1
|
Ketata I, Ellouz E. From pathological mechanisms in Krabbe disease to cutting-edge therapy: A comprehensive review. Neuropathology 2024; 44:255-277. [PMID: 38444347 DOI: 10.1111/neup.12967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/07/2024]
Abstract
Since its initial documentation by Knud Krabbe in 1916, numerous studies have scrutinized the characteristics of Krabbe disease (KD) until the identification of the mutation in the GALC gene. In alignment with that, we investigated the natural history of KD spanning eight decades to gain a deeper understanding of the evolutionary trajectory of its mechanisms. Through our comprehensive analysis, we unearthed additional novel elements in molecular biology involving the micropathological mechanism of the disease. This review offers an updated perspective on the metabolic disorder that defines KD. Recently, extracellular vesicles (EVs), autophagy impairment, and α-synuclein have emerged as pivotal players in the neuropathological processes. EVs might serve as a cellular mechanism to avoid or alleviate the detrimental impacts of excessive toxic psychosine levels, and extracting EVs could contribute to synapse dysfunction. Autophagy impairment was found to be independent of psychosine and reliant on AKT and B-cell lymphoma 2. Additionally, α-synuclein has been recognized for inducing cellular death and dysfunction in common biological pathways. Our objective is to assess the effectiveness of advanced therapies in addressing this particular condition. While hematopoietic stem cells have been a primary treatment, its administration proves challenging, particularly in the presymptomatic phase. In this review, we have compiled information from over 10 therapy trials, comparing them based on their benefits and disadvantage.
Collapse
Affiliation(s)
- Imen Ketata
- Neurology Department, University Hospital of Gabes, Gabes, Tunisia
- Sfax University, Sfax Faculty of Medicine, Sfax, Tunisia
| | - Emna Ellouz
- Neurology Department, University Hospital of Gabes, Gabes, Tunisia
- Sfax University, Sfax Faculty of Medicine, Sfax, Tunisia
| |
Collapse
|
2
|
Evans LMP, Gawron J, Sim FJ, Feltri ML, Marziali LN. Human iPSC-derived myelinating organoids and globoid cells to study Krabbe Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.604372. [PMID: 39091729 PMCID: PMC11291050 DOI: 10.1101/2024.07.19.604372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Krabbe disease (Kd) is a lysosomal storage disorder (LSD) caused by the deficiency of the lysosomal galactosylceramidase (GALC) which cleaves the myelin enriched lipid galactosylceramide (GalCer). Accumulated GalCer is catabolized into the cytotoxic lipid psychosine that causes myelinating cells death and demyelination which recruits microglia/macrophages that fail to digest myelin debris and become globoid cells. Here, to understand the pathological mechanisms of Kd, we used induced pluripotent stem cells (iPSCs) from Kd patients to produce myelinating organoids and microglia. We show that Kd organoids have no obvious defects in neurogenesis, astrogenesis, and oligodendrogenesis but manifest early myelination defects. Specifically, Kd organoids showed shorter but a similar number of myelin internodes than Controls at the peak of myelination and a reduced number and shorter internodes at a later time point. Interestingly, myelin is affected in the absence of autophagy and mTOR pathway dysregulation, suggesting lack of lysosomal dysfunction which makes this organoid model a very valuable tool to study the early events that drive demyelination in Kd. Kd iPSC-derived microglia show a marginal rate of globoid cell formation under normal culture conditions that is drastically increased upon GalCer feeding. Under normal culture conditions, Kd microglia show a minor LAMP1 content decrease and a slight increase in the autophagy protein LC3B. Upon GalCer feeding, Kd cells show accumulation of autophagy proteins and strong LAMP1 reduction that at a later time point are reverted showing the compensatory capabilities of globoid cells. Altogether, this supports the value of our cultures as tools to study the mechanisms that drive globoid cell formation and the compensatory mechanism in play to overcome GalCer accumulation in Kd.
Collapse
Affiliation(s)
- Lisa Marie P Evans
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Joseph Gawron
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Fraser J Sim
- Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - M Laura Feltri
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
- Università degli studi di Milano, Biometra department and IRCcs Carlo Besta, Milano 20133, Italy
| | - Leandro N Marziali
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
3
|
Bradbury AM, Bagel J, Swain G, Miyadera K, Pesayco JP, Assenmacher CA, Brisson B, Hendricks I, Wang XH, Herbst Z, Pyne N, Odonnell P, Shelton GD, Gelb M, Hackett N, Szabolcs P, Vite CH, Escolar M. Combination HSCT and intravenous AAV-mediated gene therapy in a canine model proves pivotal for translation of Krabbe disease therapy. Mol Ther 2024; 32:44-58. [PMID: 37952085 PMCID: PMC10787152 DOI: 10.1016/j.ymthe.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/28/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is the only approved treatment for presymptomatic infantile globoid cell leukodystrophy (GLD [Krabbe disease]). However, correction of disease is not complete, and outcomes remain poor. Herein we evaluated HSCT, intravenous (IV) adeno-associated virus rh10 vector (AAVrh10) gene therapy, and combination HSCT + IV AAVrh10 in the canine model of GLD. While HSCT alone resulted in no increase in survival as compared with untreated GLD dogs (∼16 weeks of age), combination HSCT + IV AAVrh10 at a dose of 4E13 genome copies (gc)/kg resulted in delayed disease progression and increased survival beyond 1 year of age. A 5-fold increase in AAVrh10 dose to 2E14 gc/kg, in combination with HSCT, normalized neurological dysfunction up to 2 years of age. IV AAVrh10 alone resulted in an average survival to 41.2 weeks of age. In the peripheral nervous system, IV AAVrh10 alone or in addition to HSCT normalized nerve conduction velocity, improved ultrastructure, and normalized GALC enzyme activity and psychosine concentration. In the central nervous system, only combination therapy at the highest dose was able to restore galactosylceramidase activity and psychosine concentrations to within the normal range. These data have now guided clinical translation of systemic AAV gene therapy as an addition to HSCT (NCT04693598, NCT05739643).
Collapse
Affiliation(s)
- Allison M Bradbury
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA; Abigail Wexner Research Institute, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH 43215, USA; Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH 43215, USA.
| | - Jessica Bagel
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Gary Swain
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Keiko Miyadera
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Jill P Pesayco
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92161, USA
| | - Charles-Antoine Assenmacher
- Comparative Pathology Core, Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Becky Brisson
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Ian Hendricks
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Xiao H Wang
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Zachary Herbst
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Nettie Pyne
- Abigail Wexner Research Institute, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Patricia Odonnell
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92161, USA
| | - Michael Gelb
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Neil Hackett
- Neil Hackett Consulting, New York, NY 10003, USA
| | - Paul Szabolcs
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92161, USA
| | - Charles H Vite
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Maria Escolar
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15224, USA; Forge Biologics, Grove City, OH 43123, USA
| |
Collapse
|
4
|
Shih HY, Raas Q, Bonkowsky JL. Progress in leukodystrophies with zebrafish. Dev Growth Differ 2024; 66:21-34. [PMID: 38239149 DOI: 10.1111/dgd.12907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/31/2024]
Abstract
Inherited leukodystrophies are genetic disorders characterized by abnormal white matter in the central nervous system. Although individually rare, there are more than 400 distinct types of leukodystrophies with a cumulative incidence of 1 in 4500 live births. The pathophysiology of most leukodystrophies is poorly understood, there are treatments for only a few, and there is significant morbidity and mortality, suggesting a critical need for improvements in this field. A variety of animal, cell, and induced pluripotent stem cell-derived models have been developed for leukodystrophies, but with significant limitations in all models. Many leukodystrophies lack animal models, and extant models often show no or mixed recapitulation of key phenotypes. Zebrafish (Danio rerio) have become increasingly used as disease models for studying leukodystrophies due to their early onset of disease phenotypes and conservation of molecular and neurobiological mechanisms. Here, we focus on reviewing new zebrafish disease models for leukodystrophy or models with recent progress. This includes discussion of leukodystrophy with vanishing white matter disease, X-linked adrenoleukodystrophy, Zellweger spectrum disorders and peroxisomal disorders, PSAP deficiency, metachromatic leukodystrophy, Krabbe disease, hypomyelinating leukodystrophy-8/4H leukodystrophy, Aicardi-Goutières syndrome, RNASET2-deficient cystic leukoencephalopathy, hereditary diffuse leukoencephalopathy with spheroids-1 (CSF1R-related leukoencephalopathy), and ultra-rare leukodystrophies. Zebrafish models offer important potentials for the leukodystrophy field, including testing of new variants in known genes; establishing causation of newly discovered genes; and early lead compound identification for therapies. There are also unrealized opportunities to use humanized zebrafish models which have been sparsely explored.
Collapse
Affiliation(s)
- Hung-Yu Shih
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Department of Biological Sciences, Utah Tech University, Saint George, Utah, USA
- Center for Precision & Functional Genomics, Utah Tech University, Saint George, Utah, USA
| | - Quentin Raas
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, Université de Paris, INSERM UMR 1163, Paris, France
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Center for Personalized Medicine, Primary Children's Hospital, Salt Lake City, Utah, USA
| |
Collapse
|
5
|
Braz SO, Morgado MM, Pereira MI, Monteiro AC, Golonzhka O, Jarpe M, Brites P, Sousa MM, Nogueira-Rodrigues J. HDAC-6 inhibition ameliorates the early neuropathology in a mouse model of Krabbe disease. Front Mol Neurosci 2023; 16:1231659. [PMID: 37588057 PMCID: PMC10426153 DOI: 10.3389/fnmol.2023.1231659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/12/2023] [Indexed: 08/18/2023] Open
Abstract
Introduction In Krabbe disease (KD), mutations in β-galactosylceramidase (GALC), a lysosomal enzyme responsible for the catabolism of galactolipids, leads to the accumulation of its substrates galactocerebroside and psychosine. This neurologic condition is characterized by a severe and progressive demyelination together with neuron-autonomous defects and degeneration. Twitcher mice mimic the infantile form of KD, which is the most common form of the human disease. The Twitcher CNS and PNS present demyelination, axonal loss and neuronal defects including decreased levels of acetylated tubulin, decreased microtubule stability and impaired axonal transport. Methods We tested whether inhibiting the α-tubulin deacetylase HDAC6 with a specific inhibitor, ACY-738, was able to counteract the early neuropathology and neuronal defects of Twitcher mice. Results Our data show that delivery of ACY-738 corrects the low levels of acetylated tubulin in the Twitcher nervous system. Furthermore, it reverts the loss myelinated axons in the sciatic nerve and in the optic nerve when administered from birth to postnatal day 9, suggesting that the drug holds neuroprotective properties. The extended delivery of ACY-738 to Twitcher mice delayed axonal degeneration in the CNS and ameliorated the general presentation of the disease. ACY-738 was effective in rescuing neuronal defects of Twitcher neurons, stabilizing microtubule dynamics and increasing the axonal transport of mitochondria. Discussion Overall, our results support that ACY-738 has a neuroprotective effect in KD and should be considered as an add-on therapy combined with strategies targeting metabolic correction.
Collapse
Affiliation(s)
- Sandra O. Braz
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Marlene M. Morgado
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Marta I. Pereira
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Ana C. Monteiro
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Olga Golonzhka
- Acetylon Pharmaceuticals Inc., Boston, MA, United States
| | - Matthew Jarpe
- Acetylon Pharmaceuticals Inc., Boston, MA, United States
| | - Pedro Brites
- NeuroLipid Biology Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Monica M. Sousa
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Joana Nogueira-Rodrigues
- Nerve Regeneration Group, Instituto de Biologia Molecular e Celular (IBMC), Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| |
Collapse
|
6
|
Herdt AR, Peng H, Dickson DW, Golde TE, Eckman EA, Lee CW. Brain Targeted AAV1-GALC Gene Therapy Reduces Psychosine and Extends Lifespan in a Mouse Model of Krabbe Disease. Genes (Basel) 2023; 14:1517. [PMID: 37628569 PMCID: PMC10454254 DOI: 10.3390/genes14081517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Krabbe disease (KD) is a progressive and devasting neurological disorder that leads to the toxic accumulation of psychosine in the white matter of the central nervous system (CNS). The condition is inherited via biallelic, loss-of-function mutations in the galactosylceramidase (GALC) gene. To rescue GALC gene function in the CNS of the twitcher mouse model of KD, an adeno-associated virus serotype 1 vector expressing murine GALC under control of a chicken β-actin promoter (AAV1-GALC) was administered to newborn mice by unilateral intracerebroventricular injection. AAV1-GALC treatment significantly improved body weight gain and survival of the twitcher mice (n = 8) when compared with untreated controls (n = 5). The maximum weight gain after postnatal day 10 was significantly increased from 81% to 217%. The median lifespan was extended from 43 days to 78 days (range: 74-88 days) in the AAV1-GALC-treated group. Widespread expression of GALC protein and alleviation of KD neuropathology were detected in the CNS of the treated mice when examined at the moribund stage. Functionally, elevated levels of psychosine were completely normalized in the forebrain region of the treated mice. In the posterior region, which includes the mid- and the hindbrain, psychosine was reduced by an average of 77% (range: 53-93%) compared to the controls. Notably, psychosine levels in this region were inversely correlated with body weight and lifespan of AAV1-GALC-treated mice, suggesting that the degree of viral transduction of posterior brain regions following ventricular injection determined treatment efficacy on growth and survivability, respectively. Overall, our results suggest that viral vector delivery via the cerebroventricular system can partially correct psychosine accumulation in brain that leads to slower disease progression in KD.
Collapse
Affiliation(s)
- Aimee R. Herdt
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Hui Peng
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Todd E. Golde
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
- Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Elizabeth A. Eckman
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Chris W. Lee
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| |
Collapse
|
7
|
Wang R, Qin Z, Huang L, Luo H, Peng H, Zhou X, Zhao Z, Liu M, Yang P, Shi T. SMPD1 expression profile and mutation landscape help decipher genotype-phenotype association and precision diagnosis for acid sphingomyelinase deficiency. Hereditas 2023; 160:11. [PMID: 36907956 PMCID: PMC10009935 DOI: 10.1186/s41065-023-00272-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Acid sphingomyelinase deficiency (ASMD) disorder, also known as Niemann-Pick disease (NPD) is a rare genetic disease caused by mutations in SMPD1 gene, which encodes sphingomyelin phosphodiesterase (ASM). Except for liver and spleen enlargement and lung disease, two subtypes (Type A and B) of NDP have different onset times, survival times, ASM activities, and neurological abnormalities. To comprehensively explore NPD's genotype-phenotype association and pathophysiological characteristics, we collected 144 NPD cases with strict quality control through literature mining. RESULTS The difference in ASM activity can differentiate NPD type A from other subtypes, with the ratio of ASM activity to the reference values being lower in type A (threshold 0.045 (4.45%)). Severe variations, such as deletion and insertion, can cause complete loss of ASM function, leading to type A, whereas relatively mild missense mutations generally result in type B. Among reported mutations, the p.Arg3AlafsX76 mutation is highly prevalent in the Chinese population, and the p.R608del mutation is common in Mediterranean countries. The expression profiles of SMPD1 from GTEx and single-cell RNA sequencing data of multiple fetal tissues showed that high expressions of SMPD1 can be observed in the liver, spleen, and brain tissues of adults and hepatoblasts, hematopoietic stem cells, STC2_TLX1-positive cells, mesothelial cells of the spleen, vascular endothelial cells of the cerebellum and the cerebrum of fetuses, indicating that SMPD1 dysfunction is highly likely to have a significant effect on the function of those cell types during development and the clinicians need pay attention to these organs or tissues as well during diagnosis. In addition, we also predicted 21 new pathogenic mutations in the SMPD1 gene that potentially cause the NPD, signifying that more rare cases will be detected with those mutations in SMPD1. Finally, we also analysed the function of the NPD type A cells following the extracellular milieu. CONCLUSIONS Our study is the first to elucidate the effects of SMPD1 mutation on cell types and at the tissue level, which provides new insights into the genotype-phenotype association and can help in the precise diagnosis of NPD.
Collapse
Affiliation(s)
- Ruisong Wang
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
- Affiliated Hospital of Hunan University of Arts and Science (the Maternal and Child Health Hospital), Medical college, 3150 Dongting Ave., Changde, Hunan Province, People's Republic of China, 415000
| | - Ziyi Qin
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
| | - Long Huang
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
| | - Huiling Luo
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
| | - Han Peng
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
| | - Xinyu Zhou
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
| | - Zhixiang Zhao
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
| | - Mingyao Liu
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
- Changde Research Centre for Artificial Intelligence and Biomedicine, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China
| | - Pinhong Yang
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China.
- Changde Research Centre for Artificial Intelligence and Biomedicine, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China.
| | - Tieliu Shi
- College of Life and Environmental Sciences, Hunan University of Arts and Science, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China.
- Changde Research Centre for Artificial Intelligence and Biomedicine, 3150 Dongting Ave., Changde, 415000, Hunan Province, People's Republic of China.
| |
Collapse
|