Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Am J Physiol Endocrinol Metab ; 325(5): E425-E437, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37672248

RESUMEN

Walter Cannon was a highly regarded American neurologist and physiologist with extremely broad interests. In the tradition of Cannon and his broad interests, we discuss our laboratory's multifaceted work in signal transduction over the past 40+ years. We show how our questioning of how growth hormone (GH) in the blood communicates with cells throughout the body to promote body growth and regulate body metabolism led to insight into not only body height but also important regulators of malignancy and body weight. Highlights include finding that 1) A critical initiating step in GH signal transduction is GH activating the GH receptor-associated tyrosine kinase JAK2; 2) GH activation of JAK2 leads to activation of a number of signaling proteins, including STAT transcription factors; 3) JAK2 is autophosphorylated on multiple tyrosines that regulate the activity of JAK2 and recruit signaling proteins to GH/GH receptor/JAK2 complexes; 4) Constitutively activated STAT proteins are associated with cancer; 5) GH activation of JAK2 recruits the adapter protein SH2B1 to GH/GH receptor/JAK2 complexes where it facilitates GH regulation of the actin cytoskeleton and motility; and 6) SH2B1 is recruited to other receptors in the brain, where it enhances satiety, most likely in part by regulating leptin action and neuronal connections of appetite-regulating neurons. These findings have led to increased understanding of how GH functions, as well as therapeutic interventions for certain cancer and obese individuals, thereby reinforcing the great importance of supporting basic research since one never knows ahead of time what important insight it can provide.


Asunto(s)
Hormona de Crecimiento Humana , Neoplasias , Humanos , Hormona del Crecimiento/metabolismo , Transducción de Señal/fisiología , Janus Quinasa 2/metabolismo , Hormona de Crecimiento Humana/metabolismo , Receptores de Somatotropina/metabolismo , Fosforilación , Obesidad , Proteínas Adaptadoras Transductoras de Señales/metabolismo
2.
Endocrinology ; 164(5)2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36799031

RESUMEN

Human variants of the adapter protein SH2B1 are associated with severe childhood obesity, hyperphagia, and insulin resistance-phenotypes mimicked by mice lacking Sh2b1. SH2B1ß and γ isoforms are expressed ubiquitously, whereas SH2B1α and δ isoforms are expressed primarily in the brain. Restoring SH2B1ß driven by the neuron-specific enolase promoter largely reverses the metabolic phenotype of Sh2b1-null mice, suggesting crucial roles for neuronal SH2B1ß in energy balance control. Here we test this hypothesis by using CRISPR/Cas9 gene editing to delete the ß and γ isoforms from the neurons of mice (SH2B1ßγ neuron-specific knockout [NKO] mice) or throughout the body (SH2B1ßγ knockout [KO] mice). While parameters of energy balance were normal in both male and female SH2B1ßγ NKO mice, food intake, body weight, and adiposity were increased in male (but not female) SH2B1ßγ KO mice. Analysis of long-read single-cell RNA seq data from wild-type mouse brain revealed that neurons express almost exclusively the α and δ isoforms, whereas neuroglial cells express almost exclusively the ß and γ isoforms. Our work suggests that neuronal SH2B1ß and γ are not primary regulators of energy balance. Rather, non-neuronal SH2B1ß and γ in combination with neuronal SH2B1α and δ suffice for body weight maintenance. While SH2B1ß/γ and SH2B1α/δ share some functionality, SH2B1ß/γ appears to play a larger role in promoting leanness.


Asunto(s)
Obesidad Infantil , Ratones , Masculino , Niño , Humanos , Animales , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Neuronas/metabolismo , Peso Corporal , Ratones Noqueados , Proteínas Adaptadoras Transductoras de Señales/metabolismo
3.
J Cell Sci ; 135(3)2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35019135

RESUMEN

The adapter protein SH2B1 is recruited to neurotrophin receptors, including TrkB (also known as NTRK2), the receptor for brain-derived neurotrophic factor (BDNF). Herein, we demonstrate that the four alternatively spliced isoforms of SH2B1 (SH2B1α-SH2B1δ) are important determinants of neuronal architecture and neurotrophin-induced gene expression. Primary hippocampal neurons from Sh2b1-/- [knockout (KO)] mice exhibit decreased neurite complexity and length, and BDNF-induced expression of the synapse-related immediate early genes Egr1 and Arc. Reintroduction of each SH2B1 isoform into KO neurons increases neurite complexity; the brain-specific δ isoform also increases total neurite length. Human obesity-associated variants, when expressed in SH2B1δ, alter neurite complexity, suggesting that a decrease or increase in neurite branching may have deleterious effects that contribute to the severe childhood obesity and neurobehavioral abnormalities associated with these variants. Surprisingly, in contrast to SH2B1α, SH2B1ß and SH2B1γ, which localize primarily in the cytoplasm and plasma membrane, SH2B1δ resides primarily in nucleoli. Some SH2B1δ is also present in the plasma membrane and nucleus. Nucleolar localization, driven by two highly basic regions unique to SH2B1δ, is required for SH2B1δ to maximally increase neurite complexity and BDNF-induced expression of Egr1, Arc and FosL1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neuronas/citología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Células Cultivadas , Ratones , Neuritas/metabolismo , Neuronas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
4.
Diabetes ; 70(2): 400-414, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33214137

RESUMEN

Mice lacking SH2B1 and humans with variants of SH2B1 display severe obesity and insulin resistance. SH2B1 is an adapter protein that is recruited to the receptors of multiple hormones and neurotrophic factors. Of the four known alternatively spliced SH2B1 isoforms, SH2B1ß and SH2B1γ exhibit ubiquitous expression, whereas SH2B1α and SH2B1δ are essentially restricted to the brain. To understand the roles for SH2B1α and SH2B1δ in energy balance and glucose metabolism, we generated mice lacking these brain-specific isoforms (αδ knockout [αδKO] mice). αδKO mice exhibit decreased food intake, protection from weight gain on standard and high-fat diets, and an adiposity-dependent improvement in glucose homeostasis. SH2B1 has been suggested to impact energy balance via the modulation of leptin action. However, αδKO mice exhibit leptin sensitivity that is similar to that of wild-type mice by multiple measures. Thus, decreasing the abundance of SH2B1α and/or SH2B1δ relative to the other SH2B1 isoforms likely shifts energy balance toward a lean phenotype via a primarily leptin-independent mechanism. Our findings suggest that the different alternatively spliced isoforms of SH2B1 perform different functions in vivo.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Encéfalo/metabolismo , Obesidad/genética , Isoformas de Proteínas/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Resistencia a la Insulina/genética , Leptina/farmacología , Ratones , Ratones Noqueados , Obesidad/metabolismo , Isoformas de Proteínas/metabolismo
5.
Diabetes ; 68(11): 2049-2062, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31439647

RESUMEN

Disruption of the adaptor protein SH2B1 (SH2-B, PSM) is associated with severe obesity, insulin resistance, and neurobehavioral abnormalities in mice and humans. Here, we identify 15 SH2B1 variants in severely obese children. Four obesity-associated human SH2B1 variants lie in the Pleckstrin homology (PH) domain, suggesting that the PH domain is essential for SH2B1's function. We generated a mouse model of a human variant in this domain (P322S). P322S/P322S mice exhibited substantial prenatal lethality. Examination of the P322S/+ metabolic phenotype revealed late-onset glucose intolerance. To circumvent P322S/P322S lethality, mice containing a two-amino acid deletion within the SH2B1 PH domain (ΔP317, R318 [ΔPR]) were studied. Mice homozygous for ΔPR were born at the expected Mendelian ratio and exhibited obesity plus insulin resistance and glucose intolerance beyond that attributable to their increased adiposity. These studies demonstrate that the PH domain plays a crucial role in how SH2B1 controls energy balance and glucose homeostasis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Adiposidad/genética , Metabolismo Energético/genética , Resistencia a la Insulina/genética , Obesidad Infantil/genética , Dominios Homólogos a Pleckstrina/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adolescente , Animales , Niño , Preescolar , Femenino , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Homeostasis/genética , Humanos , Masculino , Ratones , Ratones Transgénicos , Obesidad Infantil/metabolismo
6.
Mol Cell Biol ; 38(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29229648

RESUMEN

The scaffold protein SH2B1, a major regulator of body weight, is recruited to the receptors of multiple cytokines and growth factors, including nerve growth factor (NGF). The ß isoform but not the α isoform of SH2B1 greatly enhances NGF-dependent neurite outgrowth of PC12 cells. Here, we asked how the unique C-terminal tails of the α and ß isoforms modulate SH2B1 function. We compared the actions of SH2B1α and SH2B1ß to those of the N-terminal 631 amino acids shared by both isoforms. In contrast to the ß tail, the α tail inhibited the ability of SH2B1 to both cycle through the nucleus and enhance NGF-mediated neurite outgrowth, gene expression, phosphorylation of Akt and phospholipase C-gamma (PLC-γ), and autophosphorylation of the NGF receptor TrkA. These functions were restored when Tyr753 in the α tail was mutated to phenylalanine. We provide evidence that TrkA phosphorylates Tyr753 in SH2B1α, as well as tyrosines 439 and 55 in both SH2B1α and SH2B1ß. Finally, coexpression of SH2B1α but not SH2B1α with a mutation of Y to F at position 753 (Y753F) inhibited the ability of SH2B1ß to enhance neurite outgrowth. These results suggest that the C-terminal tails of SH2B1 isoforms are key determinants of the cellular role of SH2B1. Furthermore, the function of SH2B1α is regulated by phosphorylation of the α tail.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor de Crecimiento Nervioso/metabolismo , Animales , Diferenciación Celular/fisiología , Células HEK293 , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuritas , Células PC12 , Fosforilación , Dominios Proteicos , Isoformas de Proteínas , Ratas , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Transducción de Señal
7.
Cereb Cortex ; 27(3): 1732-1747, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334068

RESUMEN

KIAA0319 is a transmembrane protein associated with dyslexia with a presumed role in neuronal migration. Here we show that KIAA0319 expression is not restricted to the brain but also occurs in sensory and spinal cord neurons, increasing from early postnatal stages to adulthood and being downregulated by injury. This suggested that KIAA0319 participates in functions unrelated to neuronal migration. Supporting this hypothesis, overexpression of KIAA0319 repressed axon growth in hippocampal and dorsal root ganglia neurons; the intracellular domain of KIAA0319 was sufficient to elicit this effect. A similar inhibitory effect was observed in vivo as axon regeneration was impaired after transduction of sensory neurons with KIAA0319. Conversely, the deletion of Kiaa0319 in neurons increased neurite outgrowth in vitro and improved axon regeneration in vivo. At the mechanistic level, KIAA0319 engaged the JAK2-SH2B1 pathway to activate Smad2, which played a central role in KIAA0319-mediated repression of axon growth. In summary, we establish KIAA0319 as a novel player in axon growth and regeneration with the ability to repress the intrinsic growth potential of axons. This study describes a novel regulatory mechanism operating during peripheral nervous system and central nervous system axon growth, and offers novel targets for the development of effective therapies to promote axon regeneration.


Asunto(s)
Axones/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proyección Neuronal , Proteína Smad2/metabolismo , Envejecimiento/metabolismo , Animales , Aumento de la Célula , Línea Celular , Células Cultivadas , Femenino , Ganglios Espinales/metabolismo , Hipocampo/metabolismo , Humanos , Janus Quinasa 2/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Regeneración Nerviosa/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Dominios Proteicos , Ratas Wistar , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Médula Espinal/metabolismo
8.
Cell Metab ; 24(3): 447-461, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27546461

RESUMEN

Dietary iron intake and systemic iron balance are implicated in colorectal cancer (CRC) development, but the means by which iron contributes to CRC are unclear. Gene expression and functional studies demonstrated that the cellular iron importer, divalent metal transporter 1 (DMT1), is highly expressed in CRC through hypoxia-inducible factor 2α-dependent transcription. Colon-specific Dmt1 disruption resulted in a tumor-selective inhibitory effect of proliferation in mouse colon tumor models. Proteomic and genomic analyses identified an iron-regulated signaling axis mediated by cyclin-dependent kinase 1 (CDK1), JAK1, and STAT3 in CRC progression. A pharmacological inhibitor of DMT1 antagonized the ability of iron to promote tumor growth in a CRC mouse model and a patient-derived CRC enteroid orthotopic model. Our studies implicate a growth-promoting signaling network instigated by elevated intracellular iron levels in tumorigenesis, offering molecular insights into how a key dietary component may contribute to CRC.


Asunto(s)
Carcinogénesis/patología , Proteínas de Transporte de Catión/metabolismo , Ciclo Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Hierro/metabolismo , Quinasas Janus/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteína Quinasa CDC2/metabolismo , Carcinogénesis/metabolismo , Proteínas de Transporte de Catión/genética , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Colitis/complicaciones , Colitis/patología , Colon/patología , Neoplasias Colorrectales/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Inflamación/complicaciones , Inflamación/patología , Hierro/farmacología , Transducción de Señal/efectos de los fármacos
9.
Growth Horm IGF Res ; 28: 11-5, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26421979

RESUMEN

Over 20years ago, our laboratory showed that growth hormone (GH) signals through the GH receptor-associated tyrosine kinase JAK2. We showed that GH binding to its membrane-bound receptor enhances binding of JAK2 to the GHR, activates JAK2, and stimulates tyrosyl phosphorylation of both JAK2 and GHR. The activated JAK2/GHR complex recruits a variety of signaling proteins, thereby initiating multiple signaling pathways and cellular responses. These proteins and pathways include: 1) Stat transcription factors implicated in the expression of multiple genes, including the gene encoding insulin-like growth factor 1; 2) Shc adapter proteins that lead to activation of the grb2-SOS-Ras-Raf-MEK-ERK1,2 pathway; 3) insulin receptor substrate proteins implicated in the phosphatidylinositol-3-kinase and Akt pathway; 4) signal regulatory protein α, a transmembrane scaffold protein that recruits proteins including the tyrosine phosphatase SHP2; and 5) SH2B1, a scaffold protein that can activate JAK2 and enhance GH regulation of the actin cytoskeleton. Our recent work has focused on the function of SH2B1. We have shown that SH2B1ß is recruited to and phosphorylated by JAK2 in response to GH. SH2B1 localizes to the plasma membrane, cytoplasm and focal adhesions; it also cycles through the nucleus. SH2B1 regulates the actin cytoskeleton and promotes GH-dependent motility of RAW264.7 macrophages. Mutations in SH2B1 have been found in humans exhibiting severe early-onset childhood obesity and insulin resistance. These mutations impair SH2B1 enhancement of GH-induced macrophage motility. As SH2B1 is expressed ubiquitously and is also recruited to a variety of receptor tyrosine kinases, our results raise the possibility that effects of SH2B1 on the actin cytoskeleton in various cell types, including neurons, may play a role in regulating body weight.


Asunto(s)
Hormona del Crecimiento/metabolismo , Janus Quinasa 2/metabolismo , Receptores de Somatotropina/metabolismo , Citoesqueleto de Actina , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Movimiento Celular/genética , Humanos , Resistencia a la Insulina/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Obesidad Infantil/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal
10.
Endocrinology ; 155(9): 3219-26, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24971614

RESUMEN

We have previously reported rare variants in sarcoma (Src) homology 2 (SH2) B adaptor protein 1 (SH2B1) in individuals with obesity, insulin resistance, and maladaptive behavior. Here, we identify 4 additional SH2B1 variants by sequencing 500 individuals with severe early-onset obesity. SH2B1 has 4 alternatively spliced isoforms. One variant (T546A) lies within the N-terminal region common to all isoforms. As shown for past variants in this region, T546A impairs SH2B1ß enhancement of nerve growth factor-induced neurite outgrowth, and the individual with the T546A variant exhibits mild developmental delay. The other 3 variants (A663V, V695M, and A723V) lie in the C-terminal tail of SH2B1α. SH2B1α variant carriers were hyperinsulinemic but did not exhibit the behavioral phenotype observed in individuals with SH2B1 variants that disrupt all isoforms. In in vitro assays, SH2B1α, like SH2B1ß, enhances insulin- and leptin-induced insulin receptor substrate 2 (IRS2) phosphorylation and GH-induced cell motility. None of the variants affect SH2B1α enhancement of insulin- and leptin-induced IRS2 phosphorylation. However, T546A, A663V, and A723V all impair the ability of SH2B1α to enhance GH-induced cell motility. In contrast to SH2B1ß, SH2B1α does not enhance nerve growth factor-induced neurite outgrowth. These studies suggest that genetic variants that disrupt isoforms other than SH2B1ß may be functionally significant. Further studies are needed to understand the mechanism by which the individual isoforms regulate energy homeostasis and behavior.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Obesidad/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adolescente , Adulto , Empalme Alternativo , Niño , Femenino , Humanos , Insulina/metabolismo , Leptina/metabolismo , Masculino , Mutación Missense , Obesidad/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Adulto Joven
11.
PLoS One ; 8(9): e75472, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086539

RESUMEN

Utilizing ENU mutagenesis, we identified a mutant mouse with elevated platelets. Genetic mapping localized the mutation to an interval on chromosome 19 that encodes the Jak2 tyrosine kinase. We identified a A3056T mutation resulting in a premature stop codon within exon 19 of Jak2 (Jak2(K915X)), resulting in a protein truncation and functionally inactive enzyme. This novel platelet phenotype was also observed in mice bearing a hemizygous targeted disruption of the Jak2 locus (Jak2(+/-)). Timed pregnancy experiments revealed that Jak2(K915X/K915X) and Jak2(-/-) displayed embryonic lethality; however, Jak2(K915X/K915X) embryos were viable an additional two days compared to Jak2(-/-) embryos. Our data suggest that perturbing JAK2 activation may have unexpected consequences in elevation of platelet number and correspondingly, important implications for treatment of hematological disorders with constitutive Jak2 activity.


Asunto(s)
Plaquetas/citología , Janus Quinasa 2/genética , Fenotipo , Animales , Western Blotting , Mapeo Cromosómico , Etilnitrosourea , Fluorouracilo , Genotipo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutagénesis/genética , Fenilhidrazinas , Mutación Puntual/genética , Análisis de Secuencia de ADN
12.
Mol Endocrinol ; 27(4): 619-34, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23449887

RESUMEN

The tyrosine kinase Janus kinase 2 (JAK2) is activated by many cytokine receptors, including receptors for GH, leptin, and erythropoietin. However, very few proteins have been identified as binding partners for JAK2. Using a yeast 2-hybrid screen, we identified steroid-sensitive gene-1 (SSG1)/coiled-coil domain-containing protein 80 (Ccdc80) as a JAK2-binding partner. We demonstrate that Ccdc80 preferentially binds activated, tyrosyl-phosphorylated JAK2 but not kinase-inactive JAK2 (K882E) in both yeast and mammalian systems. Ccdc80 is tyrosyl phosphorylated in the presence of JAK2. The binding of Ccdc80 to JAK2 occurs via 1 or more of the 3 DUDES/SRPX (DRO1-URB-DRS-Equarin-SRPUL/sushi repeat containing protein, x-linked) domain 5 domains of Ccdc80. Mutagenesis of the second DUDES domain suggests that the N-terminal third of the DUDES domain is sufficient for JAK2 binding. Ccdc80 does not alter the kinase activity of JAK2. However, Ccdc80 increases GH-dependent phosphorylation of Stat (signal transducer and activator of transcription) 5b on Tyr699 and substantially enhances both basal and GH-dependent phosphorylation/activation of Stat3 on Tyr705. Furthermore, Ccdc80 belongs to the group of proteins that function both in the intracellular compartment and are secreted. Secreted Ccdc80 associates with the extracellular matrix and is also found in the medium. A substantial portion of the Ccdc80 detected in the medium is cleaved. Finally, consistent with the DUDES domain serving as a JAK2-binding domain, we also demonstrate that another protein that contains a DUDES domain, SRPX2, binds preferentially to the activated tyrosyl-phosphorylated form of JAK2.


Asunto(s)
Janus Quinasa 2/metabolismo , Proteínas de Neoplasias/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Activación Enzimática , Matriz Extracelular/metabolismo , Humanos , Espacio Intracelular/metabolismo , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Proteínas del Tejido Nervioso/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Factores de Transcripción STAT , Proteínas Supresoras de Tumor , Técnicas del Sistema de Dos Híbridos
13.
J Cell Sci ; 126(Pt 8): 1733-43, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23444381

RESUMEN

Previous studies have shown that growth hormone (GH) recruits the adapter protein SH2B1ß to the GH-activated, GH receptor-associated tyrosine kinase JAK2, implicating SH2B1ß in GH-dependent actin cytoskeleton remodeling, and suggesting that phosphorylation at serines 161 and 165 in SH2B1ß releases SH2B1ß from the plasma membrane. Here, we examined the role of SH2B1ß in GH regulation of macrophage migration. We show that GH stimulates migration of cultured RAW264.7 macrophages, and primary cultures of peritoneal and bone marrow-derived macrophages. SH2B1ß overexpression enhances, whereas SH2B1 knockdown inhibits, GH-dependent motility of RAW macrophages. At least two independent mechanisms regulate the SH2B1ß-mediated changes in motility. In response to GH, tyrosines 439 and 494 in SH2B1ß are phosphorylated. Mutating these tyrosines in SH2B1ß decreases both basal and GH-stimulated macrophage migration. In addition, mutating the polybasic nuclear localization sequence (NLS) in SH2B1ß or creating the phosphomimetics SH2B1ß(S161E) or SH2B1ß(S165E), all of which release SH2B1ß from the plasma membrane, enhances macrophage motility. Conversely, SH2B1ß(S161/165A) exhibits increased localization at the plasma membrane and decreased macrophage migration. Mutating the NLS or the nearby serine residues does not alter GH-dependent phosphorylation on tyrosines 439 and 494 in SH2B1ß. Mutating tyrosines 439 and 494 does not affect localization of SH2B1ß at the plasma membrane or movement of SH2B1ß into focal adhesions. Taken together, these results suggest that SH2B1ß enhances GH-stimulated macrophage motility via mechanisms involving phosphorylation of SH2B1ß on tyrosines 439 and 494 and movement of SH2B1ß out of the plasma membrane (e.g. as a result of phosphorylation of serines 161 and 165).


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Membrana Celular/metabolismo , Hormona del Crecimiento/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Animales , Línea Celular , Movimiento Celular/fisiología , Adhesiones Focales/metabolismo , Ratones , Fosforilación
14.
J Clin Invest ; 122(12): 4732-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23160192

RESUMEN

Src homology 2 B adapter protein 1 (SH2B1) modulates signaling by a variety of ligands that bind to receptor tyrosine kinases or JAK-associated cytokine receptors, including leptin, insulin, growth hormone (GH), and nerve growth factor (NGF). Targeted deletion of Sh2b1 in mice results in increased food intake, obesity, and insulin resistance, with an intermediate phenotype seen in heterozygous null mice on a high-fat diet. We identified SH2B1 loss-of-function mutations in a large cohort of patients with severe early-onset obesity. Mutation carriers exhibited hyperphagia, childhood-onset obesity, disproportionate insulin resistance, and reduced final height as adults. Unexpectedly, mutation carriers exhibited a spectrum of behavioral abnormalities that were not reported in controls, including social isolation and aggression. We conclude that SH2B1 plays a critical role in the control of human food intake and body weight and is implicated in maladaptive human behavior.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Mutación del Sistema de Lectura , Mutación Missense , Obesidad/genética , Adolescente , Adulto , Agresión , Secuencia de Bases , Estudios de Casos y Controles , Movimiento Celular , Niño , Preescolar , Análisis Mutacional de ADN , Ingestión de Energía/genética , Femenino , Estudios de Asociación Genética , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Transporte de Proteínas , Aislamiento Social , Adulto Joven
15.
J Biol Chem ; 287(31): 26223-34, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22669948

RESUMEN

Gene targeting experiments have shown that the cytokine erythropoietin (EPO), its cognate erythropoietin receptor (EPO-R), and associated Janus tyrosine kinase, JAK2, are all essential for erythropoiesis. Structural-functional and murine knock-in experiments have suggested that EPO-R Tyr-343 is important in EPO-mediated mitogenesis. Although Stat5 binds to EPO-R phosphotyrosine 343, the initial Stat5-deficient mice did not have profound erythroid abnormalities suggesting that additional Src homology 2 (SH2) domain-containing effectors may bind to EPO-R Tyr-343 and couple to downstream signaling pathways. We have utilized cloning of ligand target (COLT) screening to demonstrate that EPO-R Tyr(P)-343 and Tyr(P)-401 bind to the SH2 domain-containing adaptor protein SH2B1ß. Immunoprecipitation and in vitro mixing experiments reveal that EPO-R binds to SH2B1 in an SH2 domain-dependent manner and that the sequence that confers SH2B1 binding to the EPO-R is pYXXL. Previous studies have shown that SH2B1 binds directly to JAK2, but we show that in hematopoietic cells, SH2B1ß preferentially associates with the EPO-R. SH2B1 is capable of constitutive association with EPO-R, which is necessary for its optimal SH2-dependent recruitment to EPO-R-Tyr(P)-343/Tyr(P)-401. We also demonstrate that SH2B1 is responsive to EPO stimulation and becomes phosphorylated, most likely on serines/threonines, in an EPO dose- and time-dependent manner. In the absence of SH2B1, we observe enhanced activation of signaling pathways downstream of the EPO-R, indicating that SH2B1 is a negative regulator of EPO signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Receptores de Eritropoyetina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/aislamiento & purificación , Animales , Línea Celular , Eritroblastos/metabolismo , Eritropoyetina/fisiología , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/química , Fragmentos de Péptidos/aislamiento & purificación , Fragmentos de Péptidos/metabolismo , Fosforilación , Cultivo Primario de Células , Dominios y Motivos de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/aislamiento & purificación , Transducción de Señal
16.
Mol Endocrinol ; 26(6): 1056-73, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22570334

RESUMEN

GH and GH receptors are expressed throughout life, and GH elicits a diverse range of responses, including growth and altered metabolism. It is therefore important to understand the full spectrum of GH signaling pathways and cellular responses. We applied mass spectrometry-based phosphoproteomics combined with stable isotope labeling with amino acids in cell culture to identify proteins rapidly phosphorylated in response to GH in 3T3-F442A preadipocytes. We identified 132 phosphosites in 95 proteins that exhibited rapid (5 or 15 min) GH-dependent statistically significant increases in phosphorylation by more than or equal to 50% and 96 phosphosites in 46 proteins that were down-regulated by GH by more than or equal to 30%. Several of the GH-stimulated phosphorylation sites were known (e.g. regulatory Thr/Tyr in Erks 1 and 2, Tyr in signal transducers and activators of transcription (Stat) 5a and 5b, Ser939 in tuberous sclerosis protein (TSC) 2 or tuberin). The remaining 126 GH-stimulated sites were not previously associated with GH. Kyoto Encyclopedia of Genes and Genomes pathway analysis of GH-stimulated sites indicated enrichment in proteins associated with the insulin and mammalian target of rapamycin (mTOR) pathways, regulation of the actin cytoskeleton, and focal adhesions. Akt/protein kinase A consensus sites (RXRXXS/T) were the most commonly phosphorylated consensus sites. Immunoblotting confirmed GH-stimulated phosphorylation of all seven novel GH-dependent sites tested [regulatory sites in proline-rich Akt substrate, 40 kDA (PRAS40), regulatory associated protein of mTOR, ATP-citrate lyase, Na(+)/H(+) exchanger-1, N-myc downstream regulated gene 1, and Shc]). The immunoblot results suggest that many, if not most, of the GH-stimulated phosphosites identified in this large-scale quantitative phosphoproteomics analysis, including sites in multiple proteins in the Akt/ mTOR complex 1 pathway, are phosphorylated in response to GH. Their identification significantly broadens our thinking of GH-regulated cell functions.


Asunto(s)
Hormona del Crecimiento/fisiología , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional , Proteoma/metabolismo , Células 3T3 , Secuencias de Aminoácidos , Animales , Cromatografía por Intercambio Iónico , Secuencia de Consenso , Ratones , Fragmentos de Péptidos/química , Fragmentos de Péptidos/aislamiento & purificación , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/química , Fosfoproteínas/aislamiento & purificación , Proteoma/química , Proteoma/aislamiento & purificación , Proteómica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Somatotropina/metabolismo , Transducción de Señal , Espectrometría de Masas en Tándem
17.
PLoS One ; 7(2): e31636, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22359610

RESUMEN

Leptin exerts its action by binding to and activating the long form of leptin receptors (LEPRb). LEPRb activates JAK2 that subsequently phosphorylates and activates STAT3. The JAK2/STAT3 pathway is required for leptin control of energy balance and body weight. Defects in leptin signaling lead to leptin resistance, a primary risk factor for obesity. Body weight is also regulated by nutrients, including glucose. Defects in glucose sensing also contribute to obesity. Here we report crosstalk between leptin and glucose. Glucose starvation blocked the ability of leptin to stimulate tyrosyl phosphorylation and activation of JAK2 and STAT3 in a variety of cell types. Glucose dose-dependently enhanced leptin signaling. In contrast, glucose did not enhance growth hormone-stimulated phosphorylation of JAK2 and STAT5. Glucose starvation or 2-deoxyglucose-induced inhibition of glycolysis activated AMPK and inhibited leptin signaling; pharmacological inhibition of AMPK restored the ability of leptin to stimulate STAT3 phosphorylation. Conversely, pharmacological activation of AMPK was sufficient to inhibit leptin signaling and to block the ability of glucose to enhance leptin signaling. These results suggest that glucose and/or its metabolites play a permissive role in leptin signaling, and that glucose enhances leptin sensitivity at least in part by attenuating the ability of AMPK to inhibit leptin signaling.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Glucosa/farmacología , Leptina/metabolismo , Animales , Línea Celular , Janus Quinasa 2/metabolismo , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
18.
Pediatr Endocrinol Rev ; 10(2): 243-5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23539836

RESUMEN

The inaugural edition of the FASEB Science Research Conference entitled 'The GHIPRL Family in Biology and Disease' was held this past summer at Snowmass, Colorado. This report provides an overview of the scientific content and a taste of the atmosphere of this novel meetinq in the field of Endocrinology.


Asunto(s)
Hormona de Crecimiento Humana/fisiología , Prolactina/fisiología , Investigación Biomédica , Congresos como Asunto , Hormona de Crecimiento Humana/efectos adversos , Hormona de Crecimiento Humana/uso terapéutico , Humanos , Prolactina/efectos adversos , Prolactina/uso terapéutico
19.
J Cell Sci ; 124(Pt 18): 3095-105, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21878491

RESUMEN

The adaptor protein SH2B1ß participates in regulation of the actin cytoskeleton during processes such as cell migration and differentiation. Here, we identify SH2B1ß as a new focal adhesion protein. We provide evidence that SH2B1ß is phosphorylated in response to phorbol 12-myristate 13-acetate (PMA)-induced protein kinase C (PKC) activation and show that PMA induces a rapid redistribution of SH2B1ß out of focal adhesions. We also show that growth hormone (GH) increases cycling of SH2B1ß into and out of focal adhesions. Ser161 and Ser165 in SH2B1ß fall within consensus PKC substrate motifs. Mutating these two serine residues into alanine residues abrogates PMA-induced redistribution of SH2B1ß out of focal adhesions, decreases SH2B1ß cycling into and out of focal adhesions in control and GH-stimulated cells, and increases the size of focal adhesions. By contrast, mutating Ser165 into a glutamate residue decreases the amount of SH2B1ß in focal adhesions and increases the number of focal adhesions per cell. These results suggest that activation of PKC regulates SH2B1ß focal adhesion localization through phosphorylation of Ser161 and/or Ser165. The finding that phosphorylation of SH2B1ß increases the number of focal adhesions suggests a mechanism for the stimulatory effect on cell motility of SH2B1ß.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Moléculas de Adhesión Celular/metabolismo , Adhesiones Focales/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Moléculas de Adhesión Celular/genética , Línea Celular , Movimiento Celular , Citoesqueleto/metabolismo , Adhesiones Focales/efectos de los fármacos , Adhesiones Focales/patología , Hormona del Crecimiento/farmacología , Ratones , Mutagénesis Sitio-Dirigida , Mutación/genética , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Serina/genética , Transducción de Señal/efectos de los fármacos , Ciclo del Sustrato/efectos de los fármacos , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA