Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(13): e2319429121, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38513095

RESUMEN

Polyamines are a class of small polycationic alkylamines that play essential roles in both normal and cancer cell growth. Polyamine metabolism is frequently dysregulated and considered a therapeutic target in cancer. However, targeting polyamine metabolism as monotherapy often exhibits limited efficacy, and the underlying mechanisms are incompletely understood. Here we report that activation of polyamine catabolism promotes glutamine metabolism, leading to a targetable vulnerability in lung cancer. Genetic and pharmacological activation of spermidine/spermine N1-acetyltransferase 1 (SAT1), the rate-limiting enzyme of polyamine catabolism, enhances the conversion of glutamine to glutamate and subsequent glutathione (GSH) synthesis. This metabolic rewiring ameliorates oxidative stress to support lung cancer cell proliferation and survival. Simultaneous glutamine limitation and SAT1 activation result in ROS accumulation, growth inhibition, and cell death. Importantly, pharmacological inhibition of either one of glutamine transport, glutaminase, or GSH biosynthesis in combination with activation of polyamine catabolism synergistically suppresses lung cancer cell growth and xenograft tumor formation. Together, this study unveils a previously unappreciated functional interconnection between polyamine catabolism and glutamine metabolism and establishes cotargeting strategies as potential therapeutics in lung cancer.


Asunto(s)
Neoplasias Pulmonares , Humanos , Glutamina , Poliaminas/metabolismo , Pulmón/metabolismo , Muerte Celular , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Espermina/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(12): e2122245119, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-35302894

RESUMEN

High-performance metabolic analysis is emerging in the diagnosis and prognosis of breast cancer (BrCa). Still, advanced tools are in demand to deliver the application potentials of metabolic analysis. Here, we used fast nanoparticle-enhanced laser desorption/ionization mass spectrometry (NPELDI-MS) to record serum metabolic fingerprints (SMFs) of BrCa in seconds, achieving high reproducibility and low consumption of direct serum detection without treatment. Subsequently, machine learning of SMFs generated by NPELDI-MS functioned as an efficient readout to distinguish BrCa from non-BrCa with an area under the curve of 0.948. Furthermore, a metabolic prognosis scoring system was constructed using SMFs with effective prediction performance toward BrCa (P < 0.005). Finally, we identified a biomarker panel of seven metabolites that were differentially enriched in BrCa serum and their related pathways. Together, our findings provide an efficient serum metabolic tool to characterize BrCa and highlight certain metabolic signatures as potential diagnostic and prognostic factors of diseases including but not limited to BrCa.


Asunto(s)
Neoplasias de la Mama , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Espectrometría de Masas/métodos , Pronóstico , Reproducibilidad de los Resultados
3.
Hepatology ; 78(2): 503-517, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-36999531

RESUMEN

BACKGROUND AND AIMS: The aim of the study was to investigate the role and mechanisms of tuberous sclerosis complex 1 (TSC1) and mechanistic target of rapamycin complex 1 (mTORC1) in alcohol-associated liver disease. APPROACH AND RESULTS: Liver-specific Tsc1 knockout (L- Tsc1 KO) mice and their matched wild-type mice were subjected to Gao-binge alcohol. Human alcoholic hepatitis (AH) samples were also used for immunohistochemistry staining, western blot, and quantitative real-time PCR (q-PCR) analysis. Human AH and Gao-binge alcohol-fed mice had decreased hepatic TSC1 and increased mTORC1 activation. Gao-binge alcohol markedly increased liver/body weight ratio and serum alanine aminotransferase levels in L- Tsc1 KO mice compared with Gao-binge alcohol-fed wild-type mice. Results from immunohistochemistry staining, western blot, and q-PCR analysis revealed that human AH and Gao-binge alcohol-fed L- Tsc1 KO mouse livers had significantly increased hepatic progenitor cells, macrophages, and neutrophils but decreased HNF4α-positive cells. Gao-binge alcohol-fed L- Tsc1 KO mice also developed severe inflammation and liver fibrosis. Deleting Tsc1 in cholangiocytes but not in hepatocytes promoted cholangiocyte proliferation and aggravated alcohol-induced ductular reactions, fibrosis, inflammation, and liver injury. Pharmacological inhibition of mTORC1 partially reversed hepatomegaly, ductular reaction, fibrosis, inflammatory cell infiltration, and liver injury in alcohol-fed L- Tsc1 KO mice. CONCLUSIONS: Our findings indicate that persistent activation of mTORC1 due to the loss of cholangiocyte TSC1 promotes liver cell repopulation, ductular reaction, inflammation, fibrosis, and liver injury in Gao-binge alcohol-fed L- Tsc1 KO mice, which phenocopy the pathogenesis of human AH.


Asunto(s)
Hepatitis Alcohólica , Hepatopatías Alcohólicas , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteína 1 del Complejo de la Esclerosis Tuberosa , Animales , Humanos , Ratones , Etanol , Fibrosis , Hepatitis Alcohólica/patología , Inflamación/patología , Hígado/patología , Hepatopatías Alcohólicas/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Noqueados , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo
4.
Cell ; 138(2): 328-39, 2009 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-19632182

RESUMEN

Here we identify a component of the nuclear RNA cap-binding complex (CBC), Ars2, that is important for miRNA biogenesis and critical for cell proliferation. Unlike other components of the CBC, Ars2 expression is linked to the proliferative state of the cell. Deletion of Ars2 is developmentally lethal, and deletion in adult mice led to bone marrow failure whereas parenchymal organs composed of nonproliferating cells were unaffected. Depletion of Ars2 or CBP80 from proliferating cells impaired miRNA-mediated repression and led to alterations in primary miRNA processing in the nucleus. Ars2 depletion also reduced the levels of several miRNAs, including miR-21, let-7, and miR-155, that are implicated in cellular transformation. These findings provide evidence for a role for Ars2 in RNA interference regulation during cell proliferation.


Asunto(s)
Proliferación Celular , Complejo Proteico Nuclear de Unión a la Caperuza/metabolismo , Proteínas Nucleares/metabolismo , Interferencia de ARN , Animales , Arsénico/toxicidad , Línea Celular , Guanosina/análogos & derivados , Guanosina/metabolismo , Humanos , Ratones , MicroARNs
5.
Mol Cell ; 61(5): 667-676, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26942671

RESUMEN

Decades ago, Otto Warburg observed that cancers ferment glucose in the presence of oxygen, suggesting that defects in mitochondrial respiration may be the underlying cause of cancer. We now know that the genetic events that drive aberrant cancer cell proliferation also alter biochemical metabolism, including promoting aerobic glycolysis, but do not typically impair mitochondrial function. Mitochondria supply energy; provide building blocks for new cells; and control redox homeostasis, oncogenic signaling, innate immunity, and apoptosis. Indeed, mitochondrial biogenesis and quality control are often upregulated in cancers. While some cancers have mutations in nuclear-encoded mitochondrial tricarboxylic acid (TCA) cycle enzymes that produce oncogenic metabolites, there is negative selection for pathogenic mitochondrial genome mutations. Eliminating mtDNA limits tumorigenesis, and rare human tumors with mutant mitochondrial genomes are relatively benign. Thus, mitochondria play a central and multifunctional role in malignant tumor progression, and targeting mitochondria provides therapeutic opportunities.


Asunto(s)
Metabolismo Energético , Mitocondrias/metabolismo , Neoplasias/metabolismo , Animales , Antineoplásicos/uso terapéutico , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , ADN Mitocondrial/genética , ADN Mitocondrial/metabolismo , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Predisposición Genética a la Enfermedad , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Terapia Molecular Dirigida , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Transducción de Señal
6.
Mol Cell ; 61(5): 720-733, 2016 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-26942676

RESUMEN

TRIM21 is a RING finger domain-containing ubiquitin E3 ligase whose expression is elevated in autoimmune disease. While TRIM21 plays an important role in immune activation during pathogen infection, little is known about its inherent cellular function. Here we show that TRIM21 plays an essential role in redox regulation by directly interacting with SQSTM1/p62 and ubiquitylating p62 at lysine 7 (K7) via K63-linkage. As p62 oligomerizes and sequesters client proteins in inclusions, the TRIM21-mediated p62 ubiquitylation abrogates p62 oligomerization and sequestration of proteins including Keap1, a negative regulator of antioxidant response. TRIM21-deficient cells display an enhanced antioxidant response and reduced cell death in response to oxidative stress. Genetic ablation of TRIM21 in mice confers protection from oxidative damages caused by arsenic-induced liver insult and pressure overload heart injury. Therefore, TRIM21 plays an essential role in p62-regulated redox homeostasis and may be a viable target for treating pathological conditions resulting from oxidative damage.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Choque Térmico/metabolismo , Estrés Oxidativo , Ribonucleoproteínas/metabolismo , Ubiquitinación , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Trióxido de Arsénico , Arsenicales , Muerte Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Proteínas del Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Células HEK293 , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/prevención & control , Proteínas de Choque Térmico/genética , Homeostasis , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Hígado/enzimología , Hígado/patología , Lisina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/enzimología , Miocardio/patología , Oxidación-Reducción , Óxidos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Interferencia de ARN , Ribonucleoproteínas/deficiencia , Ribonucleoproteínas/genética , Proteína Sequestosoma-1 , Transducción de Señal , Factores de Tiempo , Transfección
7.
Arch Toxicol ; 98(3): 709-734, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38182913

RESUMEN

With the rapid development of new generations of antitumor therapies, the average survival time of cancer patients is expected to be continuously prolonged. However, these therapies often lead to cardiotoxicity, resulting in a growing number of tumor survivors with cardiovascular disease. Therefore, a new interdisciplinary subspecialty called "cardio-oncology" has emerged, aiming to detect and treat cardiovascular diseases associated with tumors and antitumor therapies. Recent studies have highlighted the role of ferroptosis in both cardiovascular and neoplastic diseases. The balance between intracellular oxidative stress and antioxidant defense is crucial in regulating ferroptosis. Tumor cells can evade ferroptosis by upregulating multiple antioxidant defense pathways, while many antitumor therapies rely on downregulating antioxidant defense and promoting ferroptosis in cancer cells. Unfortunately, these ferroptosis-inducing antitumor therapies often lack tissue specificity and can also cause injury to the heart, resulting in ferroptosis-induced cardiotoxicity. A range of cardioprotective agents exert cardioprotective effects by inhibiting ferroptosis. However, these cardioprotective agents might diminish the efficacy of antitumor treatment due to their antiferroptotic effects. Most current research on ferroptosis only focuses on either tumor treatment or heart protection but rarely considers both in concert. Therefore, further research is needed to study how to protect the heart during antitumor therapies by regulating ferroptosis. In this review, we summarized the role of ferroptosis in the treatment of neoplastic diseases and cardiovascular diseases and also attempted to propose further research directions for ferroptosis in the field of cardio-oncology.


Asunto(s)
Enfermedades Cardiovasculares , Ferroptosis , Humanos , Antioxidantes , Cardiooncología , Cardiotónicos , Cardiotoxicidad , Enfermedades Cardiovasculares/inducido químicamente
8.
J Biochem Mol Toxicol ; 37(12): e23506, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37598318

RESUMEN

Endocrine-disrupting chemicals pose a growing threat to human health through their increasing presence in the environment and their potential interactions with the mammalian endocrine systems. Due to their structural similarity to hormones like estrogen, these chemicals can interfere with endocrine signaling, leading to many deleterious effects. Exposure to estrogenic endocrine-disrupting compounds (EDC) is a suggested risk factor for the development of breast cancer, one of the most frequently diagnosed cancers in women. However, the mechanisms through which EDCs contribute to breast cancer development remain elusive. To rapidly proliferate, cancer cells undertake distinct metabolic programs to utilize existing nutrients in the tumor microenvironment and synthesize macromolecules de novo. EDCs are known to dysregulate cell signaling pathways related to cellular metabolism, which may be an important mechanism through which they exert their cancer-promoting effects. These altered pathways can be studied via metabolomic analysis, a new advancement in -omics technologies that can interrogate molecular pathways that favor cancer development and progression. This review will summarize recent discoveries regarding EDCs and the metabolic reprogramming that they may induce to facilitate the development of breast cancer.


Asunto(s)
Neoplasias de la Mama , Disruptores Endocrinos , Animales , Humanos , Femenino , Neoplasias de la Mama/inducido químicamente , Estrógenos , Transducción de Señal , Factores de Riesgo , Disruptores Endocrinos/toxicidad , Mamíferos , Microambiente Tumoral
9.
Mol Cell ; 58(2): 255-68, 2015 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-25866245

RESUMEN

PTEN is proposed to function at the plasma membrane, where receptor tyrosine kinases are activated. However, the majority of PTEN is located throughout the cytoplasm. Here, we show that cytoplasmic PTEN is distributed along microtubules, tethered to vesicles via phosphatidylinositol 3-phosphate (PI(3)P), the signature lipid of endosomes. We demonstrate that the non-catalytic C2 domain of PTEN specifically binds PI(3)P through the CBR3 loop. Mutations render this loop incapable of PI(3)P binding and abrogate PTEN-mediated inhibition of PI 3-kinase/AKT signaling. This loss of function is rescued by fusion of the loop mutant PTEN to FYVE, the canonical PI(3)P binding domain, demonstrating the functional importance of targeting PTEN to endosomal membranes. Beyond revealing an upstream activation mechanism of PTEN, our data introduce the concept of PI 3-kinase signal activation on the vast plasma membrane that is contrasted by PTEN-mediated signal termination on the small, discrete surfaces of internalized vesicles.


Asunto(s)
Fosfohidrolasa PTEN/química , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Vesículas Transportadoras/metabolismo , Animales , Sitios de Unión , Ratones , Microtúbulos/enzimología , Modelos Moleculares , Células 3T3 NIH , Estructura Secundaria de Proteína , Transducción de Señal
10.
J Hepatol ; 76(3): 639-651, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34710483

RESUMEN

BACKGROUND & AIMS: Either activation of mTORC1 due to loss of Tsc1 (tuberous sclerosis complex 1) or defective hepatic autophagy due to loss of Atg5 leads to spontaneous liver tumorigenesis in mice. The purpose of this study was to investigate the mechanisms by which autophagy contributes to the hepatic metabolic changes and tumorigenesis mediated by mTORC1 activation. METHODS: Atg5 Flox/Flox (Atg5F/F) and Tsc1F/F mice were crossed with albumin-Cre mice to generate liver-specific Atg5 knockout (L-Atg5 KO), L-Tsc1 KO and L-Atg5/Tsc1 double KO (DKO) mice. These mice were crossed with p62/Sqstm1F/F (p62) and whole body Nrf2 KO mice to generate L-Atg5/Tsc1/p62 and L-Atg5/Tsc1-Nrf2 triple KO mice. These mice were housed for various periods up to 12 months, and blood and liver tissues were harvested for biochemical and histological analysis RESULTS: Deletion of Atg5 in L-Tsc1 KO mice inhibited liver tumorigenesis but increased mortality and was accompanied by drastically enhanced hepatic ductular reaction (DR), hepatocyte degeneration and metabolic reprogramming. Deletion of p62 reversed DR, hepatocyte degeneration and metabolic reprogramming as well as the mortality of L-Atg5/Tsc1 DKO mice, but unexpectedly promoted liver tumorigenesis via activation of a group of oncogenic signaling pathways. Nrf2 ablation markedly improved DR with increased hepatocyte population and improved metabolic reprogramming and survival of the L-Atg5/Tsc1 DKO mice without tumor formation. Decreased p62 and increased mTOR activity were also observed in a subset of human hepatocellular carcinomas. CONCLUSIONS: These results reveal previously undescribed functions of hepatic p62 in suppressing tumorigenesis and regulating liver cell repopulation and metabolic reprogramming resulting from persistent mTORC1 activation and defective autophagy. LAY SUMMARY: Metabolic liver disease and viral hepatitis are common chronic liver diseases and risk factors of hepatocellular carcinoma, which are often associated with impaired hepatic autophagy and increased mTOR activation. Using multiple genetically engineered mouse models of defective hepatic autophagy and persistent mTOR activation, we dissected the complex mechanisms behind this observation. Our results uncovered an unexpected novel tumor suppressor function of p62/Sqstm1, which regulated liver cell repopulation, ductular reaction and metabolic reprogramming in liver tumorigenesis.


Asunto(s)
Autofagia/fisiología , Conductos Biliares Intrahepáticos/efectos de los fármacos , Hígado/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/farmacología , Animales , Autofagia/genética , Modelos Animales de Enfermedad , Hígado/fisiopatología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados/metabolismo
11.
Lab Invest ; 101(11): 1467-1474, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34504306

RESUMEN

The mortality rates among patients who initially survive sepsis are, in part, associated with a high risk of secondary lung infections and respiratory failure. Given that phagolysosomes are important for intracellular killing of pathogenic microbes, we investigated how severe lung infections associated with post-sepsis immunosuppression affect phagolysosome biogenesis. In mice with P. aeruginosa-induced pneumonia, we found a depletion of both phagosomes and lysosomes, as evidenced by decreased amounts of microtubule associated protein light chain 3-II (LC3-II) and lysosomal-associated membrane protein (LAMP1). We also found a loss of transcription factor E3 (TFE3) and transcription factor EB (TFEB), which are important activators for transcription of genes encoding autophagy and lysosomal proteins. These events were associated with increased expression of ZKSCAN3, a repressor for transcription of genes encoding autophagy and lysosomal proteins. Zkscan3-/- mice had increased expression of genes involved in the autophagy-lysosomal pathway along with enhanced killing of P. aeruginosa in the lungs, as compared to wild-type mice. These findings highlight the involvement of ZKSCAN3 in response to severe lung infection, including susceptibility to secondary bacterial infections due to immunosuppression.


Asunto(s)
Fagosomas/fisiología , Neumonía Bacteriana/complicaciones , Infecciones por Pseudomonas/complicaciones , Sepsis/inmunología , Factores de Transcripción/deficiencia , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Tolerancia Inmunológica , Pulmón/metabolismo , Masculino , Ratones Endogámicos C57BL , Neumonía Bacteriana/metabolismo , Infecciones por Pseudomonas/metabolismo , Pseudomonas aeruginosa , Sepsis/microbiología
12.
Nature ; 527(7576): 105-9, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26524528

RESUMEN

Macroautophagy (hereafter referred to as autophagy) is a catabolic membrane trafficking process that degrades a variety of cellular constituents and is associated with human diseases. Although extensive studies have focused on autophagic turnover of cytoplasmic materials, little is known about the role of autophagy in degrading nuclear components. Here we report that the autophagy machinery mediates degradation of nuclear lamina components in mammals. The autophagy protein LC3/Atg8, which is involved in autophagy membrane trafficking and substrate delivery, is present in the nucleus and directly interacts with the nuclear lamina protein lamin B1, and binds to lamin-associated domains on chromatin. This LC3-lamin B1 interaction does not downregulate lamin B1 during starvation, but mediates its degradation upon oncogenic insults, such as by activated RAS. Lamin B1 degradation is achieved by nucleus-to-cytoplasm transport that delivers lamin B1 to the lysosome. Inhibiting autophagy or the LC3-lamin B1 interaction prevents activated RAS-induced lamin B1 loss and attenuates oncogene-induced senescence in primary human cells. Our study suggests that this new function of autophagy acts as a guarding mechanism protecting cells from tumorigenesis.


Asunto(s)
Autofagia , Lámina Nuclear/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Familia de las Proteínas 8 Relacionadas con la Autofagia , Transformación Celular Neoplásica , Células Cultivadas , Senescencia Celular , Cromatina/química , Cromatina/metabolismo , Citoplasma/metabolismo , Fibroblastos , Células HEK293 , Humanos , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Lisosomas/metabolismo , Ratones , Proteínas de Microfilamentos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Unión Proteica , Proteolisis
13.
Mol Cell ; 50(1): 29-42, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23434372

RESUMEN

Autophagy is an evolutionarily conserved membrane trafficking process. Induction of autophagy in response to nutrient limitation or cellular stress occurs by similar mechanisms in organisms from yeast to mammals. Unlike yeast, metazoan cells rely more on growth factor signaling for a wide variety of cellular activities including nutrient uptake. How growth factor availability regulates autophagy is poorly understood. Here we show that, upon growth factor limitation, the p110ß catalytic subunit of the class IA phosphoinositide 3-kinases (PI3Ks) dissociates from growth factor receptor complexes and increases its interaction with the small GTPase Rab5. This p110ß-Rab5 association maintains Rab5 in its guanosine triphosphate (GTP)-bound state and enhances the Rab5-Vps34 interaction that promotes autophagy. p110ß mutants that fail to interact with Rab5 are defective in autophagy promotion. Hence, in mammalian cells, p110ß acts as a molecular sensor for growth factor availability and induces autophagy by activating a Rab5-mediated signaling cascade.


Asunto(s)
Autofagia , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Animales , Fosfatidilinositol 3-Quinasa Clase I/deficiencia , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Guanosina Trifosfato/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Mutación , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal , Transfección
14.
Anal Chem ; 92(7): 4829-4837, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32125145

RESUMEN

A large proportion of the complexity and redundancy of LC-MS metabolomics data comes from adduct formation. To reduce such redundancy, many tools have been developed to recognize and annotate adduct ions. These tools rely on predefined adduct lists that are generated empirically from reversed-phase LC-MS studies. In addition, hydrophilic interaction chromatography (HILIC) is gaining popularity in metabolomics studies due to its enhanced performance over other methods for polar compounds. HILIC methods typically use high concentrations of buffer salts to improve chromatographic performance. Therefore, it is necessary to analyze adduct formation in HILIC metabolomics. To this end, we developed covariant ion analysis (COVINA) to investigate metabolite adduct formation. Using this tool, we completely annotated 201 adduct and fragment ions from 10 metabolites. Many of the metabolite adduct ions were found to contain cluster ions corresponding to mobile phase additives. We further utilized COVINA to find the major ionized forms of metabolites. Our results show that for some metabolites, the adduct ion signals can be >200-fold higher than the signals from the deprotonated form, offering better sensitivity for targeted metabolomics analysis. Finally, we developed an in-source CID ramping (InCIDR) method to analyze the intensity changes of the adduct and fragment ions from metabolites. Our analysis demonstrates a promising method to distinguish the protonated and deprotonated ions of metabolites from the adduct and fragment ions.


Asunto(s)
Metabolómica , Línea Celular Tumoral , Cromatografía Líquida de Alta Presión , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Espectrometría de Masas
15.
Hepatology ; 70(6): 2142-2155, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31095752

RESUMEN

Autophagy is a lysosomal degradation pathway that degrades cytoplasmic proteins and organelles. Absence of autophagy in hepatocytes has been linked to promoting liver injury and tumorigenesis; however, the mechanisms behind why a lack of autophagy induces these complications are not fully understood. The role of mammalian target of rapamycin (mTOR) in impaired autophagy-induced liver pathogenesis and tumorigenesis was investigated by using liver-specific autophagy related 5 knockout (L-ATG5 KO) mice, L-ATG5/mTOR, and L-ATG5/Raptor double knockout (DKO) mice. We found that deletion of mTOR or Raptor in L-ATG5 KO mice at 2 months of age attenuated hepatomegaly, cell death, and inflammation but not fibrosis. Surprisingly, at 6 months of age, L-ATG5/mTOR DKO and L-ATG5/Raptor DKO mice also had increased hepatic inflammation, fibrosis, and liver injury, similar to the L-ATG5 KO mice. Moreover, more than 50% of L-ATG5/mTOR DKO and L-ATG5/Raptor DKO mice already developed spontaneous tumors, but none of the L-ATG5 KO mice had developed any tumors at 6 months of age. At 9 months of age, all L-ATG5/mTOR DKO and L-ATG5/Raptor DKO had developed liver tumors. Mechanistically, L-ATG5/mTOR DKO and L-ATG5/Raptor DKO mice had decreased levels of hepatic ubiquitinated proteins and persistent nuclear erythroid 2 p45-related factor 2 activation but had increased Akt activation compared with L-ATG5 KO mice. Conclusion: Loss of mTOR signaling attenuates the liver pathogenesis in mice with impaired hepatic autophagy but paradoxically promotes tumorigenesis in mice at a relatively young age. Therefore, the balance of mTOR is critical in regulating the liver pathogenesis and tumorigenesis in mice with impaired hepatic autophagy.


Asunto(s)
Proteína 5 Relacionada con la Autofagia/fisiología , Autofagia/fisiología , Neoplasias Hepáticas/etiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Carcinogénesis , Hepatomegalia/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 2 Relacionado con NF-E2/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteína Reguladora Asociada a mTOR/fisiología
16.
Semin Cell Dev Biol ; 62: 170-177, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27637160

RESUMEN

Human SERPINB3 and SERPINB4 are evolutionary duplicated serine/cysteine protease inhibitors. Genomic analysis indicates that these paralogous genes were encoded from independent loci arising from tandem gene duplication. Although the two molecules share 92% identity of their amino acid sequences, they are distinct in the Reactive Center Loop (RCL) including a hinge region and catalytic sequences which accounts for altered substrate specificity. Elevated expression of the two molecules has been reported to contribute to numerous pathological conditions such as inflammatory diseases and cancer. In this review, we focus on summarizing the biochemical features of SERPINB3/B4 and discussing the mechanistic basis for their biological functions and implications in human diseases.


Asunto(s)
Fenómenos Bioquímicos , Serpinas/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Carcinogénesis/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Serpinas/genética , Distribución Tisular
17.
J Cell Sci ; 130(12): 2007-2017, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28455411

RESUMEN

The GTPase Rab5 and phosphatidylinositol-3 phosphate [PI(3)P] coordinately regulate endosome trafficking. Rab5 recruits Vps34, the class III phosphoinositide 3-kinase (PI3K), to generate PI(3)P and recruit PI(3)P-binding proteins. Loss of Rab5 and loss of Vps34 have opposite effects on endosome size, suggesting that our understanding of how Rab5 and PI(3)P cooperate is incomplete. Here, we report a novel regulatory loop whereby Caenorhabditis elegans VPS-34 inactivates RAB-5 via recruitment of the TBC-2 Rab GTPase-activating protein. We found that loss of VPS-34 caused a phenotype with large late endosomes, as with loss of TBC-2, and that Rab5 activity (mice have two Rab5 isoforms, Rab5a and Rab5b) is increased in Vps34-knockout mouse embryonic fibroblasts (Vps34 is also known as PIK3C3 in mammals). We found that VPS-34 is required for TBC-2 endosome localization and that the pleckstrin homology (PH) domain of TBC-2 bound PI(3)P. Deletion of the PH domain enhanced TBC-2 localization to endosomes in a VPS-34-dependent manner. Thus, PI(3)P binding of the PH domain might be permissive for another PI(3)P-regulated interaction that recruits TBC-2 to endosomes. Therefore, VPS-34 recruits TBC-2 to endosomes to inactivate RAB-5 to ensure the directionality of endosome maturation.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Endosomas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de Unión al GTP rab5/metabolismo , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Membrana Celular/metabolismo , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Liposomas/química , Ratones , Ratones Noqueados , Mutación , Fenotipo , Plásmidos/metabolismo , Dominios Proteicos , Interferencia de ARN
19.
J Cell Sci ; 129(23): 4424-4435, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27793976

RESUMEN

The class III phosphoinositide 3-kinase (PI3K) Vps34 (also known as PIK3C3 in mammals) produces phosphatidylinositol 3-phosphate [PI(3)P] on both early and late endosome membranes to control membrane dynamics. We used Vps34-deficient cells to delineate whether Vps34 has additional roles in endocytic trafficking. In Vps34-/- mouse embryonic fibroblasts (MEFs), transferrin recycling and EEA1 membrane localization were unaffected despite elevated Rab5-GTP levels. Strikingly, a large increase in Rab7-GTP levels, an accumulation of enlarged late endosomes, and decreased EGFR degradation were observed in Vps34-deficient cells. The hyperactivation of Rab7 in Vps34-deficient cells stemmed from the failure to recruit the Rab7 GTPase-activating protein (GAP) Armus (also known as TBC1D2), which binds to PI(3)P, to late endosomes. Protein-lipid overlay and liposome-binding assays reveal that the putative pleckstrin homology (PH) domain in Armus can directly bind to PI(3)P. Elevated Rab7-GTP led to the failure of intraluminal vesicle (ILV) formation and lysosomal maturation. Rab7 silencing and Armus overexpression alleviated the vacuolization seen in Vps34-deficient cells. Taken together, these results demonstrate that Vps34 has a previously unknown role in regulating Rab7 activity and late endosomal trafficking.


Asunto(s)
Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Endocitosis , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Autofagia , Biocatálisis , Endosomas/metabolismo , Endosomas/ultraestructura , Fibroblastos/metabolismo , Células HeLa , Humanos , Lisosomas/metabolismo , Lisosomas/ultraestructura , Ratones Noqueados , Fosfatos de Fosfatidilinositol/metabolismo , Transporte de Proteínas , Serina-Treonina Quinasas TOR/metabolismo , Vacuolas/metabolismo , Vacuolas/ultraestructura , Proteínas de Unión a GTP rab7
20.
J Cardiovasc Pharmacol ; 69(4): 198-211, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28376509

RESUMEN

Receptors that activate the heterotrimeric G protein Gαq are thought to play a role in the development of heart failure. Dysregulation of autophagy occurs in some pathological cardiac conditions including heart failure, but whether Gαq is involved in this process is unknown. We used a cardiomyocyte-specific transgenic mouse model of inducible Gαq activation (termed GαqQ209L) to address this question. After 7 days of Gαq activation, GαqQ209L hearts contained more autophagic vacuoles than wild type hearts. Increased levels of proteins involved in autophagy, especially p62 and LC3-II, were also seen. LysoTracker staining and western blotting showed that the number and size of lysosomes and lysosomal protein levels were increased in GαqQ209L hearts, indicating enhanced lysosomal degradation activity. Importantly, an autophagic flux assay measuring LC3-II turnover in isolated adult cardiomyocytes indicated that autophagic activity is enhanced in GαqQ209L hearts. GαqQ209L hearts exhibited elevated levels of the autophagy initiation complex, which contains the Class III phosphoinositide 3-kinase Vps34. As a consequence, Vps34 activity and phosphatidylinositol 3-phosphate levels were higher in GαqQ209L hearts than wild type hearts, thus accounting for the higher abundance of autophagic vacuoles. These results indicate that an increase in autophagy is an early response to Gαq activation in the heart.


Asunto(s)
Autofagia/fisiología , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Células Cultivadas , Activación Enzimática/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA