Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Int J Biol Macromol ; 267(Pt 1): 131492, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38604418

RESUMO

Human heavy chain ferritin (HFn) protein cage has been explored as a nanocarrier for targeted anticancer drug delivery. Here, we introduced a matrix metalloproteinases (MMPs)-cleavable sequence into the DE loop of HFn, creating an MMP-responsive variant, MR-HFn, for localized and extracellular drug release. The crystal structure of MR-HFn revealed that the addition of the MMPs recognition sequence did not affect the self-assembly of HFn but presented a surface-exposed loop susceptible to MMPs cleavage. Biochemical analysis indicated that this engineered protein cage is responsive to MMPs, enabling the targeted release of encapsulated drugs. To evaluate the therapeutic potential of this engineered protein cage, monosubstituted ß-carboxy phthalocyanine zinc (CPZ), a type of photosensitizer, was loaded inside this protein cage. The prepared CPZ@MR-HFn showed higher uptake and stronger phototoxicity in MMPs overexpressed tumor cells, as well as enhanced penetration into multicellular tumor spheroids compared with its counterpart CPZ@HFn in vitro. In vivo, CPZ@MR-HFn displayed a higher tumor inhibitory rate than CPZ@HFn under illumination. These results indicated that MR-HFn is a promising nanocarrier for anticancer drug delivery and the MMP-responsive strategy here can also be adapted for other stimuli.


Assuntos
Antineoplásicos , Liberação Controlada de Fármacos , Metaloproteinases da Matriz , Engenharia de Proteínas , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/química , Metaloproteinases da Matriz/metabolismo , Animais , Linhagem Celular Tumoral , Camundongos , Ferritinas/química , Ferritinas/metabolismo , Indóis/química , Indóis/farmacologia , Portadores de Fármacos/química , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/química
2.
Cardiovasc Res ; 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38546342

RESUMO

AIMS: Alteplase is a cornerstone thrombolytic agent in clinical practice, but presents a potential bleeding risk. Stroke patients need pre-screening to exclude hemorrhagic stroke before using Alteplase. In this study, we develop a new thrombolytic agent citPA5, characterized by an enhanced safety profile and minimal bleeding tendency. METHODS AND RESULTS: A clot lysis agent, named citPA5, is developed based on rtPA with point mutations to completely suppress its proteolytic activity in the absence of fibrin. In the presence of fibrin, citPA5 exhibited significantly higher fibrinolytic activity (a 15.8-fold increase of kcat/Km). Furthermore, citPA5 showed resistance to endogenous fibrinolysis inhibitor, PAI-1, resulting in enhanced potency. In a series of safety evaluation experiments, including thrombelastography (TEG) assay, mice tail bleeding assay, and a murine intracerebral hemorrhage (ICH) model, citPA5 did not cause systemic bleeding or worsen intracerebral hemorrhage compared to Alteplase. This highlights the low risk of bleeding associated with citPA5. Finally, we found that citPA5 effectively improved cerebral blood flow and reduced infarct volume in a carotid embolism-induced stroke (CES) model. CONCLUSIONS: This clot lysis agent, citPA5, not only exhibits a low risk of bleeding but also demonstrates highly effective thrombolysis capabilities. As a result, citPA5 shows great potential for administration prior to the classification of stroke types, making it possible for use in ambulances at the onset of stroke when symptoms are identified. The findings presented in this study also suggest that this strategy could be applied to develop a new generation of fibrinolytic drugs that offer greater safety and specificity in targeting fibrin.

3.
Int J Biol Macromol ; 257(Pt 2): 128618, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38070813

RESUMO

Administration of recombinant tPA (rtPA, or trade name Alteplase®) is an FDA-approved therapy for acute ischemic stroke (AIS), but poses the risk of hemorrhagic complications. Recombinant tPA can be rapidly inactivated by the endogenous inhibitor, plasminogen activator inhibitor 1 (PAI-1). In this work, we study a novel treatment approach that combines a PAI-1 inhibitor, PAItrap4, with a reduced dose of rtPA to address the hemorrhagic concern of rtPA. PAItrap4 is a highly specific and very potent protein-based inhibitor of PAI-1, comprising of a variant of uPA serine protease domain, human serum albumin, and a cyclic RGD peptide. PAItrap4 efficiently targets and inhibits PAI-1 on activated platelets, and also possesses a long half-life in vivo. Our results demonstrate that PAItrap4 effectively counteracts the inhibitory effects of PAI-1 on rtPA, preserving rtPA activity based on amidolytic and clot lysis assays. In an in vivo murine stroke model, PAItrap4, together with low-dose rtPA, enhances the blood perfusion in the stroke-affected areas, reduces infarct size, and promotes neurological recovery in mice. Importantly, such treatment does not increase the amount of cerebral hemorrhage, thus reducing the risk of cerebral hemorrhage. In addition, PAItrap4 does not compromise the normal blood coagulation function in mice, demonstrating its safety as a therapeutic agent. These findings highlight this combination therapy as a promising alternative for the treatment of ischemic stroke, offering improved safety and efficacy.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Humanos , Camundongos , Animais , Ativador de Plasminogênio Tecidual/farmacologia , Ativador de Plasminogênio Tecidual/uso terapêutico , Inibidor 1 de Ativador de Plasminogênio , AVC Isquêmico/complicações , AVC Isquêmico/tratamento farmacológico , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/complicações , Hemorragia Cerebral/complicações , Hemorragia Cerebral/tratamento farmacológico , Fibrinolíticos/farmacologia , Fibrinolíticos/uso terapêutico
4.
J Biomol Struct Dyn ; : 1-12, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38111151

RESUMO

Cancer remains one of the most pressing challenges to global healthcare, exerting a significant impact on patient life expectancy. Cancer metastasis is a critical determinant of the lethality and treatment resistance of cancer. The urokinase-type plasminogen activator receptor (uPAR) shows great potential as a target for anticancer and antimetastatic therapies. In this work, we aimed to identify potential uPAR inhibitors by structural dynamics-based virtual screenings against a natural product library on four representative apo-uPAR structural models recently derived from long-timescale molecular dynamics (MD) simulations. Fifteen potential inhibitors (NP1-NP15) were initially identified through molecular docking, consensus scoring, and visual inspection. Subsequently, we employed MD-based molecular mechanics-generalized Born surface area (MM-GBSA) calculations to evaluate their binding affinities to uPAR. Structural dynamics analyses further indicated that all of the top 6 compounds exhibited stable binding to uPAR and interacted with the critical residues in the binding interface between uPAR and its endogenous ligand uPA, suggesting their potential as uPAR inhibitors by interrupting the uPAR-uPA interaction. We finally predicted the ADMET properties of these compounds. The natural products NP5, NP12, and NP14 with better binding affinities to uPAR than the uPAR inhibitors previously discovered by us were proven to be potentially orally active in humans. This work offers potential uPAR inhibitors that may contribute to the development of novel effective anticancer and antimetastatic therapeutics.Communicated by Ramaswamy H. Sarma.

5.
Int J Mol Sci ; 24(22)2023 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-38003338

RESUMO

Chitinase-3-like protein 1 (CHI3L1), a chitinase-like protein family member, is a secreted glycoprotein that mediates macrophage polarization, inflammation, apoptosis, angiogenesis, and carcinogenesis. Abnormal CHI3L1 expression has been associated with multiple metabolic and neurological disorders, including diabetes, atherosclerosis, and Alzheimer's disease. Aberrant CHI3L1 expression is also reportedly associated with tumor migration and metastasis, as well as contributions to immune escape, playing important roles in tumor progression. However, the physiological and pathophysiological roles of CHI3L1 in the development of metabolic and neurodegenerative diseases and cancer remain unclear. Understanding the polarization relationship between CHI3L1 and macrophages is crucial for disease progression. Recent research has uncovered the complex mechanisms of CHI3L1 in different diseases, highlighting its close association with macrophage functional polarization. In this article, we review recent findings regarding the various disease types and summarize the relationship between macrophages and CHI3L1. Furthermore, this article also provides a brief overview of the various mechanisms and inhibitors employed to inhibit CHI3L1 and disrupt its interaction with receptors. These endeavors highlight the pivotal roles of CHI3L1 and suggest therapeutic approaches targeting CHI3L1 in the development of metabolic diseases, neurodegenerative diseases, and cancers.


Assuntos
Quitinases , Neoplasias , Doenças Neurodegenerativas , Humanos , Inflamação/metabolismo , Macrófagos/metabolismo , Proteína 1 Semelhante à Quitinase-3
6.
Biophys J ; 122(21): 4135-4143, 2023 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-37731243

RESUMO

Lysophospholipids (lysoPLs) are crucial metabolites involved in various physiological and pathological cellular processes. Understanding their binding interactions, particularly with human serum albumin (HSA), is essential due to their role in regulating lysoPLs-induced cytotoxicity. However, the precise mechanism of lysoPLs binding to HSA remains elusive. In this study, we employed fluorescence quenching and optical interferometry assays to demonstrate direct binding between lysophosphatidylcholine (LPC) and HSA (KD = 25 µM). Furthermore, we determined crystal structures of HSA in complex with LPC, both in the absence and the presence of the endogenous fatty acid myristate (14:0). The crystal structure of binary HSA:LPC revealed that six LPC molecules are bound to HSA at the primary fatty acid binding sites. Interestingly, the ternary HSA:Myr:LPC structure demonstrated the continued binding of three LPC molecules to HSA at binding sites 1, 3, and 5 in the presence of myristate. These findings support HSA's role as a carrier protein for lysoPLs in blood plasma and provide valuable insights into the structural basis of their binding mechanisms.


Assuntos
Lisofosfatidilcolinas , Albumina Sérica Humana , Humanos , Albumina Sérica Humana/química , Albumina Sérica Humana/metabolismo , Albumina Sérica/química , Ligação Proteica , Miristatos , Modelos Moleculares , Ácidos Graxos/metabolismo
7.
J Hazard Mater ; 460: 132454, 2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37703742

RESUMO

The excessive use of quaternary ammonium compounds (QACs) following the COVID-19 pandemic has raised substantial concerns regarding their biosafety. Overuse of QACs has been associated with chronic biological adverse effects, including genotoxicity or carcinogenicity. In particular, inadvertent intravascular administration or oral ingestion of QACs can lead to fatal acute toxicity. To enhance the biosafety and antimicrobial efficacy of QACs, this study reports a new series of QACs, termed as PACs, with the alkyl chain of benzalkonium substituted by a phthalocyanine moiety. Firstly, the rigid phthalocyanine moiety enhances the selectivity of QACs to bacteria over human cells and reduces alkyl chain's entropic penalty of binding to bacterial membranes. Furthermore, phthalocyanine neutralizes hemolysis and cytotoxicity of QACs by binding with albumin in plasma. Our experimental results demonstrate that PACs inherit the optical properties of phthalocyanine and validate the broad-spectrum antibacterial activity of PACs in vitro. Moreover, the intravascular administration of the most potent PAC, PAC1a, significantly reduced bacterial burden and ameliorated inflammation level in a bacteria-induced septic mouse model. This study presents a new strategy to improve the antimicrobial efficacy and biosafety of QACs, thus expanding their range of applications to the treatment of systemic infections.


Assuntos
COVID-19 , Desinfetantes , Animais , Camundongos , Humanos , Antibacterianos/toxicidade , Compostos de Amônio Quaternário/toxicidade , Contenção de Riscos Biológicos , Pandemias , Indóis/toxicidade
8.
Molecules ; 28(12)2023 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-37375194

RESUMO

Photodynamic therapy (PDT) is recognized as a powerful method to inactivate cells. However, the photosensitizer (PS), a key component of PDT, has suffered from undesired photobleaching. Photobleaching reduces reactive oxygen species (ROS) yields, leading to the compromise of and even the loss of the photodynamic effect of the PS. Therefore, much effort has been devoted to minimizing photobleaching in order to ensure that there is no loss of photodynamic efficacy. Here, we report that a type of PS aggregate showed neither photobleaching nor photodynamic action. Upon direct contact with bacteria, the PS aggregate was found to fall apart into PS monomers and thus possessed photodynamic inactivation against bacteria. Interestingly, the disassembly of the bound PS aggregate in the presence of bacteria was intensified by illumination, generating more PS monomers and leading to an enhanced antibacterial photodynamic effect. This demonstrated that on a bacterial surface, the PS aggregate photo-inactivated bacteria via PS monomer during irradiation, where the photodynamic efficiency was retained without photobleaching. Further mechanistic studies showed that PS monomers disrupted bacterial membranes and affected the expression of genes related to cell wall synthesis, bacterial membrane integrity, and oxidative stress. The results obtained here are applicable to other types of PSs in PDT.


Assuntos
Isoindóis , Compostos Organometálicos , Fotodegradação , Fotoquimioterapia , Fármacos Fotossensibilizantes , Compostos de Zinco , Compostos de Zinco/química , Fármacos Fotossensibilizantes/química , Isoindóis/química , Escherichia coli/efeitos dos fármacos , Escherichia coli/efeitos da radiação , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/efeitos da radiação
9.
Int J Pharm ; 637: 122878, 2023 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-36958614

RESUMO

Recombinant tissue-type plasminogen activator (rtPA, or Alteplase) is the first approved thrombolytic drug for acute ischemic stroke, but suffers from a short half-life and poor resistance to plasminogen activator inhibitor (PAI-1), limiting its clinical use. The development of novel thrombolytic agents with improved benefit/risk balance has always been of great significance. In this study, we identified a mutant of serine protease domain of tPA (named ΔtPAA146V) capable of escaping the inhibition by endogenous PAI-1 with 66-fold increased resistance compared to the wild type tPA. Based on this mutant, we generated a triple fusion ΔtPA (TriF-ΔtPA) containing albumin and fibrin binding peptide(FBP). The fusion with albumin effectively prolonged the plasma half-life of ΔtPA in mice to 144 min, which is much longer than ΔtPA and did not affect its thrombolytic activity. Furthermore, FBP rendered fibrin specificity of the fusion protein, giving a dissociation constant of âˆ¼ 25 ± 0.9 µM. In a novel murine carotid embolism-induced stroke (CES) model, i.v. administration of TriF-ΔtPA promoted vascular recanalization, reduced infarct volume, and mitigated neurobehavioral deficits more significantly compared to ΔtPA-HSA or Alteplase, showing little bleeding risk. Together, this long-acting PAI-1-resistant thrombolytic agent holds great potential for clinical applications.


Assuntos
Embolia , AVC Isquêmico , Acidente Vascular Cerebral , Camundongos , Animais , Ativador de Plasminogênio Tecidual/química , Inibidor 1 de Ativador de Plasminogênio/química , AVC Isquêmico/tratamento farmacológico , Fibrinolíticos/farmacologia , Acidente Vascular Cerebral/tratamento farmacológico , Fibrina , Terapia Trombolítica , Embolia/tratamento farmacológico
10.
J Med Chem ; 66(8): 5415-5426, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-36854648

RESUMO

Diltiazem and glibenclamide are commonly used hypotensive and antidiabetic drugs. This study reports the discovery of the potential antitumor and antimetastatic effects of these two drugs using a structural dynamics-driven virtual screening targeting urokinase receptor (uPAR). Owing to uPAR's high flexibility, currently resolved crystal structures of uPAR, all in ligand-bound states, provide limited representations of its physiological conformation. To improve the accuracy of screening, we performed a long-timescale molecular dynamics simulation and obtained the representative conformations of apo-uPAR as the targets for our screening. Experimentally, we demonstrated that diltiazem and glibenclamide bound uPAR with KD values in the micromolar range. In addition, both compounds effectively suppressed tumor growth and metastasis in a uPAR-dependent manner in vitro and in vivo. This work not only provides two potent uPAR inhibitors but also reports a proof-of-concept study on the potential off-label antitumor and antimetastatic uses of diltiazem and glibenclamide.


Assuntos
Neoplasias , Ativador de Plasminogênio Tipo Uroquinase , Humanos , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Diltiazem , Glibureto , Neoplasias/patologia , Ligantes
11.
Int J Pharm ; 634: 122636, 2023 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-36696930

RESUMO

Urokinase plasminogen activator receptor (uPAR) is a key participant in extracellular proteolysis, tissue remodeling and cell motility. uPAR overexpresses in most solid tumors and several hematologic malignancies, but has low levels on normal tissues, thus is advocated as a molecular target for cancer therapy. One of the obstacles for the evaluation of uPAR targeting agents in preclinical study is the species specificity, where targeting agents for human uPAR  usually not bind to murine uPAR. Here, we develop a targeting agent that binds to both murine and human uPAR. This targeting agent is genetically fused to human serum albumin, a commonly used drug carrier, and the final construct is named as uPAR targeting carrier (uPARTC). uPARTC binds specifically to uPAR-overexpressing 293T/huPAR and 293T/muPAR as demonstrated by flow cytometry. A cytotoxic compound, celastrol, is embedded into uPARTC non-covalently. The resulting macromolecular complex show effective proliferation inhibition on both murine and human uPAR overexpressing cells, and exhibit potent antitumor efficacy on hepatoma H22-bearing mice. This work demonstrates that uPARTC is a promising tumor targeting drug carrier, which address the species-specificity challenge of uPAR targeting agents and can be used to load other cytotoxic compounds.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Camundongos , Animais , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Portadores de Fármacos , Receptores de Albumina , Albuminas
12.
Mol Pharm ; 20(2): 905-917, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36463525

RESUMO

Triple-negative breast cancer (TNBC) is highly aggressive and causes a higher proportion of metastatic cases. However, therapies directed to specific molecular targets have rarely achieved clinically meaningful improvements in the outcome of TNBC therapy. A urokinase-type plasminogen activator (uPA), one of the best-validated biomarkers of breast cancer, is an extracellular proteolytic serine protease involved in many pathological and physiological processes, including tumor cell invasion and metastasis. Nafamostat mesylate (NM) is a synthetic compound that inhibits various serine proteases and has been used as a therapeutic agent for the treatment of TNBC. Nevertheless, NM has poor specificity for serine proteases and is easy be hydrolyzed; moreover, the inhibitory mechanism of TNBC therapy is unclear. In this study, we combine NM with a macromolecular drug delivery vehicle, mouse amino-terminal fragment of urokinase-human serum albumin (mATF-HSA), to form a complex (mATF-HSA:NM) using the dilution-incubation-purification method. mATF specifically targets uPAR overexpressed on the surface of TNBC cells; moreover, HSA prevents NM from being hydrolyzed by numerous serine proteases. mATF-HSA:NM showed stronger inhibitory effects on the proliferation and metastasis of TNBC in vitro and in vivo without significant cytotoxicity on normal cells and tissues. In addition, we demonstrated that NM mediates metastasis of TNBC cells through inhibition of uPA using a stable uPA knockdown cell line (MDA-MB231 shuPA). Overall, we have developed a macromolecular complex targeted to treat high uPAR-expressing tumor types, and mATF-HSA can potentially be used to load other types of drugs with tumor-targeting specificity for mouse tumor models and is a promising tool to study tumor biology in mouse tumor models.


Assuntos
Neoplasias de Mama Triplo Negativas , Ativador de Plasminogênio Tipo Uroquinase , Humanos , Camundongos , Animais , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/patologia , Albumina Sérica Humana , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Resultado do Tratamento
13.
ACS Appl Mater Interfaces ; 14(41): 47003-47013, 2022 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-36214495

RESUMO

Air pollution caused by bacteria and viruses has posed a serious threat to public health. Commercial air purifiers based on dense fibrous filters can remove particulate matter, including airborne pathogens, but do not kill them efficiently. Here, we developed a double-grafted antibacterial fiber material for the high-efficiency capture and inactivation of airborne microorganisms. Tetracarboxyl phthalocyanine zinc, a photosensitizer, was first grafted onto the polyester (PET) fiber, followed by coating with chitosan on the surface of PET fiber to make a double-grafted fiber material. Under the irradiation of light with a specific wavelength (680 nm), double-grafted fiber materials killed up to 99.99% of Gram-positive bacteria and Gram-negative bacteria and had a significant antibacterial effect on drug-resistant bacteria. The double-grafted PET fiber showed broad-spectrum antibacterial activities and was capable to inactivate drug-resistant bacteria. Notably, in filtration experiments for airborne bacteria, this double-grafted PET fiber demonstrated a high bacteria capture efficiency (95.68%) better than the untreated PET fiber (64.87%). Besides, the double-grafted PET fiber was capable of efficiently killing airborne bacteria. This work provides a new idea for the development of air filtration materials that can efficiently kill airborne pathogen and has good biosafety.


Assuntos
Quitosana , Poliésteres , Fármacos Fotossensibilizantes/farmacologia , Material Particulado , Bactérias , Antibacterianos/farmacologia , Zinco , Tomografia por Emissão de Pósitrons
14.
J Clean Prod ; 379: 134632, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36246409

RESUMO

Quaternary ammonium compounds (QACs) are inexpensive and readily available disinfectants, and have been widely used, especially since the COVID-19 outbreak. The toxicity of QACs to humans has raised increasing concerns in recent years. Here, a new type of QACs was synthesized by replacing the alkyl chain with zinc phthalocyanine (ZnPc), which consists of a large aromatic ring and is hydrophobic in nature, similar to the alkyl chain of QACs. Three ZnPc-containing disinfectants were synthesized and fully characterized. These compounds showed 15-16 fold higher antimicrobial effect against Gram-negative bacteria than the well-known QACs with half-maximal inhibitory (IC50) values of 1.43 µM, 2.70 µM, and 1.31 µM, respectively. With the assistance of 680 nm light, compounds 4 and 6 had much higher bactericidal toxicities at nanomolar concentrations. Compound 6 had a bactericidal efficacy of close to 6 logs (99.9999% kill rate) at 1 µM to Gram-positive bacteria, including MRSA, under light illumination. Besides, these compounds were safe for mammalian cells. In a mouse model, compound 6 was effective in healing wound infection. Importantly, compound 6 was easily degraded at working concentrations under sunlight illumination, and is environmentally friendly. Thus, compound 6 is a novel and promising disinfectant.

15.
Biophys J ; 121(20): 3940-3949, 2022 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-36039386

RESUMO

Nafamostat mesylate (NM) is a synthetic compound that inhibits various serine proteases produced during the coagulation cascade and inflammation. Previous studies showed that NM was a highly safe drug for the treatment of different cancers, but the precise functions and mechanisms of NM are not clear. In this study, we determined a series of crystal structures of NM and its hydrolysates in complex with a serine protease (urokinase-type plasminogen activator [uPA]). These structures reveal that NM was cleaved by uPA and that a hydrolyzed product (4-guanidinobenzoic acid [GBA]) remained covalently linked to Ser195 of uPA, and the other hydrolyzed product (6-amidino-2-naphthol [6A2N]) released from uPA. Strikingly, in the inactive uPA (uPA-S195A):NM structure, the 6A2N side of intact NM binds to the specific pocket of uPA. Molecular dynamics simulations and end-point binding free-energy calculations show that the conf1 of NM (6A2N as P1 group) in the uPA-S195A:NM complex may be more stable than conf2 of NM (GBA as P1 group). Moreover, in the structure of uPA:NM complex, the imidazole group of His57 flips further away from Ser195 and disrupts the stable canonical catalytic triad conformation. These results not only reveal the inhibitory mechanism of NM as an efficient serine protease inhibitor but also might provide the structural basis for the further development of serine protease inhibitors.


Assuntos
Inibidores de Serina Proteinase , Ativador de Plasminogênio Tipo Uroquinase , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Inibidores de Serina Proteinase/farmacologia , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/metabolismo , Serina Proteases , Imidazóis
16.
Biomater Adv ; 135: 212728, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35929206

RESUMO

ZnO is an important component in skin-protection products and wound-care medicines. However, ZnO's antibacterial activity is moderate. We developed two types of ZnO microparticles loading with phthalocyanine-type photosensitizers (ZnO/PSs) introducing the photodynamic effects. These photosensitive ZnO microparticles exhibited long-term while moderate antimicrobial effects by continuously releasing Zn2+ ions. The antimicrobial efficacies were remarkably enhanced by triggering the photodynamic antimicrobial effects. Compared to the sole ZnO which showed non-measurable antimicrobial activity at a concentration of 10 mg/L, both ZnO/PSs demonstrated antimicrobial rates ranged 99%-99.99% against Escherichia coli, normal and drug-resistant Staphylococcus aureus. In a dorsal wound infection mouse model, treatment with ZnO/PSs significantly accelerated the wound recovery rates. ZnO/PSs promoted wound healing by a dual effect: 1) the release of Zn2+ ions from ZnO facilitating tissue remodeling; 2) the photodynamic effect efficiently eliminates pathogens avoiding infection. Notably, ZnO/PSs inherited the high biosafety of ZnO without causing noticeable toxicity against erythrocyte and endothelial cells. This study not only provides a highly safe and efficient antimicrobial ZnO material for skin cares and wound modulations, but also proposes a strategy to functionalize ZnO materials.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Óxido de Zinco , Animais , Antibacterianos/farmacologia , Células Endoteliais , Escherichia coli , Camundongos , Staphylococcus aureus , Óxido de Zinco/farmacologia
17.
Int J Biol Macromol ; 216: 172-178, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35788007

RESUMO

Metallodrugs are important for anticancer treatments. They bind mainly to human serum albumin (HSA) in blood circulation, greatly modulating their pharmacokinetics and anticancer efficacy. Fatty acid (FA) is one of the most important endogenous ligands of HSA with tight binding to HSA and affecting its conformation. However, the effect of fatty acids on metallodrugs interaction with HSA is unknown. Here we identify the binding sites of a widely used metallodrug, cisplatin, in HSA in the presence or absence of a representative fatty acid, myristate, by X-ray crystallography. Our crystal structures indicate that the sidechain of residue Met548 becomes more exposed to solvent in the presence of fatty acid, and is the main Pt binding site together with Met329 in HSA:Myr:cisplatin ternary structure. An undoubted new Pt binding site is detected at His338 in the presence of fatty acid, and additional two sites are also identified at His146 and His440 + K436. In addition, we revealed the mechanism of cisplatin-induced HSA aggregation, which is due to the crosslinking between Met298 and His510 of two HSA molecules.


Assuntos
Cisplatino , Albumina Sérica Humana , Sítios de Ligação , Cisplatino/farmacologia , Ácidos Graxos/química , Humanos , Modelos Moleculares , Ligação Proteica , Albumina Sérica/química , Albumina Sérica Humana/metabolismo
18.
Pharmacol Res ; 182: 106331, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35772646

RESUMO

The Angiopoietin (Ang)-Tyrosine kinase with immunoglobulin-like and EGF-like domains (Tie) axis is an endothelial cell-specific ligand-receptor signaling pathway necessary for vascular and lymphatic development. The Ang-Tie axis is involved in regulating angiogenesis, vascular remodeling, vascular permeability, and inflammation to maintain vascular quiescence. Disruptions in the Ang-Tie axis are involved in many vascular and lymphatic diseases and play an important role in physiological and pathological vascular processes. Given recent advances in the Ang-Tie axis in the vascular and lymphatic systems, this review focuses on the multiple functions of the Ang-Tie axis in inflammation-induced vascular permeability, vascular remodeling, atherosclerosis, ocular angiogenesis, tumor angiogenesis, and metastasis. A summary of relevant therapeutic approaches to the Ang-Tie axis, including therapeutic antibodies, recombinant proteins and small molecule drugs are also discussed. The purpose of this review is to provide new hypotheses and identify potential therapeutic strategies based on the Ang-Tie signaling axis for the treatment of vascular and lymphatic-related diseases.


Assuntos
Angiopoietinas , Receptor TIE-2 , Angiopoietina-1 , Angiopoietinas/metabolismo , Humanos , Inflamação , Sistema Linfático/metabolismo , Neovascularização Patológica , Receptor TIE-2/metabolismo
19.
Drug Deliv ; 29(1): 1824-1835, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35674505

RESUMO

Thrombosis occurs in both macrovasculature and microvasculature, causing various cardio-cerebral vascular diseases. The lack of effective and safe antithrombotic drugs leads to a public health crisis. Mounting evidence suggests that protein disulfide isomerase (PDI) plays a critical role in the initial stage of thrombus formation, motivating the research of the feasibility of PDI inhibitors as novel anti-thrombotics. Rutin, one of the most potent PDI inhibitors, was reported to suppress platelet aggregation and thrombosis in animal models, but further studies and clinical translation were restricted due to its low aqueous solubility and oral bioavailability. In this work, we fabricated rutin-loaded lipid-based nano-formulation (NanoR) and characterized their physical-chemical properties, release profiles, pharmacokinetic process, and pharmacodynamic function against thrombosis in macrovessels and microvessels. NanoR provided increased solubility and dissolution of rutin to achieve earlier Tmax and higher Cmax than the sodium salt of rutin (NaR) after oral gavage. Ex vivo studies demonstrated that NanoR significantly inhibited thrombin generation and clot formation in the plasma of mice. Importantly, such effect was reversed by exogenous recombinant PDI, demonstrating the specificity of the NanoR. In direct current-induced arterial thrombosis model and ferric chloride-induced microvascular thrombosis model, NanoR exhibited greatly enhanced antithrombotic activity compared with NaR. NanoR also showed good safety performance according to tail bleeding assay, global coagulation tests, and histological analysis. Overall, our current results indicated that NanoR offers a promising antithrombotic treatment with potential for clinical translation.


Assuntos
Isomerases de Dissulfetos de Proteínas , Trombose , Animais , Plaquetas/metabolismo , Fibrinolíticos/farmacologia , Fibrinolíticos/uso terapêutico , Lipídeos , Camundongos , Isomerases de Dissulfetos de Proteínas/metabolismo , Isomerases de Dissulfetos de Proteínas/uso terapêutico , Rutina/farmacologia , Trombose/tratamento farmacológico
20.
J Biol Chem ; 298(7): 102120, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35697069

RESUMO

Aflatoxins are a series of highly toxic and carcinogenic secondary metabolites that are synthesized by Aspergillus species. The degradation of aflatoxin enzymes is an important regulatory mechanism which modulates mycotoxin producing. The retromer complex is responsible for the retrograde transport of specific biomolecules and the vacuolar fusion in the intracellular transport. Late endosomal-associated GTPase (Rab7) has been shown to be a downstream effector protein of the retromer complex. A deficiency in the retromer complex or Rab7 results in several cellular trafficking problems in yeast and humans, like protein abnormal accumulation. However, whether retromer dysfunction is involved in aflatoxin synthesis remains unclear. Here, we report that the core retromer complex, which comprises three vacuolar protein sorting-associated proteins (AflVps26-AflVps29-AflVps35), is essential for the development of dormant and resistant fungal forms such as conidia (asexual reproductive spore) and sclerotia (hardened fungal mycelium), as well as aflatoxin production and pathogenicity, in Aspergillus flavus. In particular, we show the AflVps26-AflVps29-AflVps35 complex is negatively correlated with aflatoxin exportation. Structural simulation, site-specific mutagenesis, and coimmunoprecipitation experiments showed that interactions among AflVps26, AflVps29, and AflVps35 played crucial roles in the retromer complex executing its core functions. We further found an intrinsic connection between AflRab7 and the retromer involved in vesicle-vacuole fusion, which in turn affected the accumulation of aflatoxin synthesis-associated enzymes, suggesting that they work together to regulate the production of toxins. Overall, these results provide mechanistic insights that contribute to our understanding of the regulatory role of the core retromer complex in aflatoxin metabolism.


Assuntos
Aflatoxinas , Aspergillus flavus , Aflatoxinas/metabolismo , Aspergillus/metabolismo , Aspergillus flavus/genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Humanos , Metabolismo Secundário , Esporos Fúngicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...