Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(10): 1884-1899, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39327500

RESUMEN

TCF1high progenitor CD8+ T cells mediate the efficacy of immunotherapy; however, the mechanisms that govern their generation and maintenance are poorly understood. Here, we show that targeting glycolysis through deletion of pyruvate kinase muscle 2 (PKM2) results in elevated pentose phosphate pathway (PPP) activity, leading to enrichment of a TCF1high progenitor-exhausted-like phenotype and increased responsiveness to PD-1 blockade in vivo. PKM2KO CD8+ T cells showed reduced glycolytic flux, accumulation of glycolytic intermediates and PPP metabolites and increased PPP cycling as determined by 1,2-13C glucose carbon tracing. Small molecule agonism of the PPP without acute glycolytic impairment skewed CD8+ T cells toward a TCF1high population, generated a unique transcriptional landscape and adoptive transfer of agonist-treated CD8+ T cells enhanced tumor control in mice in combination with PD-1 blockade and promoted tumor killing in patient-derived tumor organoids. Our study demonstrates a new metabolic reprogramming that contributes to a progenitor-like T cell state promoting immunotherapy efficacy.


Asunto(s)
Linfocitos T CD8-positivos , Factor Nuclear 1-alfa del Hepatocito , Inmunoterapia , Vía de Pentosa Fosfato , Proteínas de Unión a Hormona Tiroide , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Ratones , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Inmunoterapia/métodos , Glucólisis , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Ratones Noqueados , Hormonas Tiroideas/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones Endogámicos C57BL , Neoplasias/inmunología , Neoplasias/terapia , Neoplasias/metabolismo , Piruvato Quinasa
2.
Nat Rev Mol Cell Biol ; 19(1): 31-44, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28974775

RESUMEN

The mechanism of insulin action is a central theme in biology and medicine. In addition to the rather rare condition of insulin deficiency caused by autoimmune destruction of pancreatic ß-cells, genetic and acquired abnormalities of insulin action underlie the far more common conditions of type 2 diabetes, obesity and insulin resistance. The latter predisposes to diseases ranging from hypertension to Alzheimer disease and cancer. Hence, understanding the biochemical and cellular properties of insulin receptor signalling is arguably a priority in biomedical research. In the past decade, major progress has led to the delineation of mechanisms of glucose transport, lipid synthesis, storage and mobilization. In addition to direct effects of insulin on signalling kinases and metabolic enzymes, the discovery of mechanisms of insulin-regulated gene transcription has led to a reassessment of the general principles of insulin action. These advances will accelerate the discovery of new treatment modalities for diabetes.


Asunto(s)
Insulina/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Transcripción Genética/fisiología
3.
Cell ; 159(2): 318-32, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25303528

RESUMEN

Increased adipose tissue lipogenesis is associated with enhanced insulin sensitivity. Mice overexpressing the Glut4 glucose transporter in adipocytes have elevated lipogenesis and increased glucose tolerance despite being obese with elevated circulating fatty acids. Lipidomic analysis of adipose tissue revealed the existence of branched fatty acid esters of hydroxy fatty acids (FAHFAs) that were elevated 16- to 18-fold in these mice. FAHFA isomers differ by the branched ester position on the hydroxy fatty acid (e.g., palmitic-acid-9-hydroxy-stearic-acid, 9-PAHSA). PAHSAs are synthesized in vivo and regulated by fasting and high-fat feeding. PAHSA levels correlate highly with insulin sensitivity and are reduced in adipose tissue and serum of insulin-resistant humans. PAHSA administration in mice lowers ambient glycemia and improves glucose tolerance while stimulating GLP-1 and insulin secretion. PAHSAs also reduce adipose tissue inflammation. In adipocytes, PAHSAs signal through GPR120 to enhance insulin-stimulated glucose uptake. Thus, FAHFAs are endogenous lipids with the potential to treat type 2 diabetes.


Asunto(s)
Tejido Adiposo/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ésteres/metabolismo , Ácidos Grasos/metabolismo , Adulto , Animales , Diabetes Mellitus Tipo 2/dietoterapia , Dieta , Ésteres/administración & dosificación , Ésteres/análisis , Ácidos Grasos/administración & dosificación , Ácidos Grasos/análisis , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Humanos , Inflamación/dietoterapia , Insulina/metabolismo , Resistencia a la Insulina , Lipogénesis , Masculino , Espectrometría de Masas , Ratones Endogámicos C57BL , Persona de Mediana Edad , Receptores Acoplados a Proteínas G/metabolismo
4.
J Cell Sci ; 136(23)2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-38126809

RESUMEN

Regulation of glucose transport, which is central for control of whole-body metabolism, is determined by the amount of GLUT4 glucose transporter (also known as SLC2A4) in the plasma membrane (PM) of fat and muscle cells. Physiologic signals [such as activated insulin receptor or AMP-activated protein kinase (AMPK)] increase PM GLUT4. Here, we show that the distribution of GLUT4 between the PM and interior of human muscle cells is dynamically maintained, and that AMPK promotes PM redistribution of GLUT4 by regulating exocytosis and endocytosis. Stimulation of exocytosis by AMPK is mediated by Rab10 and the Rab GTPase-activating protein TBC1D4. APEX2 proximity mapping reveals that GLUT4 traverses both PM-proximal and PM-distal compartments in unstimulated muscle cells, further supporting retention of GLUT4 by a constitutive retrieval mechanism. AMPK-stimulated translocation involves GLUT4 redistribution among the same compartments traversed in unstimulated cells, with a significant recruitment of GLUT4 from the Golgi and trans-Golgi network compartments. Our comprehensive proximal protein mapping provides an integrated, high-density, whole-cell accounting of the localization of GLUT4 at a resolution of ∼20 nm that serves as a structural framework for understanding the molecular mechanisms regulating GLUT4 trafficking downstream of different signaling inputs in a physiologically relevant cell type.


Asunto(s)
Transportador de Glucosa de Tipo 4 , Células Musculares , Proteoma , Humanos , Proteínas Quinasas Activadas por AMP , Membrana Celular , Músculos , Transportador de Glucosa de Tipo 4/metabolismo
5.
Cell Commun Signal ; 21(1): 58, 2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36915197

RESUMEN

BACKGROUND: PD-L1, a transmembrane ligand for immune checkpoint receptor PD1, has been successfully targeted to activate an anti-tumor immune response in a variety of solid tumors, including non-small cell lung cancer (NSCLC). Despite the success of targeting PD-L1, only about 20% of patients achieve a durable response. The reasons for the heterogeneity in response are not understood, although some molecular subtypes (e.g., mutant EGF receptor tumors) are generally poor responders. Although PD-L1 is best characterized as a transmembrane PD1 ligand, the emerging view is that PD-L1 has functions independent of activating PD1 signaling. It is not known whether these cell-intrinsic functions of PD-L1 are shared among non-transformed and transformed cells, if they vary among cancer molecular subtypes, or if they are impacted by anti-PD-L1 therapy. METHODS: Here we use quantitative microscopy techniques and APEX2 proximity mapping to describe the behavior of PD-L1 and to identify PD-L1's proximal proteome in human lung epithelial cells. RESULTS: Our data reveal growth factor control of PD-L1 recycling as a mechanism for acute and reversible regulation of PD-L1 density on the plasma membrane. In addition, we describe novel PD-L1 biology restricted to mutant EGFR cells. Anti-PD-L1 antibody treatment of mutant EGFR cells perturbs cell intrinsic PD-L1 functions, leading to reduced cell migration, increased half-life of EGFR and increased extracellular vesicle biogenesis, whereas anti-PD-L1 antibody does not induce these changes in wild type EGFR cells. CONCLUSIONS: Growth factor acute regulation of PD-L1 trafficking, by contributing to the control of plasma membrane density, might contribute to the regulation of PD-L1's immune checkpoint activity, whereas the specific effects of anti-PD-L1 on mutant EGFR cells might contribute to the poor anti-PD-L1 response of mutant EGFR tumors. Video Abstract.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteoma , Ligandos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Pulmón/metabolismo , Antígeno B7-H1/metabolismo , Mutación
6.
Lancet Oncol ; 22(6): 824-835, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34015311

RESUMEN

BACKGROUND: Previous phase 2 trials of neoadjuvant anti-PD-1 or anti-PD-L1 monotherapy in patients with early-stage non-small-cell lung cancer have reported major pathological response rates in the range of 15-45%. Evidence suggests that stereotactic body radiotherapy might be a potent immunomodulator in advanced non-small-cell lung cancer (NSCLC). In this trial, we aimed to evaluate the use of stereotactic body radiotherapy in patients with early-stage NSCLC as an immunomodulator to enhance the anti-tumour immune response associated with the anti-PD-L1 antibody durvalumab. METHODS: We did a single-centre, open-label, randomised, controlled, phase 2 trial, comparing neoadjuvant durvalumab alone with neoadjuvant durvalumab plus stereotactic radiotherapy in patients with early-stage NSCLC, at NewYork-Presbyterian and Weill Cornell Medical Center (New York, NY, USA). We enrolled patients with potentially resectable early-stage NSCLC (clinical stages I-IIIA as per the 7th edition of the American Joint Committee on Cancer) who were aged 18 years or older with an Eastern Cooperative Oncology Group performance status of 0 or 1. Eligible patients were randomly assigned (1:1) to either neoadjuvant durvalumab monotherapy or neoadjuvant durvalumab plus stereotactic body radiotherapy (8 Gy × 3 fractions), using permuted blocks with varied sizes and no stratification for clinical or molecular variables. Patients, treating physicians, and all study personnel were unmasked to treatment assignment after all patients were randomly assigned. All patients received two cycles of durvalumab 3 weeks apart at a dose of 1·12 g by intravenous infusion over 60 min. Those in the durvalumab plus radiotherapy group also received three consecutive daily fractions of 8 Gy stereotactic body radiotherapy delivered to the primary tumour immediately before the first cycle of durvalumab. Patients without systemic disease progression proceeded to surgical resection. The primary endpoint was major pathological response in the primary tumour. All analyses were done on an intention-to-treat basis. This trial is registered with ClinicalTrial.gov, NCT02904954, and is ongoing but closed to accrual. FINDINGS: Between Jan 25, 2017, and Sept 15, 2020, 96 patients were screened and 60 were enrolled and randomly assigned to either the durvalumab monotherapy group (n=30) or the durvalumab plus radiotherapy group (n=30). 26 (87%) of 30 patients in each group had their tumours surgically resected. Major pathological response was observed in two (6·7% [95% CI 0·8-22·1]) of 30 patients in the durvalumab monotherapy group and 16 (53·3% [34·3-71·7]) of 30 patients in the durvalumab plus radiotherapy group. The difference in the major pathological response rates between both groups was significant (crude odds ratio 16·0 [95% CI 3·2-79·6]; p<0·0001). In the 16 patients in the dual therapy group with a major pathological response, eight (50%) had a complete pathological response. The second cycle of durvalumab was withheld in three (10%) of 30 patients in the dual therapy group due to immune-related adverse events (grade 3 hepatitis, grade 2 pancreatitis, and grade 3 fatigue and thrombocytopaenia). Grade 3-4 adverse events occurred in five (17%) of 30 patients in the durvalumab monotherapy group and six (20%) of 30 patients in the durvalumab plus radiotherapy group. The most frequent grade 3-4 events were hyponatraemia (three [10%] patients in the durvalumab monotherapy group) and hyperlipasaemia (three [10%] patients in the durvalumab plus radiotherapy group). Two patients in each group had serious adverse events (pulmonary embolism [n=1] and stroke [n=1] in the durvalumab monotherapy group, and pancreatitis [n=1] and fatigue [n=1] in the durvalumab plus radiotherapy group). No treatment-related deaths or deaths within 30 days of surgery were reported. INTERPRETATION: Neoadjuvant durvalumab combined with stereotactic body radiotherapy is well tolerated, safe, and associated with a high major pathological response rate. This neoadjuvant strategy should be validated in a larger trial. FUNDING: AstraZeneca.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antígeno B7-H1/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Adolescente , Adulto , Anciano , Anticuerpos Monoclonales/efectos adversos , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Terapia Combinada , Femenino , Humanos , Masculino , Persona de Mediana Edad , Terapia Neoadyuvante/efectos adversos , Estadificación de Neoplasias , Radiocirugia/métodos , Adulto Joven
7.
Mol Cell ; 49(6): 1167-75, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23453806

RESUMEN

Thioredoxin-interacting protein (TXNIP) is an α-arrestin family protein that is induced in response to glucose elevation. It has been shown to provide a negative feedback loop to regulate glucose uptake into cells, though the biochemical mechanism of action has been obscure. Here, we report that TXNIP suppresses glucose uptake directly, by binding to the glucose transporter GLUT1 and inducing GLUT1 internalization through clathrin-coated pits, as well as indirectly, by reducing the level of GLUT1 messenger RNA (mRNA). In addition, we show that energy stress results in the phosphorylation of TXNIP by AMP-dependent protein kinase (AMPK), leading to its rapid degradation. This suppression of TXNIP results in an acute increase in GLUT1 function and an increase in GLUT1 mRNA (hence the total protein levels) for long-term adaptation. The glucose influx through GLUT1 restores ATP-to-ADP ratios in the short run and ultimately induces TXNIP protein production to suppress glucose uptake once energy homeostasis is reestablished.


Asunto(s)
Adenilato Quinasa/fisiología , Proteínas Portadoras/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Glucosa/metabolismo , Proteolisis , Secuencia de Aminoácidos , Animales , Vesículas Cubiertas por Clatrina/metabolismo , Secuencia Conservada , Endocitosis , Células Hep G2 , Humanos , Ratones , Datos de Secuencia Molecular , Transporte de Proteínas , Imagen de Lapso de Tiempo
8.
J Biol Chem ; 294(30): 11369-11381, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31175156

RESUMEN

A pivotal metabolic function of insulin is the stimulation of glucose uptake into muscle and adipose tissues. The discovery of the insulin-responsive glucose transporter type 4 (GLUT4) protein in 1988 inspired its molecular cloning in the following year. It also spurred numerous cellular mechanistic studies laying the foundations for how insulin regulates glucose uptake by muscle and fat cells. Here, we reflect on the importance of the GLUT4 discovery and chronicle additional key findings made in the past 30 years. That exocytosis of a multispanning membrane protein regulates cellular glucose transport illuminated a novel adaptation of the secretory pathway, which is to transiently modulate the protein composition of the cellular plasma membrane. GLUT4 controls glucose transport into fat and muscle tissues in response to insulin and also into muscle during exercise. Thus, investigation of regulated GLUT4 trafficking provides a major means by which to map the essential signaling components that transmit the effects of insulin and exercise. Manipulation of the expression of GLUT4 or GLUT4-regulating molecules in mice has revealed the impact of glucose uptake on whole-body metabolism. Remaining gaps in our understanding of GLUT4 function and regulation are highlighted here, along with opportunities for future discoveries and for the development of therapeutic approaches to manage metabolic disease.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Animales , Transporte Biológico , Clonación Molecular , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Transducción de Señal
9.
J Cosmet Laser Ther ; 20(4): 211-214, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28498021

RESUMEN

BACKGROUND: Becker's Nevus is an aesthetically troublesome condition secondary to hyperpigmentation and hypertrichosis. Although several lasers have been employed with varying degrees of success, the exact devices and treatment parameters have yet to be elucidated. OBJECTIVE: To assess the combination Nd:YAG and alexandrite laser as a safe and efficacious treatment for Becker's Nevus. METHODS: In a 20-year-old Fitzpatrick Skin Type IV male, a Becker's Nevus was treated with six sessions of long-pulsed 1064 nm Nd:YAG laser at six-week intervals followed by five sessions of long-pulsed 755 nm alexandrite laser at three-month intervals. RESULTS: This patient experienced a significant reduction in both hyperpigmentation and hypertrichosis after these treatment sessions. No serious adverse events were reported. CONCLUSION: This case supports the use of combination long-pulsed 1064 nm laser and 755 nm laser as a safe and efficacious treatment for Becker's Nevus.


Asunto(s)
Láseres de Estado Sólido/uso terapéutico , Nevo/cirugía , Neoplasias Cutáneas/cirugía , Humanos , Hiperpigmentación/complicaciones , Hiperpigmentación/cirugía , Hipertricosis/complicaciones , Hipertricosis/cirugía , Masculino , Nevo/complicaciones , Neoplasias Cutáneas/complicaciones , Adulto Joven
10.
J Biol Chem ; 291(18): 9648-56, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-26917725

RESUMEN

Insulin resistance and ß cell dysfunction contribute to the pathogenesis of type 2 diabetes. Unlike insulin resistance, ß cell dysfunction remains difficult to predict and monitor, because of the inaccessibility of the endocrine pancreas, the integrated relationship with insulin sensitivity, and the paracrine effects of incretins. The goal of our study was to survey the plasma response to a metabolic challenge in order to identify factors predictive of ß cell dysfunction. To this end, we combined (i) the power of unbiased iTRAQ (isobaric tag for relative and absolute quantification) mass spectrometry with (ii) direct sampling of the portal vein following an intravenous glucose/arginine challenge (IVGATT) in (iii) mice with a genetic ß cell defect. By so doing, we excluded the effects of peripheral insulin sensitivity as well as those of incretins on ß cells, and focused on the first phase of insulin secretion to capture the early pathophysiology of ß cell dysfunction. We compared plasma protein profiles with ex vivo islet secretome and transcriptome analyses. We detected changes to 418 plasma proteins in vivo, and detected changes to 262 proteins ex vivo The impairment of insulin secretion was associated with greater overall changes in the plasma response to IVGATT, possibly reflecting metabolic instability. Reduced levels of proteins regulating redox state and neuronal stress markers, as well as increased levels of coagulation factors, antedated the loss of insulin secretion in diabetic mice. These results suggest that a reduced complement of antioxidants in response to a mixed secretagogue challenge is an early correlate of future ß cell failure.


Asunto(s)
Antioxidantes/metabolismo , Proteínas Sanguíneas/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Tipo 2/sangre , Células Secretoras de Insulina/metabolismo , Animales , Proteínas Sanguíneas/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Incretinas/sangre , Incretinas/genética , Células Secretoras de Insulina/patología , Ratones , Ratones Noqueados
11.
J Cell Sci ; 128(16): 3143-54, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26116575

RESUMEN

The key proteins mediating store-operated Ca(2+) entry (SOCE) are the endoplasmic reticulum (ER) Ca(2+) sensor STIM1 and the plasma membrane Ca(2+)-selective channel Orai1. Here, we quantitatively dissect Orai1 trafficking dynamics and show that Orai1 recycles rapidly at the plasma membrane (Kex≃0.1 min(-1)), with ∼40% of the total Orai1 pool localizing to the plasma membrane at steady state. A subset of intracellular Orai1 localizes to a sub-plasmalemal compartment. Store depletion is coupled to Orai1 plasma membrane enrichment in a STIM1-dependent fashion. This is due to trapping of Orai1 into cortical ER STIM1 clusters, leading to its removal from the recycling pool and enrichment at the plasma membrane. Interestingly, upon high STIM1 expression, Orai1 is trapped into STIM1 clusters intracellularly, thus preventing its plasma membrane enrichment following store depletion. Consistent with this, STIM1 knockdown prevents trapping of excess Orai1 into limiting STIM1 clusters in the cortical ER. SOCE-dependent Ca(2+) influx shows a similar biphasic dependence on the Orai1:STIM1 ratio. Therefore, a STIM1-dependent Orai1 'trafficking trap' mechanism controls Orai1 plasma membrane enrichment and SOCE levels, thus modulating the SOCE 'bandwidth' for downstream signaling.


Asunto(s)
Canales de Calcio/genética , Señalización del Calcio/genética , Calcio/metabolismo , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Animales , Células CHO , Canales de Calcio/biosíntesis , Membrana Celular/metabolismo , Cricetulus , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/biosíntesis , Proteínas de Neoplasias/biosíntesis , Proteína ORAI1 , Transporte de Proteínas/genética , ARN Interferente Pequeño , Transducción de Señal , Molécula de Interacción Estromal 1
12.
Dermatol Surg ; 43 Suppl 1: S70-S74, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27631461

RESUMEN

BACKGROUND: Successful treatment of acne scars is challenging. Many modalities have been used with marginal success. OBJECTIVE: The authors describe a combination of a superficial chemical peel, subcision, and fractional carbon dioxide (CO2) laser resurfacing in a single-treatment session as a novel treatment approach for rolling acne scars. METHODS: A total of 114 patients (75 women and 39 men) with predominantly rolling acne scars were treated with a 20% trichloracetic acid chemical peel, subcision with a novel dissecting instrument, and the CO2 laser under tumescent anesthesia. This was a retrospective, uncontrolled, unblinded study. RESULTS: The mean improvement after a single treatment was 2.9 on a scale of 1 to 4 with few complications. Ninety percent of the patients were satisfied with their final result and desired no additional treatment. CONCLUSION: The combination of a trichloracetic acid 20% chemical peel, subcision, and fractional CO2 laser resurfacing combined with tumescent anesthesia is both safe and effective in the treatment of rolling acne scars.


Asunto(s)
Acné Vulgar/complicaciones , Quimioexfoliación/métodos , Cicatriz/terapia , Fármacos Dermatológicos/administración & dosificación , Terapia por Láser/métodos , Ácido Tricloroacético/administración & dosificación , Administración Tópica , Adulto , Anestesia , Cicatriz/etiología , Cicatriz/cirugía , Técnicas Cosméticas , Femenino , Humanos , Láseres de Gas , Masculino , Estudios Retrospectivos , Resultado del Tratamiento
13.
Biochem J ; 468(3): 425-34, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25856301

RESUMEN

Protein kinase B (Akt) kinases are critical signal transducers mediating insulin action. Genetic studies revealed that Akt1 and Akt2 signalling differentially contribute to sustain lipid and glucose homoeostasis; however Akt isoform-specific effectors remain elusive due to the lack of a suitable model system to mechanistically interrogate Akt isoform-specific signalling. To overcome those technical limitations we developed a novel model system that provides acute and specific control of signalling by Akt isoforms. We generated mutants of Akt1 and Akt2 resistant to the allosteric Akt inhibitor MK-2206. We then developed adipocyte cell lines, in which endogenous Akt1 or Akt2 has been replaced by their corresponding drug-resistant Akt mutant. Treatment of those cells with MK-2206 allowed for acute and specific control of either Akt1 or Akt2 function. Our data showed that Akt1(W80A) and Akt2(W80A) mutants are resistant to MK-2206, dynamically regulated by insulin and able to signal to Akt downstream effectors. Analyses of insulin action in this cellular system showed that Akt1 and Akt2 are both able to mediate insulin regulation of the transcription factor forkhead box O1 (FoxO1) and the glucose transporter 4 (GLUT4), revealing a redundant role for these Akt kinases in the control of glucose transport into fat cells. In contrast, Akt1 signalling is uniquely required for adipogenesis, by controlling the mitotic clonal expansion (MCE) of pre-adipocytes that precedes white adipose cell differentiation. Our data provide new insights into the role of Akt kinases in glucose transport and adipogenesis and support our model system as a valuable tool for the biochemical characterization of signalling by specific Akt isoforms.


Asunto(s)
Adipocitos Blancos/enzimología , Adipogénesis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células 3T3-L1 , Adipocitos Blancos/citología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Adipogénesis/efectos de los fármacos , Regulación Alostérica/efectos de los fármacos , Sustitución de Aminoácidos , Animales , Resistencia a Medicamentos , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Resistencia a la Insulina , Ratones , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Interferencia de ARN , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transducción de Señal/efectos de los fármacos
14.
Dermatol Surg ; 42 Suppl 2: S139-49, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27128240

RESUMEN

BACKGROUND: Acne scarring can be classified into atrophic icepick, boxcar, and rolling scars in addition to keloidal and hypertrophic scars. Additionally, these scars can be erythematous, hyperpigmented, and/or hypopigmented. Each scar type has a different structural cause warranting a customized approach. Many cosmetic options exist to address these changes individually, but little literature exists about the safety and efficacy of combining such procedures and devices. METHODS: A Medline search was performed on combination treatments because it relates to facial acne scarring, and results are summarized. Practical applications for these combinations of procedures are also discussed. RESULTS: Studies examining the efficacy and safety of ablative, nonablative, fractionated, and nonfractionated lasers, dermabrasion, chemical peels, needling, subcision, radiofrequency, stem cell therapy, fat transplantation, platelet-rich plasma, and hyaluronic acid dermal fillers for acne scars were found. The authors review their experience in combining these techniques. CONCLUSION: Review of the literature revealed multiple single options for facial acne scarring treatment with minimal evidence in the literature found on the safety and efficacy of combining such procedures and devices. The authors' experience is that combining acne scar treatment techniques can be performed safely and synergistically with optimal patient outcomes.


Asunto(s)
Acné Vulgar/complicaciones , Cicatriz/terapia , Quimioexfoliación , Cicatriz/etiología , Cicatriz/patología , Cicatriz Hipertrófica/terapia , Terapia Combinada , Ultrasonido Enfocado de Alta Intensidad de Ablación , Humanos , Tratamiento de Luz Pulsada Intensa , Queloide/terapia , Terapia por Láser , Agujas , Trastornos de la Pigmentación/etiología , Trastornos de la Pigmentación/terapia
15.
Adv Exp Med Biol ; 890: 75-110, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26703800

RESUMEN

The tumor microenvironment (TME) represents a milieu that enables tumor cells to acquire the hallmarks of cancer. The TME is heterogeneous in composition and consists of cellular components, growth factors, proteases, and extracellular matrix. Concerted interactions between genetically altered tumor cells and genetically stable intratumoral stromal cells result in an "activated/reprogramed" stroma that promotes carcinogenesis by contributing to inflammation, immune suppression, therapeutic resistance, and generating premetastatic niches that support the initiation and establishment of distant metastasis. The lungs present a unique milieu in which tumors progress in collusion with the TME, as evidenced by regions of aberrant angiogenesis, acidosis and hypoxia. Inflammation plays an important role in the pathogenesis of lung cancer, and pulmonary disorders in lung cancer patients such as chronic obstructive pulmonary disease (COPD) and emphysema, constitute comorbid conditions and are independent risk factors for lung cancer. The TME also contributes to immune suppression, induces epithelial-to-mesenchymal transition (EMT) and diminishes efficacy of chemotherapies. Thus, the TME has begun to emerge as the "Achilles heel" of the disease, and constitutes an attractive target for anti-cancer therapy. Drugs targeting the components of the TME are making their way into clinical trials. Here, we will focus on recent advances and emerging concepts regarding the intriguing role of the TME in lung cancer progression, and discuss future directions in the context of novel diagnostic and therapeutic opportunities.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinogénesis/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Comunicación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Humanos , Enfermedades Pulmonares Obstructivas/complicaciones , Enfermedades Pulmonares Obstructivas/tratamiento farmacológico , Enfermedades Pulmonares Obstructivas/genética , Enfermedades Pulmonares Obstructivas/metabolismo , Neoplasias Pulmonares/complicaciones , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neovascularización Patológica/prevención & control , Enfisema Pulmonar/complicaciones , Enfisema Pulmonar/tratamiento farmacológico , Enfisema Pulmonar/genética , Enfisema Pulmonar/metabolismo , Microambiente Tumoral/genética
16.
Proc Natl Acad Sci U S A ; 108(25): 10162-7, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21646544

RESUMEN

Insulin resistance is a component of the metabolic syndrome and Type 2 diabetes. It has been recently shown that in liver insulin resistance is not complete. This so-called selective insulin resistance is characterized by defective insulin inhibition of hepatic glucose output while insulin-induced lipogenesis is maintained. How this occurs and whether uncoupled insulin action develops in other tissues is unknown. Here we show in a model of chronic hyperinsulinemia that adipocytes develop selective insulin resistance in which translocation of the GLUT4 glucose transporter to the cell surface is blunted yet nuclear exclusion of the FoxO1 transcription factor is preserved, rendering uncoupled insulin-controlled carbohydrate and lipid metabolisms. We found that in adipocytes FoxO1 nuclear exclusion has a lower half-maximal insulin dose than GLUT4 translocation, and it is because of this inherent greater sensitivity that control of FoxO1 by physiological insulin concentrations is maintained in adipocytes with compromised insulin signaling. Pharmacological and genetic interventions revealed that insulin regulates GLUT4 and FoxO1 through the PI3-kinase isoform p110α, although FoxO1 showed higher sensitivity to p110α activity than GLUT4. Transient down-regulation and overexpression of Akt isoforms in adipocytes demonstrated that insulin-activated PI3-kinase signals to GLUT4 primarily through Akt2 kinase, whereas Akt1 and Akt2 signal to FoxO1. We propose that the lower threshold of insulin activity for FoxO1's nuclear exclusion is in part due to its regulation by both Akt isoforms. Identification of uncoupled insulin action in adipocytes suggests this condition might be a general phenomenon of insulin target tissues contributing to insulin resistance's pathophysiology.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Hiperinsulinismo/fisiopatología , Insulina/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Transportador de Glucosa de Tipo 4/genética , Humanos , Insulina/farmacología , Resistencia a la Insulina/fisiología , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología
17.
Cell Rep Med ; 5(3): 101438, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38401548

RESUMEN

In early-stage non-small cell lung cancer, the combination of neoadjuvant anti-PD-L1 and subablative stereotactic body radiation therapy (SBRT) is associated with higher rates of major pathologic response compared to anti-PD-L1 alone. Here, we identify a 140-gene set, enriched in genes characteristic of highly proliferating cells, associated with response to the dual therapy. Analysis of on-treatment transcriptome data indicate roles for T and B cells in response. The 140-gene set is associated with disease-free survival when applied to the combined trial arms. This 140-gene set identifies a subclass of tumors in all 7 of The Cancer Genome Atlas tumor types examined. Worse survival is associated with the 140-gene signature in 5 of these tumor types. Collectively, our data support that this 140-gene set, discovered in association with response to combined anti-PD-L1 and SBRT, identifies a clinically aggressive subclass of solid tumors that may be more likely to respond to immunotherapies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Supervivencia sin Progresión , Proliferación Celular/genética
18.
medRxiv ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38947080

RESUMEN

Circulating tumor cells (CTCs) captured from the bloodstream of patients with solid tumors have the potential to accelerate precision oncology by providing insight into tumor biology, disease progression and response to treatment. However, their potential is hampered by the lack of standardized CTC enrichment platforms across tumor types. EpCAM-based CTC enrichment, the most commonly used platform, is limited by EpCAM downregulation during metastasis and the low EpCAM expression in certain tumor types, including the highly prevalent and lethal NSCLC. In this study we demonstrate that Transferrin Receptor (TfR) is a selective, efficient biomarker for CTC identification and capture in patients with prostate, pancreatic and NSCLC. TfR identifies significantly higher CTC counts than EpCAM, and TfR + -CTC enumeration correlates with disease progression in metastatic prostate and pancreatic cancers, and overall survival and osimetrinib-resistance in non-small cell lung cancer (NSCLC). Profiling of TfR + -CTCs provides a snapshot of the molecular landscape of each respective tumor type and identifies potential mechanisms underlying treatment response to EGFR TKi and immune checkpoint inhibitors in NSCLC. One sentence summary: Transferrin Receptor identifies circulating tumor cells in solid tumors.

19.
Nat Commun ; 15(1): 8980, 2024 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-39420001

RESUMEN

The canonical G406R mutation that increases Ca2+ influx through the CACNA1C-encoded CaV1.2 Ca2+ channel underlies the multisystem disorder Timothy syndrome (TS), characterized by life-threatening arrhythmias. Severe episodic hypoglycemia is among the poorly characterized non-cardiac TS pathologies. While hypothesized from increased Ca2+ influx in pancreatic beta cells and consequent hyperinsulinism, this hypoglycemia mechanism is undemonstrated because of limited clinical data and lack of animal models. We generated a CaV1.2 G406R knockin mouse model that recapitulates key TS features, including hypoglycemia. Unexpectedly, these mice do not show hyperactive beta cells or hyperinsulinism in the setting of normal intrinsic beta cell function, suggesting dysregulated glucose homeostasis. Patient data confirm the absence of hyperinsulinism. We discover multiple alternative contributors, including perturbed counterregulatory hormone responses with defects in glucagon secretion and abnormal hypothalamic control of glucose homeostasis. These data provide new insights into contributions of CaV1.2 channels and reveal integrated consequences of the mutant channels driving life-threatening events in TS.


Asunto(s)
Trastorno Autístico , Canales de Calcio Tipo L , Modelos Animales de Enfermedad , Hipoglucemia , Células Secretoras de Insulina , Síndrome de QT Prolongado , Sindactilia , Animales , Canales de Calcio Tipo L/metabolismo , Canales de Calcio Tipo L/genética , Hipoglucemia/metabolismo , Hipoglucemia/genética , Células Secretoras de Insulina/metabolismo , Sindactilia/genética , Sindactilia/metabolismo , Sindactilia/patología , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Ratones , Humanos , Masculino , Glucagón/metabolismo , Femenino , Mutación , Glucosa/metabolismo , Calcio/metabolismo , Técnicas de Sustitución del Gen , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Insulina/metabolismo , Homeostasis , Hipotálamo/metabolismo , Glucemia/metabolismo
20.
Cancer Immunol Res ; 11(7): 866-874, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37290119

RESUMEN

Programmed death-ligand 1 (PD-L1) is a transmembrane ligand for the programmed cell death protein 1 (PD-1), a receptor that inhibits T-cell activity. The PD-L1/PD-1 immune checkpoint axis has been successfully targeted to enhance antitumor immune responses. Tethering PD-L1 to the membrane spatially restricts its ability to inhibit immune responses, and it provides for the acute and reversible modulation of PD-L1 plasma membrane density by regulation of its trafficking. PD-L1 has functions that are independent of its role as a ligand for PD-1, and control of PD-L1 residence in different intracellular compartments might contribute to the regulation of those activities. Thus, control of PD-L1 trafficking is emerging as a key feature of its biology. Herein, we focus on current understating of PD-L1 trafficking and review current attempts to therapeutically target this process in cancer cells to enhance antitumor immunity.


Asunto(s)
Antígeno B7-H1 , Receptor de Muerte Celular Programada 1 , Ligandos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA