutic strategies to promote ultimate axon and RGC regeneration and functional vision recovery following injury.
Collapse
|
72
|
Han Q, Xu XM. Mitochondrial integrity in neuronal injury and repair. Neural Regen Res 2021; 16:674-675. [PMID: 33063720 PMCID: PMC8067945 DOI: 10.4103/1673-5374.295317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/25/2020] [Accepted: 08/08/2020] [Indexed: 11/25/2022] Open
Affiliation(s)
- Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute; Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
73
|
Cheng XT, Sheng ZH. Developmental regulation of microtubule-based trafficking and anchoring of axonal mitochondria in health and diseases. Dev Neurobiol 2021; 81:284-299. [PMID: 32302463 PMCID: PMC7572491 DOI: 10.1002/dneu.22748] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/09/2020] [Accepted: 04/09/2020] [Indexed: 12/20/2022]
Abstract
Mitochondria are cellular power plants that supply most of the ATP required in the brain to power neuronal growth, function, and regeneration. Given their extremely polarized structures and extended long axons, neurons face an exceptional challenge to maintain energy homeostasis in distal axons, synapses, and growth cones. Anchored mitochondria serve as local energy sources; therefore, the regulation of mitochondrial trafficking and anchoring ensures that these metabolically active areas are adequately supplied with ATP. Chronic mitochondrial dysfunction is a hallmark feature of major aging-related neurodegenerative diseases, and thus, anchored mitochondria in aging neurons need to be removed when they become dysfunctional. Investigations into the regulation of microtubule (MT)-based trafficking and anchoring of axonal mitochondria under physiological and pathological circumstances represent an important emerging area. In this short review article, we provide an updated overview of recent in vitro and in vivo studies showing (1) how mitochondria are transported and positioned in axons and synapses during neuronal developmental and maturation stages, and (2) how altered mitochondrial motility and axonal energy deficits in aging nervous systems link to neurodegeneration and regeneration in a disease or injury setting. We also highlight a major role of syntaphilin as a key MT-based regulator of axonal mitochondrial trafficking and anchoring in mature neurons.
Collapse
Affiliation(s)
- Xiu-Tang Cheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, Maryland 20892-3706, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, Maryland 20892-3706, USA
| |
Collapse
|
74
|
Lee N, Lee MY, Lee J, Kwon SH, Seung H, Lim J, Kim S. Hepatocyte growth factor induces pErk and pSTAT3 (Ser 727) to promote mitochondrial activity and neurite outgrowth in primary dorsal root ganglion cultures. Neuroreport 2021; 32:525-530. [PMID: 33788814 DOI: 10.1097/wnr.0000000000001622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Hepatocyte growth factor (HGF) promotes the neurite outgrowth of sensory neurons in developmental stages, but its role in injured peripheral nerves in adult mice remains largely been unexplored. In this study, we investigated the role of HGF in the regeneration of injured peripheral nerves using cultured dorsal root ganglions (DRGs). When cells were treated with HGF protein, the length of the neurite was increased 1.4-fold compared to the untreated control group. HGF greatly increased the level of phosphorylated STAT3 at serine 727 [pSTAT3 (Ser 727)], thereby translocating the protein to the mitochondria. HGF treatment increased the activity of mitochondrial complex I. When DRGs were cultured in the presence of U0126, a pharmacological inhibitor of Erk, the HGF-mediated increase in neurite outgrowth and the level of pSTAT3 (Ser 727) were both suppressed. Taken together, these results suggest that the HGF/c-met pathway might promote neurite outgrowth by controlling mitochondrial activity through the HGF/Erk/STAT3 axis.
Collapse
Affiliation(s)
- Nayeon Lee
- Department of Biological Sciences, Seoul National University.,Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Mi-Young Lee
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Junghun Lee
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Sang Ho Kwon
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Hana Seung
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Jaegook Lim
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| | - Sunyoung Kim
- Division of Gene Therapy, Helixmith Co Ltd., Seoul, Korea
| |
Collapse
|
75
|
Wang B, Huang M, Shang D, Yan X, Zhao B, Zhang X. Mitochondrial Behavior in Axon Degeneration and Regeneration. Front Aging Neurosci 2021; 13:650038. [PMID: 33762926 PMCID: PMC7982458 DOI: 10.3389/fnagi.2021.650038] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are organelles responsible for bioenergetic metabolism, calcium homeostasis, and signal transmission essential for neurons due to their high energy consumption. Accumulating evidence has demonstrated that mitochondria play a key role in axon degeneration and regeneration under physiological and pathological conditions. Mitochondrial dysfunction occurs at an early stage of axon degeneration and involves oxidative stress, energy deficiency, imbalance of mitochondrial dynamics, defects in mitochondrial transport, and mitophagy dysregulation. The restoration of these defective mitochondria by enhancing mitochondrial transport, clearance of reactive oxidative species (ROS), and improving bioenergetic can greatly contribute to axon regeneration. In this paper, we focus on the biological behavior of axonal mitochondria in aging, injury (e.g., traumatic brain and spinal cord injury), and neurodegenerative diseases (Alzheimer's disease, AD; Parkinson's disease, PD; Amyotrophic lateral sclerosis, ALS) and consider the role of mitochondria in axon regeneration. We also compare the behavior of mitochondria in different diseases and outline novel therapeutic strategies for addressing abnormal mitochondrial biological behavior to promote axonal regeneration in neurological diseases and injuries.
Collapse
Affiliation(s)
- Biyao Wang
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Minghao Huang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Dehao Shang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xu Yan
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Baohong Zhao
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Xinwen Zhang
- Center of Implant Dentistry, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
76
|
Axonal Organelles as Molecular Platforms for Axon Growth and Regeneration after Injury. Int J Mol Sci 2021; 22:ijms22041798. [PMID: 33670312 PMCID: PMC7918155 DOI: 10.3390/ijms22041798] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Investigating the molecular mechanisms governing developmental axon growth has been a useful approach for identifying new strategies for boosting axon regeneration after injury, with the goal of treating debilitating conditions such as spinal cord injury and vision loss. The picture emerging is that various axonal organelles are important centers for organizing the molecular mechanisms and machinery required for growth cone development and axon extension, and these have recently been targeted to stimulate robust regeneration in the injured adult central nervous system (CNS). This review summarizes recent literature highlighting a central role for organelles such as recycling endosomes, the endoplasmic reticulum, mitochondria, lysosomes, autophagosomes and the proteasome in developmental axon growth, and describes how these organelles can be targeted to promote axon regeneration after injury to the adult CNS. This review also examines the connections between these organelles in developing and regenerating axons, and finally discusses the molecular mechanisms within the axon that are required for successful axon growth.
Collapse
|
77
|
Schaeffer J, Delpech C, Albert F, Belin S, Nawabi H. Adult Mouse Retina Explants: From ex vivo to in vivo Model of Central Nervous System Injuries. Front Mol Neurosci 2020; 13:599948. [PMID: 33324161 PMCID: PMC7723849 DOI: 10.3389/fnmol.2020.599948] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/16/2020] [Indexed: 12/14/2022] Open
Abstract
In mammals, adult neurons fail to regenerate following any insult to adult central nervous system (CNS), which leads to a permanent and irreversible loss of motor and cognitive functions. For a long time, much effort has been deployed to uncover mechanisms of axon regeneration in the CNS. Even if some cases of functional recovery have been reported, there is still a discrepancy regarding the functionality of a neuronal circuit upon lesion. Today, there is a need not only to identify new molecules implicated in adult CNS axon regeneration, but also to decipher the fine molecular mechanisms associated with regeneration failure. Here, we propose to use cultures of adult retina explants to study all molecular and cellular mechanisms that occur during CNS regeneration. We show that adult retinal explant cultures have the advantages to (i) recapitulate all the features observed in vivo, including axon regeneration induced by intrinsic factors, and (ii) be an ex vivo set-up with high accessibility and many downstream applications. Thanks to several examples, we demonstrate that adult explants can be used to address many questions, such as axon guidance, growth cone formation and cytoskeleton dynamics. Using laser guided ablation of a single axon, axonal injury can be performed at a single axon level, which allows to record early and late molecular events that occur after the lesion. Our model is the ideal tool to study all molecular and cellular events that occur during CNS regeneration at a single-axon level, which is currently not doable in vivo. It is extremely valuable to address unanswered questions of neuroprotection and neuroregeneration in the context of CNS lesion and neurodegenerative diseases.
Collapse
|
78
|
Yang LQ, Chen M, Ren DL, Hu B. Dual Oxidase Mutant Retards Mauthner-Cell Axon Regeneration at an Early Stage via Modulating Mitochondrial Dynamics in Zebrafish. Neurosci Bull 2020; 36:1500-1512. [PMID: 33123984 DOI: 10.1007/s12264-020-00600-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/25/2020] [Indexed: 12/17/2022] Open
Abstract
Dual oxidase (duox)-derived reactive oxygen species (ROS) have been correlated with neuronal polarity, cerebellar development, and neuroplasticity. However, there have not been many comprehensive studies of the effect of individual duox isoforms on central-axon regeneration in vivo. Here, we explored this question in zebrafish, an excellent model organism for central-axon regeneration studies. In our research, mutation of the duox gene with CRISPR/Cas9 significantly retarded the single-axon regeneration of the zebrafish Mauthner cell in vivo. Using deep transcriptome sequencing, we found that the expression levels of related functional enzymes in mitochondria were down-regulated in duox mutant fish. In vivo imaging showed that duox mutants had significantly disrupted mitochondrial transport and redox state in single Mauthner-cell axon. Our research data provide insights into how duox is involved in central-axon regeneration by changing mitochondrial transport.
Collapse
Affiliation(s)
- Lei-Qing Yang
- Eye Center, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Biomedical Sciences, University of Science and Technology of China, Hefei, 230026, China
| | - Min Chen
- Eye Center, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Biomedical Sciences, University of Science and Technology of China, Hefei, 230026, China
| | - Da-Long Ren
- Eye Center, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Biomedical Sciences, University of Science and Technology of China, Hefei, 230026, China.
- Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China.
| | - Bing Hu
- Eye Center, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Chinese Academy of Sciences Key Laboratory of Brain Function and Disease, School of Life Sciences, Division of Biomedical Sciences, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
79
|
Hwang J, Namgung U. Phosphorylation of STAT3 by axonal Cdk5 promotes axonal regeneration by modulating mitochondrial activity. Exp Neurol 2020; 335:113511. [PMID: 33098871 DOI: 10.1016/j.expneurol.2020.113511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/14/2020] [Accepted: 10/17/2020] [Indexed: 01/03/2023]
Abstract
Cyclin-dependent kinase 5 (Cdk5) is involved in neural organization and synaptic functions in developing and adult brains, yet its role in axonal regeneration is not known well. Here, we characterize Cdk5 function for axonal regeneration after peripheral nerve injury. Levels of Cdk5 and p25 were elevated in sciatic nerve axons after injury. Cdk5 activity was concomitantly induced from injured nerve and increased the phosphorylation of signal transducer and activator of transcription 3 (STAT3) on the serine 727 residue. Pharmacological and genetic blockades of Cdk5 activity phosphorylating STAT3 resulted in the inhibition of axonal regeneration as evidenced by reduction of retrograde labeling of dorsal root ganglion (DRG) sensory neurons and spinal motor neurons and also of neurite outgrowth of preconditioned DRG neurons in culture. Cdk5 and STAT3 were found in mitochondrial membranes of the injured sciatic nerve. Cdk5-GFP, which was translocated into the mitochondria by the mitochondrial target sequence (MTS), induced STAT3 phosphorylation in transfected DRG neurons and was sufficient to induce neurite outgrowth. In the mitochondria, Cdk5 activity was positively correlated with increased mitochondrial membrane potential as measured by fluorescence intensity of JC-1 aggregates. Our data suggest that Cdk5 may play a role in modulating mitochondrial activity through STAT3 phosphorylation, thereby promoting axonal regeneration.
Collapse
Affiliation(s)
- Jinyeon Hwang
- Neurophysiology Laboratory, Department of Oriental Medicine, Institute of Bioscience and Integrative Medicine, Daejeon University, Daehak-ro 62, Daejeon 34520, South Korea
| | - Uk Namgung
- Neurophysiology Laboratory, Department of Oriental Medicine, Institute of Bioscience and Integrative Medicine, Daejeon University, Daehak-ro 62, Daejeon 34520, South Korea.
| |
Collapse
|
80
|
Gallo G. The bioenergetics of neuronal morphogenesis and regeneration: Frontiers beyond the mitochondrion. Dev Neurobiol 2020; 80:263-276. [PMID: 32750228 DOI: 10.1002/dneu.22776] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/27/2022]
Abstract
The formation of axons and dendrites during development, and their regeneration following injury, are energy intensive processes. The underlying assembly and dynamics of the cytoskeleton, axonal transport mechanisms, and extensive signaling networks all rely on ATP and GTP consumption. Cellular ATP is generated through oxidative phosphorylation (OxP) in mitochondria, glycolysis and "regenerative" kinase systems. Recent investigations have focused on the role of the mitochondrion in axonal development and regeneration emphasizing the importance of this organelle and OxP in axon development and regeneration. In contrast, the understanding of alternative sources of ATP in neuronal morphogenesis and regeneration remains largely unexplored. This review focuses on the current state of the field of neuronal bioenergetics underlying morphogenesis and regeneration and considers the literature on the bioenergetics of non-neuronal cell motility to emphasize the potential contributions of non-mitochondrial energy sources.
Collapse
Affiliation(s)
- Gianluca Gallo
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Shriners Hospitals Pediatric Research Center, Temple University, Philadelphia, PA, USA
| |
Collapse
|
81
|
A High-Content Screen Identifies TPP1 and Aurora B as Regulators of Axonal Mitochondrial Transport. Cell Rep 2020; 28:3224-3237.e5. [PMID: 31533043 PMCID: PMC6937139 DOI: 10.1016/j.celrep.2019.08.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/12/2019] [Accepted: 08/09/2019] [Indexed: 12/22/2022] Open
Abstract
Dysregulated axonal trafficking of mitochondria is linked to neurodegenerative disorders. We report a high-content screen for small-molecule regulators of the axonal transport of mitochondria. Six compounds enhanced mitochondrial transport in the sub-micromolar range, acting via three cellular targets: F-actin, Tripeptidyl peptidase 1 (TPP1), or Aurora Kinase B (AurKB). Pharmacological inhibition or small hairpin RNA (shRNA) knockdown of each target promotes mitochondrial axonal transport in rat hippocampal neurons and induced pluripotent stem cell (iPSC)-derived human cortical neurons and enhances mitochondrial transport in iPSC-derived motor neurons from an amyotrophic lateral sclerosis (ALS) patient bearing one copy of SOD1A4V mutation. Our work identifies druggable regulators of axonal transport of mitochondria, provides broadly applicable methods for similar image-based screens, and suggests that restoration of proper axonal trafficking of mitochondria can be achieved in human ALS neurons. Shlevkov et al. establish a high-content screen for enhancers of axonal mitochondrial trafficking. Identified compounds act through three cellular targets: F-Actin, Tripeptidyl peptidase 1, and Aurora Kinase B. Motor neurons derived from a SOD1+/A4VALS patient have decreased mitochondrial motility, which can be reversed by inhibitors of these targets.
Collapse
|
82
|
Jara JS, Agger S, Hollis ER. Functional Electrical Stimulation and the Modulation of the Axon Regeneration Program. Front Cell Dev Biol 2020; 8:736. [PMID: 33015031 PMCID: PMC7462022 DOI: 10.3389/fcell.2020.00736] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/15/2020] [Indexed: 01/07/2023] Open
Abstract
Neural injury in mammals often leads to persistent functional deficits as spontaneous repair in the peripheral nervous system (PNS) is often incomplete, while endogenous repair mechanisms in the central nervous system (CNS) are negligible. Peripheral axotomy elicits growth-associated gene programs in sensory and motor neurons that can support reinnervation of peripheral targets given sufficient levels of debris clearance and proximity to nerve targets. In contrast, while damaged CNS circuitry can undergo a limited amount of sprouting and reorganization, this innate plasticity does not re-establish the original connectivity. The utility of novel CNS circuitry will depend on effective connectivity and appropriate training to strengthen these circuits. One method of enhancing novel circuit connectivity is through the use of electrical stimulation, which supports axon growth in both central and peripheral neurons. This review will focus on the effects of CNS and PNS electrical stimulation in activating axon growth-associated gene programs and supporting the recovery of motor and sensory circuits. Electrical stimulation-mediated neuroplasticity represents a therapeutically viable approach to support neural repair and recovery. Development of appropriate clinical strategies employing electrical stimulation will depend upon determining the underlying mechanisms of activity-dependent axon regeneration and the heterogeneity of neuronal subtype responses to stimulation.
Collapse
Affiliation(s)
| | - Sydney Agger
- Burke Neurological Institute, White Plains, NY, United States
| | - Edmund R Hollis
- Burke Neurological Institute, White Plains, NY, United States.,Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
83
|
Pleiotropic Mitochondria: The Influence of Mitochondria on Neuronal Development and Disease. J Neurosci 2020; 39:8200-8208. [PMID: 31619488 DOI: 10.1523/jneurosci.1157-19.2019] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/09/2019] [Accepted: 08/10/2019] [Indexed: 02/08/2023] Open
Abstract
Mitochondria play many important biological roles, including ATP production, lipid biogenesis, ROS regulation, and calcium clearance. In neurons, the mitochondrion is an essential organelle for metabolism and calcium homeostasis. Moreover, mitochondria are extremely dynamic and able to divide, fuse, and move along microtubule tracks to ensure their distribution to the neuronal periphery. Mitochondrial dysfunction and altered mitochondrial dynamics are observed in a wide range of conditions, from impaired neuronal development to various neurodegenerative diseases. Novel imaging techniques and genetic tools provide unprecedented access to the physiological roles of mitochondria by visualizing mitochondrial trafficking, morphological dynamics, ATP generation, and ultrastructure. Recent studies using these new techniques have unveiled the influence of mitochondria on axon branching, synaptic function, calcium regulation with the ER, glial cell function, neurogenesis, and neuronal repair. This review provides an overview of the crucial roles played by mitochondria in the CNS in physiological and pathophysiological conditions.
Collapse
|
84
|
Zhu J, Li P, Zhou YG, Ye J. Altered Energy Metabolism During Early Optic Nerve Crush Injury: Implications of Warburg-Like Aerobic Glycolysis in Facilitating Retinal Ganglion Cell Survival. Neurosci Bull 2020; 36:761-777. [PMID: 32277382 PMCID: PMC7340706 DOI: 10.1007/s12264-020-00490-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023] Open
Abstract
Neurons, especially axons, are metabolically demanding and energetically vulnerable during injury. However, the exact energy budget alterations that occur early after axon injury and the effects of these changes on neuronal survival remain unknown. Using a classic mouse model of optic nerve-crush injury, we found that traumatized optic nerves and retinas harbor the potential to mobilize two primary energetic machineries, glycolysis and oxidative phosphorylation, to satisfy the robustly increased adenosine triphosphate (ATP) demand. Further exploration of metabolic activation showed that mitochondrial oxidative phosphorylation was amplified over other pathways, which may lead to decreased retinal ganglion cell (RGC) survival despite its supplement to ATP production. Gene set enrichment analysis of a microarray (GSE32309) identified significant activation of oxidative phosphorylation in injured retinas from wild-type mice compared to those from mice with deletion of phosphatase and tensin homolog (PTEN), while PTEN-/- mice had more robust RGC survival. Therefore, we speculated that the oxidation-favoring metabolic pattern after optic nerve-crush injury could be adverse for RGC survival. After redirecting metabolic flux toward glycolysis (magnifying the Warburg effect) using the drug meclizine, we successfully increased RGC survival. Thus, we provide novel insights into a potential bioenergetics-based strategy for neuroprotection.
Collapse
Affiliation(s)
- Jingyi Zhu
- Department of Ophthalmology, Army Medical Center of the People's Liberation Army (PLA), Army Medical University, Chongqing, 400042, China
| | - Ping Li
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery, Army Medical Center of the PLA, Army Medical University, Chongqing, 400042, China
| | - Yuan-Guo Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery, Army Medical Center of the PLA, Army Medical University, Chongqing, 400042, China.
| | - Jian Ye
- Department of Ophthalmology, Army Medical Center of the People's Liberation Army (PLA), Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
85
|
Yang SG, Li CP, Peng XQ, Teng ZQ, Liu CM, Zhou FQ. Strategies to Promote Long-Distance Optic Nerve Regeneration. Front Cell Neurosci 2020; 14:119. [PMID: 32477071 PMCID: PMC7240020 DOI: 10.3389/fncel.2020.00119] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Mammalian retinal ganglion cells (RGCs) in the central nervous system (CNS) often die after optic nerve injury and surviving RGCs fail to regenerate their axons, eventually resulting in irreversible vision loss. Manipulation of a diverse group of genes can significantly boost optic nerve regeneration of mature RGCs by reactivating developmental-like growth programs or suppressing growth inhibitory pathways. By injury of the vision pathway near their brain targets, a few studies have shown that regenerated RGC axons could form functional synapses with targeted neurons but exhibited poor neural conduction or partial functional recovery. Therefore, the functional restoration of eye-to-brain pathways remains a greatly challenging issue. Here, we review recent advances in long-distance optic nerve regeneration and the subsequent reconnecting to central targets. By summarizing our current strategies for promoting functional recovery, we hope to provide potential insights into future exploration in vision reformation after neural injuries.
Collapse
Affiliation(s)
- Shu-Guang Yang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chang-Ping Li
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xue-Qi Peng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhao-Qian Teng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Chang-Mei Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.,The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
86
|
Morales-Hernández A, Benaksas C, Chabot A, Caprio C, Ferdous M, Zhao X, Kang G, McKinney-Freeman S. GPRASP proteins are critical negative regulators of hematopoietic stem cell transplantation. Blood 2020; 135:1111-1123. [PMID: 32027737 PMCID: PMC7118811 DOI: 10.1182/blood.2019003435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/21/2020] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cell (HSC) transplantation (HSCT) is often exploited to treat hematologic disease. Donor HSCs must survive, proliferate, and differentiate in the damaged environment of the reconstituting niche. Illuminating molecular mechanisms regulating the activity of transplanted HSCs will inform efforts to improve HSCT. Here, we report that G-protein-coupled receptor-associated sorting proteins (GPRASPs) function as negative regulators of HSCT. Silencing of Gprasp1 or Gprasp2 increased the survival, quiescence, migration, niche retention, and hematopoietic repopulating activity of hematopoietic stem and progenitor cells (HSPCs) posttransplant. We further show that GPRASP1 and GPRASP2 promote the degradation of CXCR4, a master regulator of HSC function during transplantation. CXCR4 accumulates in Gprasp-deficient HSPCs, boosting their function posttransplant. Thus, GPRASPs negatively regulate CXCR4 stability in HSCs. Our work reveals GPRASP proteins as negative regulators of HSCT and CXCR4 activity. Disruption of GPRASP/CXCR4 interactions could be exploited in the future to enhance the efficiency of HSCT.
Collapse
Affiliation(s)
| | - Chaïma Benaksas
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
- Paris Diderot University, Paris, France; and
| | - Ashley Chabot
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Claire Caprio
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Maheen Ferdous
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Xiwen Zhao
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN
| | | |
Collapse
|
87
|
Regenerative Potential of Carbon Monoxide in Adult Neural Circuits of the Central Nervous System. Int J Mol Sci 2020; 21:ijms21072273. [PMID: 32218342 PMCID: PMC7177523 DOI: 10.3390/ijms21072273] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/17/2020] [Accepted: 03/23/2020] [Indexed: 01/04/2023] Open
Abstract
Regeneration of adult neural circuits after an injury is limited in the central nervous system (CNS). Heme oxygenase (HO) is an enzyme that produces HO metabolites, such as carbon monoxide (CO), biliverdin and iron by heme degradation. CO may act as a biological signal transduction effector in CNS regeneration by stimulating neuronal intrinsic and extrinsic mechanisms as well as mitochondrial biogenesis. CO may give directions by which the injured neurovascular system switches into regeneration mode by stimulating endogenous neural stem cells and endothelial cells to produce neurons and vessels capable of replacing injured neurons and vessels in the CNS. The present review discusses the regenerative potential of CO in acute and chronic neuroinflammatory diseases of the CNS, such as stroke, traumatic brain injury, multiple sclerosis and Alzheimer’s disease and the role of signaling pathways and neurotrophic factors. CO-mediated facilitation of cellular communications may boost regeneration, consequently forming functional adult neural circuits in CNS injury.
Collapse
|
88
|
Han Q, Xie Y, Ordaz JD, Huh AJ, Huang N, Wu W, Liu N, Chamberlain KA, Sheng ZH, Xu XM. Restoring Cellular Energetics Promotes Axonal Regeneration and Functional Recovery after Spinal Cord Injury. Cell Metab 2020; 31:623-641.e8. [PMID: 32130884 PMCID: PMC7188478 DOI: 10.1016/j.cmet.2020.02.002] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 11/24/2019] [Accepted: 01/31/2020] [Indexed: 01/30/2023]
Abstract
Axonal regeneration in the central nervous system (CNS) is a highly energy-demanding process. Extrinsic insults and intrinsic restrictions lead to an energy crisis in injured axons, raising the question of whether recovering energy deficits facilitates regeneration. Here, we reveal that enhancing axonal mitochondrial transport by deleting syntaphilin (Snph) recovers injury-induced mitochondrial depolarization. Using three CNS injury mouse models, we demonstrate that Snph-/- mice display enhanced corticospinal tract (CST) regeneration passing through a spinal cord lesion, accelerated regrowth of monoaminergic axons across a transection gap, and increased compensatory sprouting of uninjured CST. Notably, regenerated CST axons form functional synapses and promote motor functional recovery. Administration of the bioenergetic compound creatine boosts CST regenerative capacity in Snph-/- mice. Our study provides mechanistic insights into intrinsic regeneration failure in CNS and suggests that enhancing mitochondrial transport and cellular energetics are promising strategies to promote regeneration and functional restoration after CNS injuries.
Collapse
Affiliation(s)
- Qi Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yuxiang Xie
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Josue D Ordaz
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrew J Huh
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ning Huang
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Wei Wu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Naikui Liu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kelly A Chamberlain
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Xiao-Ming Xu
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
89
|
Tang NH, Kim KW, Xu S, Blazie SM, Yee BA, Yeo GW, Jin Y, Chisholm AD. The mRNA Decay Factor CAR-1/LSM14 Regulates Axon Regeneration via Mitochondrial Calcium Dynamics. Curr Biol 2020; 30:865-876.e7. [PMID: 31983639 DOI: 10.1016/j.cub.2019.12.061] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/26/2019] [Accepted: 12/19/2019] [Indexed: 12/11/2022]
Abstract
mRNA decay factors regulate mRNA turnover by recruiting non-translating mRNAs and targeting them for translational repression and mRNA degradation. How mRNA decay pathways regulate cellular function in vivo with specificity is poorly understood. Here, we show that C. elegans mRNA decay factors, including the translational repressors CAR-1/LSM14 and CGH-1/DDX6, and the decapping enzymes DCAP-1/DCP1, function in neurons to differentially regulate axon development, maintenance, and regrowth following injury. In neuronal cell bodies, CAR-1 fully colocalizes with CGH-1 and partially colocalizes with DCAP-1, suggesting that mRNA decay components form at least two types of cytoplasmic granules. Following axon injury in adult neurons, loss of CAR-1 or CGH-1 results in increased axon regrowth and growth cone formation, whereas loss of DCAP-1 or DCAP-2 results in reduced regrowth. To determine how CAR-1 inhibits regrowth, we analyzed mRNAs bound to pan-neuronally expressed GFP::CAR-1 using a crosslinking and immunoprecipitation-based approach. Among the putative mRNA targets of CAR-1, we characterized the roles of micu-1, a regulator of the mitochondrial calcium uniporter MCU-1, in axon injury. We show that loss of car-1 results increased MICU-1 protein levels, and that enhanced axon regrowth in car-1 mutants is dependent on micu-1 and mcu-1. Moreover, axon injury induces transient calcium influx into axonal mitochondria, dependent on MCU-1. In car-1 loss-of-function mutants and in micu-1 overexpressing animals, the axonal mitochondrial calcium influx is more sustained, which likely underlies enhanced axon regrowth. Our data uncover a novel pathway that controls axon regrowth through axonal mitochondrial calcium uptake.
Collapse
Affiliation(s)
- Ngang Heok Tang
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kyung Won Kim
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Suhong Xu
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephen M Blazie
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brian A Yee
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yishi Jin
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrew D Chisholm
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
90
|
Laredo F, Plebanski J, Tedeschi A. Pericytes: Problems and Promises for CNS Repair. Front Cell Neurosci 2019; 13:546. [PMID: 31866833 PMCID: PMC6908836 DOI: 10.3389/fncel.2019.00546] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022] Open
Abstract
Microvascular complications are often associated with slow and progressive damage of various organs. Pericytes are multi-functional mural cells of the microcirculation that control blood flow, vascular permeability and homeostasis. Whereas accumulating evidence suggests that these cells are also implicated in a variety of diseases, pericytes represent promising targets that can be manipulated for therapeutic gain. Here, we review the role of pericytes in angiogenesis, blood-brain barrier (BBB) function, neuroinflammation, tissue fibrosis, axon regeneration failure, and neurodegeneration. In addition, we outline strategies altering pericyte behavior to point out problems and promises for axon regeneration and central nervous system (CNS) repair following injury or disease.
Collapse
Affiliation(s)
- Fabio Laredo
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Department of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Julia Plebanski
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Andrea Tedeschi
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, Columbus, OH, United States.,Discovery Theme on Chronic Brain Injury, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
91
|
Rewiring Neuronal Glycerolipid Metabolism Determines the Extent of Axon Regeneration. Neuron 2019; 105:276-292.e5. [PMID: 31786011 PMCID: PMC6975164 DOI: 10.1016/j.neuron.2019.10.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/11/2019] [Accepted: 10/03/2019] [Indexed: 12/21/2022]
Abstract
How adult neurons coordinate lipid metabolism to regenerate axons remains elusive. We found that depleting neuronal lipin1, a key enzyme controlling the balanced synthesis of glycerolipids through the glycerol phosphate pathway, enhanced axon regeneration after optic nerve injury. Axotomy elevated lipin1 in retinal ganglion cells, which contributed to regeneration failure in the CNS by favorably producing triglyceride (TG) storage lipids rather than phospholipid (PL) membrane lipids in neurons. Regrowth induced by lipin1 depletion required TG hydrolysis and PL synthesis. Decreasing TG synthesis by deleting neuronal diglyceride acyltransferases (DGATs) and enhancing PL synthesis through the Kennedy pathway promoted axon regeneration. In addition, peripheral neurons adopted this mechanism for their spontaneous axon regeneration. Our study reveals a critical role of lipin1 and DGATs as intrinsic regulators of glycerolipid metabolism in neurons and indicates that directing neuronal lipid synthesis away from TG synthesis and toward PL synthesis may promote axon regeneration. Injury-elevated Lipin1 and DGAT in retinal ganglion cells suppress regeneration Neuronal lipin1 and DGATs increase triglyceride and decrease phospholipids Redirecting triacylglyceride to phospholipid synthesis promotes axon regeneration
Collapse
|
92
|
Andrusiak MG, Sharifnia P, Lyu X, Wang Z, Dickey AM, Wu Z, Chisholm AD, Jin Y. Inhibition of Axon Regeneration by Liquid-like TIAR-2 Granules. Neuron 2019; 104:290-304.e8. [PMID: 31378567 PMCID: PMC6813885 DOI: 10.1016/j.neuron.2019.07.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 05/19/2019] [Accepted: 07/02/2019] [Indexed: 12/21/2022]
Abstract
Phase separation into liquid-like compartments is an emerging property of proteins containing prion-like domains (PrLDs), yet the in vivo roles of phase separation remain poorly understood. TIA proteins contain a C-terminal PrLD, and mutations in the PrLD are associated with several diseases. Here, we show that the C. elegans TIAR-2/TIA protein functions cell autonomously to inhibit axon regeneration. TIAR-2 undergoes liquid-liquid phase separation in vitro and forms granules with liquid-like properties in vivo. Axon injury induces a transient increase in TIAR-2 granule number. The PrLD is necessary and sufficient for granule formation and inhibiting regeneration. Tyrosine residues within the PrLD are important for granule formation and inhibition of regeneration. TIAR-2 is also serine phosphorylated in vivo. Non-phosphorylatable TIAR-2 variants do not form granules and are unable to inhibit axon regeneration. Our data demonstrate an in vivo function for phase-separated TIAR-2 and identify features critical for its function in axon regeneration.
Collapse
Affiliation(s)
- Matthew G Andrusiak
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Panid Sharifnia
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Xiaohui Lyu
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zhiping Wang
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrea M Dickey
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zilu Wu
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Andrew D Chisholm
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yishi Jin
- Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
93
|
Wang S, Smith GM, Selzer ME, Li S. Emerging molecular therapeutic targets for spinal cord injury. Expert Opin Ther Targets 2019; 23:787-803. [PMID: 31460807 DOI: 10.1080/14728222.2019.1661381] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Introduction: Spinal cord injury (SCI) is a complicated and devastating neurological disorder. Patients with SCI usually have dramatically reduced quality of life. In recent years, numerous studies have reported advances in understanding the pathophysiology of SCI and developing preclinical therapeutic strategies for SCI, including various molecular therapies, and yet there is still no cure. Areas covered: After SCI, tissue damage, responses and repair involve interactions among many cellular components, including neurons, axons, glia, leukocytes, and other cells. Accordingly, numerous cellular genes and molecules have become therapeutic targets for neural tissue repair, circuit reconstruction, and behavioral restoration. Here, we review the major recent advances in biological and molecular strategies to enhance neuroprotection, axon regeneration, remyelination, neuroplasticity and functional recovery in preclinical studies of SCI. Expert opinion: Researchers have made tremendous progress in identifying individual and combined molecular therapies in animal studies. It is very important to identify additional highly effective treatments for early neuroprotective intervention and for functionally meaningful axon regeneration and neuronal reconnections. Because multiple mechanisms contribute to the functional loss after SCI, combining the most promising approaches that target different pathophysiological and molecular mechanisms should exhibit synergistic actions for maximal functional restoration. [Databases searched: PubMed; inclusive dates: 6/27/2019].
Collapse
Affiliation(s)
- Shuo Wang
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia , PA , USA.,Department of Anatomy and Cell Biology, Temple University School of Medicine , Philadelphia , PA , USA
| | - George M Smith
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia , PA , USA.,Department of Neuroscience, Temple University School of Medicine , Philadelphia , PA , USA
| | - Michael E Selzer
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia , PA , USA.,Department of Neurology, Temple University School of Medicine , Philadelphia , PA , USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Temple University School of Medicine , Philadelphia , PA , USA.,Department of Anatomy and Cell Biology, Temple University School of Medicine , Philadelphia , PA , USA
| |
Collapse
|
94
|
Abstract
Permanent disabilities following CNS injuries result from the failure of injured axons to regenerate and rebuild functional connections with their original targets. By contrast, injury to peripheral nerves is followed by robust regeneration, which can lead to recovery of sensory and motor functions. This regenerative response requires the induction of widespread transcriptional and epigenetic changes in injured neurons. Considerable progress has been made in recent years in understanding how peripheral axon injury elicits these widespread changes through the coordinated actions of transcription factors, epigenetic modifiers and, to a lesser extent, microRNAs. Although many questions remain about the interplay between these mechanisms, these new findings provide important insights into the pivotal role of coordinated gene expression and chromatin remodelling in the neuronal response to injury.
Collapse
Affiliation(s)
- Marcus Mahar
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Valeria Cavalli
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
95
|
Chamberlain KA, Sheng ZH. Mechanisms for the maintenance and regulation of axonal energy supply. J Neurosci Res 2019; 97:897-913. [PMID: 30883896 PMCID: PMC6565461 DOI: 10.1002/jnr.24411] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/04/2019] [Accepted: 02/18/2019] [Indexed: 12/25/2022]
Abstract
The unique polarization and high-energy demand of neurons necessitates specialized mechanisms to maintain energy homeostasis throughout the cell, particularly in the distal axon. Mitochondria play a key role in meeting axonal energy demand by generating adenosine triphosphate through oxidative phosphorylation. Recent evidence demonstrates how axonal mitochondrial trafficking and anchoring are coordinated to sense and respond to altered energy requirements. If and when these mechanisms are impacted in pathological conditions, such as injury and neurodegenerative disease, is an emerging research frontier. Recent evidence also suggests that axonal energy demand may be supplemented by local glial cells, including astrocytes and oligodendrocytes. In this review, we provide an updated discussion of how oxidative phosphorylation, aerobic glycolysis, and oligodendrocyte-derived metabolic support contribute to the maintenance of axonal energy homeostasis.
Collapse
Affiliation(s)
- Kelly Anne Chamberlain
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, Maryland 20892-3706, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, Maryland 20892-3706, USA
| |
Collapse
|
96
|
Devine MJ, Kittler JT. Mitochondria at the neuronal presynapse in health and disease. Nat Rev Neurosci 2019; 19:63-80. [PMID: 29348666 DOI: 10.1038/nrn.2017.170] [Citation(s) in RCA: 394] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Synapses enable neurons to communicate with each other and are therefore a prerequisite for normal brain function. Presynaptically, this communication requires energy and generates large fluctuations in calcium concentrations. Mitochondria are optimized for supplying energy and buffering calcium, and they are actively recruited to presynapses. However, not all presynapses contain mitochondria; thus, how might synapses with and without mitochondria differ? Mitochondria are also increasingly recognized to serve additional functions at the presynapse. Here, we discuss the importance of presynaptic mitochondria in maintaining neuronal homeostasis and how dysfunctional presynaptic mitochondria might contribute to the development of disease.
Collapse
Affiliation(s)
- Michael J Devine
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Josef T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| |
Collapse
|
97
|
Smith GA, Lin TH, Sheehan AE, Van der Goes van Naters W, Neukomm LJ, Graves HK, Bis-Brewer DM, Züchner S, Freeman MR. Glutathione S-Transferase Regulates Mitochondrial Populations in Axons through Increased Glutathione Oxidation. Neuron 2019; 103:52-65.e6. [PMID: 31101394 PMCID: PMC6616599 DOI: 10.1016/j.neuron.2019.04.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/13/2018] [Accepted: 04/10/2019] [Indexed: 01/14/2023]
Abstract
Mitochondria are essential in long axons to provide metabolic support and sustain neuron integrity. A healthy mitochondrial pool is maintained by biogenesis, transport, mitophagy, fission, and fusion, but how these events are regulated in axons is not well defined. Here, we show that the Drosophila glutathione S-transferase (GST) Gfzf prevents mitochondrial hyperfusion in axons. Gfzf loss altered redox balance between glutathione (GSH) and oxidized glutathione (GSSG) and initiated mitochondrial fusion through the coordinated action of Mfn and Opa1. Gfzf functioned epistatically with the thioredoxin peroxidase Jafrac1 and the thioredoxin reductase 1 TrxR-1 to regulate mitochondrial dynamics. Altering GSH:GSSG ratios in mouse primary neurons in vitro also induced hyperfusion. Mitochondrial changes caused deficits in trafficking, the metabolome, and neuronal physiology. Changes in GSH and oxidative state are associated with neurodegenerative diseases like Alzheimer's. Our demonstration that GSTs are key in vivo regulators of axonal mitochondrial length and number provides a potential mechanistic link.
Collapse
Affiliation(s)
- Gaynor A Smith
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA; UK Dementia Research Institute, School of Medicine, Cardiff University, Cardiff CF24 4HQ, UK.
| | - Tzu-Huai Lin
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amy E Sheehan
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Lukas J Neukomm
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne VD, Switzerland
| | - Hillary K Graves
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Dana M Bis-Brewer
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Stephan Züchner
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA; Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Marc R Freeman
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Vollum Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
98
|
Chang CY, Liang MZ, Chen L. Current progress of mitochondrial transplantation that promotes neuronal regeneration. Transl Neurodegener 2019; 8:17. [PMID: 31210929 PMCID: PMC6567446 DOI: 10.1186/s40035-019-0158-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background Mitochondria are the major source of intracellular adenosine triphosphate (ATP) and play an essential role in a plethora of physiological functions, including the regulation of metabolism and the maintenance of cellular homeostasis. Mutations of mitochondrial DNA, proteins and impaired mitochondrial function have been implicated in the neurodegenerative diseases, stroke and injury of the central nervous system (CNS). The dynamic feature of mitochondrial fusion, fission, trafficking and turnover have also been documented in these diseases. Perspectives A major bottleneck of traditional approach to correct mitochondria-related disorders is the difficulty of drugs or gene targeting agents to arrive at specific sub-compartments of mitochondria. Moreover, the diverse nature of mitochondrial mutations among patients makes it impossible to develop one drug for one disease. To this end, mitochondrial transplantation presents a new paradigm of therapeutic intervention that benefits neuronal survival and regeneration for neurodegenerative diseases, stroke, and CNS injury. Supplement of healthy mitochondria to damaged neurons has been reported to promote neuronal viability, activity and neurite re-growth. In this review, we provide an overview of the recent advance and development on mitochondrial therapy. Conclusion Key parameters for the success of mitochondrial transplantation depend on the source and quality of isolated mitochondria, delivery protocol, and cellular uptake of supplemented mitochondria. To expedite clinical application of the mitochondrial transplantation, current isolation protocol needs optimization to obtain high percentage of functional mitochondria, isolated mitochondria may be packaged by biomaterials for successful delivery to brain allowing for efficient neuronal uptake.
Collapse
Affiliation(s)
- Chu-Yuan Chang
- 1Institute of Molecular Medicine, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| | - Min-Zong Liang
- 1Institute of Molecular Medicine, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan
| | - Linyi Chen
- 1Institute of Molecular Medicine, National Tsing Hua University, 101, Section 2, Kuang-Fu Road, Hsinchu, 30013 Taiwan.,2Department of Medical Science, National Tsing Hua University, Hsinchu, 30013 Taiwan
| |
Collapse
|
99
|
Kalinski AL, Kar AN, Craver J, Tosolini AP, Sleigh JN, Lee SJ, Hawthorne A, Brito-Vargas P, Miller-Randolph S, Passino R, Shi L, Wong VSC, Picci C, Smith DS, Willis DE, Havton LA, Schiavo G, Giger RJ, Langley B, Twiss JL. Deacetylation of Miro1 by HDAC6 blocks mitochondrial transport and mediates axon growth inhibition. J Cell Biol 2019; 218:1871-1890. [PMID: 31068376 PMCID: PMC6548128 DOI: 10.1083/jcb.201702187] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 02/15/2018] [Accepted: 04/15/2019] [Indexed: 02/08/2023] Open
Abstract
Inhibition of histone deacetylase 6 (HDAC6) was shown to support axon growth on the nonpermissive substrates myelin-associated glycoprotein (MAG) and chondroitin sulfate proteoglycans (CSPGs). Though HDAC6 deacetylates α-tubulin, we find that another HDAC6 substrate contributes to this axon growth failure. HDAC6 is known to impact transport of mitochondria, and we show that mitochondria accumulate in distal axons after HDAC6 inhibition. Miro and Milton proteins link mitochondria to motor proteins for axon transport. Exposing neurons to MAG and CSPGs decreases acetylation of Miro1 on Lysine 105 (K105) and decreases axonal mitochondrial transport. HDAC6 inhibition increases acetylated Miro1 in axons, and acetyl-mimetic Miro1 K105Q prevents CSPG-dependent decreases in mitochondrial transport and axon growth. MAG- and CSPG-dependent deacetylation of Miro1 requires RhoA/ROCK activation and downstream intracellular Ca2+ increase, and Miro1 K105Q prevents the decrease in axonal mitochondria seen with activated RhoA and elevated Ca2+ These data point to HDAC6-dependent deacetylation of Miro1 as a mediator of axon growth inhibition through decreased mitochondrial transport.
Collapse
Affiliation(s)
- Ashley L Kalinski
- Department of Biology, Drexel University, Philadelphia, PA.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Amar N Kar
- Department of Biological Sciences, University of South Carolina, Columbia, SC
| | - John Craver
- Department of Biological Sciences, University of South Carolina, Columbia, SC
| | - Andrew P Tosolini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - James N Sleigh
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute, University College London, London, UK
| | - Seung Joon Lee
- Department of Biological Sciences, University of South Carolina, Columbia, SC
| | | | - Paul Brito-Vargas
- Department of Biological Sciences, University of South Carolina, Columbia, SC
| | | | - Ryan Passino
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI
| | - Liang Shi
- Department of Biological Sciences, University of South Carolina, Columbia, SC
| | | | | | - Deanna S Smith
- Department of Biological Sciences, University of South Carolina, Columbia, SC
| | | | - Leif A Havton
- Departments of Neurology and Neurobiology, University of California, Los Angeles, Los Angeles, CA
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute, University College London, London, UK.,Discoveries Centre for Regenerative and Precision Medicine, University College London Campus, London, UK
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI
| | | | - Jeffery L Twiss
- Department of Biological Sciences, University of South Carolina, Columbia, SC
| |
Collapse
|
100
|
Abstract
Traumatic brain and spinal cord injuries cause permanent disability. Although progress has been made in understanding the cellular and molecular mechanisms underlying the pathophysiological changes that affect both structure and function after injury to the brain or spinal cord, there are currently no cures for either condition. This may change with the development and application of multi-layer omics, new sophisticated bioinformatics tools, and cutting-edge imaging techniques. Already, these technical advances, when combined, are revealing an unprecedented number of novel cellular and molecular targets that could be manipulated alone or in combination to repair the injured central nervous system with precision. In this review, we highlight recent advances in applying these new technologies to the study of axon regeneration and rebuilding of injured neural circuitry. We then discuss the challenges ahead to translate results produced by these technologies into clinical application to help improve the lives of individuals who have a brain or spinal cord injury.
Collapse
Affiliation(s)
- Andrea Tedeschi
- Department of Neuroscience and Discovery Themes Initiative, College of Medicine, Ohio State University, Columbus, Ohio, 43210, USA
| | - Phillip G Popovich
- Center for Brain and Spinal Cord Repair, Institute for Behavioral Medicine Research, Ohio State University, Columbus, Ohio, 43210, USA
| |
Collapse
|