Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Bioconjug Chem ; 35(6): 855-866, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38789102

RESUMO

Antibody effector functions including antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) are mediated through the interaction of the antibody Fc region with Fcγ receptors present on immune cells. Several approaches have been used to modulate antibody Fc-Fcγ interactions with the goal of driving an effective antitumor immune response, including Fc point mutations and glycan modifications. However, robust antibody-Fcγ engagement and immune cell binding of Fc-enhanced antibodies in the periphery can lead to the unwanted induction of systemic cytokine release and other dose-limiting infusion-related reactions. Creating a balance between effective engagement of Fcγ receptors that can induce antitumor activity without incurring systemic immune activation is an ongoing challenge in the field of antibody and immuno-oncology therapeutics. Herein, we describe a method for the reversible chemical modulation of antibody-Fcγ interactions using simple poly(ethylene glycol) (PEG) linkers conjugated to antibody interchain disulfides with maleimide attachments. This method enables dosing of a therapeutic with muted Fcγ engagement that is restored in vivo in a time-dependent manner. The technology was applied to an effector function enhanced agonist CD40 antibody, SEA-CD40, and experiments demonstrate significant reductions in Fc-induced immune activation in vitro and in mice and nonhuman primates despite showing retained efficacy and improved pharmacokinetics compared to the parent antibody. We foresee that this simple, modular system can be rapidly applied to antibodies that suffer from systemic immune activation due to peripheral FcγR binding immediately upon infusion.


Assuntos
Receptores de IgG , Animais , Camundongos , Receptores de IgG/imunologia , Humanos , Polietilenoglicóis/química , Citotoxicidade Celular Dependente de Anticorpos , Fagocitose/efeitos dos fármacos
2.
Pharm Res ; 40(4): 873-887, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35352281

RESUMO

Exosomes are extracellular vesicles secreted by cells with a particle size of 30-150 nm in diameter. Exosomes can be used as natural drug carriers. The treatment of cancer with drug-loaded exosomes is an area of high interest. This review introduces the composition, function, isolation and characterization of exosomes, and briefly describes the selection of exosome donor cells and methods for drug loading. Through studies on therapies with drug-loaded exosomes in gastric cancer, lung cancer, brain cancer and other cancers, the advantages and disadvantages of drug-loaded exosomes have been analyzed.


Assuntos
Neoplasias Encefálicas , Exossomos , Neoplasias Pulmonares , Humanos , Sistemas de Liberação de Medicamentos/métodos , Portadores de Fármacos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico
3.
J Allergy Clin Immunol ; 139(2): 508-518.e4, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27394914

RESUMO

BACKGROUND: TH2 responses are implicated in asthma pathobiology. Epidemiologic studies have found a positive association between asthma and exposure to staphylococcal enterotoxins. OBJECTIVE: We used a mouse model of asthma to determine whether staphylococcal enterotoxins promote TH2 differentiation of allergen-specific CD4 conventional T (Tcon) cells and asthma by activating allergen-nonspecific regulatory T (Treg) cells to create a TH2-polarizing cytokine milieu. METHODS: Ovalbumin (OVA)-specific, staphylococcal enterotoxin A (SEA)-nonreactive naive CD4 Tcon cells were cocultured with SEA-reactive allergen-nonspecific Treg or CD4 Tcon cells in the presence of OVA and SEA. The OVA-specific CD4 T cells were then analyzed for IL-13 and IFN-γ expression. SEA-activated Treg cells were analyzed for the expression of the TH2-polarizing cytokine IL-4 and the T-cell activation markers CD69 and CD62L. For asthma induction, mice were intratracheally sensitized with OVA or cat dander extract (CDE) alone or together with SEA and then challenged with OVA or CDE. Mice were also subject to transient Treg cell depletion before sensitization with OVA plus SEA. Asthma features and TH2 differentiation in these mice were analyzed. RESULTS: SEA-activated Treg cells induced IL-13 but suppressed IFN-γ expression in OVA-specific CD4 Tcon cells. SEA-activated Treg cells expressed IL-4, upregulated CD69, and downregulated CD62L. Sensitization with OVA plus SEA but not OVA alone induced asthma, and SEA exacerbated asthma induced by CDE. Depletion of Treg cells abolished these effects of SEA and IL-13 expression in OVA-specific T cells. CONCLUSION: SEA promoted TH2 responses of allergen-specific T cells and asthma pathogenesis by activating Treg cells.


Assuntos
Asma/imunologia , Enterotoxinas/imunologia , Linfócitos T Reguladores/imunologia , Células Th2/imunologia , Alérgenos/imunologia , Animais , Diferenciação Celular , Células Cultivadas , Modelos Animais de Doenças , Humanos , Interferon gama/metabolismo , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina/imunologia , Traqueia/metabolismo
4.
Proc Natl Acad Sci U S A ; 110(14): 5404-9, 2013 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-23493549

RESUMO

The key role played by fucose in glycoprotein and cellular function has prompted significant research toward identifying recombinant and biochemical strategies for blocking its incorporation into proteins and membrane structures. Technologies surrounding engineered cell lines have evolved for the inhibition of in vitro fucosylation, but they are not applicable for in vivo use and drug development. To address this, we screened a panel of fucose analogues and identified 2-fluorofucose and 5-alkynylfucose derivatives that depleted cells of GDP-fucose, the substrate used by fucosyltransferases to incorporate fucose into protein and cellular glycans. The inhibitors were used in vitro to generate fucose-deficient antibodies with enhanced antibody-dependent cellular cytotoxicity activities. When given orally to mice, 2-fluorofucose inhibited fucosylation of endogenously produced antibodies, tumor xenograft membranes, and neutrophil adhesion glycans. We show that oral 2-fluorofucose treatment afforded complete protection from tumor engraftment in a syngeneic tumor vaccine model, inhibited neutrophil extravasation, and delayed the outgrowth of tumor xenografts in immune-deficient mice. The results point to several potential therapeutic applications for molecules that selectively block the endogenous generation of fucosylated glycan structures.


Assuntos
Anticorpos Monoclonais/metabolismo , Vacinas Anticâncer/farmacologia , Fucose/farmacologia , Fucosiltransferases/antagonistas & inibidores , Guanosina Difosfato Fucose/metabolismo , Polissacarídeos/metabolismo , Animais , Células CHO , Linhagem Celular Tumoral , Cromatografia Líquida , Cricetinae , Cricetulus , Desenho de Fármacos , Feminino , Fucose/química , Humanos , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos BALB C , Estrutura Molecular , Neutrófilos/metabolismo
5.
Blood ; 122(8): 1455-63, 2013 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-23770776

RESUMO

Outcomes in acute myeloid leukemia (AML) remain unsatisfactory, and novel treatments are urgently needed. One strategy explores antibodies and their drug conjugates, particularly those targeting CD33. Emerging data with gemtuzumab ozogamicin (GO) demonstrate target validity and activity in some patients with AML, but efficacy is limited by heterogeneous drug conjugation, linker instability, and a high incidence of multidrug resistance. We describe here the development of SGN-CD33A, a humanized anti-CD33 antibody with engineered cysteines conjugated to a highly potent, synthetic DNA cross-linking pyrrolobenzodiazepine dimer via a protease-cleavable linker. The use of engineered cysteine residues at the sites of drug linker attachment results in a drug loading of approximately 2 pyrrolobenzodiazepine dimers per antibody. In preclinical testing, SGN-CD33A is more potent than GO against a panel of AML cell lines and primary AML cells in vitro and in xenotransplantation studies in mice. Unlike GO, antileukemic activity is observed with SGN-CD33A in AML models with the multidrug-resistant phenotype. Mechanistic studies indicate that the cytotoxic effects of SGN-CD33A involve DNA damage with ensuing cell cycle arrest and apoptotic cell death. Together, these data suggest that SGN-CD33A has CD33-directed antitumor activity and support clinical testing of this novel therapeutic in patients with AML.


Assuntos
Anticorpos Monoclonais Humanizados/química , Benzodiazepinas/química , Resistencia a Medicamentos Antineoplásicos , Imunoconjugados/química , Leucemia Mieloide Aguda/tratamento farmacológico , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/química , Animais , Apoptose , Ciclo Celular , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacologia , Cisteína/genética , Dimerização , Desenho de Fármacos , Células HEK293 , Células HL-60 , Humanos , Leucemia Mieloide Aguda/imunologia , Camundongos
6.
J Neurotrauma ; 41(7-8): 844-861, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38047531

RESUMO

Traumatic brain injury (TBI) is a leading cause of death and disability, primarily caused by falls and motor vehicle collisions (MVCs). Although many TBIs are preventable, there is a notable lack of studies exploring the association of geographically defined TBI hotspots with social deprivation. Geographic information systems (GIS) can be used to identify at-risk neighborhoods (hotspots) for targeted interventions. This study aims to determine the spatial distribution of TBI by major causes and to explore the sociodemographic and economic characteristics of TBI hotspots and cold spots in Nova Scotia. Patient data for TBIs from 2003 to 2019 were obtained from the Nova Scotia Trauma Registry. Residential postal codes were geocoded and assigned to dissemination areas (DA). Area-based risk factors and deprivation status (residential instability [RI], economic dependency [ED], ethnocultural composition [EC], and situational vulnerability [SV]) from the national census data were linked to DAs. Spatial autocorrelation was assessed using Moran's I, and hotspot analysis was performed using Getis-Ord Gi* statistic. Differences in risk factors between hot and cold spots were evaluated using the Mann-Whitney U test for numerical variables and the χ2 test or Fisher's exact test for categorical variables. A total of 5394 TBI patients were eligible for inclusion in the study. The distribution of hotspots for falls exhibited no significant difference between urban and rural areas (p = 0.71). Conversely, hotspots related to violence were predominantly urban (p = 0.001), whereas hotspots for MVCs were mostly rural (p < 0.001). Distinct dimensions of deprivation were associated with falls, MVCs, and violent hotspots. Fall hotspots were significantly associated with areas characterized by higher RI (p < 0.001) and greater ethnocultural diversity (p < 0.001). Conversely, the same domains exhibited an inverse relationship with MVC hotspots; areas with low RI and ethnic homogeneity displayed a higher proportion of MVC hotspots. ED and SV exhibited a strong gradient with MVC hotspots; the most deprived quintiles displayed the highest proportion of MVC hotspots compared with cold spots (ED; p = 0.002, SV; p < 0.001). Areas with the highest levels of ethnocultural diversity were found to have a significantly higher proportion of violence-related hotspots than cold spots (p = 0.005). This study offers two significant contributions to spatial epidemiology. First, it demonstrates the distribution of TBI hotspots by major injury causes using the smallest available geographical unit. Second, we disentangle the various pathways through which deprivation impacts the risk of main mechanisms of TBI. These findings provide valuable insights for public health officials to design targeted injury prevention strategies in high-risk areas.


Assuntos
Lesões Encefálicas Traumáticas , Humanos , Nova Escócia/epidemiologia , Lesões Encefálicas Traumáticas/epidemiologia , Análise Espacial , Fatores de Risco , Características de Residência
7.
Eur J Med Res ; 29(1): 270, 2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38704575

RESUMO

BACKGROUND: This study aims to investigate the effects of a conditioned medium (CM) from human umbilical cord mesenchymal stem cells (HuMSCs) cultivated in gelatin sponge (GS-HuMSCs-CM) on hair growth in a mouse model. METHODS: CM was collected from the HuMSCs cultivated in a monolayer or in a gelatin sponge. Vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), keratinocyte growth factor (KGF), and hepatocyte growth factor (HGF) levels in CMs were measured by enzyme-linked immunosorbent assays (ELISAs). A hair loss model by a C57 BL/6J mouse was prepared. The effects of GS-HuMSCs-CM and HuMSCs on hair regrowth in mice were investigated by intradermal injection in the depilated back skin with normal saline (NS) as the control. The time for hair regrowth and full covering in depilated areas was observed, and the hair growth was evaluated histologically and by grossly measuring hair length and diameter. RESULTS: Compared with monolayer cultured cells, the three-dimensional (3D) culture of HuMSCs in gelatin sponge drastically increased VEGF, IGF-1, KGF, and HGF production. GS-HuMSCs-CM and HuMSCs injection both promoted hair regeneration in mice, while GS-HuMSCs-CM presented more enhanced effects in hair length, hair diameter, and growth rate. GS-HuMSCs-CM significantly promoted angiogenesis in injected skin areas, which might also contribute to faster hair regrowth. CONCLUSION: GS-HuMSCs-CM exerted significant effects on inducing hair growth and promoted skin angiogenesis in C57BL/6J mice.


Assuntos
Cabelo , Fator de Crescimento Insulin-Like I , Células-Tronco Mesenquimais , Cordão Umbilical , Animais , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Humanos , Meios de Cultivo Condicionados/farmacologia , Camundongos , Cordão Umbilical/citologia , Cabelo/crescimento & desenvolvimento , Cabelo/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Gelatina/química , Alicerces Teciduais/química , Camundongos Endogâmicos C57BL , Células Cultivadas , Fator 7 de Crescimento de Fibroblastos/metabolismo
8.
Mol Cancer Ther ; 23(1): 68-83, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-37775098

RESUMO

Brentuximab vedotin, a CD30-directed antibody-drug conjugate (ADC), is approved for clinical use in multiple CD30-expressing lymphomas. The cytotoxic payload component of brentuximab vedotin is monomethyl auristatin E (MMAE), a highly potent microtubule-disrupting agent. Preclinical results provided here demonstrate that treatment of cancer cells with brentuximab vedotin or free MMAE leads to a catastrophic disruption of the microtubule network eliciting a robust endoplasmic reticulum (ER) stress response that culminates in the induction of the classic hallmarks of immunogenic cell death (ICD). In accordance with the induction of ICD, brentuximab vedotin-killed lymphoma cells drove innate immune cell activation in vitro and in vivo. In the "gold-standard" test of ICD, vaccination of mice with brentuximab vedotin or free MMAE-killed tumor cells protected animals from tumor rechallenge; in addition, T cells transferred from previously vaccinated animals slowed tumor growth in immunodeficient mice. Immunity acquired from killed tumor cell vaccination was further amplified by the addition of PD-1 blockade. In a humanized model of CD30+ B-cell tumors, treatment with brentuximab vedotin drove the expansion and recruitment of autologous Epstein-Barr virus-reactive CD8+ T cells potentiating the activity of anti-PD-1 therapy. Together, these data support the ability of brentuximab vedotin and MMAE to drive ICD in tumor cells resulting in the activation of antigen-presenting cells and augmented T-cell immunity. These data provide a strong rationale for the clinical combination of brentuximab vedotin and other MMAE-based ADCs with checkpoint inhibitors.


Assuntos
Infecções por Vírus Epstein-Barr , Imunoconjugados , Animais , Camundongos , Brentuximab Vedotin , Morte Celular Imunogênica , Antígeno Ki-1 , Herpesvirus Humano 4/metabolismo , Imunoconjugados/uso terapêutico , Microtúbulos/metabolismo
9.
Immunology ; 140(1): 31-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23668241

RESUMO

T helper type 2 (Th2) cells are critical to host defence against helminth infection and the pathogenesis of allergic diseases. The differentiation of Th2 cells from naive CD4 T cells is controlled by intricate transcriptional mechanisms. At the precursor stage of naive CD4 T cells, transcriptional mechanisms maintain the potential and in the meantime prevent spontaneous differentiation to Th2 fate. In addition, intrachromosomal interactions important for co-ordinated expression of Th2 cytokines pre-exist in naive CD4 T cells. Upon T-cell receptor (TCR) engagement, naive CD4 T cells are induced by polarizing signals of the interleukin-4/Stat6 and Jagged/Notch pathways to up-regulate the expression of GATA-3. Once up-regulated, GATA-3 drives Th2 and suppresses Th1 differentiation in a cell autonomous fashion. In this stage of differentiation, the Th2 cytokine locus, as well as the interferon-γ locus, undergoes chromatin remodelling and epigenetic modifications that contribute to the somatic memory of Th2 cytokine gene expression pattern. Once differentiated, Th2 effector cells promptly produce Th2 cytokines upon TCR stimulation, which is regulated by concerted actions of GATA-3, TCR signalling, enhancers and the Th2 locus control region. This review provides a detailed account of the transcriptional regulatory events at these different stages of Th2 differentiation.


Assuntos
Células Th2/citologia , Células Th2/imunologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Montagem e Desmontagem da Cromatina , Mapeamento Cromossômico , Citocinas/genética , Epigênese Genética , Fator de Transcrição GATA3/metabolismo , Redes Reguladoras de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Interleucina-4/metabolismo , Proteína Jagged-1 , Ativação Linfocitária , Proteínas de Membrana/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores Notch/metabolismo , Fator de Transcrição STAT6/metabolismo , Proteínas Serrate-Jagged , Transdução de Sinais , Células Th1/citologia , Células Th1/imunologia , Células Th2/metabolismo
10.
Am J Pathol ; 181(5): 1504-12, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22959669

RESUMO

Upon insult, such as infection or tissue injury, the innate and adaptive immune systems initiate a series of responses to defend the body. Recent studies from immune cell-specific androgen receptor (AR) knockout mice demonstrated that androgen and its receptor (androgen/AR) play significant roles in both immune regulations. In the innate immunity, androgen/AR is required for generation and proper function of neutrophils; androgen/AR also regulates wound healing processes through macrophage recruitment and proinflammatory cytokine production. In adaptive immunity, androgen/AR exerts suppressive effects on development and activation of T and B cells. Removal of such suppression causes thymic enlargement and excessive export of immature B cells. Altogether, androgen/AR plays distinct roles in individual immune cells, and targeting androgen/AR may help in treatment and management of immune-related diseases.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Receptores Androgênicos/deficiência , Animais , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Modelos Imunológicos , Receptores Androgênicos/metabolismo , Transdução de Sinais
11.
Bioconjug Chem ; 24(7): 1256-63, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23808985

RESUMO

A highly cytotoxic DNA cross-linking pyrrolobenzodiazepine (PBD) dimer with a valine-alanine dipeptide linker was conjugated to the anti-CD70 h1F6 mAb either through endogenous interchain cysteines or, site-specifically, through engineered cysteines at position 239 of the heavy chains. The h1F6239C-PBD conjugation strategy proved to be superior to interchain cysteine conjugation, affording an antibody-drug conjugate (ADC) with high uniformity in drug-loading and low levels of aggregation. In vitro cytotoxicity experiments demonstrated that the h1F6239C-PBD was potent and immunologically specific on CD70-positive renal cell carcinoma (RCC) and non-Hodgkin lymphoma (NHL) cell lines. The conjugate was resistant to drug loss in plasma and in circulation, and had a pharmacokinetic profile closely matching that of the parental h1F6239C antibody capped with N-ethylmaleimide (NEM). Evaluation in CD70-positive RCC and NHL mouse xenograft models showed pronounced antitumor activities at single or weekly doses as low as 0.1 mg/kg of ADC. The ADC was tolerated at 2.5 mg/kg. These results demonstrate that PBDs can be effectively used for antibody-targeted therapy.


Assuntos
Benzodiazepinas/química , Ligante CD27/química , Imunoconjugados/farmacologia , Animais , Dimerização , Desenho de Fármacos , Feminino , Meia-Vida , Imunoconjugados/química , Camundongos , Camundongos Endogâmicos BALB C
12.
PeerJ ; 11: e14517, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36684668

RESUMO

The Sevatian of the late Norian is one of the key intervals in biotic turnover and in changes of paleoclimate and paleoenvironments. Conodont faunas recovered from two sections of upper Norian strata of the Dashuitang and Nanshuba formations near Baoshan City in western Yunnan province provide new insights into the diversity and biostratigraphy of the Sevatian conodonts within China as well as globally. A lower Mockina (M.) bidentata Zone and an upper Parvigondolella (P.) andrusovi Zone are identified in this area according to the first occurrences of M. bidentata and of P. andrusovi. Rich conodont fauna of M. zapfei is detailed and presents various intraspecific forms. A total of 19 forms of P1 elements are presented, which, when combined with the reported conodonts in the M. bidentata Zone, suggest that there was a peak in conodont diversity within the M. bidentata Zone. A biotic crisis in the uppermost M. bidentata Zone is recognized from the contrast between the diverse conodont fauna in the M. bidentata Zone and the rare conodonts in the P. andrusovi Zone. The conodont turnover during the middle Sevatian highlights the fact that the prolonged phases of the end-Triassic mass extinction probably began in the transition interval from M. bidentata Zone to P. andrusovi Zone.


Assuntos
Extinção Biológica , Fósseis , China
13.
Adv Healthc Mater ; 12(26): e2300970, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37379527

RESUMO

Mesenchymal stem cell (MSC) therapies have been brought forward as a promising treatment modality for cutaneous wound healing. However, current approaches for stem cell delivery have many drawbacks, such as lack of targetability and cell loss, leading to poor efficacy of stem cell therapy. To overcome these problems, in the present study, an in situ cell electrospinning system is developed as an attractive approach for stem cell delivery. MSCs have a high cell viability of over 90% even with a high applied voltage of 15 kV post-cell electrospinning process. In addition, cell electrospinning does not show any negative effect on the surface marker expression and differentiation capacity of MSCs. In vivo studies demonstrate that in situ cell electrospinning treatment can promote cutaneous wound healing through direct deposition of bioactive fish gelatin fibers and MSCs onto wound sites, leading to a synergic therapeutic effect. The approach enhances extracellular matrix remodeling by increasing collagen deposition, promotes angiogenesis by increasing the expression of vascular endothelial growth factor (VEGF) and forming small blood vessels, and dramatically reduces the expression of interleukin-6 (IL-6) during wound healing. The use of in situ cell electrospinning system potentially provides a rapid, no touch, personalized treatment for cutaneous wound healing.


Assuntos
Células-Tronco Mesenquimais , Fator A de Crescimento do Endotélio Vascular , Animais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Cicatrização , Colágeno/metabolismo , Pele
14.
J Cancer Res Clin Oncol ; 149(14): 12881-12896, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37466793

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) with high incidence and mortality is one of the most common malignant cancers worldwide. Increasing evidence has reported that N6-methyladenosine (m6A) modification has been considered as a major contribution to the occurrence and development of tumors. METHOD: In our study, we comprehensively analyzed the connection between m6A regulatory factors and cancer stem cells (CSCs) of HCC to establish a clinical tool for predicting its outcome. First, we concluded that the expression level of m6A regulatory factors was related with the stemness of hepatocellular carcinoma. Subsequently, we gained a ten hub regulatory factors that were associated with prognosis of hepatocellular carcinoma by overall survival (OS) analysis using ICGC and TCGA datasets, and these regulatory factors included YTHDF1, IGF2BP1, METTL3, IGF2BP3, HNRNPA2B1, IGF2BP2, RBM15B, HNRNPC, RBMX, and LRPPR. Next, we found that these ten hub m6A regulatory factors were highly expressed in CSCs, and CSCs related pathways were also enriched by the gene set variation analysis (GSVA). Then, correlation, consensus clustering and PCA analysis were performed to reveal potential therapeutic benefits of HCC. Moreover, univariate Cox regression (UNICOX), LASSON and multivariate Cox regression (MULTICOX) analyses were adopted to establish HCC prognosis prediction signature. RESULTS: Four regulatory factors RBM15B, LRPPRC, IGF2BP1, and IGF2BP3 were picked as valuable prognostic indicators. CONCLUSION: In summary, these ten hub regulatory factors would be useful therapeutic targets for HCC treatment, and RBM15B/LRPPRC/IGF2BP1/IGF2BP3 prognostic indicators can be used to guide therapy for HCC patients.

15.
Front Immunol ; 14: 1280986, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38022590

RESUMO

TIGIT is an immune checkpoint receptor expressed on activated and memory T cells, immunosuppressive T regulatory cells, and natural killer (NK) cells. TIGIT has emerged as an attractive target for antitumor therapies, due to its proposed immunosuppressive effects on lymphocyte function and T cell activation. We generated an anti-TIGIT monoclonal antibody (mAb) that binds with high affinity to human, non-human primate, and murine TIGIT and through multiple experimental methodologies demonstrated that checkpoint blockade alone is insufficient for antitumor activity. Generating anti-TIGIT mAbs with various Fc backbones we show that muting the Fc-Fcγ receptor (FcγR) interaction failed to drive antitumor activity, while mAbs with Fc functional backbones demonstrate substantial antitumor activity, mediated through activation of antigen-presenting cells (APCs), T cell priming, and NK-mediated depletion of suppressive Tregs and exhausted T cells. Further, nonfucosylation of the Fc backbone resulted in enhanced immune responses and antitumor activity relative to the intact IgG1 backbone. The improved activity correlated with the biased FcγR interaction profile of the nonfucosylated anti-TIGIT mAb, which supports that FcγRIIIa binding with decreased FcγRIIb binding favorably activates APCs and enhances tumor-specific CD8+ T cell responses. The anti-TIGIT mAbs with intact FcγR interacting backbones also demonstrated synergistic enhancement of other standard antitumor treatments, including anti-PD-1 treatment and a model monomethyl auristatin E antibody-drug conjugate. These findings highlight the importance of the anti-TIGIT mAb's Fc backbone to its antitumor activity and the extent to which this activity can be enhanced through nonfucosylation of the backbone.


Assuntos
Neoplasias , Receptores de IgG , Camundongos , Animais , Receptores Imunológicos/metabolismo , Anticorpos Monoclonais/farmacologia , Imunidade Inata
16.
Life Sci ; 293: 120336, 2022 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-35065166

RESUMO

AIMS: Individuals with nonalcoholic hepatosteatosis (NAFLD) have a worse atherogenic lipoprotein profile and are susceptible to cardiovascular diseases. The MEK-ERK signaling cascades are central regulators of the levels of LDL receptor (LDLR), a major determinant of circulating cholesterol. It is elusive how hepatic steatosis contributes to dyslipidemia, especially hypercholesterolemia. MAIN METHODS: The effects of BChE on signaling pathways were determined by immunoblotting in a BChE knockout hepatocyte cell line. DiI-LDL probe was used to explore the effect of BChE expression on LDL internalization. Co-immunoprecipitation and LC-MS were used to explore the interacting proteins with BChE. Finally, a hepatocyte-restricted BChE silencing mouse model was established by AAV8-Tbg-shRNA, and the hypercholesterolemia was induced by 65% kcal% high-fat, high-sucrose diet feeding. MAIN FINDINGS: Here we demonstrate that butyrylcholinesterase (BChE) governs the LDL receptor levels and LDL uptake capacity through the MEK-ERK signaling cascades to promote Ldlr transcription. BChE interacts and co-localizes with PRMT5, a protein methylation modifier controlling the ERK signaling. PRMT5 regulates LDLR-dependent LDL uptake and is a substrate of chaperone-mediated autophagy (CMA). BChE deficiency induces the PRTM5 degradation dependent on CMA activity, possibly through facilitating the HSC70 (Heat shock cognate 71 kDa) recognition of PRMT5. Remarkably, in vivo hepatocyte-restricted BChE silencing reduces plasma cholesterol levels substantially. In contrast, the BChE knockout mice are predisposed to hypercholesterolemia. SIGNIFICANCE: Taken together, these findings outline a regulatory role for the BChE-PRMT5-ERK-LDLR axis in hepatocyte cholesterol metabolism, and suggest that targeting liver BChE is an effective therapeutic strategy to treat hypercholesterolemia.


Assuntos
Butirilcolinesterase/deficiência , Hepatócitos/metabolismo , Hipercolesterolemia/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Receptores de LDL/metabolismo , Transcrição Gênica/fisiologia , Sequência de Aminoácidos , Animais , Butirilcolinesterase/genética , Tetracloreto de Carbono/toxicidade , Células Hep G2 , Humanos , Hipercolesterolemia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Proteína-Arginina N-Metiltransferases/genética , Receptores de LDL/genética
17.
J Immunol ; 183(6): 3583-90, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19710452

RESUMO

The mechanism for controlling Th cytokine expression in natural regulatory T (nTreg) cells is unclear. Here, it was found that under polarizing conditions Foxp3 did not affect Th1 cell, partially inhibited Th17 cell, but greatly inhibited Th2 cell differentiation of conventional CD4 T cells. Under the polarizing conditions, nTreg cells failed to differentiate into Th2 and Th17 cells, but differentiated into IFN-gamma-producing cells. Such Foxp3-transduced CD4 T cells and nTreg cells expressed T-bet, GATA-3, or retinoic acid-related orphan receptor (ROR)gammat, and retroviral GATA-3 and RORgammat could not induce Th2 and Th17 differentiation from nTreg cells. However, regardless of their cytokine profiles, the Foxp3-transduced CD4 T cells and nTreg cells remained immune suppressive. These results suggested that it is possible to convert pathogenic Th cells to Treg-like cells for therapeutic application. In conclusion, our studies show that Foxp3 is sufficient for immune suppression, whereas the inhibition of cytokine expression requires additional mechanisms.


Assuntos
Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/fisiologia , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Reguladores/citologia , Animais , Citocinas/antagonistas & inibidores , Imunidade , Interferon gama/biossíntese , Interleucina-17 , Camundongos , Camundongos Endogâmicos BALB C , Células Th1/citologia , Células Th2/citologia
18.
J Immunol ; 183(6): 3770-7, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19710474

RESUMO

Rheumatoid arthritis (RA) is characterized by inflammation and cellular proliferation in the synovial lining of joints that result in cartilage and bone destruction. Although the etiology of RA is unclear, activated lymphocytes and proinflammatory molecules, in particular TNF superfamily members, have been implicated in the disease pathology. A TNF superfamily member, CD70, is found on activated lymphocytes and shown to be important in memory and effector responses of lymphocytes. CD70 is expressed at high levels on chronically activated T cells in patients with autoimmune disorders, including RA. The involvement of CD70 in the progression of RA, however, remains unknown. In this study, we report effects of targeting CD70 on disease pathogenesis by using an anti-mouse CD70 Ab in a murine model of collagen-induced arthritis (CIA). In addition to blocking CD70 binding to its receptor CD27, the anti-CD70 Ab used also engages Fc-dependent effector functions including Ab-dependent cellular cytotoxicity, phagocytosis, and complement fixation. Treatment of mice with anti-CD70 Ab both before the onset or after the established disease in CIA model resulted in marked improvements in disease severity and significant reduction in the production of autoantibodies. Histopathological analyses of the joints of mice revealed a substantial reduction of inflammation, and bone and cartilage destruction in response to the anti-CD70 Ab treatment. These results uncover a novel role for CD27-CD70 interactions in the regulation of in vivo inflammatory response leading to arthritis, and provide a molecular basis to support the rationale for anti-CD70 therapy for autoimmune and inflammatory diseases.


Assuntos
Anticorpos Monoclonais/farmacologia , Artrite Experimental/tratamento farmacológico , Ligante CD27/antagonistas & inibidores , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/antagonistas & inibidores , Animais , Artrite Experimental/patologia , Autoanticorpos/análise , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Cartilagem/efeitos dos fármacos , Cartilagem/patologia , Progressão da Doença , Inflamação/prevenção & controle , Artropatias , Masculino , Camundongos
19.
Artigo em Inglês | MEDLINE | ID: mdl-33662568

RESUMO

Cholinesterases act as bio scavengers to clear organophosphorus (OP) compounds and prodrugs. The butyrylcholinesterase (BChE) gene has been found in several types of teleost fish but this gene has yet to be identified in cyprinid fish. Indeed, BChE homologs have not been found in the zebrafish (Danio rerio) genomic database. Here, we demonstrate that BChE activity is present in zebrafish, in line with other groups' findings. Using in-gel native-PAGE enzymatic activity staining and LC-MS/MS technique, an atypical BChE-like protein was identified in zebrafish. The si:ch211-93f2.1 gene was cloned, and His-tagged recombinant protein was expressed using the Pichia yeast system. The purified protein (molecular weight ~ 180 kDa) showed BChE activity, and degraded acetylcholinesterase (ACh) at a higher rate than BCh. However, phylogram analysis shows that this novel cholinesterase shared an evolutionary origin with carboxylic esterase rather than BChE. The zebrafish BChE-like protein shares structural characteristics with cholinesterase and carboxylesterase. The 2-arachidonoylglycerol (2-AG), nicosulfuron, and triacetin exhibited a higher binding affinity to the zebrafish BChE-like protein than BCh and ACh. With the identification of BChE-like protein in zebrafish, this study could shed light on the origin of BChE and may contribute towards the development of a BChE knockout zebrafish model for sensitive drug or toxin screening.


Assuntos
Hidrolases de Éster Carboxílico , Clonagem Molecular , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Hidrolases de Éster Carboxílico/biossíntese , Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/genética , Domínios Proteicos , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
20.
Front Cell Dev Biol ; 9: 711149, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34977001

RESUMO

Integrin ß1 (ITGB1), which acts as an extracellular matrix (ECM) receptor, has gained increasing attention as a therapeutic target for the treatment of hepatocellular carcinoma (HCC). However, the underpinning mechanism of how ITGB1 drives HCC progression remains elusive. In this study, we first found that ITGB1 expression was significantly higher in HCC tissues than in normal controls by bioinformatics analysis. Furthermore, bioinformatics analysis revealed that paxillin (PXN) and 14-3-3 protein zeta (YWHAZ) are the molecules participating in ITGB1-regulated HCC tumor cell cycle progression. Indeed, immunohistochemistry (IHC) revealed that ITGB1, paxillin, and YWHAZ were strongly upregulated in paired HCC tissue compared with adjacent normal tissues. Notably, the inhibition of ITGB1 expression by small interfering RNA (siRNA) resulted in the downregulated expression of PXN and YWHAZ in primary HCC cells, as assessed by western blot and immunostaining. In addition, ITGB1 knockdown markedly impaired the aggressive behavior of HCC tumor cells and delayed cell cycle progression as determined by cell migration assay, drug-resistance analysis, colony formation assay, quantitative real-time polymerase chain reaction (qRT-PCR), and cell cycle analysis as well as cell viability measurements. More importantly, we proved that xenograft ITGB1high tumors grew more rapidly than ITGB1low tumors. Altogether, our study showed that the ITGB1/PXN/YWHAZ/protein kinase B (AKT) axis enhances HCC progression by accelerating the cell cycle process, which offers a promising approach to halt HCC tumor growth.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa