Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 16(8): e1008733, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32817694

RESUMO

Staphylococcus aureus (S. aureus) is one of the most common bacterial infections worldwide, and antibiotic resistant strains such as Methicillin-Resistant S. aureus (MRSA) are a major threat and burden to public health. MRSA not only infects immunocompromised patients but also healthy individuals and has rapidly spread from the healthcare setting to the outside community. However, all vaccines tested in clinical trials to date have failed. Immunocompromised individuals such as patients with HIV or decreased levels of CD4+ T cells are highly susceptible to S. aureus infections, and they are also at increased risk of developing fungal infections. We therefore wondered whether stimulation of antifungal immunity might promote the type of immune responses needed for effective host defense against S. aureus. Here we show that vaccination of mice with a fungal ß-glucan particle (GP) loaded with S. aureus antigens provides protective immunity to S. aureus. We generated glucan particles loaded with the four S. aureus proteins ClfA, IsdA, MntC, and SdrE, creating the 4X-SA-GP vaccine. Vaccination of mice with three doses of 4X-SA-GP promoted protection in a systemic model of S. aureus infection with a significant reduction in the bacterial burden in the spleen and kidneys. 4X-SA-GP vaccination induced antigen-specific Th1 and Th17 CD4+ T cell and antibody responses and provided long-term protection. This work suggests that the GP vaccine system has potential as a novel approach to developing vaccines for S. aureus.


Assuntos
Saccharomyces cerevisiae/imunologia , Infecções Estafilocócicas/imunologia , Vacinas Antiestafilocócicas/imunologia , Staphylococcus aureus/imunologia , Animais , Anticorpos Antibacterianos/imunologia , Coagulase/administração & dosagem , Coagulase/genética , Coagulase/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Saccharomyces cerevisiae/química , Infecções Estafilocócicas/microbiologia , Vacinas Antiestafilocócicas/administração & dosagem , Vacinas Antiestafilocócicas/genética , Staphylococcus aureus/genética , Células Th1/imunologia , Células Th17/imunologia , Vacinação , beta-Glucanas/administração & dosagem , beta-Glucanas/imunologia
2.
PLoS Pathog ; 14(9): e1007260, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30235351

RESUMO

The gastrointestinal microbiota influences immune function throughout the body. The gut-lung axis refers to the concept that alterations of gut commensal microorganisms can have a distant effect on immune function in the lung. Overgrowth of intestinal Candida albicans has been previously observed to exacerbate allergic airways disease in mice, but whether subtler changes in intestinal fungal microbiota can affect allergic airways disease is less clear. In this study we have investigated the effects of the population expansion of commensal fungus Wallemia mellicola without overgrowth of the total fungal community. Wallemia spp. are commonly found as a minor component of the commensal gastrointestinal mycobiota in both humans and mice. Mice with an unaltered gut microbiota community resist population expansion when gavaged with W. mellicola; however, transient antibiotic depletion of gut microbiota creates a window of opportunity for expansion of W. mellicola following delivery of live spores to the gastrointestinal tract. This phenomenon is not universal as other commensal fungi (Aspergillus amstelodami, Epicoccum nigrum) do not expand when delivered to mice with antibiotic-depleted microbiota. Mice with Wallemia-expanded gut mycobiota experienced altered pulmonary immune responses to inhaled aeroallergens. Specifically, after induction of allergic airways disease with intratracheal house dust mite (HDM) antigen, mice demonstrated enhanced eosinophilic airway infiltration, airway hyperresponsiveness (AHR) to methacholine challenge, goblet cell hyperplasia, elevated bronchoalveolar lavage IL-5, and enhanced serum HDM IgG1. This phenomenon occurred with no detectable Wallemia in the lung. Targeted amplicon sequencing analysis of the gastrointestinal mycobiota revealed that expansion of W. mellicola in the gut was associated with additional alterations of bacterial and fungal commensal communities. We therefore colonized fungus-free Altered Schaedler Flora (ASF) mice with W. mellicola. ASF mice colonized with W. mellicola experienced enhanced severity of allergic airways disease compared to fungus-free control ASF mice without changes in bacterial community composition.


Assuntos
Basidiomycota/imunologia , Basidiomycota/patogenicidade , Microbioma Gastrointestinal/imunologia , Micobioma/imunologia , Hipersensibilidade Respiratória/etiologia , Alérgenos/administração & dosagem , Animais , Antibacterianos/efeitos adversos , Antígenos de Dermatophagoides/administração & dosagem , Basidiomycota/crescimento & desenvolvimento , Modelos Animais de Doenças , Microbiologia Ambiental , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/genética , Vida Livre de Germes/imunologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Micobioma/genética , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/microbiologia , Simbiose/imunologia
3.
J Immunol ; 198(10): 4036-4045, 2017 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-28411186

RESUMO

Type I IFNs are a cytokine family essential for antiviral defense. More recently, type I IFNs were shown to be important during bacterial infections. In this article, we show that, in addition to known cytokine functions, IFN-ß is antimicrobial. Parts of the IFN-ß molecular surface (especially helix 4) are cationic and amphipathic, both classic characteristics of antimicrobial peptides, and we observed that IFN-ß can directly kill Staphylococcus aureus Further, a mutant S. aureus that is more sensitive to antimicrobial peptides was killed more efficiently by IFN-ß than was the wild-type S. aureus, and immunoblotting showed that IFN-ß interacts with the bacterial cell surface. To determine whether specific parts of IFN-ß are antimicrobial, we synthesized IFN-ß helix 4 and found that it is sufficient to permeate model prokaryotic membranes using synchrotron x-ray diffraction and that it is sufficient to kill S. aureus These results suggest that, in addition to its well-known signaling activity, IFN-ß may be directly antimicrobial and be part of a growing family of cytokines and chemokines, called kinocidins, that also have antimicrobial properties.


Assuntos
Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Interferon beta/fisiologia , Staphylococcus aureus/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacologia , Humanos , Interferon beta/química , Interferon beta/metabolismo , Interferon beta/farmacologia , Camundongos , Testes de Sensibilidade Microbiana , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/genética , Difração de Raios X
4.
Curr Opin Gastroenterol ; 34(6): 398-403, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30299290

RESUMO

PURPOSE OF REVIEW: The intestinal microbiota plays a central role in inflammatory diseases of the gut. Although most investigations regarding how the mucosal immune system interacts with the microbiota have focused on bacteria, recent studies are elucidating the additional role of commensal fungi in health and disease in the gut. RECENT FINDINGS: New technical approaches are defining the makeup of the fungal communities in the intestines of humans and mice. The reported composition of these communities is influenced by the approaches used to define the fungi. Changes in the intestinal mycobiota are associated with gut inflammation in patients with inflammatory bowel disease and in mouse models of colitis. Recent studies are beginning to elucidate the mechanisms by which the mucosal immune system interacts with and is influenced by intestinal fungi. SUMMARY: Studies clearly demonstrate the presence of intestinal fungi and document the ability of the mucosal immune system to recognize and respond to fungi. Future studies will further investigate whether intestinal fungi directly influence intestinal disease and what cellular, molecular, and genetic mechanisms contribute.


Assuntos
Fungos/imunologia , Microbioma Gastrointestinal/imunologia , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/imunologia , Interações entre Hospedeiro e Microrganismos , Humanos , Imunidade nas Mucosas , Doenças Inflamatórias Intestinais/microbiologia , Mucosa Intestinal/microbiologia
5.
Proc Natl Acad Sci U S A ; 111(47): E5076-85, 2014 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-25385646

RESUMO

The mammalian target of rapamycin (mTOR) is a kinase that functions in two distinct complexes, mTORC1 and mTORC2. In peripheral B cells, complete deletion of mTOR suppresses germinal center B-cell responses, including class switching and somatic hypermutation. The allosteric mTORC1 inhibitor rapamycin blocks proliferation and differentiation, but lower doses can promote protective IgM responses. To elucidate the complexity of mTOR signaling in B cells further, we used ATP-competitive mTOR kinase inhibitors (TOR-KIs), which inhibit both mTORC1 and mTORC2. Although TOR-KIs are in clinical development for cancer, their effects on mature lymphocytes are largely unknown. We show that high concentrations of TOR-KIs suppress B-cell proliferation and differentiation, yet lower concentrations that preserve proliferation increase the fraction of B cells undergoing class switching in vitro. Transient treatment of mice with the TOR-KI compound AZD8055 increased titers of class-switched high-affinity antibodies to a hapten-protein conjugate. Mechanistic investigation identified opposing roles for mTORC1 and mTORC2 in B-cell differentiation and showed that TOR-KIs enhance class switching in a manner dependent on forkhead box, subgroup O (FoxO) transcription factors. These observations emphasize the distinct actions of TOR-KIs compared with rapamycin and suggest that TOR-KIs might be useful to enhance production of class-switched antibodies following vaccination.


Assuntos
Switching de Imunoglobulina/efeitos dos fármacos , Complexos Multiproteicos/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Regulação Alostérica , Animais , Imunoglobulina G/biossíntese , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Sirolimo/farmacologia
6.
Eur J Immunol ; 40(7): 1890-6, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20449867

RESUMO

A fundamental component of signaling initiated by the BCR and CD19 is the activation of phosphoinositide 3-kinase. Downstream of phosphoinositide 3-kinase, the protein kinase AKT phosphorylates several substrates, including members of the forkhead box subgroup O (Foxo) transcription factor family. Among the Foxo proteins, Foxo1 has unique functions in bone marrow B-cell development and peripheral B-cell function. Here, we report a previously unrecognized role for Foxo1 in controlling the ratio of mature B-cell subsets in the spleen. Conditional deletion of Foxo1 in B cells resulted in an increased percentage of marginal zone B cells and a decrease in follicular (FO) B cells. In addition, Foxo1 deficiency corrected the absence of marginal zone B cells that occurs in CD19-deficient mice. These findings show that Foxo1 regulates the balance of mature B-cell subsets and is required for the marginal zone B-cell deficiency phenotype of mice lacking CD19.


Assuntos
Antígenos CD19/metabolismo , Linfócitos B/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Células Precursoras de Linfócitos B/metabolismo , Baço/imunologia , Animais , Antígenos CD19/genética , Antígenos CD19/imunologia , Linfócitos B/imunologia , Linfócitos B/patologia , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Citometria de Fluxo , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Switching de Imunoglobulina/genética , Camundongos , Camundongos Knockout , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/patologia , Baço/embriologia , Baço/patologia
7.
J Leukoc Biol ; 109(1): 161-172, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32941658

RESUMO

Malassezia spp. are common eukaryotic yeasts that colonize mammalian skin. Recently, the authors and others have observed that Malassezia globosa and Malassezia restricta can be found in the intestines in the context of certain diseases, including Crohn's disease and pancreatic cancer. In order to better understand the nature of innate inflammatory responses to these yeasts, inflammatory responses induced by M. restricta and M. globosa in mouse bone marrow-derived Mϕs (BMDM) and dendritic cells (BMDC) are evaluated. While Malassezia yeasts induce proinflammatory cytokine production from both Mϕs and dendritic cells, the levels of production from BMDC were more pronounced. Both M. restricta and M. globosa activated inflammatory cytokine production from BMDC in large part through Dectin2 and CARD9 signaling, although additional receptors appear to be involved in phagocytosis and activation of reactive oxygen production in response to the yeasts. Both M. restricta and M. globosa stimulate production of pro-IL-1ß as well as activation of the NLRP3 inflammasome. NLRP3 inflammasome activation by Malassezia fungi requires SYK signaling, potassium efflux and actin rearrangement. Together, the data further the understanding of the coordinated involvement of multiple innate immune receptors in recognizing Malassezia globosa and Malassezia restricta and orchestrating phagocyte inflammatory and antimicrobial responses.


Assuntos
Inflamassomos/imunologia , Inflamação/imunologia , Malassezia/imunologia , Micoses/imunologia , Fagócitos/imunologia , Animais , Citocinas/imunologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia
8.
Cancer Cell ; 39(9): 1202-1213.e6, 2021 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-34329585

RESUMO

Studies suggest that the efficacy of cancer chemotherapy and immunotherapy is influenced by intestinal bacteria. However, the influence of the microbiome on radiation therapy is not as well understood, and the microbiome comprises more than bacteria. Here, we find that intestinal fungi regulate antitumor immune responses following radiation in mouse models of breast cancer and melanoma and that fungi and bacteria have opposite influences on these responses. Antibiotic-mediated depletion or gnotobiotic exclusion of fungi enhances responsiveness to radiation, whereas antibiotic-mediated depletion of bacteria reduces responsiveness and is associated with overgrowth of commensal fungi. Further, elevated intratumoral expression of Dectin-1, a primary innate sensor of fungi, is negatively associated with survival in patients with breast cancer and is required for the effects of commensal fungi in mouse models of radiation therapy.


Assuntos
Antifúngicos/administração & dosagem , Bactérias/classificação , Neoplasias da Mama/terapia , Fungos/efeitos dos fármacos , Lectinas Tipo C/genética , Melanoma/terapia , Animais , Antifúngicos/farmacologia , Bactérias/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/microbiologia , Terapia Combinada , Regulação para Baixo , Feminino , Fungos/classificação , Fungos/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Melanoma/imunologia , Melanoma/microbiologia , Camundongos , Simbiose , Linfócitos T/metabolismo , Macrófagos Associados a Tumor/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cell Host Microbe ; 25(3): 377-388.e6, 2019 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-30850233

RESUMO

Inflammatory bowel disease (IBD) is characterized by alterations in the intestinal microbiota and altered immune responses to gut microbiota. Evidence is accumulating that IBD is influenced by not only commensal bacteria but also commensal fungi. We characterized fungi directly associated with the intestinal mucosa in healthy people and Crohn's disease patients and identified fungi specifically abundant in patients. One of these, the common skin resident fungus Malassezia restricta, is also linked to the presence of an IBD-associated polymorphism in the gene for CARD9, a signaling adaptor important for anti-fungal defense. M. restricta elicits innate inflammatory responses largely through CARD9 and is recognized by Crohn's disease patient anti-fungal antibodies. This yeast elicits strong inflammatory cytokine production from innate cells harboring the IBD-linked polymorphism in CARD9 and exacerbates colitis via CARD9 in mouse models of disease. Collectively, these results suggest that targeting specific commensal fungi may be a therapeutic strategy for IBD.


Assuntos
Colite/patologia , Colite/fisiopatologia , Doença de Crohn/patologia , Doença de Crohn/fisiopatologia , Trato Gastrointestinal/microbiologia , Malassezia/crescimento & desenvolvimento , Malassezia/isolamento & purificação , Animais , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Camundongos
10.
Cell Host Microbe ; 22(2): 156-165, 2017 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-28799901

RESUMO

Fungi are increasingly being recognized as common members of the microbiomes found on nearly all mucosal surfaces, and interest is growing in understanding how these organisms may contribute to health and disease. In this review, we investigate recent developments in our understanding of the fungal microbiota or "mycobiota" including challenges faced in characterizing it, where these organisms are found, their diversity, and how they interact with host immunity. Growing evidence indicates that, like the bacterial microbiota, the fungal microbiota is often altered in disease states, and increasingly studies are being designed to probe the functional consequences of such fungal dysbiosis on health and disease.


Assuntos
Fungos/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Simbiose/imunologia , Bactérias/imunologia , DNA Fúngico , Disbiose/imunologia , Disbiose/microbiologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Humanos , Imunidade nas Mucosas , Pneumopatias/imunologia , Pneumopatias/microbiologia , Síndrome Metabólica/imunologia , Síndrome Metabólica/microbiologia , Interações Microbianas , Microbiota/imunologia , Sistema Respiratório/imunologia , Sistema Respiratório/microbiologia , Pele/microbiologia , Dermatopatias/imunologia , Dermatopatias/microbiologia , Sistema Urogenital/imunologia , Sistema Urogenital/microbiologia
11.
Annu Rev Pathol ; 12: 359-385, 2017 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-28068483

RESUMO

Fungi are ubiquitous in our environment, and a healthy immune system is essential to maintain adequate protection from fungal infections. When this protection breaks down, superficial and invasive fungal infections cause diseases that range from irritating to life-threatening. Millions of people worldwide develop invasive infections during their lives, and mortality for these infections often exceeds 50%. Nevertheless, we are normally colonized with many of the same disease-causing fungi (e.g., on the skin or in the gut). Recent research is dramatically expanding our understanding of the mechanisms by which our immune systems interact with these organisms in health and disease. In this review, we discuss what is currently known about where and how the immune system interacts with common fungi.


Assuntos
Imunidade Adaptativa/imunologia , Doença/etiologia , Fungos/imunologia , Imunidade Inata/imunologia , Micoses/complicações , Micoses/imunologia , Animais , Humanos
12.
Cell Host Microbe ; 19(6): 865-73, 2016 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-27237365

RESUMO

Compared to bacteria, the role of fungi within the intestinal microbiota is poorly understood. In this study we investigated whether the presence of a "healthy" fungal community in the gut is important for modulating immune function. Prolonged oral treatment of mice with antifungal drugs resulted in increased disease severity in acute and chronic models of colitis, and also exacerbated the development of allergic airway disease. Microbiota profiling revealed restructuring of fungal and bacterial communities. Specifically, representation of Candida spp. was reduced, while Aspergillus, Wallemia, and Epicoccum spp. were increased. Oral supplementation with a mixture of three fungi found to expand during antifungal treatment (Aspergillus amstelodami, Epicoccum nigrum, and Wallemia sebi) was sufficient to recapitulate the exacerbating effects of antifungal drugs on allergic airway disease. Taken together, these results indicate that disruption of commensal fungal populations can influence local and peripheral immune responses and enhance relevant disease states.


Assuntos
Antifúngicos/efeitos adversos , Disbiose/induzido quimicamente , Disbiose/imunologia , Fungos/efeitos dos fármacos , Fungos/imunologia , Intestinos/microbiologia , Anfotericina B/efeitos adversos , Anfotericina B/farmacologia , Animais , Antifúngicos/farmacologia , Bactérias/efeitos dos fármacos , Sequência de Bases , Colite/imunologia , Colite/microbiologia , Suplementos Nutricionais , Hipersensibilidade a Drogas/imunologia , Hipersensibilidade a Drogas/microbiologia , Fluconazol/efeitos adversos , Fluconazol/farmacologia , Fungos/genética , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Hipersensibilidade/imunologia , Hipersensibilidade/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
13.
Front Immunol ; 3: 228, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22888331

RESUMO

Activation of phosphoinositide 3-kinase (PI3K) is required for B cell proliferation and survival. PI3K signaling also controls key aspects of B cell differentiation. Upon engagement of the B cell receptor (BCR), PI3K activation promotes Ca(2+) mobilization and activation of NFκB-dependent transcription, events which are essential for B cell proliferation. PI3K also initiates a distinct signaling pathway involving the Akt and mTOR serine/threonine kinases. It has been generally assumed that activation of Akt and mTOR downstream of PI3K is essential for B cell function. However, Akt and mTOR have complex roles in B cell fate decisions and suppression of this pathway can enhance certain B cell responses while repressing others. In this review we will discuss genetic and pharmacological studies of Akt and mTOR function in normal B cells, and in malignancies of B cell origin.

14.
Sci Signal ; 3(134): pe25, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20699473

RESUMO

The B cell receptor (BCR) and the pre-BCR control cell fate at many stages of B cell development, survival, and antigen response. Most of these processes require the activation of phosphatidylinositol 3-kinase (PI3K). Previous work has pointed to p110delta as the key catalytic isoform of PI3K for many B cell responses. A study of mice with different combinations of PI3K mutations confirms the central role of p110delta in agonist-mediated signaling, while identifying an unexpected function for the p110alpha isoform in tonic signaling by the pre-BCR and mature BCR.


Assuntos
Linfócitos B/imunologia , Modelos Imunológicos , Fosfatidilinositol 3-Quinases/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Transdução de Sinais/imunologia , Animais , Classe I de Fosfatidilinositol 3-Quinases , Genes RAG-1/imunologia , Camundongos , Mutação/genética , Fosfatidilinositol 3-Quinases/genética , Receptores de Antígenos de Linfócitos B/metabolismo
15.
Nat Med ; 16(2): 205-13, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20072130

RESUMO

Targeting the mammalian target of rapamycin (mTOR) protein is a promising strategy for cancer therapy. The mTOR kinase functions in two complexes, TORC1 (target of rapamycin complex-1) and TORC2 (target of rapamycin complex-2); however, neither of these complexes is fully inhibited by the allosteric inhibitor rapamycin or its analogs. We compared rapamycin with PP242, an inhibitor of the active site of mTOR in both TORC1 and TORC2 (hereafter referred to as TORC1/2), in models of acute leukemia harboring the Philadelphia chromosome (Ph) translocation. We demonstrate that PP242, but not rapamycin, causes death of mouse and human leukemia cells. In vivo, PP242 delays leukemia onset and augments the effects of the current front-line tyrosine kinase inhibitors more effectively than does rapamycin. Unexpectedly, PP242 has much weaker effects than rapamycin on the proliferation and function of normal lymphocytes. PI-103, a less selective TORC1/2 inhibitor that also targets phosphoinositide 3-kinase (PI3K), is more immunosuppressive than PP242. These findings establish that Ph(+) transformed cells are more sensitive than normal lymphocytes to selective TORC1/2 inhibitors and support the development of such inhibitors for leukemia therapy.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Leucemia/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Animais , Humanos , Camundongos , Sirolimo/farmacologia
16.
Am J Physiol Endocrinol Metab ; 293(4): E941-9, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17623749

RESUMO

The aims of this investigation were 1) to determine whether endurance exercise training could reverse impairments in insulin-stimulated compartmentalization and/or activation of aPKCzeta/lambda and Akt2 in skeletal muscle from high-fat-fed rodents and 2) to assess whether the PPARgamma agonist rosiglitazone could reverse impairments in skeletal muscle insulin signaling typically observed after high-fat feeding. Sprague-Dawley rats were placed on chow (NORCON, n = 16) or high-fat (n = 64) diets for 4 wk. During a subsequent 4-wk experimental period, high-fat-fed rats were allocated (n = 16/group) to either sedentary control (HFC), exercise training (HFX), rosiglitazone treatment (HFRSG), or a combination of both exercise training and rosiglitazone (HFRX). Following the 4-wk experimental period, animals underwent hindlimb perfusions. Insulin-stimulated plasma membrane-associated aPKCzeta and -lambda protein concentration, aPKCzeta/lambda activity, GLUT4 protein concentration, cytosolic Akt2, and aPKCzeta/lambda activities were reduced (P < 0.05) in HFC compared with NORCON. Cytosolic Akt2, aPKCzeta, and aPKClambda protein concentrations were not affected in HFC compared with NORCON. Exercise training reversed the deleterious effects of the high-fat diet such that insulin-stimulated compartmentalization and activation of components of the insulin-signaling cascade in HFX were normalized to NORCON. High-fat diet-induced impairments to skeletal muscle glucose metabolism were not reversed by rosiglitazone administration, nor did rosiglitazone augment the effect of exercise. Our findings indicate that chronic exercise training, but not rosiglitazone, reverses high-fat diet induced impairments in compartmentalization and activation of components of the insulin-signaling cascade in skeletal muscle.


Assuntos
Compartimento Celular , Dieta Aterogênica , Insulina/metabolismo , Músculo Esquelético/metabolismo , Condicionamento Físico Animal/fisiologia , Tecido Adiposo/anatomia & histologia , Tecido Adiposo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Compartimento Celular/efeitos dos fármacos , Epididimo/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Proteínas Substratos do Receptor de Insulina , Masculino , Músculo Esquelético/efeitos dos fármacos , Fosfoproteínas/metabolismo , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Rosiglitazona , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tiazolidinedionas/farmacologia
17.
J Physiol ; 565(Pt 2): 627-36, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15802290

RESUMO

Several recent reports using cell lines have suggested that both Akt and atypical protein kinase C (aPKC) zeta/lambda are translocated to the plasma membrane (PM) in response to insulin. However, it has yet to be determined in skeletal muscle whether: (1) insulin increases PM-associated Akt2, aPKC zeta and/or lambda protein concentration, (2) the activity of these kinases is altered by insulin at the PM, and (3) high fat feeding alters the insulin-stimulated PM concentration and/or activity of Akt2 and aPKC zeta/lambda. Sprague-Dawley rats were randomly assigned to either normal (n=16) or high fat (n=16) dietary groups. Following a 12 week dietary period, animals were subjected to hind limb perfusions in the presence (n=8 per group) or absence (n=8 per group) of insulin. In normal skeletal muscle, total PI3-kinase, Akt2 and aPKC zeta/lambda activities were increased by insulin. PM-associated aPKC zeta and lambda, and aPKC zeta/lambda activity, but not Akt2 or Akt2 activity, were increased by insulin in normal muscle. High fat feeding did not alter total skeletal muscle Akt2, aPKC zeta or aPKC lambda protein concentration. Insulin-stimulated total PI3-kinase, Akt2 and aPKC zeta/lambda activities were reduced in the high fat fed animals. Insulin-stimulated PM aPKC zeta, aPKC lambda, aPKC zeta/lambda activity and GLUT4 protein concentration were also reduced in high fat fed animals. These findings suggest that in skeletal muscle, insulin stimulates translocation of aPKC zeta and lambda, but not Akt2, to the PM. In addition, high fat feeding impairs insulin-stimulated activation of total aPKC zeta/lambda and Akt2, as well as PM association and activation of aPKC zeta and lambda.


Assuntos
Membrana Celular/enzimologia , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Músculo Esquelético/enzimologia , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Tecido Adiposo/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Gorduras na Dieta/farmacologia , Epididimo/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 4 , Proteínas Substratos do Receptor de Insulina , Isoenzimas/metabolismo , Masculino , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteínas Musculares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa