mucolipidosis type iv
Recently Published Documents


TOTAL DOCUMENTS

106
(FIVE YEARS 13)

H-INDEX

30
(FIVE YEARS 2)

2021 ◽  
Author(s):  
Taylor F. Minckley ◽  
Anna M. Dischler ◽  
Dylan H. Fudge ◽  
Ebrahim Ghazvini Zadeh ◽  
Wen-hong Li ◽  
...  

AbstractAccurate delivery of cargo over long distances through axonal transport requires precise spatiotemporal regulation. Here we discover that Zn2+, either released from lysosomes through TRPML1 or influx via depolarization, inhibits axonal transport. Zn2+-mediated inhibition is neither selective for cargo nor for cell type because elevated Zn2+ (IC50 ≈ 5 nM) reduces both lysosomal and mitochondrial motility in primary rat hippocampal neurons and HeLa cells. We further reveal that Zn2+ directly binds to microtubules and inhibits movement of kinesin motors. Loss of TRPML1 function, which causes Mucolipidosis Type IV (MLIV) disease, impairs lysosomal Zn2+ release, disrupts Zn2+-mediated regulation of axonal transport, and increases overall organellar motility. In addition, MLIV patient mutations in TRPML1 have decreased Zn2+ permeability, which parallels disease severity. Our results reveal that Zn2+ acts as a critical signal to locally pause axonal transport by directly blocking the progression of motor proteins on microtubules.Significance StatementDisruptions in proper axonal transport have been linked to neurodevelopmental and neurodegenerative diseases. Here we discover that activation of the lysosomal channel TRPML1 arrests lysosomal trafficking. Such lysosome self-regulation mechanism is mediated via TRPML1-mediated Zn2+, not Ca2+. We further reveal that Zn2+ acts as a critical brake signal to pause axonal transport locally by directly decorating microtubules and blocking the movement of motor proteins. Dysfunction of TRPML1, the genetic cause of Mucolipidosis type IV (MLIV), blocks lysosomal Zn2+ release, causing loss of fine-tuning of lysosomal motility. Overall, this study implicates the importance of Zn2+ signals and axonal transport in the pathology of MLIV and reveals new signaling roles for Zn2+ in regulating cell processes involved with microtubule-based transport.


2021 ◽  
Vol 132 ◽  
pp. S122
Author(s):  
David Gibson ◽  
Natario Couser ◽  
Vikram Brar ◽  
Rachel Li ◽  
Arun Kalra ◽  
...  

Ophthalmology ◽  
2021 ◽  
Vol 128 (4) ◽  
pp. 521
Author(s):  
Gilles C. Martin ◽  
Antoinette Bernabe-Gelot ◽  
Eric Gabison

2021 ◽  
Author(s):  
Andrea Hadjikyriacou ◽  
Sangeetha Iyer ◽  
Joshua D. Mast ◽  
Nina DiPrimio ◽  
John Concannon ◽  
...  

AbstractInvertebrate model organisms (the nematode Caenorhabditis elegans and the fruit fly Drosophila melanogaster) are valuable tools to bridge the gap between traditional in vitro discovery and preclinical animal models. Invertebrate model organisms are poised to serve as better disease models than 2D cellular monocultures for drug discovery, as well as easier and more cost-effective to scale up than 3D organoids/assembloids or co-cultures. A strength of model organisms is the opportunity to probe conserved biology such as lysosomal function and autophagy in a physiological setting. However, invertebrate models are not without pharmacokinetic and pharmacodynamic challenges, such as poor tissue penetration and confidence in a compound’s mechanism of action. To confront those challenges, we took advantage of the Novartis mechanism-of-action box (MoA Box), a compound library of well-annotated and drug-like chemical probes. Curious as to how the MoA Box, comprised of chemical probes optimized for mammalian targets, would fare in an invertebrate setting we screened the MoA Box across three different models of the lysosomal storage disease mucolipidosis Type IV (MLIV). MLIV is caused by mutations in the lysosomal transient receptor potential ion channel mucolipin-1 (TRPML1) resulting in hyper-acidic lysosomes and dysregulated autophagy. The overlap of screening hits from worm, fly, and patient fibroblast screens identified cyclin-dependent kinase (CDK) inhibition as an evolutionarily conserved disease modifier and potential drug repurposing strategy.Summary statementA trio of phenotypic screens across Drosophila, C. elegans, and H. sapiens models of mucolipidosis IV was performed and identified overlapping hits including cyclin-dependent kinase inhibitors.


Author(s):  
Cristina Bostan ◽  
Grant Mitchell ◽  
Benjamin Ellezam ◽  
Jean-François Soucy ◽  
Mona Harissi-Dagher ◽  
...  

2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Ayelet Vardi ◽  
Amir Pri-Or ◽  
Noa Wigoda ◽  
Yulia Grishchuk ◽  
Anthony H. Futerman

Abstract Background Mucolipidosis type IV (MLIV), an ultra-rare neurodevelopmental and neurodegenerative disorder, is caused by mutations in the MCOLN1 gene, which encodes the late endosomal/lysosomal transient receptor potential channel TRPML1 (mucolipin 1). The precise pathophysiogical pathways that cause neurological disease in MLIV are poorly understood. Recently, the first post-mortem brain sample became available from a single MLIV patient, and in the current study we performed mass spectrometry (MS)-based proteomics on this tissue with a view to delineating pathological pathways, and to compare with previously-published data on MLIV, including studies using the Mcoln1−/− mouse. Results A number of pathways were altered in two brain regions from the MLIV patient, including those related to the lysosome, lipid metabolism, myelination, cellular trafficking and autophagy, mTOR and calmodulin, the complement system and interferon signaling. Of these, levels of some proteins not known previously to be associated with MLIV were altered, including APOD, PLIN4, ATG and proteins related to interferon signaling. Moreover, when proteins detected by proteomics in the human brain were compared with their orthologs detected in the Mcoln1−/− mouse by RNAseq, the results were remarkably similar. Finally, analysis of proteins in human and mouse CSF suggest that calbindin 1 and calbindin 2 might be useful as biomarkers to help chart the course of disease development. Conclusions Despite the sample size limitations, our findings are consistent with the relatively general changes in lysosomal function previously reported in MLIV, and shed light on new pathways of disease pathophysiology, which is required in order to understand the course of disease development and to determine the efficacy of therapies when they become available for this devastating disease.


2020 ◽  
Vol 63 (7) ◽  
pp. 103927 ◽  
Author(s):  
Ben Pode-Shakked ◽  
Yael Finezilber ◽  
Yonit Levi ◽  
Shiri Liber ◽  
Nicole Fleischer ◽  
...  

2020 ◽  
Vol 13 (7) ◽  
pp. dmm044230 ◽  
Author(s):  
Molly Mepyans ◽  
Livia Andrzejczuk ◽  
Jahree Sosa ◽  
Sierra Smith ◽  
Shawn Herron ◽  
...  

ABSTRACTMucolipidosis type IV (MLIV) is a lysosomal disease caused by mutations in the MCOLN1 gene that encodes the endolysosomal transient receptor potential channel mucolipin-1, or TRPML1. MLIV results in developmental delay, motor and cognitive impairments, and vision loss. Brain abnormalities include thinning and malformation of the corpus callosum, white-matter abnormalities, accumulation of undegraded intracellular ‘storage’ material and cerebellar atrophy in older patients. Identification of the early events in the MLIV course is key to understanding the disease and deploying therapies. The Mcoln1−/− mouse model reproduces all major aspects of the human disease. We have previously reported hypomyelination in the MLIV mouse brain. Here, we investigated the onset of hypomyelination and compared oligodendrocyte maturation between the cortex/forebrain and cerebellum. We found significant delays in expression of mature oligodendrocyte markers Mag, Mbp and Mobp in the Mcoln1−/− cortex, manifesting as early as 10 days after birth and persisting later in life. Such delays were less pronounced in the cerebellum. Despite our previous finding of diminished accumulation of the ferritin-bound iron in the Mcoln1−/− brain, we report no significant changes in expression of the cytosolic iron reporters, suggesting that iron-handling deficits in MLIV occur in the lysosomes and do not involve broad iron deficiency. These data demonstrate very early deficits of oligodendrocyte maturation and critical regional differences in myelination between the forebrain and cerebellum in the mouse model of MLIV. Furthermore, they establish quantitative readouts of the MLIV impact on early brain development, useful to gauge efficacy in pre-clinical trials.


Sign in / Sign up

Export Citation Format

Share Document