Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Microbiol Biol Educ ; 25(1): e0006723, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38661421

RESUMEN

Reading, presenting, and discussing peer-reviewed scientific reports, case studies, and reviews are essential to modern biology education. These exercises model crucial aspects of students' future professional activities and introduce the students to the current scientific concepts and methodology, data analysis, and presentation. A common format for working with primary literature is a journal club: presenting and discussing research literature in front of peers, which has many merits. However, in large modern classrooms, this format is very time-consuming and stressful, especially since presenting is not a commonly taught skill. We argue that student groups for whom the current educational and professional paradigms present a challenge due to a historical lack of representation or wellness issues are deprived of a key educational opportunity. To solve this problem, we formulated an approach called Peer-Reviewed Presentation Exchange (PRPE), which focuses on collaborative analysis, presentation, and review of research literature that includes (i) voice-narrated research presentations by students, (ii) checklists generated by the instructor to establish expectations for an informative presentation or review, and (iii) presentation assignment and peer review process. We tested this approach in an undergraduate cell biology class over 3 years. Pre- and post-assessments show significant gains in self-efficacy and knowledge not only by students who presented but also by the students who reviewed the presentations; therefore, peer-reviewed presentations are an effective tool for learning. Exit surveys show that the approach is seen as beneficial by most students. Our approach allows every student to speak and ask questions in a low-stress creative environment. It is an excellent customizable, trackable, and scalable low-stakes assessment tool.

2.
Glia ; 72(2): 433-451, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37870193

RESUMEN

Mitochondria support the energetic demands of the cells. Autophagic turnover of mitochondria serves as a critical pathway for mitochondrial homeostasis. It is unclear how bioenergetics and autophagy are functionally connected. Here, we identify an endolysosomal membrane protein that facilitates autophagy to regulate ATP production in glia. We determined that Drosophila tweety (tty) is highly expressed in glia and localized to endolysosomes. Diminished fusion between autophagosomes and endolysosomes in tty-deficient glia was rescued by expressing the human Tweety Homolog 1 (TTYH1). Loss of tty in glia attenuated mitochondrial turnover, elevated mitochondrial oxidative stress, and impaired locomotor functions. The cellular and organismal defects were partially reversed by antioxidant treatment. We performed live-cell imaging of genetically encoded metabolite sensors to determine the impact of tty and autophagy deficiencies on glial bioenergetics. We found that tty-deficient glia exhibited reduced mitochondrial pyruvate consumption accompanied by a shift toward glycolysis for ATP production. Likewise, genetic inhibition of autophagy in glia resulted in a similar glycolytic shift in bioenergetics. Furthermore, the survival of mutant flies became more sensitive to starvation, underlining the significance of tty in the crosstalk between autophagy and bioenergetics. Together, our findings uncover the role for tty in mitochondrial homeostasis via facilitating autophagy, which determines bioenergetic balance in glia.


Asunto(s)
Autofagia , Drosophila , Metabolismo Energético , Mitocondrias , Animales , Humanos , Adenosina Trifosfato/metabolismo , Autofagia/genética , Drosophila/genética , Drosophila/metabolismo , Metabolismo Energético/genética , Homeostasis , Mitocondrias/metabolismo , Neuroglía/metabolismo
3.
Nat Metab ; 5(12): 2184-2205, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37996701

RESUMEN

Barth syndrome (BTHS) is a life-threatening genetic disorder with unknown pathogenicity caused by mutations in TAFAZZIN (TAZ) that affect remodeling of mitochondrial cardiolipin (CL). TAZ deficiency leads to accumulation of mono-lyso-CL (MLCL), which forms a peroxidase complex with cytochrome c (cyt c) capable of oxidizing polyunsaturated fatty acid-containing lipids. We hypothesized that accumulation of MLCL facilitates formation of anomalous MLCL-cyt c peroxidase complexes and peroxidation of polyunsaturated fatty acid phospholipids as the primary BTHS pathogenic mechanism. Using genetic, biochemical/biophysical, redox lipidomic and computational approaches, we reveal mechanisms of peroxidase-competent MLCL-cyt c complexation and increased phospholipid peroxidation in different TAZ-deficient cells and animal models and in pre-transplant biopsies from hearts of patients with BTHS. A specific mitochondria-targeted anti-peroxidase agent inhibited MLCL-cyt c peroxidase activity, prevented phospholipid peroxidation, improved mitochondrial respiration of TAZ-deficient C2C12 myoblasts and restored exercise endurance in a BTHS Drosophila model. Targeting MLCL-cyt c peroxidase offers therapeutic approaches to BTHS treatment.


Asunto(s)
Síndrome de Barth , Animales , Humanos , Síndrome de Barth/genética , Síndrome de Barth/patología , Citocromos c , Fosfolípidos , Cardiolipinas , Ácidos Grasos Insaturados , Peroxidasas
4.
Biomaterials ; 302: 122316, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37738741

RESUMEN

Intracortical microelectrodes that can record and stimulate brain activity have become a valuable technique for basic science research and clinical applications. However, long-term implantation of these microelectrodes can lead to progressive neurodegeneration in the surrounding microenvironment, characterized by elevation in disease-associated markers. Dysregulation of autophagy-lysosomal degradation, a major intracellular waste removal process, is considered a key factor in the onset and progression of neurodegenerative diseases. It is plausible that similar dysfunctions in autophagy-lysosomal degradation contribute to tissue degeneration following implantation-induced focal brain injury, ultimately impacting recording performance. To understand how the focal, persistent brain injury caused by long-term microelectrode implantation impairs autophagy-lysosomal pathway, we employed two-photon microscopy and immunohistology. This investigation focused on the spatiotemporal characterization of autophagy-lysosomal activity near the chronically implanted microelectrode. We observed an aberrant accumulation of immature autophagy vesicles near the microelectrode over the chronic implantation period. Additionally, we found deficits in autophagy-lysosomal clearance proximal to the chronic implant, which was associated with an accumulation of autophagy cargo and a reduction in lysosomal protease level during the chronic period. Furthermore, our evidence demonstrates reactive astrocytes have myelin-containing lysosomes near the microelectrode, suggesting its role of myelin engulfment during acute implantation period. Together, this study sheds light on the process of brain tissue degeneration caused by long-term microelectrode implantation, with a specific focus on impaired intracellular waste degradation.


Asunto(s)
Lesiones Encefálicas , Enfermedades Neurodegenerativas , Humanos , Microelectrodos , Electrodos Implantados/efectos adversos , Autofagia/fisiología , Lisosomas
6.
bioRxiv ; 2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37425842

RESUMEN

Tumor growth and proliferation are regulated by numerous mechanisms. Communication between intracellular organelles has recently been shown to regulate cellular proliferation and fitness. The way lysosomes and mitochondria communicate with each other (lysosomal/mitochondrial interaction) is emerging as a major determinant of tumor proliferation and growth. About 30% of squamous carcinomas (including squamous cell carcinoma of the head and neck, SCCHN) overexpress TMEM16A, a calcium-activated chloride channel, which promotes cellular growth and negatively correlates with patient survival. TMEM16A has recently been shown to drive lysosomal biogenesis, but its impact on mitochondrial function is unclear. Here, we show that (1) patients with high TMEM16A SCCHN display increased mitochondrial content specifically complex I; (2) In vitro and in vivo models uniquely depend on mitochondrial complex I activity for growth and survival; (3) ß-catenin/NRF2 signaling is a critical linchpin that drives mitochondrial biogenesis, and (4) mitochondrial complex I and lysosomal function are codependent for proliferation. Taken together, our data demonstrate that LMI drives tumor proliferation and facilitates a functional interaction between lysosomes and mitochondria. Therefore, inhibition of LMI may serve as a therapeutic strategy for patients with SCCHN.

7.
J Microbiol Biol Educ ; 23(2)2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36061331

RESUMEN

Research in a range of disciplines shows that many undergraduate students struggle with aggregating complex knowledge components into a complete picture and incorporating research literature into the learning process. To build and improve on the practice of project-based approaches to teaching cell biology, we transformed an undergraduate cell biology class by introducing the concept of storylines that are selected by groups of students for development throughout the semester. Each storyline integrates molecular and organellar concepts discussed during the semester into the cell- and tissue-level functions, conditions, or diseases shared and discussed during online poster sessions. Three semester-long studies conducted with an undergraduate cell biology class utilized pre- and postassignment assessments of self-efficacy and content knowledge (administered three times during the semester), and these studies showed that both parameters were significantly improved following each assignment. Specifically, student self-efficacy showed large gains, preassignment to postassignment (pre-post) [F (1,13) = 47.8, P < .001], and content knowledge showed moderate pre-post gains [F (1,12) = 14.5, P < 0.002]. Attitude surveys administered at the end of the semester suggest that the approach is seen as beneficial and enriching. We conclude that it is possible to integrate multiple levels of material in a complicated class by using storytelling and that such integration is positive and useful.

8.
Brain ; 145(12): 4202-4209, 2022 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-35953447

RESUMEN

Hypomyelinating leukodystrophies comprise a subclass of genetic disorders with deficient myelination of the CNS white matter. Here we report four unrelated families with a hypomyelinating leukodystrophy phenotype harbouring variants in TMEM163 (NM_030923.5). The initial clinical presentation resembled Pelizaeus-Merzbacher disease with congenital nystagmus, hypotonia, delayed global development and neuroimaging findings suggestive of significant and diffuse hypomyelination. Genomic testing identified three distinct heterozygous missense variants in TMEM163 with two unrelated individuals sharing the same de novo variant. TMEM163 is highly expressed in the CNS particularly in newly myelinating oligodendrocytes and was recently revealed to function as a zinc efflux transporter. All the variants identified lie in highly conserved residues in the cytoplasmic domain of the protein, and functional in vitro analysis of the mutant protein demonstrated significant impairment in the ability to efflux zinc out of the cell. Expression of the mutant proteins in an oligodendroglial cell line resulted in substantially reduced mRNA expression of key myelin genes, reduced branching and increased cell death. Our findings indicate that variants in TMEM163 cause a hypomyelinating leukodystrophy and uncover a novel role for zinc homeostasis in oligodendrocyte development and myelin formation.


Asunto(s)
Enfermedad de Pelizaeus-Merzbacher , Humanos , Enfermedad de Pelizaeus-Merzbacher/genética , Mutación Missense , Vaina de Mielina/metabolismo , Zinc/metabolismo , Proteínas de la Membrana/genética
9.
Proc Natl Acad Sci U S A ; 119(12): e2100670119, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35286200

RESUMEN

Squamous cell carcinoma of the head and neck (SCCHN) is a devastating disease that continues to have low cure rates despite the recent advances in therapies. Cisplatin is the most used chemotherapy agent, and treatment failure is largely driven by resistance to this drug. Amplification of chromosomal band 11q13 occurs in ∼30% of SCCHN tumors. This region harbors the ANO1 gene that encodes the TMEM16A ion channel, which is responsible for calcium-activated chloride transport in epithelial tissues. TMEM16A overexpression is associated with cisplatin resistance, and high TMEM16A levels correlate with decreased survival. However, the mechanistic underpinning of this effect remains unknown. Lysosomal biogenesis and exocytosis have been implicated in cancer because of their roles in the clearance of damaged organelles and exocytosis of chemotherapeutic drugs and toxins. Here, we show that TMEM16A overexpression promotes lysosomal biogenesis and exocytosis, which is consistent with the expulsion of intracellular cisplatin. Using a combination of genetic and pharmacologic approaches, we find that TMEM16A promotes lysosomal flux in a manner that requires reactive oxygen species, TRPML1, and the activation of the ß-catenin­melanocyte-inducing transcription factor pathway. The lysosomal inhibitor hydroxychloroquine (HCQ) synergizes with cisplatin in killing SCCHN cells in vitro. Using a murine model of SCCHN, we show that HCQ and cisplatin retard the growth of cisplatin-resistant patient-derived xenografts in vivo. We propose that TMEM16A enables cell survival by the up-regulation of lysosomal sequestration and exocytosis of the cytotoxic drugs. These results uncover a model of treatment for resistance in cancer, its reversal, and a role for TMEM16A.


Asunto(s)
Anoctamina-1 , Antineoplásicos , Cisplatino , Neoplasias de Cabeza y Cuello , Proteínas de Neoplasias , Anoctamina-1/genética , Anoctamina-1/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Canales de Cloruro , Cisplatino/farmacología , Humanos , Lisosomas/metabolismo , Proteínas de Neoplasias/metabolismo
10.
Clin Cancer Res ; 27(22): 6250-6264, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34407971

RESUMEN

PURPOSE: Human papillomavirus (HPV) infection drives the development of some head and neck squamous cell carcinomas (HNSCC). This disease is rapidly increasing in incidence worldwide. Although these tumors are sensitive to treatment, approximately 10% of patients fail therapy. However, the mechanisms that underlie treatment failure remain unclear. EXPERIMENTAL DESIGN: We performed RNA sequencing (RNA-seq) on tissues from matched primary- (pHNSCC) and metachronous-recurrent cancers (rHNSCC) to identify transcriptional differences to gain mechanistic insight into the evolutionary adaptations of metachronous-recurrent tumors. We used HPV-related HNSCC cells lines to investigate the effect of (i) NRF2 overexpression on growth in vitro and in vivo, (ii) oxidative phosphorylation (OXPHOS) inhibition using IACS-010759 on NRF2-dependent cells, and (iii) combination of cisplatin and OXPHOS inhibition. RESULTS: The OXPHOS pathway is enriched in recurrent HPV-associated HNSCC and may contribute to treatment failure. NRF2-enriched HNSCC samples from The Cancer Genome Atlas (TCGA) with enrichment in OXPHOS, fatty-acid metabolism, Myc, Mtor, reactive oxygen species (ROS), and glycolytic signaling networks exhibited worse survival. HPV-positive HNSCC cells demonstrated sensitivity to the OXPHOS inhibitor, in a NRF2-dependent manner. Further, using murine xenograft models, we identified NRF2 as a driver of tumor growth. Mechanistically, NRF2 drives ROS and mitochondrial respiration, and NRF2 is a critical regulator of redox homeostasis that can be crippled by disruption of OXPHOS. NRF2 also mediated cisplatin sensitivity in endogenously overexpressing primary HPV-related HNSCC cells. CONCLUSIONS: These results unveil a paradigm-shifting translational target harnessing NRF2-mediated metabolic reprogramming in HPV-related HNSCC.


Asunto(s)
Alphapapillomavirus , Neoplasias de Cabeza y Cuello , Infecciones por Papillomavirus , Animales , Neoplasias de Cabeza y Cuello/genética , Humanos , Ratones , Recurrencia Local de Neoplasia/genética , Fosforilación Oxidativa , Papillomaviridae/genética , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/genética
11.
Biochem J ; 478(17): 3205-3220, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34397090

RESUMEN

Recent advances in genome sequencing have led to the identification of new ion and metabolite transporters, many of which have not been characterized. Due to the variety of subcellular localizations, cargo and transport mechanisms, such characterization is a daunting task, and predictive approaches focused on the functional context of transporters are very much needed. Here we present a case for identifying a transporter localization using evolutionary rate covariation (ERC), a computational approach based on pairwise correlations of amino acid sequence evolutionary rates across the mammalian phylogeny. As a case study, we find that poorly characterized transporter SLC30A9 (ZnT9) coevolves with several components of the mitochondrial oxidative phosphorylation chain, suggesting mitochondrial localization. We confirmed this computational finding experimentally using recombinant human SLC30A9. SLC30A9 loss caused zinc mishandling in the mitochondria, suggesting that under normal conditions it acts as a zinc exporter. We therefore propose that ERC can be used to predict the functional context of novel transporters and other poorly characterized proteins.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Biología Computacional/métodos , Evolución Molecular , Mitocondrias/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Animales , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas Mitocondriales/metabolismo , Filogenia , Transfección , Secuenciación Completa del Genoma/métodos , Zinc/metabolismo
12.
Neurosci Lett ; 755: 135944, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-33965501

RESUMEN

Mucolipidosis IV (MLIV) is an autosomal-recessive disease caused by loss-of-function mutations in the MCOLN1 gene encoding the non-selective cationic lysosomal channel transient receptor potential mucolipin-1 (TRPML1). Patients with MLIV suffer from severe motor and cognitive deficits that manifest in early infancy and progressive loss of vision leading to blindness in the second decade of life. There are no therapies available for MLIV and the unmet medical need is extremely high. Here we review the spectrum of clinical presentations and the latest research in the MLIV pre-clinical model, with the aim of highlighting the progress in understanding the pathophysiology of the disease. These highlights include elucidation of the neurodevelopmental versus neurodegenerative features over the course of disease, hypomyelination as one of the major brain pathological disease hallmarks, and dysregulation of cytokines, with emerging evidence of IFN-gamma pathway upregulation in response to TRPML1 loss and pro-inflammatory activation of astrocytes and microglia. These scientific advances in the MLIV field provide a basis for future translational research, including biomarker and therapy development, that are desperately needed for this patient population.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Mucolipidosis/diagnóstico por imagen , Mucolipidosis/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Encéfalo/patología , Humanos , Lisosomas/genética , Lisosomas/metabolismo , Lisosomas/patología , Mucolipidosis/genética , Mucolipidosis/patología , Vaina de Mielina/genética , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Canales de Potencial de Receptor Transitorio/genética
13.
ACS Cent Sci ; 6(10): 1772-1788, 2020 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-33145414

RESUMEN

The development of a fluorescent probe for a specific metal has required exquisite design, synthesis, and optimization of fluorogenic molecules endowed with chelating moieties with heteroatoms. These probes are generally chelation- or reactivity-based. Catalysis-based fluorescent probes have the potential to be more sensitive; however, catalytic methods with a biocompatible fluorescence turn-on switch are rare. Here, we have exploited ligand-accelerated metal catalysis to repurpose known fluorescent probes for different metals, a new approach in probe development. We used the cleavage of allylic and propargylic ethers as platforms that were previously designed for palladium. After a single experiment that combinatorially examined >800 reactions with two variables (metal and ligand) for each ether, we discovered a platinum- or copper-selective method with the ligand effect of specific phosphines. Both metal-ligand systems were previously unknown and afforded strong signals owing to catalytic turnover. The fluorometric technologies were applied to geological, pharmaceutical, serum, and live cell samples and were used to discover that platinum accumulates in lysosomes in cisplatin-resistant cells in a manner that appears to be independent of copper distribution. The use of ligand-accelerated catalysis may present a new blueprint for engineering metal selectivity in probe development.

14.
Dis Model Mech ; 13(7)2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32586947

RESUMEN

Mucolipidosis 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.


Asunto(s)
Encéfalo/metabolismo , Diferenciación Celular , Mucolipidosis/metabolismo , Oligodendroglía/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Factores de Edad , Animales , Encéfalo/patología , Cerebelo/metabolismo , Cerebelo/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Ratones Endogámicos C57BL , Ratones Noqueados , Mucolipidosis/genética , Mucolipidosis/patología , Proteína Básica de Mielina/metabolismo , Proteínas de la Mielina/metabolismo , Glicoproteína Asociada a Mielina/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Células Precursoras de Oligodendrocitos/patología , Oligodendroglía/patología , Prosencéfalo/metabolismo , Prosencéfalo/patología , Canales de Potencial de Receptor Transitorio/genética
15.
FASEB J ; 34(5): 7192-7207, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32274853

RESUMEN

Mutations in ganglioside-induced differentiation-associated protein 1 (GDAP1) alter mitochondrial morphology and result in several subtypes of the inherited peripheral neuropathy Charcot-Marie-Tooth disease; however, the mechanism by which GDAP1 functions has remained elusive. GDAP1 contains primary sequence homology to the GST superfamily; however, the question of whether GDAP1 is an active GST has not been clearly resolved. Here, we present biochemical evidence, suggesting that GDAP1 has lost the ability to bind glutathione without a loss of substrate binding activity. We have revealed that the α-loop, located within the H-site motif is the primary determinant for substrate binding. Using structural data of GDAP1, we have found that critical residues and configurations in the G-site which canonically interact with glutathione are altered in GDAP1, rendering it incapable of binding glutathione. Last, we have found that the overexpression of GDAP1 in HeLa cells results in a mitochondrial phenotype which is distinct from oxidative stress-induced mitochondrial fragmentation. This phenotype is dependent on the presence of the transmembrane domain, as well as a unique hydrophobic domain that is not found in canonical GSTs. Together, we data point toward a non-enzymatic role for GDAP1, such as a sensor or receptor.


Asunto(s)
Glutatión Transferasa/química , Glutatión Transferasa/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Dominio Catalítico/genética , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/metabolismo , Cristalografía por Rayos X , Glutatión/metabolismo , Glutatión Transferasa/genética , Células HeLa , Humanos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Modelos Moleculares , Mutación , Proteínas del Tejido Nervioso/genética , Estrés Oxidativo , Fenotipo , Dominios Proteicos , Estructura Cuaternaria de Proteína , Especificidad por Sustrato
16.
Biochem J ; 476(24): 3705-3719, 2019 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-31790150

RESUMEN

Platinum-containing drugs such as cisplatin and carboplatin are routinely used for the treatment of many solid tumors including squamous cell carcinoma of the head and neck (SCCHN). However, SCCHN resistance to platinum compounds is well documented. The resistance to platinum has been linked to the activity of divalent transporter ATP7B, which pumps platinum from the cytoplasm into lysosomes, decreasing its concentration in the cytoplasm. Several cancer models show increased expression of ATP7B; however, the reason for such an increase is not known. Here we show a strong positive correlation between mRNA levels of TMEM16A and ATP7B in human SCCHN tumors. TMEM16A overexpression and depletion in SCCHN cell lines caused parallel changes in the ATP7B mRNA levels. The ATP7B increase in TMEM16A-overexpressing cells was reversed by suppression of NADPH oxidase 2 (NOX2), by the antioxidant N-Acetyl-Cysteine (NAC) and by copper chelation using cuprizone and bathocuproine sulphonate (BCS). Pretreatment with either chelator significantly increased cisplatin's sensitivity, particularly in the context of TMEM16A overexpression. We propose that increased oxidative stress in TMEM16A-overexpressing cells liberates the chelated copper in the cytoplasm, leading to the transcriptional activation of ATP7B expression. This, in turn, decreases the efficacy of platinum compounds by promoting their vesicular sequestration. We think that such a new explanation of the mechanism of SCCHN tumors' platinum resistance identifies novel approach to treating these tumors.


Asunto(s)
Anoctamina-1/metabolismo , Cisplatino/farmacología , ATPasas Transportadoras de Cobre/metabolismo , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Proteínas de Neoplasias/metabolismo , Anoctamina-1/genética , Antineoplásicos/farmacología , Línea Celular Tumoral , ATPasas Transportadoras de Cobre/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Neoplasias/genética , Regulación hacia Arriba
17.
Cell Rep ; 23(12): 3591-3606, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29925001

RESUMEN

In eukaryotic cells, lysosomes are distributed in the cytoplasm as individual membrane-bound compartments to degrade macromolecules and to control cellular metabolism. A fundamental yet unanswered question is whether and, if so, how individual lysosomes are organized spatially to coordinate and integrate their functions. To address this question, we analyzed their collective behavior in cultured cells using spatial statistical techniques. We found that in single cells, lysosomes maintain non-random, stable, yet distinct spatial distributions mediated by the cytoskeleton, the endoplasmic reticulum (ER), and lysosomal biogenesis. Throughout the intracellular space, lysosomes form dynamic clusters that significantly increase their interactions with endosomes. Cluster formation is associated with local increases in ER spatial density but does not depend on fusion with endosomes or spatial exclusion by mitochondria. Taken together, our findings reveal whole-cell scale spatial organization of lysosomes and provide insights into how organelle interactions are mediated and regulated across the entire intracellular space.


Asunto(s)
Lisosomas/metabolismo , Análisis Espacial , Estadística como Asunto , Citoesqueleto de Actina/metabolismo , Animales , Transporte Biológico , Células COS , Chlorocebus aethiops , Difusión , Retículo Endoplásmico/metabolismo , Endosomas/metabolismo , Fusión de Membrana , Microtúbulos/metabolismo , Mitocondrias/metabolismo
18.
Mol Neurobiol ; 55(3): 2454-2470, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28365875

RESUMEN

Traumatic brain injury (TBI) triggers endoplasmic reticulum (ER) stress and impairs autophagic clearance of damaged organelles and toxic macromolecules. In this study, we investigated the effects of the post-TBI administration of docosahexaenoic acid (DHA) on improving hippocampal autophagy flux and cognitive functions of rats. TBI was induced by cortical contusion injury in Sprague-Dawley rats, which received DHA (16 mg/kg in DMSO, intraperitoneal administration) or vehicle DMSO (1 ml/kg) with an initial dose within 15 min after the injury, followed by a daily dose for 3 or 7 days. First, RT-qPCR reveals that TBI induced a significant elevation in expression of autophagy-related genes in the hippocampus, including SQSTM1/p62 (sequestosome 1), lysosomal-associated membrane proteins 1 and 2 (Lamp1 and Lamp2), and cathepsin D (Ctsd). Upregulation of the corresponding autophagy-related proteins was detected by immunoblotting and immunostaining. In contrast, the DHA-treated rats did not exhibit the TBI-induced autophagy biogenesis and showed restored CTSD protein expression and activity. T2-weighted images and diffusion tensor imaging (DTI) of ex vivo brains showed that DHA reduced both gray matter and white matter damages in cortical and hippocampal tissues. DHA-treated animals performed better than the vehicle control group on the Morris water maze test. Taken together, these findings suggest that TBI triggers sustained stimulation of autophagy biogenesis, autophagy flux, and lysosomal functions in the hippocampus. Swift post-injury DHA administration restores hippocampal lysosomal biogenesis and function, demonstrating its therapeutic potential.


Asunto(s)
Autofagia/fisiología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Ácidos Docosahexaenoicos/uso terapéutico , Hipocampo/metabolismo , Lisosomas/fisiología , Animales , Autofagia/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/patología , Ácidos Docosahexaenoicos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/patología , Lisosomas/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
19.
FASEB J ; 32(2): 782-794, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29030399

RESUMEN

The transient receptor potential cation channel mucolipin 1 (TRPML1) channel is a conduit for lysosomal calcium efflux, and channel activity may be affected by lysosomal contents. The lysosomes of retinal pigmented epithelial (RPE) cells are particularly susceptible to build-up of lysosomal waste products because they must degrade the outer segments phagocytosed daily from adjacent photoreceptors; incomplete degradation leads to accumulation of lipid waste in lysosomes. This study asks whether stimulation of TRPML1 can release lysosomal calcium in RPE cells and whether such release is affected by lysosomal accumulations. The TRPML agonist ML-SA1 raised cytoplasmic calcium levels in mouse RPE cells, hesRPE cells, and ARPE-19 cells; this increase was rapid, robust, reversible, and reproducible. The increase was not altered by extracellular calcium removal or by thapsigargin but was eliminated by lysosomal rupture with glycyl-l-phenylalanine-ß-naphthylamide. Treatment with desipramine to inhibit acid sphingomyelinase or YM201636 to inhibit PIKfyve also reduced the cytoplasmic calcium increase triggered by ML-SA1, whereas RPE cells from TRPML1-/- mice showed no response to ML-SA1. Cotreatment with chloroquine and U18666A induced formation of neutral, autofluorescent lipid in RPE lysosomes and decreased lysosomal Ca2+ release. Lysosomal Ca2+ release was also impaired in RPE cells from the ATP-binding cassette, subfamily A, member 4-/- mouse model of Stargardt's retinal dystrophy. Neither TRPML1 mRNA nor total lysosomal calcium levels were altered in these models, suggesting a more direct effect on the channel. In summary, stimulation of TRPML1 elevates cytoplasmic calcium levels in RPE cells, but this response is reduced by lysosomal accumulation.-Gómez, N. M., Lu, W. Lim, J. C., Kiselyov, K., Campagno, K. E., Grishchuk, Y., Slaugenhaupt, S. A., Pfeffer, B., Fliesler, S. J., Mitchell, C. H. Robust lysosomal calcium signaling through channel TRPML1 is impaired by lysosomal lipid accumulation.


Asunto(s)
Señalización del Calcio , Metabolismo de los Lípidos , Lisosomas/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Humanos , Lisosomas/patología , Degeneración Macular/congénito , Degeneración Macular/genética , Degeneración Macular/metabolismo , Degeneración Macular/patología , Ratones , Ratones Noqueados , Ftalimidas/farmacología , Quinolinas/farmacología , Epitelio Pigmentado de la Retina/patología , Enfermedad de Stargardt , Canales de Potencial de Receptor Transitorio/agonistas , Canales de Potencial de Receptor Transitorio/genética
20.
Front Biosci (Landmark Ed) ; 22(8): 1330-1343, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28199205

RESUMEN

Lysosomes are emerging as important players in cellular zinc ion (Zn2+) homeostasis. The series of work on Zn2+ accumulation in the neuronal lysosomes and the mounting evidence on the role of lysosomal Zn2+ in cell death during mammary gland involution set a biological precedent for the central role of the lysosomes in cellular Zn2+ handling. Such a role appears to involve cytoprotection on the one hand, and cell death on the other. The recent series of work began to identify the molecular determinants of the lysosomal Zn2+ handling. In addition to zinc transporters (ZnT) of the solute-carrier family type 30A (SLC30A), the lysosomal ion channel TRPML1 and the poorly understood novel transporter TMEM163 have been shown to play a role in the Zn2+ uptake by the lysosomes. In this review, we summarize the current knowledge on molecular determinants of the lysosomal Zn2+ handling, uptake, and release pathways, as well as discuss their possible roles in health and disease.


Asunto(s)
Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Zinc/metabolismo , Secuencia de Aminoácidos , Animales , Humanos , Transporte Iónico , Proteínas de la Membrana/genética , Neuronas/metabolismo , Canales de Potencial de Receptor Transitorio/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...