Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Bioconjug Chem ; 34(10): 1894-1901, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37787935

RESUMO

As a pleiotropic cytokine, interleukin-2 (IL-2) can effectively regulate lymphocyte proliferation, survival, and active antitumor immune responses in tumor microenvironments. Although the ability of IL-2 to boost immune responses was reported in cancer patients, its short circulating half-life and high toxicity hinder its broad and continual clinical application. Herein, we developed a novel tumor target agent by fusing pH low insertion peptides (pHLIP) with IL-2, forming the fusion protein pHLIP-IL2. Based on the low pH insertion property of pHLIP, the pHLIP-IL2 fusion protein could be selectively delivered to the acidic tumor microenvironments and then promote the proliferation of killer immune cells to elicit tumor regression. We found that pHLIP-IL2 fusion proteins can be significantly enriched in tumor tissues and can effectively reduce tumor size in diverse tumor models, including breast cancer and melanoma, without apparent adverse effects. These data suggest that the pHLIP-IL2 fusion protein may be a promising solution for the continual and extensive application of IL-2, and pHLIP-IL2 is a potential and valuable therapeutic drug for cancer patients with antitumor immunotherapy.


Assuntos
Interleucina-2 , Melanoma , Humanos , Linhagem Celular Tumoral , Concentração de Íons de Hidrogênio , Imunoterapia , Interleucina-2/administração & dosagem , Melanoma/tratamento farmacológico , Microambiente Tumoral , Sistemas de Liberação de Medicamentos
2.
Nano Lett ; 21(6): 2588-2595, 2021 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-33650872

RESUMO

Compared with traditional chemotherapeutics, vascular disruption agents (VDAs) have the advantages of rapidly blocking the supply of nutrients and starving tumors to death. Although the VDAs are effective under certain scenarios, this treatment triggers angiogenesis in the later stage of therapy that frequently leads to tumor recurrence and treatment failure. Additionally, the nonspecific tumor targeting and considerable side effects also impede the clinical applications of VDAs. Here we develop a customized strategy that combines a VDA with an anti-angiogenic drug (AAD) using mesoporous silica nanoparticles (MSNs) coated with platelet membrane for the self-assembled tumor targeting accumulation. The tailor-made nanoparticles accumulate in tumor tissues through the targeted adhesion of platelet membrane surface to damaged vessel sites, resulting in significant vascular disruption and efficient anti-angiogenesis in animal models. This study demonstrates the promising potential of combining VDA and AAD in a single nanoplatform for tumor eradication.


Assuntos
Nanopartículas , Neoplasias , Inibidores da Angiogênese/uso terapêutico , Animais , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Dióxido de Silício/uso terapêutico
3.
Wei Sheng Yan Jiu ; 46(2): 232-236, 2017 Mar.
Artigo em Zh | MEDLINE | ID: mdl-29903099

RESUMO

OBJECTIVE: To analyze the relationship between serum ferritin(SF) level and high sensitive C reactive protein( hs-CRP) in men and the risk of gout. METHODS: We chosed 600 male patients diagnosed with gout as gout group, 600 male patients with hyperuricemia were diagnosed as hyperuricemia group, and randomly selected 600 cases of the same period of male health examination as the control group. The detection information of physical examination and related indicators of three groups were collected, such as height, weight, serum ferritin, high sensitive C reactive protein, uric acid( UA), fasting blood glucose( FPG), triglyceride(TG), total cholesterol( TC) and so on. RESULTS: Serum ferritin( SF) higher than that of hyperuricemia group 114. 45 µg/L( P<0. 05)and the control group 76. 02 µg/L( P<0. 05), while the level of hs-CRP in gout patients up to 0. 3 mg/dL, was significantly higher than that 0. 13 mg/dL in hyperuricemia group and 0. 09 mg/dL in control group( all P<0. 05). After adjusting for BMI, TG, TC, FPG and UA five confounding factors, SF was positively correlated with hs-CRP levels in the hyperuricemia group and the gout group, while there was no association between SF and hs-CRP levels in the control group. The multivariate logistic regression analysis showed that SF( ≥ 69. 01 µg/L) had significantly increased risk of HUA, after adjusting for BMI, TG, TC, FPG and UA five confounding factors, the high level of SF( ≥155. 78µg/L) had significantly increased risk of gout, with OR of 2. 678( 95% CI 1. 484-4. 833), and higher levels of hs-CRP( > 0. 9 mg/dL) was also a risk factor of gout, with OR of 3. 104( 95% CI 1. 727-5. 580). However, SF and hs-CRP were not risk factors of hyperuricemia. CONCLUSION: Serum ferritin level and high sensitive C reactive protein levels are significantly elevated in patients with gout. It is revealed that hs-CRP, SF may be involved in the pathogenesis of gout patients.


Assuntos
Proteína C-Reativa/análise , Ferritinas/sangue , Gota/diagnóstico , Proteína C-Reativa/metabolismo , Gota/sangue , Humanos , Hiperuricemia/sangue , Masculino , Ácido Úrico
4.
IUBMB Life ; 66(3): 212-219, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24659529

RESUMO

Cellular glutathione peroxidase (GPx1; EC1.11.1.9) is a major intracellular antioxidant selenoenzyme in mammals. However, the complicated expression mechanism of selenocysteine (Sec)-containing protein increases the difficulty of expressing human GPx1 (hGPx1) in Escherichia coli (E. coli). In this study, hGPx1 gene was cloned from a cDNA library of the human hepatoma cell line HepG2. The codon UGA encoding Sec49 of hGPx1 was first mutated to UGC encoding cysteine (Cys) and then biosynthetically converted to Sec during expression in an E. coli BL21(DE3)cys auxotrophic system. Seleno-GPx1Sec displayed a low GPx activity of 522 U/µmol. To improve the activity, the other five Cys residues (C2, C78, C115, C156, C202) were mutated to serine (Ser) in one hGPx1 molecule. The mutant seleno-hGPx1Ser showed a high activity of 5278 U/µmol, which was more than 10-fold enhanced as compared with seleno-GPx1Sec . The activity was the highest among all of those seleno-proteins obtained by this method so far. Kinetic analysis of seleno-hGPx1Ser showed a typical ping-pong mechanism, which was similar to those of natural GPxs. This research will be of value in overcoming the problem of limited sources of natural GPx and substantially promotes the research of the characterization of GPx. © 2014 IUBMB Life, 66(3):212-219, 2014.

5.
Transl Res ; 273: 1-15, 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38945255

RESUMO

Renal aging and the subsequent rise in kidney-related diseases are attributed to senescence in renal tubular epithelial cells (RTECs). Our study revealed that the abnormal expression of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3), a reader of RNA N6-methyladenosine, is critically involved in cisplatin-induced renal tubular senescence. In cisplatin-induced senescence of RTECs, the promoter activity and transcription of IGF2BP3 is markedly suppressed. It was due to the down regulation of MYC proto-oncogene (MYC), which regulates IGF2BP3 transcription by binding to the putative site at 1852-1863 of the IGF2BP3 promoter. Overexpression of IGF2BP3 ameliorated cisplatin-induced renal tubular senescence in vitro. Mechanistic studies revealed that IGF2BP3 inhibits cellular senescence in RTECs by enhancing cyclin-dependent kinase 6 (CDK6) mRNA stability and increasing its expression. The inhibition effect of IGF2BP3 on tubular senescence is partially reversed by the knockdown of CDK6. Further, IGF2BP3 recruits nuclear cap binding protein subunit 1 (NCBP1) and inhibits CDK6 mRNA decay, by recognizing m6A modification. Specifically, IGF2BP3 recognizes m6A motif "GGACU" at nucleotides 110-114 in the 5' untranslated region (UTR) field of CDK6 mRNA. The involvement of IGF2BP3/CDK6 in alleviating tubular senescence was confirmed in a cisplatin-induced acute kidney injury (AKI)-to-chronic kidney disease (CKD) model. Clinical data also suggests an age-related decrease in IGF2BP3 and CDK6 levels in renal tissue or serum samples from patients. These findings suggest that IGF2BP3/CDK6 may be a promising target in cisplatin-induced tubular senescence and renal failure.

6.
J Mol Recognit ; 26(1): 38-45, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23280616

RESUMO

Accumulating evidence shows that glutathione peroxidase (GPX, EC.1.11.1.9), one of the most important antioxidant selenoenzymes, plays an essential role in protecting cells and tissues against oxidative damage by catalyzing the reduction of hydrogen peroxide by glutathione. Unfortunately, because of the limited availability and poor stability of GPX, it has not been used clinically to protect against oxidative stress. To overcome these problems, it is necessary to generate mimics of GPX. In this study, we have used directed mutagenesis and the inclusion of a selenocysteine (Sec) insertion sequence to engineer the expression in eukaryotic cells of human glutathione transferase zeta1-1 (hGSTZ1-1) with Sec in the active site (seleno-hGSTZ1-1). This modification converted hGSTZ1-1 into an active GPX and is the first time this has been achieved in eukaryotic cells. The GPX activity of seleno-hGSTZ1-1 is higher than that of GPX from bovine liver, indicating Sec at the active site plays an important role in the determination of catalytic specificity and performance. Kinetic studies revealed that the ping-pong catalytic mechanism of Se-hGSTZ1-1 is similar to that of the natural GPX.


Assuntos
Glutationa Transferase/química , Glutationa Transferase/genética , Selenocisteína/química , Selenocisteína/genética , Catálise , Domínio Catalítico , Glutationa Peroxidase/química , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Glutationa Transferase/metabolismo , Células HEK293 , Humanos , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Concentração de Íons de Hidrogênio , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Selênio/química , Selênio/metabolismo , Selenocisteína/metabolismo , Temperatura
7.
IUBMB Life ; 65(2): 163-70, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23299908

RESUMO

Human glutathione transferase zeta 1c-1c (hGSTZ1c-1c) is one of the glutathione transferase isoenzymes and considered to be a protein scaffold to imitate glutathione peroxidase (GPX) owing to the natural binding site of glutathione (GSH). In this report, several residues near GSH were mutated to selenocysteine (Sec) or cysteine (Cys) residues and the impacts of the substitutions on different activities were discussed. Mutations of Ser-14 or/and Ser-15 to Cys or Sec residues resulted in dramatic loss of catalytic activity of hGSTZ1c-1c with chlorofluoroacetic acid as substrate, which indicated the importance of the hydroxyl groups in Ser-14 and Ser-15. And subsequent study by molecular modeling suggested that Ser-15 was probably involved in catalysis, while Ser-14 may play a crucial role in binding and orientation of GSH and possibly had a synergistic effect with Ser-15 in catalysis. On the contrary, the result of converting Cys-16 to Ser indicated its trivial role in catalysis. The investigations of the selenocysteine-containing hGSTZ1c-1c (seleno-hGSTZ1c-1c) and the mutant S17C implied that the substitutions of multi-Sec for Cys residues at position 16, 137, and 205 could lead to subtle change in the structure of the protein molecule and concomitant change in catalytic activity as a direct result. This finding provides overwhelming evidence that the protein scaffold containing fewer cysteines should be chosen for imitating GPX using cysteine auxotrophic strain system to avoid unexpected structural changes.


Assuntos
Substituição de Aminoácidos , Glutationa Transferase/química , Simulação de Acoplamento Molecular , Selenocisteína/química , Acetatos/química , Motivos de Aminoácidos , Domínio Catalítico , Escherichia coli , Glutationa/química , Glutationa Transferase/biossíntese , Glutationa Transferase/genética , Humanos , Mutagênese Sítio-Dirigida , Ligação Proteica
8.
IUBMB Life ; 65(11): 951-6, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24170573

RESUMO

Phospholipid hydroperoxide glutathione peroxidase (PHGPx or GPx4; EC1.11.1.12) is a selenoperoxidase that can directly reduce phospholipid and cholesterol hydroperoxides. The mature cytoplasmic GPx4 is a monomeric protein with molecular weight of 19.5 kDa. In this study, human GPx4 (hGPx4) gene was amplified from the complementary DNA library of human hepatoma cell line. Eukaryotic expression plasmid pSelExpress1-leader-GPx4 was constructed and transfected into the eukaryotic cells HEK293T. Expression of hGPx4 was detected by Western blotting, and the target protein was purified by immobilized metal affinity chromatography. The results of the activity and kinetics of the purified protein show that the obtained protein follows a "ping-pong" mechanism, which is similar to that of native cytosolic glutathione peroxidase (GPx1; EC1.11.1.9). This is the first time that hGPx4 could be expressed and purified from HEK293T cells, and this work will provide an important resource of hGPx4 for its functional study in vitro and in vivo.


Assuntos
Glutationa Peroxidase/biossíntese , Glutationa Peroxidase/metabolismo , Linhagem Celular Tumoral , Clonagem Molecular , Células HEK293 , Humanos , Cinética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Proteínas Recombinantes/biossíntese , Selenoproteínas/biossíntese , Selenoproteínas/metabolismo , Transfecção
9.
Cancer Lett ; 560: 216138, 2023 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-36934836

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal malignancy with insidious onset and early distal metastasis. Metabolic reprogramming, the autonomous changes in cellular bioenergetics driven by aberrant genetic events and crosstalk between cancer and non-cancer cells in the desmoplastic microenvironment, is pivotal for the rapid progression of PDAC. As an attractive therapeutic target, nucleoside metabolism is regulated by various anti-metabolic drugs for the clinical treatment of PDAC. Despite various challenges, such as poor drug delivery efficiency and off-target side effects, metabolic modification and intervention are emerging as promising strategies for PDAC therapy, enabled by the rapid development of nanotechnology-based drug delivery strategies. In this review, we discuss the metabolic characteristics of PDAC and highlight how the development of nanomedicine has boosted the development of new therapeutics for PDAC by modulating critical targets in metabolic reprogramming.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/patologia , Metabolismo Energético , Nanomedicina , Microambiente Tumoral , Neoplasias Pancreáticas
10.
Antioxidants (Basel) ; 12(9)2023 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-37760069

RESUMO

Important antioxidant enzymes, glutathione peroxidase (GPx) and superoxide dismutase (SOD), are involved in maintaining redox balance. They can protect each other and result in more efficiently removing excessive reactive oxygen species (ROS), protecting cells against injury, and maintaining the normal metabolism of ROS. In this study, human cytosolic GPx (hGPx1) and human phospholipid hydroperoxide GPx (hGPx4) genes were integrated into the same open reading frame with human extracellular SOD active site (SOD3-72P) genes, respectively, and several novel fusion proteins were obtained by using the UTuT6 expression system for the first time. Among them, Se-hGPx1UAG-L4-SOD3-72P is the bifunctional fusion protein with the highest GPx activity and the best anti-hydrogen peroxide inactivation ability thus far. The Se-hGPx4UAG-L3-SOD3-72P fusion protein exhibits the strongest alkali and high temperature resistance and a greater protective effect against lipoprotein peroxidation damage. Se-hGPx1UAG-L4-SOD3-72P and Se-hGPx4UAG-L3-SOD3-72P fusion proteins both have good synergistic and antioxidant abilities in H2O2-induced RBCs and liver damage models. We believe that this research will help with the development of novel bifunctional fusion proteins and the investigation of the synergistic and catalytic mechanisms of GPx and SOD, which are important in creating novel protein therapeutics.

11.
Cancer Res ; 83(17): 2924-2937, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37326467

RESUMO

Nanoparticles (NP) spanning diverse materials and properties have the potential to encapsulate and to protect a wide range of therapeutic cargos to increase bioavailability, to prevent undesired degradation, and to mitigate toxicity. Fulvestrant, a selective estrogen receptor degrader, is commonly used for treating patients with estrogen receptor (ER)-positive breast cancer, but its broad and continual application is limited by poor solubility, invasive muscle administration, and drug resistance. Here, we developed an active targeting motif-modified, intravenously injectable, hydrophilic NP that encapsulates fulvestrant to facilitate its delivery via the bloodstream to tumors, improving bioavailability and systemic tolerability. In addition, the NP was coloaded with abemaciclib, an inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6), to prevent the development of drug resistance associated with long-term fulvestrant treatment. Targeting peptide modifications on the NP surface assisted in the site-specific release of the drugs to ensure specific toxicity in the tumor tissues and to spare normal tissue. The NP formulation (PPFA-cRGD) exhibited efficient tumor cell killing in both in vitro organoid models and in vivo orthotopic ER-positive breast cancer models without apparent adverse effects, as verified in mouse and Bama miniature pig models. This NP-based therapeutic provides an opportunity for continual and extensive clinical application of fulvestrant, thus indicating its promise as a treatment option for patients with ER-positive breast cancer. SIGNIFICANCE: A smart nanomedicine encapsulating fulvestrant to improve its half-life, bioavailability, and tumor-targeting and coloaded with CDK4/6 inhibitor abemaciclib to block resistance is a safe and effective therapy for ER-positive breast cancer.


Assuntos
Neoplasias , Receptores de Estrogênio , Animais , Camundongos , Suínos , Fulvestranto/farmacologia , Fulvestranto/uso terapêutico , Receptores de Estrogênio/metabolismo , Aminopiridinas/farmacologia , Neoplasias/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Linhagem Celular Tumoral
12.
Protein Expr Purif ; 84(1): 59-63, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22561244

RESUMO

Glutathione peroxidase (GPX) is a crucial antioxidant selenocysteine (Sec) containing enzyme which plays a significant role in protecting cells against oxidative damage by catalyzing the reduction of hydroperoxides with glutathione (GSH). Several methods have been used to generate GPX mimics, however, only a few of these methods involved genetic engineering and none of them have achieved specific site-directed incorporation of Sec without other modifications, which has hampered further structure-function studies. Here, we report for the first time the conversion of human glutathione transferase Zeta (hGSTZ1-1) into seleno-hGSTZ1-1 by means of genetic engineering in eukaryotes. Fluorescence microscopy images of the expression of Seleno-GST-green fluorescent protein chimaera indicated that we successfully achieved the read-through of the UGA codon to specifically incorporate Sec. Therefore, we achieved the conversion of human glutathione transferase Zeta (hGSTZ1-1) into a seleno-GST (seleno-hGSTZ1-1) by means of genetic engineering in eukaryotes. These results show that recombinant selenoproteins with incorporation of specific selenocysteine residues may be heterologously produced in eukaryotes by using a Sec insertion sequence in the 3' untranslated region (3'-UTR) of the mRNA, and the recombinant selenoproteins is single catalytically active residue and well-characterized structure. In this case a novel GPX activity of 2050±225 U/µmol was introduced into hGSTZ1-1 by substitution of serine 15 by Sec 15. This result will lay a foundation for preparing much smaller GPX mimics with higher activity.


Assuntos
Glutationa Transferase/biossíntese , Selenocisteína/metabolismo , Western Blotting , Clonagem Molecular , Códon , Vetores Genéticos , Glutationa Transferase/química , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Células HEK293 , Humanos , NADP/química , NADP/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Selenocisteína/química , Selenocisteína/genética
13.
Antioxidants (Basel) ; 11(6)2022 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-35739980

RESUMO

Plasma glutathione peroxidase (GPx3) belongs to the GPx superfamily, and it is the only known secreted selenocysteine (Sec)-containing GPx in humans. It exists as a glycosylated homotetramer and catalyzes the reduction of hydrogen peroxide and lipid peroxides, depending on the Sec in its active center. In this study, a previously reported chimeric tRNAUTuT6 was used for the incorporation of Sec at the UAG amber codon, and the mature form of human GPx3 (hGPx3) without the signal peptide was expressed in amber-less E. coli C321.ΔA.exp. Reactive Sec-hGPx3, able to reduce H2O2 and tert-butyl hydroperoxide (t-BuOOH), was produced with high purity and yield. Study of the quaternary structure suggested that the recombinant Sec-hGPx3 contained an intra-molecular disulfide bridge but failed to form tetramer. Mutational and structural analysis of the mutants with three Cys residues, individually or jointly replaced with Ser, indicated that the formation of intra-molecular disulfide bridges involved structure conformational changes. The secondary structure containing Cys77 and Cys132 was flexible and could form a disulfide bond, or form a sulfhydryl-selenyl bond with Sec49 in relative mutants. Mutation of Cys8 and Cys132 to Sec8 and Sec132 could fix the oligomerization loop through the formation of diselenide bond, which, in turn, facilitated tetramer formation and noticeably improved the GPx activity. This research provides an important foundation for the further catalysis and functional study of hGPx3.

14.
Aging (Albany NY) ; 14(1): 443-461, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-35020600

RESUMO

Human glutathione peroxidase1 (hGPx1) is a good antioxidant and potential drug, but the limited availability and poor stability of hGPx1 have affected its development and application. To solve this problem, we prepared a hGPx1 mutant (GPx1M) with high activity in an Escherichia coli BL21(DE3)cys auxotrophic strain using a single protein production (SPP) system. In this study, the GPx1M was conjugated with methoxypolyethylene glycol-succinimidyl succinate (SS-mPEG, Mw = 5 kDa) chains to enhance its stability. SS-mPEG-GPx1M and GPx1M exhibited similar enzymatic activity and stability toward pH and temperature change, and in a few cases, SS-mPEG-GPx1M was discovered to widen the range of pH stability and increase the temperature stability. Lys 38 was confirmed as PEGylated site by liquid-mass spectrometry. H9c2 cardiomyoblast cells and Sprague-Dawley (SD) rats were used to evaluate the effects of GPx1M and SS-mPEG-GPx1M on preventing or alleviating adriamycin (ADR)-mediated cardiotoxicity, respectively. The results indicated that GPx1M and SS-mPEG-GPx1M had good antioxidant effects in vitro and in vivo, and the effect of SS-mPEG-GPx1M is more prominent than GPx1M in vivo. Thus, PEGylation might be a promising method for the application of GPx1M as an important antioxidant and potential drug.


Assuntos
Antioxidantes/farmacologia , Glutationa Peroxidase/metabolismo , Animais , Antioxidantes/química , Antioxidantes/metabolismo , Linhagem Celular , Desenho de Fármacos , Escherichia coli , Glutationa Peroxidase/química , Humanos , Concentração de Íons de Hidrogênio , Modelos Moleculares , Mutação , Miócitos Cardíacos , Polietilenoglicóis/química , Conformação Proteica , Estabilidade Proteica , Ratos , Ratos Sprague-Dawley , Succinimidas/química , Temperatura , Glutationa Peroxidase GPX1
15.
Front Cell Dev Biol ; 10: 862675, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35592244

RESUMO

Renal fibrosis is a common feature of various chronic kidney diseases (CKD). However, its underlying mechanism has not been totally clarified. C-X-C motif chemokine receptor (CXCR) family plays a role in renal fibrosis, however, detailed mechanisms have not been elucidated. Here, we report that CXCR2 has a potential role in tubular cell senescence and renal fibrosis, and is associated with ß-catenin-activated mitochondrial dysfunction. CXCR2 is one of most increased members among CXCR family in unilateral ureteral obstruction (UUO) mice. CXCR2 was expressed primarily in tubules and co-localized with p16INK4A, a cellular senescence marker, and ß-catenin. Administration of SB225002, a selective CXCR2 antagonist, significantly inhibited the activation of ß-catenin signaling, restored mitochondrial function, protected against tubular cell senescence and renal fibrosis in unilateral ureteral obstruction (UUO) mice. In unilateral ischemia-reperfusion injury (UIRI) mice, treatment with interlukin-8 (IL-8), the ligand of CXCR2, further aggravated ß-catenin activation, mitochondrial dysfunction, tubular cell senescence and renal fibrosis, whereas knockdown of p16INK4A inhibited IL-8-induced these effects. In vitro, SB225002 inhibited mitochondrial dysfunction and tubular cell senescence. Furthermore, ICG-001, a ß-catenin signaling blocker, significantly retarded CXCR2-induced cellular senescence and fibrotic changes. These results suggest that CXCR2 promotes tubular cell senescence and renal fibrosis through inducing ß-catenin-activated mitochondrial dysfunction.

16.
Adv Sci (Weinh) ; 9(20): e2200477, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35524631

RESUMO

New strategies to decrease risk of relapse after surgery are needed for improving 5-year survival rate of hepatocellular carcinoma (HCC). To address this need, a wound-targeted nanodrug is developed, that contains an immune checkpoint inhibitor (anti-PD-L1)and an angiogenesis inhibitor (sorafenib)). These nanoparticles consist of highly biocompatible mesoporous silica (MSNP) that is surface-coated with platelet membrane (PM) to achieve surgical site targeting in a self-amplified accumulation manner. Sorafenib is introduced into the MSNP pores while covalently attaching anti-PD-L1 antibody on the PM surface. The resulting nano-formulation, abbreviated as a-PM-S-MSNP, can effectively target the surgical margin when intraperitoneally (IP) administered into an immune competent murine orthotopic HCC model. Multiple administrations of a-PM-S-MSNP generate potent anti-HCC effect and significantly prolong overall mice survival. Immunophenotyping and immunochemistry staining reveal the signatures of favorable anti-HCC immunity and anti-angiogenesis effect at tumor sites. More importantly, microscopic inspection of a-PM-S-MSNP treated mice shows that 2 out 6 are histologically tumor-free, which is in sharp contrast to the control mice where tumor foci can be easily identified. The data suggest that a-PM-S-MSNP can efficiently inhibit post-surgical HCC relapse without obvious side effects and holds considerable promise for clinical translation as a novel nanodrug.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/cirurgia , Linhagem Celular Tumoral , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/cirurgia , Camundongos , Nanopartículas/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico
17.
Int J Biol Macromol ; 170: 503-513, 2021 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-33383079

RESUMO

Glutathione peroxidase 1 (GPx1) is an important antioxidant selenium enzyme and has a good prospect for drug development. However, the expression of GPx1 requires a complex expression mechanism, which makes the drug development of recombinant GPx1 (rGPx1) difficult. In the previous study, we expressed highly active rhGPx1 in amber-less Escherichia coli by using a novel chimeric tRNAUTuT6. However, the antioxidant effect of rhGPx1 at the cellular and animal levels has not been verified. In this study, we established isoproterenol (ISO)-induced oxidative stress injury models to study the antioxidant effect of rhGPx1 at the cellular and animal levels. Meanwhile, in order to more accurately reflect the antioxidant effect of rGPx1 in mice, we used the same method to express recombinant mouse GPx1 (rmGPx1) as a control for rhGPx1. The results of a study showed that rhGPx1 has a good antioxidant effect at the cellular and animal levels. However, due to species differences, rhGPx1 had immunogenicity in mice and antibodies of rhGPx1 could inhibit its antioxidant activity, so the antioxidant effect of rhGPx1 was not as good as rmGPx1 in mice. Nevertheless, this study provides a reliable theoretical basis for the development of rhGPx1 as an antioxidant drug.


Assuntos
Antioxidantes/metabolismo , Antioxidantes/farmacologia , Glutationa Peroxidase/metabolismo , Glutationa Peroxidase/farmacologia , Animais , Linhagem Celular , Escherichia coli/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Estresse Oxidativo/efeitos dos fármacos , Ratos , Selênio/metabolismo , Selênio/farmacologia , Glutationa Peroxidase GPX1
18.
Oxid Med Cell Longev ; 2021: 6632813, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34539971

RESUMO

Ischemic heart disease (IHD) is a cardiovascular disease with high fatality rate, and its pathogenesis is closely related to oxidative stress. Reactive oxygen species (ROS) in oxidative stress can lead to myocardial ischemia (MI) injury in many ways. Therefore, the application of antioxidants may be an effective way to prevent IHD. In recent years, glutathione peroxidase 4 (GPx4) has received increasing attention due to its antioxidant effect. In a previous study, we used the new chimeric tRNAUTuT6 to express highly active recombinant human GPx4 (rhGPx4) in amber-less Escherichia coli. In this study, we established an isoproterenol- (ISO-) induced MI injury model in rats and an in vitro model to research the protective effect and mechanism of rhGPx4 on MI injury. The results showed that rhGPx4 could reduce the area of myocardial infarction and ameliorate the pathological injury of heart tissue, significantly reduce ISO-induced abnormalities on electrocardiogram (ECG) and cardiac serum biomarkers, protect mitochondrial function, and attenuate cardiac oxidative stress injury. In an in vitro model, the results also confirmed that rhGPx4 could inhibit ISO-induced oxidative stress injury and cardiomyocyte apoptosis. The mechanism of action of rhGPx4 involves not only the inhibition of lipid peroxidation by eliminating ROS but also keeping a normal level of endogenous antioxidant enzymes by eliminating ROS, thereby preventing oxidative stress injury in cardiomyocytes. Additionally, rhGPx4 could inhibit cardiomyocyte apoptosis through a mitochondria-dependent pathway. In short, rhGPx4, a recombinant antioxidant enzyme, can play an important role in the prevention of IHD and may have great potential for application.


Assuntos
Infarto do Miocárdio/tratamento farmacológico , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/uso terapêutico , Substâncias Protetoras/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/sangue , Linhagem Celular , Modelos Animais de Doenças , Eletrocardiografia , Humanos , Isoproterenol/toxicidade , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Infarto do Miocárdio/induzido quimicamente , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/farmacologia , Substâncias Protetoras/farmacologia , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Troponina T/sangue
19.
Int J Pharm ; 609: 121152, 2021 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-34626796

RESUMO

Human glutathione peroxidase (GPx), as an important kind of antioxidant enzyme, is often used for the removal of reactive oxygen species. Unfortunately, the application has been hindered by its limited source and poor stability. To solve these problems, human glutathione peroxidase mutant (GPxM) with high activity and yield was obtained using Escherichia coli BL21(DE3) cys auxotrophic strain and the single-protein production system in our previous work. However, the antioxidant effect of this novel recombinant protein drug in animals has not been demonstrated, and its immunogenicity and short biological half-life as a biological macromolecule may have seriously hindered its clinical application. Therefore, it is important to find an effective strategy to address the above issues. In this work, PEGylated GPxM was prepared by conjugating the corresponding mutant with monomethoxy polyethylene glycol succinimidyl succinate (SS-mPEG). We researched the structure, stability, pharmacokinetic properties, antioxidant effect in vivo and protective mechanism against adriamycin (ADR)-mediated cardiotoxicity of modified products, and compared with the above properties of GPxM. The results revealed that GPxM had an excellent antioxidant effect in vivo, and PEGylation can enhance the stability, half-life and antioxidant effect of GPxM while reducing immunogenicity. In addition, the above improvement of PEGylated GPx1M was stronger than that of monoPEGylated GPx4M. Hence, PEGylation might be an effective method to broaden the applications of GPxM as the important antioxidant drug, especially the PEGylated GPx1M with high antioxidant effect.


Assuntos
Antioxidantes , Polietilenoglicóis , Animais , Glutationa Peroxidase/genética , Meia-Vida , Humanos , Proteínas Recombinantes/genética , Ácido Succínico
20.
Acta Pharm Sin B ; 11(7): 2059-2069, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34386338

RESUMO

Selective occlusion of tumor vasculature has proven to be an effective strategy for cancer therapy. Among vascular coagulation agents, the extracellular domain of coagulation-inducing protein tissue factor, truncated tissue factor (tTF), is the most widely used. Since the truncated protein exhibits no coagulation activity and is rapidly cleared in the circulation, free tTF cannot be used for cancer treatment on its own but must be combined with other moieties. We here developed a novel, tumor-specific tTF delivery system through coupling tTF with the DNA aptamer, AS1411, which selectively binds to nucleolin receptors overexpressing on the surface of tumor vascular endothelial cells and is specifically cytotoxic to target cells. Systemic administration of the tTF-AS1411 conjugates into tumor-bearing animals induced intravascular thrombosis solely in tumors, thus reducing tumor blood supply and inducing tumor necrosis without apparent side effects. This conjugate represents a uniquely attractive candidate for the clinical translation of vessel occlusion agent for cancer therapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA