Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(12): e2100670119, 2022 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-35286200

RESUMO

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.


Assuntos
Anoctamina-1 , Antineoplásicos , Cisplatino , Neoplasias de Cabeça e Pescoço , Proteínas de Neoplasias , Anoctamina-1/genética , Anoctamina-1/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Canais de Cloreto , Cisplatino/farmacologia , Humanos , Lisossomos/metabolismo , Proteínas de Neoplasias/metabolismo
2.
Glia ; 72(2): 433-451, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37870193

RESUMO

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.


Assuntos
Autofagia , Drosophila , Metabolismo Energético , Mitocôndrias , Animais , Humanos , Trifosfato de Adenosina/metabolismo , Autofagia/genética , Drosophila/genética , Drosophila/metabolismo , Metabolismo Energético/genética , Homeostase , Mitocôndrias/metabolismo , Neuroglia/metabolismo
3.
Brain ; 145(12): 4202-4209, 2022 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-35953447

RESUMO

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.


Assuntos
Doença de Pelizaeus-Merzbacher , Humanos , Doença de Pelizaeus-Merzbacher/genética , Mutação de Sentido Incorreto , Bainha de Mielina/metabolismo , Zinco/metabolismo , Proteínas de Membrana/genética
4.
Biochem J ; 478(17): 3205-3220, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34397090

RESUMO

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.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Biologia Computacional/métodos , Evolução Molecular , Mitocôndrias/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Proteínas Mitocondriais/metabolismo , Filogenia , Transfecção , Sequenciamento Completo do Genoma/métodos , Zinco/metabolismo
5.
FASEB J ; 34(5): 7192-7207, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32274853

RESUMO

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.


Assuntos
Glutationa Transferase/química , Glutationa Transferase/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Domínio Catalítico/genética , Doença de Charcot-Marie-Tooth/genética , Doença de Charcot-Marie-Tooth/metabolismo , Cristalografia por Raios X , Glutationa/metabolismo , Glutationa Transferase/genética , Células HeLa , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Modelos Moleculares , Mutação , Proteínas do Tecido Nervoso/genética , Estresse Oxidativo , Fenótipo , Domínios Proteicos , Estrutura Quaternária de Proteína , Especificidade por Substrato
6.
Biochem J ; 476(24): 3705-3719, 2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31790150

RESUMO

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.


Assuntos
Anoctamina-1/metabolismo , Cisplatino/farmacologia , ATPases Transportadoras de Cobre/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Proteínas de Neoplasias/metabolismo , Anoctamina-1/genética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , ATPases Transportadoras de Cobre/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas de Neoplasias/genética , Regulação para Cima
7.
FASEB J ; 32(2): 782-794, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29030399

RESUMO

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.


Assuntos
Sinalização do Cálcio , Metabolismo dos Lipídeos , Lisossomos/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Humanos , Lisossomos/patologia , Degeneração Macular/congênito , Degeneração Macular/genética , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Camundongos , Camundongos Knockout , Ftalimidas/farmacologia , Quinolinas/farmacologia , Epitélio Pigmentado da Retina/patologia , Doença de Stargardt , Canais de Potencial de Receptor Transitório/agonistas , Canais de Potencial de Receptor Transitório/genética
8.
J Cell Sci ; 127(Pt 14): 3094-103, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24829149

RESUMO

Zn(2+) is an essential micronutrient and an important ionic signal whose excess, as well as scarcity, is detrimental to cells. Free cytoplasmic Zn(2+) is controlled by a network of Zn(2+) transporters and chelating proteins. Recently, lysosomes became the focus of studies in Zn(2+) transport, as they were shown to play a role in Zn(2+)-induced toxicity by serving as Zn(2+) sinks that absorb Zn(2+) from the cytoplasm. Here, we investigated the impact of the lysosomal Zn(2+) sink on the net cellular Zn(2+) distribution and its role in cell death. We found that lysosomes played a cytoprotective role during exposure to extracellular Zn(2+). Such a role required lysosomal acidification and exocytosis. Specifically, we found that the inhibition of lysosomal acidification using Bafilomycin A1 (Baf) led to a redistribution of Zn(2+) pools and increased apoptosis. Additionally, the inhibition of lysosomal exocytosis through knockdown (KD) of the lysosomal SNARE proteins VAMP7 and synaptotagmin VII (SYT7) suppressed Zn(2+) secretion and VAMP7 KD cells had increased apoptosis. These data show that lysosomes play a central role in Zn(2+) handling, suggesting that there is a new Zn(2+) detoxification pathway.


Assuntos
Exocitose/fisiologia , Lisossomos/metabolismo , Zinco/metabolismo , Zinco/toxicidade , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Exocitose/efeitos dos fármacos , Células HeLa , Humanos , Macrolídeos/farmacologia , Transfecção
9.
Biochem J ; 470(1): 65-76, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26251447

RESUMO

Transition metal toxicity is an important factor in the pathogenesis of numerous human disorders, including neurodegenerative diseases. Lysosomes have emerged as important factors in transition metal toxicity because they handle transition metals via endocytosis, autophagy, absorption from the cytoplasm and exocytosis. Transcription factor EB (TFEB) regulates lysosomal biogenesis and the expression of lysosomal proteins in response to lysosomal and/or metabolic stresses. Since transition metals cause lysosomal dysfunction, we proposed that TFEB may be activated to drive gene expression in response to transition metal exposure and that such activation may influence transition metal toxicity. We found that transition metals copper (Cu) and iron (Fe) activate recombinant TFEB and stimulate the expression of TFEB-dependent genes in TFEB-overexpressing cells. In cells that show robust lysosomal exocytosis, TFEB was cytoprotective at moderate levels of Cu exposure, decreasing oxidative stress as reported by the expression of heme oxygenase-1 (HMOX1) gene. However, at high levels of Cu exposure, particularly in cells with low levels of lysosomal exocytosis, activation of overexpressed TFEB was toxic, increasing oxidative stress and mitochondrial damage. Based on these data, we conclude that TFEB-driven gene network is a component of the cellular response to transition metals. These data suggest limitations and disadvantages of TFEB overexpression as a therapeutic approach.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/biossíntese , Cloretos/farmacologia , Cobre/farmacologia , Compostos Férricos/farmacologia , Elementos de Transição/farmacologia , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia
10.
Pflugers Arch ; 467(6): 1143-64, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25106481

RESUMO

Transient receptor potential (TRP) channels form a diverse family of cation channels comprising 28 members in mammals. Although some TRP proteins can only be found on intracellular membranes, most of the TRP protein isoforms reach the plasma membrane where they form ion channels and control a wide number of biological processes. There, their involvement in the transport of cations such as calcium and sodium has been well documented. However, a growing number of studies have started to expand our understanding of these proteins by showing that they also transport other biologically relevant metal ions like zinc, magnesium, manganese and cobalt. In addition to this newly recognized property, the activity and expression of TRP channels can be regulated by metal ions like magnesium, gadolinium, lanthanum or cisplatin. The aim of this review is to highlight the complex relationship between metal ions and TRP channels.


Assuntos
Moduladores de Transporte de Membrana/farmacologia , Metais/farmacologia , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Humanos , Transporte de Íons/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/genética
11.
Biochem J ; 457(2): 361-8, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24192042

RESUMO

TRPML1 (transient receptor potential mucolipin 1) is a lysosomal ion channel permeable to cations, including Fe2+. Mutations in MCOLN1, the gene coding for TRPML1, cause the LSD (lysosomal storage disease) MLIV (mucolipidosis type IV). The role of TRPML1 in the cell is disputed and the mechanisms of cell deterioration in MLIV are unclear. The demonstration of Fe2+ buildup in MLIV cells raised the possibility that TRPML1 dissipates lysosomal Fe2+ and prevents its accumulation. Since Fe2+ catalyses the production of ROS (reactive oxygen species), we set out to test whether or not the loss of TRPML1 promotes ROS production by Fe2+ trapped in lysosomes. Our data show that RPE1 (retinal pigmented epithelial 1) cells develop a punctate mitochondrial phenotype within 48 h of siRNA-induced TRPML1-KD (knockdown). This mitochondrial fragmentation was aggravated by Fe2+ exposure, but was reversed by incubation with the ROS chelator α-Toc (α-tocopherol). The exposure of TRPML1-KD cells to Fe2+ led to loss of ΔΨm (mitochondrial membrane potential), ROS buildup, lipid peroxidation and increased transcription of genes responsive to cytotoxic oxidative stress in TRPML1-KD cells. These data suggest that TRPML1 redistributes Fe2+ between the lysosomes and the cytoplasm. Fe2+ buildup caused by TRPML1 loss potentiates ROS production and leads to mitochondrial deterioration. Beyond suggesting a new model for MLIV pathogenesis, these data show that TRPML1's role in the cell extends outside lysosomes.


Assuntos
Mucolipidoses/genética , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Canais de Potencial de Receptor Transitório/deficiência , Canais de Potencial de Receptor Transitório/genética , Células Cultivadas , Técnicas de Silenciamento de Genes/métodos , Humanos , Peroxidação de Lipídeos/fisiologia , Fenótipo , RNA Interferente Pequeno/genética , Epitélio Pigmentado da Retina/metabolismo
12.
J Mammary Gland Biol Neoplasia ; 19(1): 59-71, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24338187

RESUMO

Zinc plays a critical role in a vast array of cellular functions including gene transcription, protein translation, cell proliferation, differentiation, bioenergetics, and programmed cell death. The mammary gland depends upon tight coordination of these processes during development and reproduction for optimal expansion, differentiation, and involution. For example, zinc is required for activation of matrix metalloproteinases, intracellular signaling cascades such as MAPK and PKC, and the activation of both mitochondrial-mediated apoptosis and lysosomal-mediated cell death. In addition to functional needs, during lactation the mammary gland must balance providing optimal zinc for cellular requirements with the need to secrete a substantial amount of zinc into milk to meet the requirements of the developing neonate. Finally, the mammary gland exhibits the most profound example of programmed cell death, which is driven by both apoptotic and lysosomal-mediated cell death. Two families of zinc-specific transporters regulate zinc delivery for these diverse functions. Members of the ZIP family of zinc transporters (ZIP1-14) import zinc into the cytoplasm from outside the cell or from subcellular organelles, while members of the ZnT family (ZnT1-10) export zinc from the cytoplasm. Recently, the ion channel transient receptor potential mucolipin 1 (TRPML1) has also been implicated in zinc transport. Herein, we review our current understanding of the molecular mechanisms through which mammary epithelial cells utilize zinc with a focus on the transport of zinc into discrete subcellular organelles for specific cellular functions during mammary gland development, lactation, and involution.


Assuntos
Lactação/metabolismo , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Humanas/metabolismo , Zinco/metabolismo , Animais , Transporte Biológico , Células Epiteliais/metabolismo , Feminino , Humanos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Humanas/crescimento & desenvolvimento
13.
Am J Pathol ; 182(2): 474-84, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23231918

RESUMO

Mutations in the leucine-rich repeat kinase 2 (LRRK2) have been associated with familial and sporadic cases of Parkinson disease. Mutant LRRK2 causes in vitro and in vivo neurite shortening, mediated in part by autophagy, and a parkinsonian phenotype in transgenic mice; however, the underlying mechanisms remain unclear. Because mitochondrial content/function is essential for dendritic morphogenesis and maintenance, we investigated whether mutant LRRK2 affects mitochondrial homeostasis in neurons. Mouse cortical neurons expressing either LRRK2 G2019S or R1441C mutations exhibited autophagic degradation of mitochondria and dendrite shortening. In addition, mutant LRRK2 altered the ability of the neurons to buffer intracellular calcium levels. Either calcium chelators or inhibitors of voltage-gated L-type calcium channels prevented mitochondrial degradation and dendrite shortening. These data suggest that mutant LRRK2 causes a deficit in calcium homeostasis, leading to enhanced mitophagy and dendrite shortening.


Assuntos
Cálcio/metabolismo , Dendritos/metabolismo , Homeostase , Mitocôndrias/enzimologia , Proteínas Mutantes/metabolismo , Mutação/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Autofagia , Canais de Cálcio Tipo L/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Potencial da Membrana Mitocondrial , Camundongos , Mitofagia , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico
14.
Mol Genet Metab ; 111(2): 184-92, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24215843

RESUMO

Accumulation of globotriaosylceramide (Gb3) and other neutral glycosphingolipids with galactosyl residues is the hallmark of Fabry disease, a lysosomal storage disorder caused by deficiency of the enzyme alpha-galactosidase A (α-gal A). These lipids are incorporated into the plasma membrane and intracellular membranes, with a preference for lipid rafts. Disruption of raft mediated cell processes is implicated in the pathogenesis of several human diseases, but little is known about the effects of the accumulation of glycosphingolipids on raft dynamics in the context of Fabry disease. Using siRNA technology, we have generated a polarized renal epithelial cell model of Fabry disease in Madin-Darby canine kidney cells. These cells present increased levels of Gb3 and enlarged lysosomes, and progressively accumulate zebra bodies. The polarized delivery of both raft-associated and raft-independent proteins was unaffected by α-gal A knockdown, suggesting that accumulation of Gb3 does not disrupt biosynthetic trafficking pathways. To assess the effect of α-gal A silencing on lipid raft dynamics, we employed number and brightness (N&B) analysis to measure the oligomeric status and mobility of the model glycosylphosphatidylinositol (GPI)-anchored protein GFP-GPI. We observed a significant increase in the oligomeric size of antibody-induced clusters of GFP-GPI at the plasma membrane of α-gal A silenced cells compared with control cells. Our results suggest that the interaction of GFP-GPI with lipid rafts may be altered in the presence of accumulated Gb3. The implications of our results with respect to the pathogenesis of Fabry disease are discussed.


Assuntos
Proteínas de Fluorescência Verde/metabolismo , Microdomínios da Membrana/metabolismo , Modelos Biológicos , alfa-Galactosidase/metabolismo , Animais , Cães , Doença de Fabry/enzimologia , Doença de Fabry/patologia , Expressão Gênica , Glicosilfosfatidilinositóis/metabolismo , Proteínas de Fluorescência Verde/genética , Humanos , Rim/enzimologia , Rim/patologia , Lisossomos/enzimologia , Lisossomos/patologia , Células Madin Darby de Rim Canino , Microdomínios da Membrana/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Triexosilceramidas/biossíntese , alfa-Galactosidase/antagonistas & inibidores , alfa-Galactosidase/genética
15.
Biochem J ; 451(2): 155-63, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23368743

RESUMO

Zinc is critical for a multitude of cellular processes, including gene expression, secretion and enzymatic activities. Cellular zinc is controlled by zinc-chelating proteins and by zinc transporters. The recent identification of zinc permeability of the lysosomal ion channel TRPML1 (transient receptor potential mucolipin 1), and the evidence of abnormal zinc levels in cells deficient in TRPML1, suggested a role for TRPML1 in zinc transport. In the present study we provide new evidence for such a role and identify additional cellular components responsible for it. In agreement with the previously published data, an acute siRNA (small interfering RNA)-driven TRPML1 KD (knockdown) leads to the build-up of large cytoplasmic vesicles positive for LysoTracker™ and zinc staining, when cells are exposed to high concentrations of zinc. We now show that lysosomal enlargement and zinc build-up in TRPML1-KD cells exposed to zinc are ameliorated by KD of the zinc-sensitive transcription factor MTF-1 (metal-regulatory-element-binding transcription factor-1) or the zinc transporter ZnT4. TRPML1 KD is associated with a build-up of cytoplasmic zinc and with enhanced transcriptional response of mRNA for MT2a (metallothionein 2a). TRPML1 KD did not suppress lysosomal secretion, but it did delay zinc leak from the lysosomes into the cytoplasm. These results underscore a role for TRPML1 in zinc metabolism. Furthermore, they suggest that TRPML1 works in concert with ZnT4 to regulate zinc translocation between the cytoplasm and lysosomes.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Proteínas de Ligação a DNA/metabolismo , Lisossomos/metabolismo , Lisossomos/patologia , Fatores de Transcrição/metabolismo , Canais de Potencial de Receptor Transitório/genética , Zinco/metabolismo , Proteínas de Transporte de Cátions/genética , Citosol/metabolismo , Proteínas de Ligação a DNA/genética , Células HeLa/efeitos dos fármacos , Homeostase , Humanos , RNA Interferente Pequeno , Fatores de Transcrição/genética , Canais de Potencial de Receptor Transitório/deficiência , Canais de Potencial de Receptor Transitório/metabolismo , Zinco/farmacologia , Fator MTF-1 de Transcrição
16.
J Microbiol Biol Educ ; 25(1): e0006723, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38661421

RESUMO

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.

17.
J Biol Chem ; 287(11): 8082-91, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22262857

RESUMO

Mucolipidosis type IV (MLIV) is a lysosomal storage disease caused by mutations in the gene MCOLN1, which codes for the transient receptor potential family ion channel TRPML1. MLIV has an early onset and is characterized by developmental delays, motor and cognitive deficiencies, gastric abnormalities, retinal degeneration, and corneal cloudiness. The degenerative aspects of MLIV have been attributed to cell death, whose mechanisms remain to be delineated in MLIV and in most other storage diseases. Here we report that an acute siRNA-mediated loss of TRPML1 specifically causes a leak of lysosomal protease cathepsin B (CatB) into the cytoplasm. CatB leak is associated with apoptosis, which can be prevented by CatB inhibition. Inhibition of the proapoptotic protein Bax prevents TRPML1 KD-mediated apoptosis but does not prevent cytosolic release of CatB. This is the first evidence of a mechanistic link between acute TRPML1 loss and cell death.


Assuntos
Catepsina B/metabolismo , Citoplasma/metabolismo , Lisossomos/metabolismo , Mucolipidoses/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Proteína X Associada a bcl-2/metabolismo , Catepsina B/genética , Citoplasma/genética , Citoplasma/patologia , Células HeLa , Humanos , Lisossomos/genética , Lisossomos/patologia , Mucolipidoses/genética , Mucolipidoses/patologia , Canais de Potencial de Receptor Transitório/antagonistas & inibidores , Canais de Potencial de Receptor Transitório/genética , Proteína X Associada a bcl-2/genética
18.
Biomaterials ; 302: 122316, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37738741

RESUMO

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.


Assuntos
Lesões Encefálicas , Doenças Neurodegenerativas , Humanos , Microeletrodos , Eletrodos Implantados/efeitos adversos , Autofagia/fisiologia , Lisossomos
19.
bioRxiv ; 2023 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-37425842

RESUMO

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.

20.
Nat Metab ; 5(12): 2184-2205, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37996701

RESUMO

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.


Assuntos
Síndrome de Barth , Animais , Humanos , Síndrome de Barth/genética , Síndrome de Barth/patologia , Citocromos c , Fosfolipídeos , Cardiolipinas , Ácidos Graxos Insaturados , Peroxidases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA