1
|
Pandey MK. Exploring Pro-Inflammatory Immunological Mediators: Unraveling the Mechanisms of Neuroinflammation in Lysosomal Storage Diseases. Biomedicines 2023; 11:biomedicines11041067. [PMID: 37189685 DOI: 10.3390/biomedicines11041067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/28/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Lysosomal storage diseases are a group of rare and ultra-rare genetic disorders caused by defects in specific genes that result in the accumulation of toxic substances in the lysosome. This excess accumulation of such cellular materials stimulates the activation of immune and neurological cells, leading to neuroinflammation and neurodegeneration in the central and peripheral nervous systems. Examples of lysosomal storage diseases include Gaucher, Fabry, Tay–Sachs, Sandhoff, and Wolman diseases. These diseases are characterized by the accumulation of various substrates, such as glucosylceramide, globotriaosylceramide, ganglioside GM2, sphingomyelin, ceramide, and triglycerides, in the affected cells. The resulting pro-inflammatory environment leads to the generation of pro-inflammatory cytokines, chemokines, growth factors, and several components of complement cascades, which contribute to the progressive neurodegeneration seen in these diseases. In this study, we provide an overview of the genetic defects associated with lysosomal storage diseases and their impact on the induction of neuro-immune inflammation. By understanding the underlying mechanisms behind these diseases, we aim to provide new insights into potential biomarkers and therapeutic targets for monitoring and managing the severity of these diseases. In conclusion, lysosomal storage diseases present a complex challenge for patients and clinicians, but this study offers a comprehensive overview of the impact of these diseases on the central and peripheral nervous systems and provides a foundation for further research into potential treatments.
Collapse
Affiliation(s)
- Manoj Kumar Pandey
- Cincinnati Children’s Hospital Medical Center, Division of Human Genetics, Cincinnati, OH 45229-3026, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0515, USA
| |
Collapse
|
2
|
Glycosphingolipid GM3 prevents albuminuria and podocytopathy induced by anti-nephrin antibody. Sci Rep 2022; 12:16058. [PMID: 36163359 PMCID: PMC9513075 DOI: 10.1038/s41598-022-20265-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/23/2022] [Accepted: 09/12/2022] [Indexed: 11/18/2022] Open
Abstract
Podocytopathy, which is characterized by injury to podocytes, frequently causes proteinuria or nephrotic syndrome. There is currently a paucity of effective therapeutic drugs to treat proteinuric kidney disease. Recent research suggests the possibility that glycosphingolipid GM3 maintains podocyte function by acting on various molecules including nephrin, but its mechanism of action remains unknown. Here, various analyses were performed to examine the potential relationship between GM3 and nephrin, and the function of GM3 in podocytes using podocytopathy mice, GM3 synthase gene knockout mice, and nephrin injury cells. Reduced amounts of GM3 and nephrin were observed in podocytopathy mice. Intriguingly, this reduction of GM3 and nephrin, as well as albuminuria, were inhibited by administration of valproic acid. However, when the same experiment was performed using GM3 synthase gene knockout mice, valproic acid administration did not inhibit albuminuria. Equivalent results were obtained in model cells. These findings indicate that GM3 acts with nephrin in a collaborative manner in the cell membrane. Taken together, elevated levels of GM3 stabilize nephrin, which is a key molecule of the slit diaphragm, by enhancing the environment of the cell membrane and preventing albuminuria. This study provides novel insight into new drug discovery, which may offer a new therapy for kidney disease with albuminuria.
Collapse
|
3
|
Alteration of the neuronal and glial cell profiles in Neu1-deficient zebrafish. Glycoconj J 2022; 39:499-512. [PMID: 35877057 DOI: 10.1007/s10719-022-10074-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/27/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/04/2022]
Abstract
Neu1 is a glycosidase that releases sialic acids from the non-reducing ends of glycoconjugates, and its enzymatic properties are conserved among vertebrates. Recently, Neu1-KO zebrafish were generated using genome editing technology, and the KO fish showed abnormal emotional behavior, such as low schooling, low aggressiveness, and excess exploratory behavior, accompanied by the downregulation of anxiety-related genes. To examine the alteration of neuronal and glial cells in Neu1-KO zebrafish, we analyzed the molecular profiles in the zebrafish brain, focusing on the midbrain and telencephalon. Using immunohistochemistry, we found that signals of Maackia amurensis (MAM) lectin that recognizes Sia α2-3 linked glycoconjugates were highly increased in Neu1-KO zebrafish brains, accompanied by an increase in Lamp1a. Neu1-KO zebrafish suppressed the gene expression of AMPA-type glutamate receptors such as gria1a, gria2a, and gria3b, and vesicular glutamate transporter 1. Additionally, Neu1-KO zebrafish induced the hyperactivation of astrocytes accompanied by an increase in Gfap and phosphorylated ERK levels, while the mRNA levels of astrocyte glutamate transporters (eaat1a, eaat1c, and eaat2) were downregulated. The mRNA levels of sypb and ho1b, which are markers of synaptic plasticity, were also suppressed by Neu1 deficiency. Abnormal activity of microglia was also revealed by IHC, and the expressions of iNOS and IL-1β, an inflammatory cytokine, were increased in Neu1-KO zebrafish. Furthermore, drastic neuronal degeneration was detected in Neu1-KO zebrafish using Fluoro-Jade B staining. Collectively, the neuronal and glial abnormalities in Neu1-KO zebrafish may be caused by changes in the excitatory neurotransmitter glutamate and involved in the emotional abnormalities.
Collapse
|
4
|
Kuil LE, López Martí A, Carreras Mascaro A, van den Bosch JC, van den Berg P, van der Linde HC, Schoonderwoerd K, Ruijter GJG, van Ham TJ. Hexb enzyme deficiency leads to lysosomal abnormalities in radial glia and microglia in zebrafish brain development. Glia 2019; 67:1705-1718. [PMID: 31140649 PMCID: PMC6772114 DOI: 10.1002/glia.23641] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/15/2019] [Revised: 04/12/2019] [Accepted: 05/10/2019] [Indexed: 12/11/2022]
Abstract
Sphingolipidoses are severe, mostly infantile lysosomal storage disorders (LSDs) caused by defective glycosphingolipid degradation. Two of these sphingolipidoses, Tay Sachs and Sandhoff diseases, are caused by β-Hexosaminidase (HEXB) enzyme deficiency, resulting in ganglioside (GM2) accumulation and neuronal loss. The precise sequence of cellular events preceding, and leading to, neuropathology remains unclear, but likely involves inflammation and lysosomal accumulation of GM2 in multiple cell types. We aimed to determine the consequences of Hexb activity loss for different brain cell types using zebrafish. Hexb deficient zebrafish (hexb-/- ) showed lysosomal abnormalities already early in development both in radial glia, which are the neuronal and glial progenitors, and in microglia. Additionally, at 5 days postfertilization, hexb-/- zebrafish showed reduced locomotor activity. Although specific oligosaccharides accumulate in the adult brain, hexb-/- ) zebrafish are viable and apparently resistant to Hexb deficiency. In all, we identified cellular consequences of loss of Hexb enzyme activity during embryonic brain development, showing early effects on glia, which possibly underlie the behavioral aberrations. Hereby, we identified clues into the contribution of non-neuronal lysosomal abnormalities in LSDs affecting the brain and provide a tool to further study what underlies the relative resistance to Hexb deficiency in vivo.
Collapse
Affiliation(s)
- Laura E. Kuil
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Anna López Martí
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Ana Carreras Mascaro
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Jeroen C. van den Bosch
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Paul van den Berg
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Herma C. van der Linde
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Kees Schoonderwoerd
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - George J. G. Ruijter
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| |
Collapse
|
5
|
Kasitinon SY, Eskiocak U, Martin M, Bezwada D, Khivansara V, Tasdogan A, Zhao Z, Mathews T, Aurora AB, Morrison SJ. TRPML1 Promotes Protein Homeostasis in Melanoma Cells by Negatively Regulating MAPK and mTORC1 Signaling. Cell Rep 2019; 28:2293-2305.e9. [PMID: 31461647 PMCID: PMC6813770 DOI: 10.1016/j.celrep.2019.07.086] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2019] [Revised: 07/01/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022] Open
Abstract
We screen ion channels and transporters throughout the genome to identify those required by human melanoma cells but not by normal human melanocytes. We discover that Mucolipin-1 (MCOLN1), which encodes the lysosomal cation channel TRPML1, is preferentially required for the survival and proliferation of melanoma cells. Loss of MCOLN1/TRPML1 function impairs the growth of patient-derived melanomas in culture and in xenografts but does not affect the growth of human melanocytes. TRPML1 expression and macropinocytosis are elevated in melanoma cells relative to melanocytes. TRPML1 is required in melanoma cells to negatively regulate MAPK pathway and mTORC1 signaling. TRPML1-deficient melanoma cells exhibit decreased survival, proliferation, tumor growth, and macropinocytosis, as well as serine depletion and proteotoxic stress. All of these phenotypes are partially or completely rescued by mTORC1 inhibition. Melanoma cells thus increase TRPML1 expression relative to melanocytes to attenuate MAPK and mTORC1 signaling, to sustain macropinocytosis, and to avoid proteotoxic stress.
Collapse
Affiliation(s)
- Stacy Y Kasitinon
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ugur Eskiocak
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Misty Martin
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Divya Bezwada
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vishal Khivansara
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Alpaslan Tasdogan
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Thomas Mathews
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Arin B Aurora
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sean J Morrison
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
6
|
Ogawa Y, Sasanuma Y, Shitara S, Koshizuka A, Okada R, Sakuraba H, Oishi K. Abnormal organization during neurodevelopment in a mouse model of Sandhoff disease. Neurosci Res 2019; 155:12-19. [PMID: 31340161 DOI: 10.1016/j.neures.2019.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/01/2019] [Revised: 07/16/2019] [Accepted: 07/19/2019] [Indexed: 12/24/2022]
Abstract
Sandhoff disease (SD) is a genetic disorder caused by a mutation of HEXB, which is the β-subunit gene of β-hexosaminidase A and B (HexA and HexB) in humans. HEXB mutation reduces HexA and HexB enzymatic activities, and results in the massive accumulation of ganglioside GM2 in the nervous system. Severe phenotypes of SD show progressive neurodegeneration in human infants, and lysosomal dysfunction that may affect the early development of the nervous system. In a previous study, neural stem cells (NSCs) and induced pluripotent stem cells derived from SD model mice, which are Hexb-deficient (Hexb-/-), demonstrated impaired neuronal differentiation. This study investigated early neurodevelopment in vivo using Hexb-/- mice. The structure of adult cerebral cortices of Hexb-/- mice was normal. However, the expression of Sox2, an NSC-related gene, was reduced in the embryonic cerebral cortices of Hexb-/- mice. Moreover, a reduction of early neuronal migration and differentiation was observed in the embryonic cerebral cortices of Hexb-/- mice. In addition, we showed that the production of layer-specific neurons was delayed in somatosensory cerebral cortices of Hexb-/- mice. These findings suggest that the alterations observed in embryonic Hexb-/- mice may contribute to deficits in neurodevelopment of SD.
Collapse
Affiliation(s)
- Yasuhiro Ogawa
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan.
| | - Yayoi Sasanuma
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Shuhei Shitara
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Asuna Koshizuka
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Rieko Okada
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Hitoshi Sakuraba
- Department of Clinical Genetics, Meiji Pharmaceutical University, Tokyo, Japan
| | - Kazuhiko Oishi
- Department of Pharmacology, Meiji Pharmaceutical University, Tokyo, Japan
| |
Collapse
|
7
|
Seyrantepe V, Demir SA, Timur ZK, Von Gerichten J, Marsching C, Erdemli E, Oztas E, Takahashi K, Yamaguchi K, Ates N, Dönmez Demir B, Dalkara T, Erich K, Hopf C, Sandhoff R, Miyagi T. Murine Sialidase Neu3 facilitates GM2 degradation and bypass in mouse model of Tay-Sachs disease. Exp Neurol 2017; 299:26-41. [PMID: 28974375 DOI: 10.1016/j.expneurol.2017.09.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/13/2017] [Revised: 09/14/2017] [Accepted: 09/28/2017] [Indexed: 12/29/2022]
Abstract
Tay-Sachs disease is a severe lysosomal storage disorder caused by mutations in Hexa, the gene that encodes for the α subunit of lysosomal β-hexosaminidase A (HEXA), which converts GM2 to GM3 ganglioside. Unexpectedly, Hexa-/- mice have a normal lifespan and show no obvious neurological impairment until at least one year of age. These mice catabolize stored GM2 ganglioside using sialidase(s) to remove sialic acid and form the glycolipid GA2, which is further processed by β-hexosaminidase B. Therefore, the presence of the sialidase (s) allows the consequences of the Hexa defect to be bypassed. To determine if the sialidase NEU3 contributes to GM2 ganglioside degradation, we generated a mouse model with combined deficiencies of HEXA and NEU3. The Hexa-/-Neu3-/- mice were healthy at birth, but died at 1.5 to 4.5months of age. Thin-layer chromatography and mass spectrometric analysis of the brains of Hexa-/-Neu3-/- mice revealed the abnormal accumulation of GM2 ganglioside. Histological and immunohistochemical analysis demonstrated cytoplasmic vacuolation in the neurons. Electron microscopic examination of the brain, kidneys and testes revealed pleomorphic inclusions of many small vesicles and complex lamellar structures. The Hexa-/-Neu3-/- mice exhibited progressive neurodegeneration with neuronal loss, Purkinje cell depletion, and astrogliosis. Slow movement, ataxia, and tremors were the prominent neurological abnormalities observed in these mice. Furthermore, radiographs revealed abnormalities in the skeletal bones of the Hexa-/-Neu3-/- mice. Thus, the Hexa-/-Neu3-/- mice mimic the neuropathological and clinical abnormalities of the classical early-onset Tay-Sachs patients, and provide a suitable model for the future pre-clinical testing of potential treatments for this condition.
Collapse
Affiliation(s)
- Volkan Seyrantepe
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey.
| | - Secil Akyildiz Demir
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Zehra Kevser Timur
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Johanna Von Gerichten
- Lipid Pathobiochemistry Group (G131) German Cancer Research Center, INF 280, 69120 Heidelberg, Germany
| | - Christian Marsching
- Lipid Pathobiochemistry Group (G131) German Cancer Research Center, INF 280, 69120 Heidelberg, Germany; Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), Mannheim, Germany; Institute of Medical Technology (IMT) of Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsach-Str.10, 68163 Mannheim, Germany
| | - Esra Erdemli
- Departments of Histology and Embryology, Ankara University, Medical School, 06100, Sihhiye, Ankara, Turkey
| | - Emin Oztas
- Departments of Histology and Embryology, GATA Medical School, 06100 Ankara, Turkey
| | - Kohta Takahashi
- Cell Recognition Study, Tohoku Pharmaceutical University, Sendai, Japan
| | | | - Nurselin Ates
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, 35430 Izmir, Turkey
| | - Buket Dönmez Demir
- Institutes of Neurological Science and Psychiatry, University of Hacettepe, 06100 Ankara, Turkey
| | - Turgay Dalkara
- Institutes of Neurological Science and Psychiatry, University of Hacettepe, 06100 Ankara, Turkey
| | - Katrin Erich
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), Mannheim, Germany; Institute of Medical Technology (IMT) of Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsach-Str.10, 68163 Mannheim, Germany
| | - Carsten Hopf
- Center for Applied Research in Applied Biomedical Mass Spectrometry (ABIMAS), Mannheim, Germany; Institute of Medical Technology (IMT) of Heidelberg University and Mannheim University of Applied Sciences, Paul-Wittsach-Str.10, 68163 Mannheim, Germany
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group (G131) German Cancer Research Center, INF 280, 69120 Heidelberg, Germany
| | - Taeko Miyagi
- Miyagi Cancer Center Research Institute, Natori, Japan
| |
Collapse
|
8
|
Hooper AWM, Alamilla JF, Venier RE, Gillespie DC, Igdoura SA. Neuronal pentraxin 1 depletion delays neurodegeneration and extends life in Sandhoff disease mice. Hum Mol Genet 2017; 26:661-673. [PMID: 28007910 DOI: 10.1093/hmg/ddw422] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/19/2016] [Accepted: 12/09/2016] [Indexed: 01/15/2023] Open
Abstract
GM2 gangliosidoses are a group of lysosomal storage disorders which include Sandhoff disease and Tay-Sachs disease. Dysregulation of glutamate receptors has been recently postulated in the pathology of Sandhoff disease. Glutamate receptor association with neuronal pentraxins 1 and 2, and the neuronal pentraxin receptor facilitates receptor potentiation and synaptic shaping. In this study, we have observed an upregulation of a novel form of neuronal pentraxin 1 (NP1-38) in the brains of a mouse model of Sandhoff disease and Tay-Sachs disease. In order to determine the impact of NP1 on the pathophysiology of Sandhoff disease mouse models, we have generated an Np1-/-Hexb-/- double knockout mouse, and observed extended lifespan, improved righting reflex and enhanced body condition relative to Hexb-/- mice, with no effect on gliosis or apoptotic markers in the CNS. Sandhoff mouse brain slices reveals a reduction in AMPA receptor-mediated currents, and increased variability in total glutamate currents in the CA1 region of the hippocampus; Np1-/-Hexb-/- mice show a correction of this phenotype, suggesting NP1-38 may be interfering with glutamate receptor function. Indeed, some of the psychiatric aspects of Sandhoff and Tay-Sachs disease (particularly late onset) may be attributed to a dysfunctional hippocampal glutamatergic system. Our work highlights a potential role for synaptic proteins, such as NP1 and glutamate receptors in lysosomal storage diseases.
Collapse
Affiliation(s)
| | | | | | | | - Suleiman A Igdoura
- Department of Biology.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
9
|
White EJ, Trigatti BL, Igdoura SA. Suppression of NK and CD8+ T cells reduces astrogliosis but accelerates cerebellar dysfunction and shortens life span in a mouse model of Sandhoff disease. J Neuroimmunol 2017; 306:55-67. [DOI: 10.1016/j.jneuroim.2017.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/02/2016] [Revised: 02/15/2017] [Accepted: 03/06/2017] [Indexed: 01/09/2023]
|
10
|
Hooper AWM, Igdoura SA. Bi-phasic gliosis drives neuropathology in a Sandhoff disease mouse model. J Neuroimmunol 2016; 299:19-27. [PMID: 27725117 DOI: 10.1016/j.jneuroim.2016.08.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/27/2016] [Revised: 08/02/2016] [Accepted: 08/07/2016] [Indexed: 11/20/2022]
Abstract
Microgliosis and astrogliosis are known to be exacerbating factors in the progression of the lysosomal storage disorder Sandhoff disease. We have also found evidence for excitotoxicity via glutamate receptors in Sandhoff disease. To view the interaction of these cascades, we measured cerebellar expression of markers for gliosis, apoptosis, and excitatory synapses over the disease course in a Sandhoff disease mouse model. We observe a 2-stage model, with initial activation of microgliosis as early as 60days of age, followed by a later onset of astrogliosis, caspase-mediated apoptosis, and reduction in GluR1 at approximately 100days of age. These results implicate immune cells as first responders in Sandhoff disease.
Collapse
Affiliation(s)
| | - Suleiman A Igdoura
- Department of Biology, McMaster University, Hamilton, Ont. L8S 4K1, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ont. L8S 4L8, Canada.
| |
Collapse
|
11
|
Onyenwoke RU, Brenman JE. Lysosomal Storage Diseases-Regulating Neurodegeneration. J Exp Neurosci 2016; 9:81-91. [PMID: 27081317 PMCID: PMC4822725 DOI: 10.4137/jen.s25475] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/04/2015] [Revised: 11/11/2015] [Accepted: 11/16/2015] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a complex pathway regulated by numerous signaling events that recycles macromolecules and can be perturbed in lysosomal storage diseases (LSDs). The concept of LSDs, which are characterized by aberrant, excessive storage of cellular material in lysosomes, developed following the discovery of an enzyme deficiency as the cause of Pompe disease in 1963. Great strides have since been made in better understanding the biology of LSDs. Defective lysosomal storage typically occurs in many cell types, but the nervous system, including the central nervous system and peripheral nervous system, is particularly vulnerable to LSDs, being affected in two-thirds of LSDs. This review provides a summary of some of the better characterized LSDs and the pathways affected in these disorders.
Collapse
Affiliation(s)
- Rob U Onyenwoke
- Department of Pharmaceutical Science, Biomanufacturing Research Institute and Technology Enterprise (BRITE), North Carolina Central University, Durham, NC, USA
| | - Jay E Brenman
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.; Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
12
|
Loth MK, Choi J, McGlothan JL, Pletnikov MV, Pomper MG, Guilarte TR. TSPO in a murine model of Sandhoff disease: presymptomatic marker of neurodegeneration and disease pathophysiology. Neurobiol Dis 2015; 85:174-186. [PMID: 26545928 DOI: 10.1016/j.nbd.2015.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/29/2015] [Accepted: 11/02/2015] [Indexed: 10/22/2022] Open
Abstract
Translocator protein (18 kDa), formerly known as the peripheral benzodiazepine receptor (PBR), has been extensively used as a biomarker of active brain disease and neuroinflammation. TSPO expression increases dramatically in glial cells, particularly in microglia and astrocytes, as a result of brain injury, and this phenomenon is a component of the hallmark response of the brain to injury. In this study, we used a mouse model of Sandhoff disease (SD) to assess the longitudinal expression of TSPO as a function of disease progression and its relationship to behavioral and neuropathological endpoints. Focusing on the presymptomatic period of the disease, we used ex vivo [(3)H]DPA-713 quantitative autoradiography and in vivo [(125)I]IodoDPA-713 small animal SPECT imaging to show that brain TSPO levels markedly increase prior to physical and behavioral manifestation of disease. We further show that TSPO upregulation coincides with early neuronal GM2 ganglioside aggregation and is associated with ongoing neurodegeneration and activation of both microglia and astrocytes. In brain regions with increased TSPO levels, there is a differential pattern of glial cell activation with astrocytes being activated earlier than microglia during the progression of disease. Immunofluorescent confocal imaging confirmed that TSPO colocalizes with both microglia and astrocyte markers, but the glial source of the TSPO response differs by brain region and age in SD mice. Notably, TSPO colocalization with the astrocyte marker GFAP was greater than with the microglia marker, Mac-1. Taken together, our findings have significant implications for understanding TSPO glial cell biology and for detecting neurodegeneration prior to clinical expression of disease.
Collapse
Affiliation(s)
- Meredith K Loth
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Judy Choi
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Jennifer L McGlothan
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Mikhail V Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Tomás R Guilarte
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| |
Collapse
|
13
|
Rama Rao KV, Kielian T. Astrocytes and lysosomal storage diseases. Neuroscience 2015; 323:195-206. [PMID: 26037807 DOI: 10.1016/j.neuroscience.2015.05.061] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/14/2015] [Revised: 05/21/2015] [Accepted: 05/22/2015] [Indexed: 12/19/2022]
Abstract
Lysosomal storage diseases (LSDs) encompass a wide range of disorders characterized by inborn errors of lysosomal function. The majority of LSDs result from genetic defects in lysosomal enzymes, although some arise from mutations in lysosomal proteins that lack known enzymatic activity. Neuropathological abnormalities are a feature of several LSDs and when severe, represent an important determinant in disease outcome. Glial dysfunction, particularly in astrocytes, is also observed in numerous LSDs and has been suggested to impact neurodegeneration. This review will discuss the potential role of astrocytes in LSDs and highlight the possibility of targeting glia as a beneficial strategy to counteract the neuropathology associated with LSDs.
Collapse
Affiliation(s)
- K V Rama Rao
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - T Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
14
|
Fukuishi N, Murakami S, Ohno A, Yamanaka N, Matsui N, Fukutsuji K, Yamada S, Itoh K, Akagi M. Does β-hexosaminidase function only as a degranulation indicator in mast cells? The primary role of β-hexosaminidase in mast cell granules. THE JOURNAL OF IMMUNOLOGY 2014; 193:1886-94. [PMID: 25015817 DOI: 10.4049/jimmunol.1302520] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022]
Abstract
β-Hexosaminidase, which is generally present in the lysosome, is essential for glycoprotein metabolism in the maintenance of cell homeostasis. In mast cells (MCs), large amounts of β-hexosaminidase are present in the granules as opposed to the lysosome, and the biological role of MC β-hexosaminidase has yet to be fully elucidated. Therefore, we investigated the biological role of β-hexosaminidase in MC granules. Bone marrow-derived MCs from C57BL/6 (BL/6-BMMC) or β-hexosaminidase gene-deficient (hexb(-/-)-BMMC) mice were transplanted into MC-deficient (WBB6F1/J-Kit(W)/Kit(W-v) [W/W(v)]) mice to generate MC-reconstituted models. In asthma model experiments, no differences were observed in the symptoms of BL/6, W/W(v), BL/6-BMMC-reconstituted W/W(v), or hexb(-/-)-BMMC-reconstituted W/W(v) mice. In Staphylococcus epidermidis experimental infection model experiments, the severity of symptoms and frequency of death were markedly higher in W/W(v) and hexb(-/-)-BMMC-reconstituted W/W(v) mice than in BL/6 and BL/6-BMMC-reconstituted W/W(v) mice. The growth of S. epidermidis in an in vitro study was clearly inhibited by addition of BL/6-BMMC lysate, but not by addition of hexb(-/-)-BMMC lysate. Moreover, suppression of bacterial proliferation was completely recovered when bacteria were incubated with hexb(-/-)-BMMC lysate plus β-hexosaminidase. Transmission electron microscopy indicated that the cell wall of S. epidermidis was heavily degraded following coincubation of bacteria with BL/6-BMMC lysate, but not following coincubation with hexb(-/-)-BMMC lysate. These findings strongly suggest that MC granule β-hexosaminidase is crucial for defense against bacterial invasion, but is not involved in the allergic response. Our results also suggest that the bactericidal mechanism of β-hexosaminidase involves degradation of bacterial cell wall peptidoglycan.
Collapse
Affiliation(s)
- Nobuyuki Fukuishi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan;
| | - Shinya Murakami
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| | - Akane Ohno
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| | - Naoya Yamanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| | - Nobuaki Matsui
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| | - Kenji Fukutsuji
- Department of Microbiology, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan; and
| | - Sakuo Yamada
- Department of Microbiology, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan; and
| | - Kouji Itoh
- Department of Medicinal Biotechnology, Institute of Health Biosciences, University of Tokushima Graduate School, Tokushima, Tokushima 770-8503, Japan
| | - Masaaki Akagi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Tokushima, Tokushima 770-8514, Japan
| |
Collapse
|
15
|
Abstract
Tay-Sachs and Sandhoff diseases (GM2 gangliosidoses) are autosomal recessive lysosomal storage diseases caused by gene mutations in HEXA and HEXB, each encoding human lysosomal β-hexosaminidase α-subunits and β-subunits, respectively. In Tay-Sachs disease, excessive accumulation of GM2 ganglioside (GM2), mainly in the central nervous system, is caused by a deficiency of the HexA isozyme (αβ heterodimer), resulting in progressive neurologic disorders. In Sandhoff disease, combined deficiencies of HexA and HexB (ββ homodimer) cause not only the accumulation of GM2 but also of oligosaccharides carrying terminal N-acetylhexosamine residues (GlcNAc-oligosaccharides), resulting in systemic manifestations including hepatosplenomegaly as well as neurologic symptoms. Hence there is little clinically effective treatment for these GM2 gangliosidoses. Recent studies on the molecular pathogenesis in Sandhoff disease patients and disease model mice have shown the involvement of microglial activation and chemokine induction in neuroinflammation and neurodegeneration in this disease. Experimental and therapeutic approaches, including recombinant enzyme replacement, have been performed using Sandhoff disease model mice, suggesting the future application of novel techniques to treat GM2 gangliosidoses (Hex deficiencies), including Sandhoff disease as well as Tay-Sachs disease. In this study, we isolated astrocytes and microglia from the neonatal brain of Sandhoff disease model mice and demonstrated abnormalities of glial cells. Moreover, we demonstrated the therapeutic effect of an intracerebroventricular administration of novel recombinant human HexA carrying a high content of M6P residue in Sandhoff disease model mice.
Collapse
Affiliation(s)
- Daisuke Tsuji
- Department of Medicinal Biotechnology, Institute of Medicinal Resources, Graduate School of Pharmaceutical Sciences, The University of Tokushima, Tokushima, Japan.
| |
Collapse
|
16
|
Matsuoka K, Tsuji D, Taki T, Itoh K. Thymic involution and corticosterone level in Sandhoff disease model mice: new aspects the pathogenesis of GM2 gangliosidosis. J Inherit Metab Dis 2011; 34:1061-8. [PMID: 21598013 DOI: 10.1007/s10545-011-9316-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 10/29/2010] [Revised: 03/04/2011] [Accepted: 03/08/2011] [Indexed: 11/28/2022]
Abstract
Sandhoff disease (SD) is a lysosomal disease caused by a mutation of the HEXB gene associated with excessive accumulation of GM2 ganglioside (GM2) in lysosomes and neurological manifestations. Production of autoantibodies against the accumulated gangliosides has been reported to be involved in the progressive pathogenesis of GM2 gangliosidosis, although the underlying mechanism has not been fully elucidated. The thymus is the key organ in the acquired immune system including the development of autoantibodies. We showed here that thymic involution and an increase in cell death in the organ occur in SD model mice at a late stage of the pathogenesis. Dramatic increases in the populations of Annexin-V(+) cells and terminal deoxynucletidyl transferase dUTP nick end labeling (TUNEL) (+) cells were observed throughout the thymuses of 15-week old SD mice. Enhanced caspase-3/7 activation, but not that of caspase-1/4, -6 ,-8, or -9, was also demonstrated. Furthermore, the serum level of corticosterone, a potent inducer of apoptosis of thymocytes, was elevated during the same period of apoptosis. Our studies suggested that an increase in endocrine corticosterone may be one of the causes that accelerate the apoptosis of thymocytes leading to thymic involution in GM2 gangliosidosis, and thus can be used as a disease marker for evaluation of the thymic condition and disease progression.
Collapse
Affiliation(s)
- Kazuhiko Matsuoka
- Department of Medicinal Biotechnology, Institute for Medicinal Research, Graduate School of Pharmaceutical Sciences, The University of Tokushima, 1-78 Sho-machi, Tokushima, Tokushima, 770-8505, Japan
| | | | | | | |
Collapse
|
17
|
Whitehead SN, Chan KHN, Gangaraju S, Slinn J, Li J, Hou ST. Imaging mass spectrometry detection of gangliosides species in the mouse brain following transient focal cerebral ischemia and long-term recovery. PLoS One 2011; 6:e20808. [PMID: 21687673 PMCID: PMC3110773 DOI: 10.1371/journal.pone.0020808] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/26/2011] [Accepted: 05/10/2011] [Indexed: 11/18/2022] Open
Abstract
Gangliosides, a member of the glycosphingolipid family, are heterogeneously expressed in biological membranes and are particularly enriched within the central nervous system. Gangliosides consist of mono- or poly-sialylated oligosaccharide chains of variable lengths attached to a ceramide unit and are found to be intimately involved in brain disease development. The purpose of this study is to examine the spatial profile of ganglioside species using matrix-assisted laser desorption/ionization (MALDI) imaging (IMS) following middle cerebral artery occlusion (MCAO) reperfusion injury in the mouse. IMS is a powerful method to not only discriminate gangliosides by their oligosaccharide components, but also by their carbon length within their sphingosine base. Mice were subjected to a 30 min unilateral MCAO followed by long-term survival (up to 28 days of reperfusion). Brain sections were sprayed with the matrix 5-Chloro-2-mercaptobenzothiazole, scanned and analyzed for a series of ganglioside molecules using an Applied Biosystems 4800 MALDI TOF/TOF. Traditional histological and immunofluorescence techniques were performed to assess brain tissue damage and verification of the expression of gangliosides of interest. Results revealed a unique anatomical profile of GM1, GD1 and GT1b (d18∶1, d20∶1 as well as other members of the glycosphingolipid family). There was marked variability in the ratio of expression between ipsilateral and contralateral cortices for the various detected ganglioside species following MCAO-reperfusion injury. Most interestingly, MCAO resulted in the transient induction of both GM2 and GM3 signals within the ipsilateral hemisphere; at the border of the infarcted tissue. Taken together, the data suggest that brain region specific expression of gangliosides, particularly with respect to hydrocarbon length, may play a role in neuronal responses to injury.
Collapse
Affiliation(s)
- Shawn N. Whitehead
- Experimental NeuroTherapeutics Laboratory, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada
- * E-mail: (SNW); (STH)
| | - Kenneth H. N. Chan
- Mass Spectrometry Glycoanalysis Laboratory, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Sandhya Gangaraju
- Experimental NeuroTherapeutics Laboratory, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Jacqueline Slinn
- Experimental NeuroTherapeutics Laboratory, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Jianjun Li
- Mass Spectrometry Glycoanalysis Laboratory, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada
| | - Sheng T. Hou
- Experimental NeuroTherapeutics Laboratory, Institute for Biological Sciences, National Research Council of Canada, Ottawa, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
- * E-mail: (SNW); (STH)
| |
Collapse
|
18
|
He D, Luo X, Wei W, Xie M, Wang W, Yu Z. DCPIB, A Specific Inhibitor of Volume-Regulated Anion Channels (VRACs), Inhibits Astrocyte Proliferation and Cell Cycle Progression Via G1/S Arrest. J Mol Neurosci 2011; 46:249-57. [DOI: 10.1007/s12031-011-9524-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/18/2011] [Accepted: 04/18/2011] [Indexed: 10/18/2022]
|
19
|
Porter BF, Lewis BC, Edwards JF, Alroy J, Zeng BJ, Torres PA, Bretzlaff KN, Kolodny EH. Pathology of GM2 gangliosidosis in Jacob sheep. Vet Pathol 2010; 48:807-13. [PMID: 21123862 DOI: 10.1177/0300985810388522] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
The G(M2) gangliosidoses are a group of lysosomal storage diseases caused by defects in the genes coding for the enzyme hexosaminidase or the G(M2) activator protein. Four Jacob sheep from the same farm were examined over a 3-year period for a progressive neurologic disease. Two lambs were 6-month-old intact males and 2 were 8-month-old females. Clinical findings included ataxia in all 4 limbs, proprioceptive deficits, and cortical blindness. At necropsy, the nervous system appeared grossly normal. Histologically, most neurons within the brain, spinal cord, and peripheral ganglia were enlarged, and the cytoplasm was distended by foamy to granular material that stained positively with Luxol fast blue and Sudan black B stains. Other neuropathologic findings included widespread astrocytosis, microgliosis, and scattered spheroids. Electron microscopy revealed membranous cytoplasmic bodies within the cytoplasm of neurons. Biochemical and molecular genetic studies confirmed the diagnosis of G(M2) gangliosidosis. This form of G(M2) gangliosidosis in Jacob sheep is very similar to human Tay-Sachs disease and is potentially a useful animal model.
Collapse
Affiliation(s)
- B F Porter
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4467, USA.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Hemsley KM, Hopwood JJ. Lessons learnt from animal models: pathophysiology of neuropathic lysosomal storage disorders. J Inherit Metab Dis 2010; 33:363-71. [PMID: 20449662 DOI: 10.1007/s10545-010-9078-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 09/14/2009] [Revised: 03/09/2010] [Accepted: 03/16/2010] [Indexed: 11/29/2022]
Abstract
Approximately 50 inborn errors of metabolism known as lysosomal storage disorders have been discovered to date, most of which are due to a single mutation in a gene encoding a soluble lysosomal enzyme. Consequently, inadequate enzyme activity results in the accumulation of substrates for that enzyme, invariably accompanied by a wide variety of secondary pathological changes. Many of these conditions remain untreatable, and therefore, research into pathogenic processes and potential treatment strategies is intense. A key tool for researchers in this area is the availability of clinically relevant animal models in which to study disease manifestation and evaluate therapeutic outcomes. Large numbers of both naturally occurring and genetically modified animal models of neurodegenerative lysosomal storage disorders are in existence, with spontaneous models occurring in both large domestic (e.g., cat, dog, sheep) and small (e.g., mouse) animal species. Many have undergone rigorous phenotypic characterization and are now providing us with insights into neurological disease processes. The purpose of this review is to highlight some of the major lessons learnt from these studies.
Collapse
Affiliation(s)
- Kim M Hemsley
- Lysosomal Diseases Research Unit, 4th Floor Rogerson Building, Women's and Children's Hospital, 72 King William Road, North Adelaide, SA, 5006, Australia.
| | | |
Collapse
|
21
|
Kato Y, Kuan CT, Chang J, Kaneko MK, Ayriss J, Piao H, Chandramohan V, Pegram C, McLendon RE, Fredman P, Månsson JE, Bigner DD. GMab-1, a high-affinity anti-3'-isoLM1/3',6'-isoLD1 IgG monoclonal antibody, raised in lacto-series ganglioside-defective knockout mice. Biochem Biophys Res Commun 2009; 391:750-5. [PMID: 19944071 DOI: 10.1016/j.bbrc.2009.11.132] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/16/2009] [Accepted: 11/19/2009] [Indexed: 11/16/2022]
Abstract
The lacto-series gangliosides 3'-isoLM1 and 3',6'-isoLD1 have been identified as tumor-associated antigens whose formation is initiated by the Lc3-synthase. Until now, high-affinity IgG monoclonal antibodies (mAbs) against 3'-isoLM1 and 3',6'-isoLD1, which are highly expressed in gliomas, have not been developed, although mAbs against lacto-series gangliosides are powerful tools for functional studies. We previously produced the Lc3-synthase gene beta3Gn-T5 knockout mice. In this study, we immunized beta3Gn-T5 knockout mice with 3'-isoLM1/3',6'-isoLD1 and produced the anti-3'-isoLM1/3',6'-isoLD1 mAb GMab-1, of the IgG(3) subclass, which should be useful for functional analysis of lacto-series gangliosides and for antibody-based therapy of gliomas.
Collapse
Affiliation(s)
- Yukinari Kato
- Department of Pathology, Duke University Medical Center, DUMC-3156, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|