Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 487
Filtrar
1.
Cytotherapy ; 26(2): 126-135, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38043051

RESUMO

BACKGROUND AIMS: Amyotrophic lateral sclerosis (ALS) is a fatal disease associated with motor neuron degeneration, accumulation of aggregated misfolded proteins and neuroinflammation in motor regions of the central nervous system (CNS). Clinical trials using regulatory T cells (Tregs) are ongoing because of Tregs' immunomodulatory function, ability to traffic to the CNS, high numbers correlating with slower disease in ALS and disease-modifying activity in ALS mouse models. In the current study, a chimeric antigen receptor (CAR) was developed and characterized in human Tregs to enhance their immunomodulatory activity when in contact with an ALS protein aggregate. METHODS: A CAR (DG05-28-3z) consisting of a human superoxide dismutase 1 (hSOD1)-binding single-chain variable fragment, CD28 hinge, transmembrane and co-stimulatory domain and CD3ζ signaling domain was created and expressed in human Tregs. Human Tregs were isolated by either magnetic enrichment for CD4+CD25hi cells (Enr-Tregs) or cell sorting for CD4+CD25hiCD127lo cells (FP-Tregs), transduced and expanded for 17 days. RESULTS: The CAR bound preferentially to the ALS mutant G93A-hSOD1 protein relative to the wild-type hSOD1 protein. The CAR Tregs produced IL-10 when cultured with aggregated G93A-hSOD1 proteins or spinal cord explants from G93A-hSOD1 transgenic mice. Co-culturing DG05-28-3z CAR Tregs with human monocytes/macrophages inhibited production of tumor necrosis factor alpha and reactive oxygen species. Expanded FP-Tregs resulted in more robust Tregs compared with Enr-Tregs. FP-Tregs produced similar IL-10 and less interferon gamma, had lower Treg-specific demethylated region methylation and expressed higher FoxP3 and CD39. CONCLUSIONS: Taken together, this study demonstrates that gene-modified Tregs can be developed to target an aggregated ALS-relevant protein to elicit CAR-mediated Treg effector functions and provides an approach for generating Treg therapies for ALS with the goal of enhanced disease site-specific immunomodulation.


Assuntos
Esclerose Lateral Amiotrófica , Receptores de Antígenos Quiméricos , Camundongos , Animais , Humanos , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Superóxido Dismutase-1/uso terapêutico , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/terapia , Receptores de Antígenos Quiméricos/uso terapêutico , Interleucina-10/genética , Superóxido Dismutase/metabolismo , Camundongos Transgênicos , Linfócitos T CD4-Positivos/metabolismo , Imunomodulação , Modelos Animais de Doenças
2.
Commun Biol ; 6(1): 1261, 2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-38087051

RESUMO

The amphibian skin microbiome is an important component of anti-pathogen defense, but the impact of environmental change on the link between microbiome composition and host stress remains unclear. In this study, we used radiotelemetry and host translocation to track microbiome composition and function, pathogen infection, and host stress over time across natural movement paths for the forest-associated treefrog, Boana faber. We found a negative correlation between cortisol levels and putative microbiome function for frogs translocated to forest fragments, indicating strong integration of host stress response and anti-pathogen potential of the microbiome. Additionally, we observed a capacity for resilience (resistance to structural change and functional loss) in the amphibian skin microbiome, with maintenance of putative pathogen-inhibitory function despite major temporal shifts in microbiome composition. Although microbiome community composition did not return to baseline during the study period, the rate of microbiome change indicated that forest fragmentation had more pronounced effects on microbiome composition than translocation alone. Our findings reveal associations between stress hormones and host microbiome defenses, with implications for resilience of amphibians and their associated microbes facing accelerated tropical deforestation.


Assuntos
Anuros , Microbiota , Animais , Pele
3.
J Public Health Res ; 12(3): 22799036231193071, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37622100

RESUMO

Background: The COVID-19 pandemic brought changes to primary school lunches leading to concerns over nutritional quality and uptake of lunches by vulnerable children. Regional data from Tayside, Scotland, showed that only 55% of children who were eligible for free school meals took these (normal uptake pre-pandemic was 66%). The current work aimed to identify teachers' perceptions of meal provisioning in primary schools during the first year of the COVID-19 pandemic. Design and methods: A cross-sectional online survey was carried out among primary school teachers across Tayside, Scotland. Using an online survey (21 questions combining multiple choice formats and open text) and interviews, primary school teachers shared their views on food quality, quantity, meal choices and factors influencing uptake of primary school lunches. Interviews were recorded, transcribed and thematically analysed with respect to factors influencing consumption. Results: The survey was completed by 41 teachers and 8 participated in a follow up interview. Around one-third (29%) of primary school teachers believed the quality of lunches had decreased and cited poor appearance of food, use of takeaway containers and food wastage. The lunch format was viewed negatively principally relating to the substitution of hot lunches with cold sandwiches, portion sizes, choice and perceived value for money. Concerns were expressed about acceptability and how far the meals contributed to food security. Conclusions: Further work on food provisioning is needed in order to identify ways to provide a nutritional safety net for vulnerable children.

4.
Cancer Immunol Res ; 10(8): 962-977, 2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35696724

RESUMO

Chimeric-antigen receptor (CAR) T-cell therapy has shown remarkable efficacy against hematologic tumors. Yet, CAR T-cell therapy has had little success against solid tumors due to obstacles presented by the tumor microenvironment (TME) of these cancers. Here, we show that CAR T cells armored with the engineered IL-2 superkine Super2 and IL-33 were able to promote tumor control as a single-agent therapy. IFNγ and perforin were dispensable for the effects of Super2- and IL-33-armored CAR T cells. Super2 and IL-33 synergized to shift leukocyte proportions in the TME and to recruit and activate a broad repertoire of endogenous innate and adaptive immune cells including tumor-specific T cells. However, depletion of CD8+ T cells or NK cells did not disrupt tumor control, suggesting that broad immune activation compensated for loss of individual cell subsets. Thus, we have shown that Super2 and IL-33 CAR T cells can promote antitumor immunity in multiple solid tumor models and can potentially overcome antigen loss, highlighting the potential of this universal CAR T-cell platform for the treatment of solid tumors.


Assuntos
Neoplasias , Microambiente Tumoral , Humanos , Imunoterapia Adotiva , Interleucina-2 , Interleucina-33
5.
Immunohorizons ; 4(10): 597-607, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-33037097

RESUMO

The NKG2D ligand MHC class I chain-related protein A (MICA) is expressed on many varieties of malignant cells but is absent from most normal tissues, and thus represents a potential target for chimeric Ag receptor (CAR) T cell-based therapeutics. However, there are more than 100 alleles of MICA, so the ability to target a conserved site is needed for a therapy to be used in most patients. In this study, we describe a fully human anti-MICA CAR created by fusing the single-chain fragment variable B2 to the full length DAP10 protein and the traditional CD3ζ signaling domain. Human T cells expressing the B2 CAR killed MICA-positive tumor cells, produced IFN-γ when in contact with MICA-positive tumor cells or plate-bound MICA protein, and inhibited PANC-1 growth in a mouse xenograft model. To localize B2's epitope on MICA, we used novel computational methods to model potential binding modes and to design mutational variants of MICA testing these hypotheses. Flow cytometry using a commercial anti-MICA/MICB Ab indicated that the variant proteins were expressed at high levels on transduced P815 cell lines. One variant protein (R38S/K40T/K57E) showed reduced staining with a B2-IgG1 fusion protein compared with controls and did not induce IFN-γ production by human T cells expressing the B2 CAR. These results show antitumor activity of MICA-specific CAR T cells and indicate an essential role for a conserved site in the exposed loop involving aa 38-57 of MICA. This study describes a novel MICA-specific CAR and discusses its potential use as a cancer therapeutic.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Animais , Linhagem Celular Tumoral , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Future Oncol ; 13(18): 1593-1605, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28613086

RESUMO

Chimeric antigen receptors (CARs) are genetically engineered proteins that combine an extracellular antigen-specific recognition domain with one or several intracellular T-cell signaling domains. When expressed in T cells, these CARs specifically trigger T-cell activation upon antigen recognition. While the clinical proof of principle of CAR T-cell therapy has been established in hematological cancers, CAR T cells are only at the early stages of being explored to tackle solid cancers. This special report discusses the concept of exploiting natural killer cell receptors as an approach that could broaden the specificity of CAR T cells and potentially enhance the efficacy of this therapy against solid tumors. New data demonstrating feasibility of this approach in humans and supporting the ongoing clinical trial are also presented.


Assuntos
Antígenos de Neoplasias/imunologia , Imunoterapia Adotiva , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Neoplasias/imunologia , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Antígenos de Neoplasias/metabolismo , Ensaios Clínicos como Assunto , Citotoxicidade Imunológica , Avaliação Pré-Clínica de Medicamentos , Humanos , Imunoterapia Adotiva/métodos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Neoplasias/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Resultado do Tratamento
8.
J Perinatol ; 37(6): 658-661, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28206994

RESUMO

OBJECTIVE: The objective of this study is to determine the impact of postnatal age on the bias between transcutaneous (TcB) and total serum bilirubin (TSB), and evaluate a TcB screening protocol. STUDY DESIGN: Preterm and term infants had paired TcB and TSB performed on days 1 to 3 of life; a subset of preterm infants had measurements on days 4 to 7. Sensitivity and specificity of TcB (plotted on an age-specific TSB nomogram) for prediction of high-intermediate (HIR) or high-risk TSB were calculated. RESULTS: Median TcB bias was 2.6 and 2.5 mg dl-1 for term and preterm infants in the first 3 days of life, respectively. However, median bias was 2.2 mg dl-1 for preterm infants at 4 to 7 days of life. TcB in preterm infants predicted HIR or high-risk TSB with 94% sensitivity and 56% specificity. CONCLUSION: TcB screening protocols developed for term infants can be used for late preterm infants in the first 3 days of life.


Assuntos
Bilirrubina/sangue , Recém-Nascido Prematuro/sangue , Triagem Neonatal/métodos , Nascimento a Termo/sangue , Feminino , Humanos , Hiperbilirrubinemia Neonatal/sangue , Hiperbilirrubinemia Neonatal/diagnóstico , Recém-Nascido , Icterícia Neonatal/sangue , Icterícia Neonatal/diagnóstico , Masculino , Minnesota , Nomogramas , Estudos Prospectivos , Sensibilidade e Especificidade
9.
J Immunol ; 197(12): 4674-4685, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27849169

RESUMO

Targeting cancer through the use of effector T cells bearing chimeric Ag receptors (CARs) leads to elimination of tumors in animals and patients, but recognition of normal cells or excessive activation can result in significant toxicity and even death. CAR T cells based on modified NKG2D receptors are effective against many types of tumors, and their efficacy is mediated through direct cytotoxicity and cytokine production. Under certain conditions, their ligands can be expressed on nontumor cells, so a better understanding of the potential off-tumor activity of these NKG2D CAR T cells is needed. Injection of very high numbers of activated T cells expressing CARs based on murine NKG2D or DNAM1 resulted in increased serum cytokines (IFN-γ, IL-6, and MCP-1) and acute toxicity similar to cytokine release syndrome. Acute toxicity required two key effector molecules in CAR T cells-perforin and GM-CSF. Host immune cells also contributed to this toxicity, and mice with severe immune cell defects remained healthy at the highest CAR T cell dose. These data demonstrate that specific CAR T cell effector mechanisms and the host immune system are required for this cytokine release-like syndrome in murine models.


Assuntos
Vacinas Anticâncer/imunologia , Imunoterapia Adotiva/métodos , Linfoma de Células T/terapia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Linfócitos T/fisiologia , Animais , Complexo CD3/genética , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Ativação Linfocitária , Linfoma de Células T/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Neoplasias Experimentais , Perforina/metabolismo , Proteínas Recombinantes de Fusão/genética , Linfócitos T/transplante
10.
Nutr Metab Cardiovasc Dis ; 26(5): 374-9, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27033025

RESUMO

BACKGROUND AND AIMS: To assess the efficacy and safety of saxagliptin 2.5 and 5 mg/d in patients with type 2 diabetes mellitus (T2DM) and high risk of coronary heart disease (CHD) or stroke as estimated by the United Kingdom Prospective Diabetes Study (UKPDS) risk engine. METHODS AND RESULTS: Post hoc analysis of data pooled from 5 previously reported phase 3, randomized, placebo-controlled, 24-week studies was conducted. Patients were stratified into subgroups by UKPDS 10-year CHD and/or stroke risk ≥20% and CHD and stroke risk <20%. End points were adjusted mean change from baseline in glycated hemoglobin (HbA1c), fasting plasma glucose (FPG), 120-min postprandial glucose (PPG), and body weight and the proportion of patients achieving HbA1c <7% and ≤8% at week 24. Pooled safety data were analyzed for adverse events (AEs) and hypoglycemia. Both doses of saxagliptin reduced HbA1c, FPG, and PPG to a greater extent than placebo regardless of UKPDS risk score. The proportions of patients achieving HbA1c <7% and ≤8% were greater with saxagliptin than placebo and consistent across risk score groups. AE profile and hypoglycemia incidence were similar for saxagliptin and placebo across UKPDS risk score groups. CONCLUSION: Saxagliptin was well tolerated and improved glycemic control in patients with T2DM regardless of their CHD and stroke UKPDS risk score. Clinical trial registration numbers: Clinicaltrials.gov NCT00121641, NCT00316082, NCT00121667, NCT00313313, and NCT00295633.


Assuntos
Adamantano/análogos & derivados , Glicemia/efeitos dos fármacos , Doenças Cardiovasculares/prevenção & controle , Diabetes Mellitus Tipo 2/tratamento farmacológico , Dipeptídeos/uso terapêutico , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Adamantano/efeitos adversos , Adamantano/uso terapêutico , Idoso , Biomarcadores/sangue , Glicemia/metabolismo , Doenças Cardiovasculares/etiologia , Ensaios Clínicos Fase III como Assunto , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/enzimologia , Dipeptídeos/efeitos adversos , Inibidores da Dipeptidil Peptidase IV/efeitos adversos , Feminino , Hemoglobinas Glicadas/metabolismo , Humanos , Hipoglicemia/sangue , Hipoglicemia/induzido quimicamente , Masculino , Pessoa de Meia-Idade , Ensaios Clínicos Controlados Aleatórios como Assunto , Medição de Risco , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
11.
Nanotechnology ; 25(47): 475101, 2014 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-25371538

RESUMO

Iron oxide nanoparticles (IONPs) hold great potential for cancer therapy. Actively targeting IONPs to tumor cells can further increase therapeutic efficacy and decrease off-target side effects. To target tumor cells, a natural killer (NK) cell activating receptor, NKG2D, was utilized to develop pan-tumor targeting IONPs. NKG2D ligands are expressed on many tumor types and its ligands are not found on most normal tissues under steady state conditions. The data showed that mouse and human fragment crystallizable (Fc)-fusion NKG2D (Fc-NKG2D) coated IONPs (NKG2D/NPs) can target multiple NKG2D ligand positive tumor types in vitro in a dose dependent manner by magnetic cell sorting. Tumor targeting effect was robust even under a very low tumor cell to normal cell ratio and targeting efficiency correlated with NKG2D ligand expression level on tumor cells. Furthermore, the magnetic separation platform utilized to test NKG2D/NP specificity has the potential to be developed into high throughput screening strategies to identify ideal fusion proteins or antibodies for targeting IONPs. In conclusion, NKG2D/NPs can be used to target multiple tumor types and magnetic separation platform can facilitate the proof-of-concept phase of tumor targeting IONP development.


Assuntos
Nanopartículas de Magnetita/química , Subfamília K de Receptores Semelhantes a Lectina de Células NK/química , Neoplasias/tratamento farmacológico , Animais , Humanos , Nanopartículas de Magnetita/uso terapêutico , Camundongos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/uso terapêutico
12.
Cell Death Differ ; 21(10): 1600-12, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24902899

RESUMO

Both receptor-interacting protein kinase 1 (RIPK1) and RIPK3 can signal cell death following death receptor ligation. To study the requirements for RIPK-triggered cell death in the absence of death receptor signaling, we engineered inducible versions of RIPK1 and RIPK3 that can be activated by dimerization with the antibiotic coumermycin. In the absence of TNF or other death ligands, expression and dimerization of RIPK1 was sufficient to cause cell death by caspase- or RIPK3-dependent mechanisms. Dimerized RIPK3 induced cell death by an MLKL-dependent mechanism but, surprisingly, also induced death mediated by FADD, caspase 8 and RIPK1. Catalytically active RIPK3 kinase domains were essential for MLKL-dependent but not for caspase 8-dependent death. When RIPK1 or RIPK3 proteins were dimerized, the mode of cell death was determined by the availability of downstream molecules such as FADD, caspase 8 and MLKL. These observations imply that rather than a 'switch' operating between the two modes of cell death, the final mechanism depends on levels of the respective signaling and effector proteins.


Assuntos
Apoptose/genética , Multimerização Proteica/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteínas Recombinantes/metabolismo , Aminocumarinas/metabolismo , Animais , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular , Proteína de Domínio de Morte Associada a Fas/metabolismo , Camundongos , Camundongos Knockout , Proteínas Quinases/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Transdução de Sinais/genética , Fator de Necrose Tumoral alfa/metabolismo
13.
Cell Death Dis ; 5: e1086, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24577084

RESUMO

Necroptosis is a mechanism by which cells can kill themselves that does not require caspase activity or the presence of the pro-apoptotic Bcl-2 family members Bax or Bak. It has been reported that RIPK3 (receptor interacting protein kinase 3) activates MLKL (mixed lineage kinase domain-like) to cause cell death that requires dynamin-related protein 1 (Drp1), because survival was increased in cells depleted of Drp1 or treated with the Drp1 inhibitor mdivi-1. To analyze necroptosis in a system that does not require addition of tumor necrosis factor (TNF), we used a construct that allows RIPK3 to be induced in cells, and then dimerized via an E. coli gyrase domain fused to its carboxyl-terminus, using the dimeric gyrase binding antibiotic coumermycin. We have previously shown elsewhere that RIPK3 dimerized in this manner not only induces necroptosis but also apoptosis, which can be inhibited by the broad-spectrum caspase inhibitor Q-VD-OPh (QVD). In response to RIPK3 dimerization, wild-type mouse embryonic fibroblasts (MEFs) underwent cell death that was reduced but not completely blocked by QVD. In contrast, death upon dimerization of RIPK3 in Mlkl(-/-) MEFs was completely inhibited with QVD, confirming that MLKL is required for necroptosis. Similar to wild-type MEFs, most Drp1(-/-) MEFs died when RIPK3 was activated, even in the presence of QVD. Furthermore, overexpression of wild-type MLKL or dominant active mutants of MLKL (Q343A or S345E/S347E) caused death of wild-type and Drp1(-/-) MEFs that was not inhibited with QVD. These results indicate that necroptosis caused by RIPK3 requires MLKL but not Drp1.


Assuntos
Apoptose , Dinaminas/metabolismo , Fibroblastos/enzimologia , Proteínas Quinases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais , Aminocumarinas/farmacologia , Animais , Antibacterianos/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Caspase/farmacologia , Linhagem Celular , Dinaminas/deficiência , Dinaminas/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Camundongos , Camundongos Knockout , Mutação , Necrose , Proteínas Quinases/deficiência , Proteínas Quinases/genética , Multimerização Proteica , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Fator de Necrose Tumoral alfa/farmacologia
15.
Glycobiology ; 23(10): 1192-203, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23893788

RESUMO

The methylotrophic yeast, Pichia pastoris, is an important organism used for the production of therapeutic proteins. Previously, we have reported the glycoengineering of this organism to produce human-like N-linked glycans but up to now no one has addressed engineering the O-linked glycosylation pathway. Typically, O-linked glycans produced by wild-type P. pastoris are linear chains of four to five α-linked mannose residues, which may be capped with ß- or phospho-mannose. Previous genetic engineering of the N-linked glycosylation pathway of P. pastoris has eliminated both of these two latter modifications, resulting in O-linked glycans which are linear α-linked mannose structures. Here, we describe a method for the co-expression of an α-1,2-mannosidase, which reduces these glycans to primarily a single O-linked mannose residue. In doing so, we have reduced the potential of these glycans to interact with carbohydrate-binding proteins, such as dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin. Furthermore, the introduction of the enzyme protein-O-linked-mannose ß-1,2-N-acetylglucosaminyltransferase 1, resulted in the capping of the single O-linked mannose residues with N-acetylglucosamine. Subsequently, this glycoform was extended into human-like sialylated glycans, similar in structure to α-dystroglycan-type glycoforms. As such, this represents the first example of sialylated O-linked glycans being produced in yeast and extends the utility of the P. pastoris production platform beyond N-linked glycosylated biotherapeutics to include molecules possessing O-linked glycans.


Assuntos
Manose/biossíntese , Engenharia Metabólica/métodos , Pichia/metabolismo , alfa-Manosidase/metabolismo , Pichia/crescimento & desenvolvimento , Engenharia de Proteínas , alfa-Manosidase/genética
16.
PLoS One ; 8(7): e68325, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23840891

RESUMO

The methylotrophic yeast, Pichiapastoris, is an important organism used for the production of therapeutic proteins. However, the presence of fungal-like glycans, either N-linked or O-linked, can elicit an immune response or enable the expressed protein to bind to mannose receptors, thus reducing their efficacy. Previously we have reported the elimination of ß-linked glycans in this organism. In the current report we have focused on reducing the O-linked mannose content of proteins produced in P. pastoris, thereby reducing the potential to bind to mannose receptors. The initial step in the synthesis of O-linked glycans in P. pastoris is the transfer of mannose from dolichol-phosphomannose to a target protein in the yeast secretory pathway by members of the protein-O-mannosyltransferase (PMT) family. In this report we identify and characterize the members of the P. pastoris PMT family. Like Candida albicans, P. pastoris has five PMT genes. Based on sequence homology, these PMTs can be grouped into three sub-families, with both PMT1 and PMT2 sub-families possessing two members each (PMT1 and PMT5, and PMT2 and PMT6, respectively). The remaining sub-family, PMT4, has only one member (PMT4). Through gene knockouts we show that PMT1 and PMT2 each play a significant role in O-glycosylation. Both, by gene knockouts and the use of Pmt inhibitors we were able to significantly reduce not only the degree of O-mannosylation, but also the chain-length of these glycans. Taken together, this reduction of O-glycosylation represents an important step forward in developing the P. pastoris platform as a suitable system for the production of therapeutic glycoproteins.


Assuntos
Manosiltransferases/genética , Pichia/enzimologia , Pichia/genética , Candida albicans/enzimologia , Candida albicans/genética , Técnicas de Inativação de Genes , Genes Fúngicos , Glicosilação , Manosiltransferases/metabolismo , Filogenia , Pichia/metabolismo , Polissacarídeos/metabolismo
17.
Cell Death Dis ; 4: e465, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23328672

RESUMO

Ligation of tumor necrosis factor receptor 1 (TNFR1) can cause cell death by caspase 8 or receptor-interacting protein kinase 1 (RIPK1)- and RIPK3-dependent mechanisms. It has been assumed that because RIPK1 bears a death domain (DD), but RIPK3 does not, RIPK1 is necessary for recruitment of RIPK3 into signaling and death-inducing complexes. To test this assumption, we expressed elevated levels of RIPK3 in murine embryonic fibroblasts (MEFs) from wild-type (WT) and gene-deleted mice, and exposed them to TNF. Neither treatment with TNF nor overexpression of RIPK3 alone caused MEFs to die, but when levels of RIPK3 were increased, addition of TNF killed WT, Ripk1(-/-), caspase 8(-/-), and Bax(-/-)/Bak(-/-) MEFs, even in the presence of the broad-spectrum caspase inhibitor Q-VD-OPh. In contrast, Tnfr1(-/-) and Tradd(-/-) MEFs did not die. These results show for the first time that in the absence of RIPK1, TNF can activate RIPK3 to induce cell death both by a caspase 8-dependent mechanism and by a separate Bax/Bak- and caspase-independent mechanism. RIPK1 is therefore not essential for TNF to activate RIPK3 to induce necroptosis nor for the formation of a functional ripoptosome/necrosome.


Assuntos
Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Caspase 8/genética , Caspase 8/metabolismo , Inibidores de Caspase/farmacologia , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Camundongos , Necrose , Quinolinas/farmacologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Proteína de Domínio de Morte Associada a Receptor de TNF/deficiência , Proteína de Domínio de Morte Associada a Receptor de TNF/genética , Proteína de Domínio de Morte Associada a Receptor de TNF/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteína Killer-Antagonista Homóloga a bcl-2/deficiência , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína Killer-Antagonista Homóloga a bcl-2/metabolismo , Proteína X Associada a bcl-2/deficiência , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
18.
J Virol ; 87(4): 2058-71, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23221564

RESUMO

Myeloid-derived suppressor cells (MDSCs) have been characterized in several disease settings, especially in many tumor systems. Compared to their involvement in tumor microenvironments, however, MDSCs have been less well studied in their responses to infectious disease processes, in particular to retroviruses that induce immunodeficiency. Here, we demonstrate for the first time the development of a highly immunosuppressive MDSC population that is dependent on infection by the LP-BM5 retrovirus, which causes murine acquired immunodeficiency. These MDSCs express a cell surface marker signature (CD11b(+) Gr-1(+) Ly6C(+)) characteristic of monocyte-type MDSCs. Such MDSCs profoundly inhibit immune responsiveness by a cell dose- and substantially inducible nitric oxide synthase (iNOS)-dependent mechanism that is independent of arginase activity, PD-1-PD-L1 expression, and interleukin 10 (IL-10) production. These MDSCs display levels of immunosuppressive function in parallel with the extent of disease in LP-BM5-infected wild-type (w.t.) versus knockout mouse strains that are differentially susceptible to pathogenesis. These MDSCs suppressed not only T-cell but also B-cell responses, which are an understudied target for MDSC inhibition. The MDSC immunosuppression of B-cell responses was confirmed by the use of purified B responder cells, multiple B-cell stimuli, and independent assays measuring B-cell expansion. Retroviral load measurements indicated that the suppressive Ly6G(low/±) Ly6C(+) CD11b(+)-enriched MDSC subset was positive for LP-BM5, albeit at a significantly lower level than that of nonfractionated splenocytes from LP-BM5-infected mice. These results, including the strong direct MDSC inhibition of B-cell responsiveness, are novel for murine retrovirus-induced immunosuppression and, as this broadly suppressive function mirrors that of the LP-BM5-induced disease syndrome, support a possible pathogenic effector role for these retrovirus-induced MDSCs.


Assuntos
Síndrome da Imunodeficiência Adquirida/virologia , Linfócitos B/imunologia , Tolerância Imunológica , Monócitos/imunologia , Retroviridae/patogenicidade , Linfócitos T/imunologia , Síndrome da Imunodeficiência Adquirida/imunologia , Animais , Antígenos Ly/análise , Antígeno CD11b/análise , Imunofenotipagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Quimiocinas/análise , Retroviridae/imunologia
19.
J Perinatol ; 32(11): 851-5, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22343396

RESUMO

OBJECTIVE: To determine the effect of universal transcutaneous bilirubin (TcB) screening on total serum bilirubin (TSB) blood draws and phototherapy usage. STUDY DESIGN: The subjects were infants ≥ 36 weeks gestation. In period 1, TSB was ordered based on clinical factors. In period 2, all infants underwent predischarge TcB measurement; infants with adjusted TcB values in the high-intermediate or high-risk zones had TSB ordered. Data were extracted through chart review. RESULT: TSB measurements per 1000 infants decreased from 717 to 713 (P=0.008) between period 1 and 2, with more outpatient and less inpatient blood draws in period 2. Between periods 1 and 2, total phototherapy decreased from 59 to 39 per 1000 infants (P<0.0001), with less inpatient and more readmission phototherapy. CONCLUSION: Universal TcB screening was implemented without increasing total blood draws or phototherapy treatment; however, it was associated with a shift in blood draws and phototherapy usage from inpatients to outpatients.


Assuntos
Bilirrubina/análise , Hiperbilirrubinemia Neonatal/epidemiologia , Hiperbilirrubinemia Neonatal/terapia , Triagem Neonatal , Flebotomia/estatística & dados numéricos , Fototerapia , Humanos , Recém-Nascido , Laboratórios Hospitalares , Triagem Neonatal/tendências , Fototerapia/estatística & dados numéricos , Guias de Prática Clínica como Assunto , Estudos Retrospectivos
20.
Cell Death Differ ; 19(5): 808-15, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22095280

RESUMO

Binding of TNF to TNF receptor-1 can give a pro-survival signal through activation of p65/RelA NF-κB, but also signals cell death. To determine the roles of FLICE-inhibitory protein (FLIP) and caspase-8 in TNF-induced activation of NF-κB and apoptosis, we used mouse embryonic fibroblasts derived from FLIP and caspase-8 gene-deleted mice, and treated them with TNF and a smac-mimetic compound that causes degradation of cellular inhibitor of apoptosis proteins (cIAPs). In cells treated with smac mimetic, TNF and Fas Ligand caused wild-type and FLIP(-/-) MEFs to die, whereas caspase-8(-/-) MEFs survived, indicating that caspase-8 is necessary for death of MEFs triggered by these ligands when IAPs are degraded. By contrast, neither caspase-8 nor FLIP was required for TNF to activate p65/RelA NF-κB, because IκB was degraded, p65 translocated to the nucleus, and an NF-κB reporter gene activated normally in caspase-8(-/-) or FLIP(-/-) MEFs. Reconstitution of FLIP(-/-) MEFs with the FLIP isoforms FLIP-L, FLIP-R, or FLIP-p43 protected these cells from dying when treated with TNF or FasL, whether or not cIAPs were depleted. These results show that in MEFs, caspase-8 is necessary for TNF- and FasL-induced death, and FLIP is needed to prevent it, but neither caspase-8 nor FLIP is required for TNF to activate NF-κB.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Caspase 8/metabolismo , NF-kappa B/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Caspase 8/genética , Morte Celular/efeitos dos fármacos , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Imunoprecipitação , Camundongos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...