Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(17)2023 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-37686063

RESUMEN

Amino acid availability is crucial for cancer cells' survivability. Leukemia and colorectal cancer cells have been shown to resist asparagine depletion by utilizing GSK3-dependent proteasomal degradation, termed the Wnt-dependent stabilization of proteins (Wnt/STOP), to replenish their amino acid pool. The inhibition of GSK3α halts the sourcing of amino acids, which subsequently leads to cancer cell vulnerability toward asparaginase therapy. However, resistance toward GSK3α-mediated protein breakdown can occur, whose underlying mechanism is poorly understood. Here, we set out to define the mechanisms driving dependence toward this degradation machinery upon asparagine starvation in cancer cells. We show the independence of known stress response pathways including the integrated stress response mediated with GCN2. Additionally, we demonstrate the independence of changes in cell cycle progression and expression levels of the asparagine-synthesizing enzyme ASNS. Instead, RNA sequencing revealed that GSK3α inhibition and asparagine starvation leads to the temporally dynamic downregulation of distinct ribosomal proteins, which have been shown to display anti-proliferative functions. Using a CRISPR/Cas9 viability screen, we demonstrate that the downregulation of these specific ribosomal proteins can rescue cell death upon GSK3α inhibition and asparagine starvation. Thus, our findings suggest the vital role of the previously unrecognized regulation of ribosomal proteins in bridging GSK3α activity and tolerance of asparagine starvation.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Neoplasias , Aminoácidos , Asparagina , Glucógeno Sintasa Quinasa 3/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinasas , Proteínas Ribosómicas/genética , Humanos
2.
Mol Cell ; 82(15): 2858-2870.e8, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35732190

RESUMEN

The tolerance of amino acid starvation is fundamental to robust cellular fitness. Asparagine depletion is lethal to some cancer cells, a vulnerability that can be exploited clinically. We report that resistance to asparagine starvation is uniquely dependent on an N-terminal low-complexity domain of GSK3α, which its paralog GSK3ß lacks. In response to depletion of specific amino acids, including asparagine, leucine, and valine, this domain mediates supramolecular assembly of GSK3α with ubiquitin-proteasome system components in spatially sequestered cytoplasmic bodies. This effect is independent of mTORC1 or GCN2. In normal cells, GSK3α promotes survival during essential amino acid starvation. In human leukemia, GSK3α body formation predicts asparaginase resistance, and sensitivity to asparaginase combined with a GSK3α inhibitor. We propose that GSK3α body formation provides a cellular mechanism to maximize the catalytic efficiency of proteasomal protein degradation in response to amino acid starvation, an adaptive response co-opted by cancer cells for asparaginase resistance.


Asunto(s)
Asparaginasa , Leucemia , Aminoácidos/metabolismo , Asparaginasa/genética , Asparaginasa/metabolismo , Asparaginasa/farmacología , Asparagina , Humanos , Proteínas Serina-Treonina Quinasas
3.
Gastroenterology ; 159(6): 2130-2145.e5, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32805279

RESUMEN

BACKGROUND & AIMS: After birth, the immune system matures via interactions with microbes in the gut. The S100 calcium binding proteins S100A8 and S100A9, and their extracellular complex form, S100A8-A9, are found in high amounts in human breast milk. We studied levels of S100A8-A9 in fecal samples (also called fecal calprotectin) from newborns and during infancy, and their effects on development of the intestinal microbiota and mucosal immune system. METHODS: We collected stool samples (n = 517) from full-term (n = 72) and preterm infants (n = 49) at different timepoints over the first year of life (days 1, 3, 10, 30, 90, 180, and 360). We measured levels of S100A8-A9 by enzyme-linked immunosorbent assay and analyzed fecal microbiomes by 16S sRNA gene sequencing. We also obtained small and large intestine biopsies from 8 adults and 10 newborn infants without inflammatory bowel diseases (controls) and 8 infants with necrotizing enterocolitis and measured levels of S100A8 by immunofluorescence microscopy. Children were followed for 2.5 years and anthropometric data and medical information on infections were collected. We performed studies with newborn C57BL/6J wild-type and S100a9-/- mice (which also lack S100A8). Some mice were fed or given intraperitoneal injections of S100A8 or subcutaneous injections of Staphylococcus aureus. Blood and intestine, mesenterial and celiac lymph nodes were collected; cells and cytokines were measured by flow cytometry and studied in cell culture assays. Colon contents from mice were analyzed by culture-based microbiology assays. RESULTS: Loss of S100A8 and S100A9 in mice altered the phenotypes of colonic lamina propria macrophages, compared with wild-type mice. Intestinal tissues from neonatal S100-knockout mice had reduced levels of CX3CR1 protein, and Il10 and Tgfb1 mRNAs, compared with wild-type mice, and fewer T-regulatory cells. S100-knockout mice weighed 21% more than wild-type mice at age 8 weeks and a higher proportion developed fatal sepsis during the neonatal period. S100-knockout mice had alterations in their fecal microbiomes, with higher abundance of Enterobacteriaceae. Feeding mice S100 at birth prevented the expansion of Enterobacteriaceae, increased numbers of T-regulatory cells and levels of CX3CR1 protein and Il10 mRNA in intestine tissues, and reduced body weight and death from neonatal sepsis. Fecal samples from term infants, but not preterm infants, had significantly higher levels of S100A8-A9 during the first 3 months of life than fecal samples from adults; levels decreased to adult levels after weaning. Fecal samples from infants born by cesarean delivery had lower levels of S100A8-A9 than from infants born by vaginal delivery. S100 proteins were expressed by lamina propria macrophages in intestinal tissues from infants, at higher levels than in intestinal tissues from adults. High fecal levels of S100 proteins, from 30 days to 1 year of age, were associated with higher abundance of Actinobacteria and Bifidobacteriaceae, and lower abundance of Gammaproteobacteria-particularly opportunistic Enterobacteriaceae. A low level of S100 proteins in infants' fecal samples associated with development of sepsis and obesity by age 2 years. CONCLUSION: S100A8 and S100A9 regulate development of the intestinal microbiota and immune system in neonates. Nutritional supplementation with these proteins might aide in development of preterm infants and prevent microbiota-associated disorders in later years.


Asunto(s)
Calgranulina A/metabolismo , Calgranulina B/metabolismo , Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Adulto , Animales , Biopsia , Calgranulina A/administración & dosificación , Calgranulina A/análisis , Calgranulina B/análisis , Calgranulina B/genética , Preescolar , Colon/microbiología , Colon/patología , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Disbiosis/microbiología , Disbiosis/prevención & control , Enterocolitis Necrotizante/epidemiología , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Heces/química , Heces/microbiología , Femenino , Estudios de Seguimiento , Microbioma Gastrointestinal/genética , Humanos , Inmunidad Mucosa , Lactante , Recién Nacido , Recien Nacido Prematuro/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Noqueados , Obesidad/epidemiología , Obesidad/inmunología , Obesidad/microbiología , Obesidad/prevención & control , ARN Ribosómico 16S/genética , Sepsis/epidemiología , Sepsis/inmunología , Sepsis/microbiología , Sepsis/prevención & control
4.
Cancer Discov ; 10(11): 1690-1705, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32703769

RESUMEN

Colorectal cancer is driven by mutations that activate canonical WNT/ß-catenin signaling, but inhibiting WNT has significant on-target toxicity, and there are no approved therapies targeting dominant oncogenic drivers. We recently found that activating a ß-catenin-independent branch of WNT signaling that inhibits GSK3-dependent protein degradation induces asparaginase sensitivity in drug-resistant leukemias. To test predictions from our model, we turned to colorectal cancer because these cancers can have WNT-activating mutations that function either upstream (i.e., R-spondin fusions) or downstream (APC or ß-catenin mutations) of GSK3, thus allowing WNT/ß-catenin and WNT-induced asparaginase sensitivity to be unlinked genetically. We found that asparaginase had little efficacy in APC or ß-catenin-mutant colorectal cancer, but was profoundly toxic in the setting of R-spondin fusions. Pharmacologic GSK3α inhibition was sufficient for asparaginase sensitization in APC or ß-catenin-mutant colorectal cancer, but not in normal intestinal progenitors. Our findings demonstrate that WNT-induced therapeutic vulnerabilities can be exploited for colorectal cancer therapy. SIGNIFICANCE: Solid tumors are thought to be asparaginase-resistant via de novo asparagine synthesis. In leukemia, GSK3α-dependent protein degradation, a catabolic amino acid source, mediates asparaginase resistance. We found that asparaginase is profoundly toxic to colorectal cancers with WNT-activating mutations that inhibit GSK3. Aberrant WNT activation can provide a therapeutic vulnerability in colorectal cancer.See related commentary by Davidsen and Sullivan, p. 1632.This article is highlighted in the In This Issue feature, p. 1611.


Asunto(s)
Asparaginasa/metabolismo , Neoplasias Colorrectales/genética , Vía de Señalización Wnt/genética , Línea Celular Tumoral , Humanos
7.
Nat Immunol ; 18(6): 622-632, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28459433

RESUMEN

The high risk of neonatal death from sepsis is thought to result from impaired responses by innate immune cells; however, the clinical observation of hyperinflammatory courses of neonatal sepsis contradicts this concept. Using transcriptomic, epigenetic and immunological approaches, we demonstrated that high amounts of the perinatal alarmins S100A8 and S100A9 specifically altered MyD88-dependent proinflammatory gene programs. S100 programming prevented hyperinflammatory responses without impairing pathogen defense. TRIF-adaptor-dependent regulatory genes remained unaffected by perinatal S100 programming and responded strongly to lipopolysaccharide, but were barely expressed. Steady-state expression of TRIF-dependent genes increased only gradually during the first year of life in human neonates, shifting immune regulation toward the adult phenotype. Disruption of this critical sequence of transient alarmin programming and subsequent reprogramming of regulatory pathways increased the risk of hyperinflammation and sepsis. Collectively these data suggest that neonates are characterized by a selective, transient microbial unresponsiveness that prevents harmful hyperinflammation in the delicate neonate while allowing for sufficient immunological protection.


Asunto(s)
Calgranulina A/inmunología , Calgranulina B/inmunología , Inmunidad Innata/inmunología , Monocitos/inmunología , Sepsis Neonatal/inmunología , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Animales Recién Nacidos , Calgranulina A/efectos de los fármacos , Calgranulina B/efectos de los fármacos , Epigénesis Genética , Sangre Fetal , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inmunidad Innata/efectos de los fármacos , Immunoblotting , Recién Nacido , Inflamación , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Monocitos/efectos de los fármacos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Sepsis Neonatal/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Toll-Like 4/inmunología
8.
Pancreas ; 41(2): 218-21, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21946813

RESUMEN

OBJECTIVES: This study aimed to identify microRNAs as novel biomarkers for improved diagnosis, prognosis prediction, and as a therapeutic target for pancreatic cancer. microRNAs may have a general role by acting as superordinated key regulators of tumorigenesis. METHODS: Individual cellular molecules of multiple pathways associated with pancreatic cancer were analyzed for common microRNA binding sites, thereby enabling the identification of key regulating microRNAs. The potential of the identified microRNAs was subsequently determined in cell culture experiments. RESULTS: Using bioinformatic pathway analyses, miR-548d was identified to target multiple components of pancreatic cancer-related pathways. The effect of microRNA on pancreatic cells was determined by overexpression studies using PANC-1 cells, resulting in impaired cell proliferation because of increased apoptosis and cell cycle arrest. In addition, miR-548d overexpression led to a sensitization to gemcitabine. CONCLUSIONS: MicroRNA miR-548d was identified as a potential superior regulator for the development and progression of pancreatic cancer by targeting multiple factors of crucial pathways. Therapeutically, microRNAs with superordinate function, such as miR-548d, may be promising diagnostic and therapeutic tools for the future treatment of pancreatic cancer.


Asunto(s)
MicroARNs/metabolismo , Neoplasias Pancreáticas/genética , Antimetabolitos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Sitios de Unión , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Biología Computacional , Bases de Datos Genéticas , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Transfección , Gemcitabina
9.
Int J Cancer ; 129(12): 2797-806, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21618216

RESUMEN

The expression of microRNAs is altered in various cancer types, leading to their definition as onco- and tumor-suppressor microRNAs. In our study, we investigated the role of miR-335 in the formation of sporadic human breast cancer and its involvement in the regulatory network of the breast cancer susceptibility gene BRCA1. To validate single components of the BRCA1 cascade, microRNA overexpression was performed in a cell culture model with subsequent protein analysis and luciferase reporter assays. Here, we were able to identify miR-335 as simultaneously regulating the known BRCA1 activators ERα, IGF1R, SP1 and the repressor ID4, including a feedback regulation of miR-335 expression by estrogens. Overexpression of miR-335 resulted in an upregulation of BRCA1 mRNA expression, suggesting a functional dominance of ID4 signaling. The relevance of the miR-335 regulation for human breast cancer was confirmed in primary sporadic breast cancer specimens with significantly decreased miR-335 levels (p < 0.05) in comparison to normal controls. Interestingly, the microRNA expression level correlated positively to the BRCA1 transcript level, supporting the hypothesis of a miR-335-mediated regulation of the tumor suppressor gene. Functionally, overexpression of miR-335 led to decreased cell viability and an increase in apoptosis, supporting its tumor-suppressive function. In summary, our data indicate that miR-335 affects different targets in the upstream BRCA1-regulatory cascade with impact on key cellular functions such as proliferation and apoptosis. Deregulation of the microRNA during breast cancer development and progression may thereby lead to an increased tumorigenic potential by inactivating crucial tumor-suppressive signals.


Asunto(s)
Neoplasias de la Mama/genética , Genes BRCA1 , MicroARNs/fisiología , Anciano , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Estrógenos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Transducción de Señal , Regulación hacia Arriba
10.
Blood ; 108(2): 493-500, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16551967

RESUMEN

Severe congenital neutropenia (SCN) and cyclic neutropenia (CyN) are sporadic or inherited hematologic disorders of myelopoiesis. Heterozygous mutations in the gene encoding neutrophil elastase (ELA2) have been reported in both diseases. We used an inducible system to express a panel of ELA2 mutations and found for almost all mutants disruption of intracellular neutrophil elastase (HNE) protein processing at different levels. This disruption resulted in cytoplasmic accumulation of a nonfunctional protein, thereby preventing its physiologic transport to azurophil granules. Furthermore, the secretory capacity of the mutant proteins was greatly diminished, indicating alteration of the regulated and the constitutive pathways. Through analysis of primary granulocytes from SCN patients carrying ELA2 mutations, we found an identical pattern of intracellular accumulation of mutant HNE protein in the cytoplasm. Moreover, cells expressing mutant HNE protein exhibited a significant increase in apoptosis associated with up-regulation of the master ER chaperone BiP, indicating that disturbance of intracellular trafficking results in activation of the mammalian unfolded protein response.


Asunto(s)
Elastasa de Leucocito/genética , Neutropenia/congénito , Neutropenia/etiología , Apoptosis , Citoplasma/química , Chaperón BiP del Retículo Endoplásmico , Proteínas de Choque Térmico/genética , Humanos , Elastasa de Leucocito/metabolismo , Chaperonas Moleculares/genética , Proteínas Mutantes , Mutación , Pliegue de Proteína , Transporte de Proteínas , Regulación hacia Arriba/genética
11.
Oncogene ; 24(12): 1982-93, 2005 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-15674325

RESUMEN

The proto-oncoprotein c-Myc functions as a transcriptional regulator that controls different aspects of cell behavior, including proliferation, differentiation, and apoptosis. In addition, Myc proteins have the potential to transform cells and are deregulated in the majority of human cancers. Several Myc-interacting factors have been described that mediate part of Myc's functions in the control of cell behavior. Here, we describe the isolation of a novel 150 kDa protein, designated PARP-10, that interacts with Myc. PARP-10 possesses domains with homology to RNA recognition motifs and to poly(ADP-ribose) polymerases (PARP). Molecular modeling and biochemical analysis define a PARP domain that is capable of ADP-ribosylating PARP-10 itself and core histones, but neither Myc nor Max. PARP-10 is localized to the nuclear and cytoplasmic compartments that is controlled at least in part by a Leu-rich nuclear export sequence (NES). Functionally, PARP-10 inhibits c-Myc- and E1A-mediated cotransformation of rat embryo fibroblasts, a function that is independent of PARP activity but that depends on a functional NES. Together, our findings define a novel PARP enzyme involved in the control of cell proliferation.


Asunto(s)
Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Secuencia de Aminoácidos , División Celular , Línea Celular , Mapeo Cromosómico , Humanos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular , Plásmidos , Biosíntesis de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...