Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Arterioscler Thromb Vasc Biol ; 44(6): e172-e195, 2024 Jun.
Article En | MEDLINE | ID: mdl-38572649

BACKGROUND: Pulmonary hypertension (PH) is a progressive and life-threatening disease characterized by pulmonary vascular remodeling, which involves aberrant proliferation and apoptosis resistance of the pulmonary arterial smooth muscle cells (PASMCs), resembling the hallmark characteristics of cancer. In cancer, the HMGB2 (high-mobility group box 2) protein promotes the pro-proliferative/antiapoptotic phenotype. However, the function of HMGB2 in PH remains uninvestigated. METHODS: Smooth muscle cell (SMC)-specific HMGB2 knockout or HMGB2-OE (HMGB2 overexpression) mice and HMGB2 silenced rats were used to establish hypoxia+Su5416 (HySu)-induced PH mouse and monocrotaline-induced PH rat models, respectively. The effects of HMGB2 and its underlying mechanisms were subsequently elucidated using RNA-sequencing and cellular and molecular biology analyses. Serum HMGB2 levels were measured in the controls and patients with pulmonary arterial (PA) hypertension. RESULTS: HMGB2 expression was markedly increased in the PAs of patients with PA hypertension and PH rodent models and was predominantly localized in PASMCs. SMC-specific HMGB2 deficiency or silencing attenuated PH development and pulmonary vascular remodeling in hypoxia+Su5416-induced mice and monocrotaline-treated rats. SMC-specific HMGB2 overexpression aggravated hypoxia+Su5416-induced PH. HMGB2 knockdown inhibited PASMC proliferation in vitro in response to PDGF-BB (platelet-derived growth factor-BB). In contrast, HMGB2 protein stimulation caused the hyperproliferation of PASMCs. In addition, HMGB2 promoted PASMC proliferation and the development of PH by RAGE (receptor for advanced glycation end products)/FAK (focal adhesion kinase)-mediated Hippo/YAP (yes-associated protein) signaling suppression. Serum HMGB2 levels were significantly increased in patients with PA hypertension, and they correlated with disease severity, predicting worse survival. CONCLUSIONS: Our findings indicate that targeting HMGB2 might be a novel therapeutic strategy for treating PH. Serum HMGB2 levels could serve as a novel biomarker for diagnosing PA hypertension and determining its prognosis.


Disease Models, Animal , HMGB2 Protein , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Pulmonary Artery , Vascular Remodeling , Animals , HMGB2 Protein/genetics , HMGB2 Protein/metabolism , Humans , Male , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Pulmonary Artery/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Rats , Mice , Cell Proliferation , Severity of Illness Index , Signal Transduction , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/physiopathology , Rats, Sprague-Dawley , Female , Cells, Cultured , Middle Aged , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology
2.
Int J Biol Macromol ; 259(Pt 2): 129289, 2024 Feb.
Article En | MEDLINE | ID: mdl-38211910

FS145, a protein containing a WGD motif, was previously described from the salivary transcriptome of the flea Xenopsylla cheopis. Nevertheless, its biological function and complete structure are still uncertain. Herein, FS145 was confirmed to adopt a common αßß structure with the WGD motif exposed on its surface and located right at the top of a loop composed of residues 72-81. Furthermore, FS145 dose-dependently inhibited the proliferation, adhesion, migration, and tube formation of HUVECs by not only binding to integrin αvß3 but also by subsequently inactivating the FAK/Src/MAPK pathway along with the reduction of the expression of MMP-2, MMP-9, VEGFA, bFGF, Ang2, Tie2, HIF-1α, and FAK. Moreover, FS145 also inhibited aortic vessel sprout and showed strong anti-angiogenic activities as assessed ex vivo, by employing the rat aortic ring assay, chick embryo chorioallantoic membrane, and zebrafish embryo models. Altogether, our results suggest that FS145 suppresses angiogenesis ex vivo and in vitro by blocking integrin αvß3. The current study reveals the first anti-angiogenesis disintegrin with WGD motif from invertebrates and provides a beneficial pharmacological activity to inhibit abnormal angiogenesis.


Disintegrins , Siphonaptera , Chick Embryo , Rats , Animals , Disintegrins/pharmacology , Disintegrins/chemistry , Integrin alphaVbeta3/metabolism , Siphonaptera/metabolism , Angiogenesis , Zebrafish/metabolism , Cells, Cultured , Neovascularization, Physiologic , Cell Movement , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry
3.
Article En | MEDLINE | ID: mdl-37702834

BACKGROUND: Atrial fibrillation (AF) is a prevalent and chronic cardiovascular disorder associated with various pathophysiological alterations, including atrial electrical and structural remodeling, disrupted calcium handling, autonomic nervous system dysfunction, aberrant energy metabolism, and immune dysregulation. Emerging evidence suggests that long non-coding RNAs (lncRNAs) play a significant role in the pathogenesis of AF. OBJECTIVE: This discussion aims to elucidate the involvement of AF-related lncRNAs, with a specific focus on their role as miRNA sponges that modulate crucial signaling pathways, contributing to the progression of AF. We also address current limitations in AF-related lncRNA research and explore potential future directions in this field. Additionally, we summarize feasible strategies and promising delivery systems for targeting lncRNAs in AF therapy. CONCLUSION: In conclusion, targeting AF-related lncRNAs holds substantial promise for future investigations and represents a potential therapeutic avenue for managing AF.

4.
Curr Med Sci ; 43(3): 445-455, 2023 Jun.
Article En | MEDLINE | ID: mdl-37191939

OBJECTIVE: Acute lung injury (ALI) is an acute clinical syndrome characterized by uncontrolled inflammation response, which causes high mortality and poor prognosis. The present study determined the protective effect and underlying mechanism of Periplaneta americana extract (PAE) against lipopolysaccharide (LPS)-induced ALI. METHODS: The viability of MH-S cells was measured by MTT. ALI was induced in BALB/c mice by intranasal administration of LPS (5 mg/kg), and the pathological changes, oxidative stress, myeloperoxidase activity, lactate dehydrogenase activity, inflammatory cytokine expression, edema formation, and signal pathway activation in lung tissues and bronchoalveolar lavage fluid (BALF) were examined by H&E staining, MDA, SOD and CAT assays, MPO assay, ELISA, wet/dry analysis, immunofluorescence staining and Western blotting, respectively. RESULTS: The results revealed that PAE obviously inhibited the release of proinflammatory TNF-α, IL-6 and IL-1ß by suppressing the activation of MAPK/Akt/NF-κB signaling pathways in LPS-treated MH-S cells. Furthermore, PAE suppressed the neutrophil infiltration, permeability increase, pathological changes, cellular damage and death, pro-inflammatory cytokines expression, and oxidative stress upregulation, which was associated with its blockage of the MAPK/Akt/NF-κB pathway in lung tissues of ALI mice. CONCLUSION: PAE may serve as a potential agent for ALI treatment due to its anti-inflammatory and anti-oxidative properties, which correlate to the blockage of the MAPK/NF-κB and AKT signaling pathways.


Acute Lung Injury , Periplaneta , Mice , Animals , Lipopolysaccharides/toxicity , NF-kappa B/metabolism , Periplaneta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Cytokines/metabolism , Oxidative Stress , Mice, Inbred BALB C
5.
Acta Pharm ; 73(1): 145-155, 2023 Mar 01.
Article En | MEDLINE | ID: mdl-36692462

Voltage-gated K+ (Kv) channels play a role in the cellular processes of various cancer cells, including lung cancer cells. We previously identified and reported a salivary protein from the Xenopsylla cheopis, FS48, which exhibited inhibitory activity against Kv1.1-1.3 channels when assayed in HEK 293T cells. However, whether FS48 has an inhibitory effect on cancer cells expressing Kv channels is unclear. The present study aims to reveal the effects of FS48 on the Kv channels and the NCI-H460 human lung cancer cells through patch clamp, MTT, wound healing, transwell, gelatinase zymography, qRT-PCR and WB assays. The results demonstrated that FS48 can be effective in suppressing the Kv currents, migration, and invasion of NCI-H460 cells in a dose-dependent manner, despite the failure to inhibit the proliferation. Moreover, the expression of Kv1.1 and Kv1.3 mRNA and protein were found to be significantly reduced. Finally, FS48 decreases the mRNA level of MMP-9 while increasing TIMP-1 mRNA level. The present study highlights for the first time that blood-sucking arthropod saliva-derived protein can inhibit the physiological activities of tumour cells via the Kv channels. Furthermore, FS48 can be taken as a hit compound against the tumour cells expressing Kv channels.


Neoplasms , Potassium Channels, Voltage-Gated , Xenopsylla , Animals , Humans , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Xenopsylla/genetics , Xenopsylla/metabolism , Salivary Glands/metabolism , RNA, Messenger/metabolism
6.
Front Cardiovasc Med ; 9: 911393, 2022.
Article En | MEDLINE | ID: mdl-35966522

Backgrounds: The understanding of death in patients with atrial fibrillation (AF) in China is limited. This study aimed to assess the contemporary survival of AF patients in China and to explore risk factors for deaths. Methods: This was a prospective community-based cohort study including 559 AF patients, who were followed-up from July 2015 to December 2020. Results: During 66-month follow-up, there were 200 deaths (56.5% cardiovascular, 40.0% non-cardiovascular, and 3.5% unknown causes) among 559 AF patients with the median age of 76 years. The top three causes of death were heart failure (33.0%), ischemic stroke (17.0%) and cancer (16.5%). Multivariate Cox regression analysis indicated baseline variables positively associated with all-cause death were age (HR: 1.10, 95% CI: 1.08-1.13), AF subtype (HR: 1.37, 95% CI: 1.08-1.73), prior myocardial infarction (HR: 3.40, 95% CI: 1.48-7.78), previous tumor (HR: 2.61, 95% CI: 1.37-4.98), hypoglycemic therapy at baseline (HR: 1.81, 95% CI: 1.13-2.91), but body weight (HR: 0.98, 95% CI: 0.97-1.00) and use of calcium channel blocker (CCB) (HR: 0.62, 95% CI: 0.41-0.95) played a protective role to all-cause death. Of patients who were alive at the end of follow-up, 24.0% were on oral anticoagulants (OAC) alone, 4.5% on dual antithrombotic therapy, 33.1% on antiplatelet agents alone and 38.4% weren't on any antithrombotic medication. Conclusion: Ischemic stroke still remains one of the leading causes of death and OAC is seriously underused in AF patients in China. Independent risk factors for death are age, AF subtype, previous tumor, prior myocardial infarction, hypoglycemic therapy, low body weight and no CCB use. Clinical Trial Registration: http://www.chictr.org.cn/ (ChiCTR-ICR-15007036).

7.
Curr Pharm Biotechnol ; 23(15): 1873-1882, 2022.
Article En | MEDLINE | ID: mdl-35249479

BACKGROUND: Bacterial resistance to all currently available conventional antibiotics has caused a global public health crisis and led to an imperative search for new agents. Antimicrobial peptides (AMPs) are essential components of host innate immune defense against microbial invasions. OBJECTIVES: The objective of this study was to report a novel AMP, brevinin-2KP, from the skin of the black Kaloula pulchra frog and describe its structural and biological characterization. MATERIALS AND METHODS: The physical and chemical parameters of brevinin-2KP were predicted with the ExPASy Bioinformatics Resource Portal. The assembled sequences were aligned with ClustalW, and the phylogenetic tree was constructed using Mega. Circular dichroism (CD) experiments were carried out to identify the secondary structure and the stability of peptide in different solvent environments. The cytotoxicity of brevinin-2KP was evaluated by the MTT test. To determine antibacterial activity of brevinin- 2KP, a standard two-fold broth dilution method was used. SEM was carried out to observe the morphological change in the bacterial treated by brevinin-2KP. The live/dead bacterial viability was measured with a LIVE/DEAD® BacLight kit. Histamine release and mast cell degranulation assays were performed. RESULTS: The precursor of brevinin-2KP contains 72 amino acid residues, including a conserved signal peptide, acidic propeptide with KR residues, and mature peptide with a sequence of GVITDALKGAAKTVAAELLKKAHCKLTNSC. Phylogenetic analysis based on the amino acid sequences of 34 brevinin-2 peptides from 30 anuran species demonstrates that K. pulchra is genetically closely related to the genus Hylarana. The CD spectra analysis indicates that brevinin-2KP adopts random coil in the water and an organized α-helical conformation in SDS solution. Further, this secondary structure is stable under high salt and high-temperature conditions. Brevinin-2KP is weakly active towards the tested Gram-positive and Gram-negative bacteria as well as fungi due to its membranolytic action. Moreover, brevinin-2KP inhibits the proliferation of several mammal cells with IC50 values ranging from 3.27 to 59.75 µM. In addition, brevinin-2KP promotes degranulation and histamine release of mast cells, indicating that it is involved in the inflammatory response. CONCLUSION: This is the first report on AMP identified from the skin of K. pulchra. Brevinin-2KP adopts a typical amphipathic α-helix conformation in membrane mimic environment and shows antimicrobial and antitumor activities by potential membranolytic mechanism. In addition, brevinin-2KP can promote degranulation and histamine release of mast cells. Brevinin-2KP is expected to become a good drug temple molecule.


Anti-Infective Agents , Antimicrobial Cationic Peptides , Animals , Amino Acids , Anti-Bacterial Agents/metabolism , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Peptides , Anura/metabolism , Cloning, Molecular , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/metabolism , Mammals/metabolism , Microbial Sensitivity Tests , Phylogeny , Protein Sorting Signals , Skin/metabolism , Solvents , Water
8.
J Biol Chem ; 298(1): 101497, 2022 01.
Article En | MEDLINE | ID: mdl-34919963

The Kv1.3 channel has been widely demonstrated to play crucial roles in the activation and proliferation of T cells, which suggests that selective blockers could serve as potential therapeutics for autoimmune diseases mediated by T cells. We previously described that the toxin mimic FS48 from salivary gland of Xenopsylla cheopis downregulates the secretion of proinflammatory factors by Raw 264.7 cells by blocking the Kv1.3 channel and the subsequent inactivation of the proinflammatory MAPK/NF-κB pathways. However, the effects of FS48 on human T cells and autoimmune diseases are unclear. Here, we described its immunomodulatory effects on human T cells derived from suppression of Kv1.3 channel. Kv1.3 currents in Jurkat T cells were recorded by whole-cell patch-clamp, and Ca2+ influx, cell proliferation, and TNF-α and IL-2 secretion were measured using Fluo-4, CCK-8, and ELISA assays, respectively. The in vivo immunosuppressive activity of FS48 was evaluated with a rat DTH model. We found that FS48 reduced Kv1.3 currents in Jurkat T cells in a concentration-dependent manner with an IC50 value of about 1.42 µM. FS48 also significantly suppressed Kv1.3 protein expression, Ca2+ influx, MAPK/NF-κB/NFATc1 pathway activation, and TNF-α and IL-2 production in activated Jurkat T cells. Finally, we show that FS48 relieved the DTH response in rats. We therefore conclude that FS48 can block the Kv1.3 channel and inhibit human T cell activation, which most likely contributes to its immunomodulatory actions and highlights the great potential of this evolutionary-guided peptide as a drug template in future studies.


Autoimmune Diseases , Kv1.3 Potassium Channel , Scorpion Venoms , T-Lymphocytes , Xenopsylla , Adjuvants, Immunologic/pharmacology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Humans , Immunologic Factors/pharmacology , Interleukin-2/metabolism , Kv1.3 Potassium Channel/immunology , Lymphocyte Activation/drug effects , NF-kappa B/metabolism , Potassium Channel Blockers/immunology , Rats , Salivary Glands/chemistry , Scorpion Venoms/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Xenopsylla/chemistry
9.
J Biol Chem ; 296: 100670, 2021.
Article En | MEDLINE | ID: mdl-33864815

The voltage-gated potassium (Kv) 1.3 channel plays a crucial role in the immune responsiveness of T-lymphocytes and macrophages, presenting a potential target for treatment of immune- and inflammation related-diseases. FS48, a protein from the rodent flea Xenopsylla cheopis, shares the three disulfide bond feature of scorpion toxins. However, its three-dimensional structure and biological function are still unclear. In the present study, the structure of FS48 was evaluated by circular dichroism and homology modeling. We also described its in vitro ion channel activity using patch clamp recording and investigated its anti-inflammatory activity in LPS-induced Raw 264.7 macrophage cells and carrageenan-induced paw edema in mice. FS48 was found to adopt a common αßß structure and contain an atypical dyad motif. It dose-dependently exhibited the Kv1.3 channel in Raw 264.7 and HEK 293T cells, and its ability to block the channel pore was demonstrated by the kinetics of activation and competition binding with tetraethylammonium. FS48 also downregulated the secretion of proinflammatory molecules NO, IL-1ß, TNF-α, and IL-6 by Raw 264.7 cells in a manner dependent on Kv1.3 channel blockage and the subsequent inactivation of the MAPK/NF-κB pathways. Finally, we observed that FS48 inhibited the paw edema formation, tissue myeloperoxidase activity, and inflammatory cell infiltrations in carrageenan-treated mice. We therefore conclude that FS48 identified from the flea saliva is a novel potassium channel inhibitor displaying anti-inflammatory activity. This discovery will promote understanding of the bloodsucking mechanism of the flea and provide a new template molecule for the design of Kv1.3 channel blockers.


Anti-Inflammatory Agents/pharmacology , Edema/drug therapy , Inflammation/drug therapy , Kv1.3 Potassium Channel/antagonists & inhibitors , Macrophages/drug effects , Salivary Glands/metabolism , Scorpion Venoms/chemistry , Animals , Edema/immunology , Edema/metabolism , Edema/pathology , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , NF-kappa B/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Xenopsylla
10.
Elife ; 102021 04 20.
Article En | MEDLINE | ID: mdl-33875135

Antimicrobial peptides form part of the innate immune response and play a vital role in host defense against pathogens. Here we report a new antimicrobial peptide belonging to the cathelicidin family, cathelicidin-MH (cath-MH), from the skin of Microhyla heymonsivogt frog. Cath-MH has a single α-helical structure in membrane-mimetic environments and is antimicrobial against fungi and bacteria, especially Gram-negative bacteria. In contrast to other cathelicidins, cath-MH suppresses coagulation by affecting the enzymatic activities of tissue plasminogen activator, plasmin, ß-tryptase, elastase, thrombin, and chymase. Cath-MH protects against lipopolysaccharide (LPS)- and cecal ligation and puncture-induced sepsis, effectively ameliorating multiorgan pathology and inflammatory cytokine through its antimicrobial, LPS-neutralizing, coagulation suppressing effects as well as suppression of MAPK signaling. Taken together, these data suggest that cath-MH is an attractive candidate therapeutic agent for the treatment of septic shock.


Amphibian Proteins/pharmacology , Anti-Infective Agents/pharmacology , Anura , Cathelicidins/pharmacology , Sepsis/drug therapy , Amino Acid Sequence , Amphibian Proteins/chemistry , Animals , Anti-Infective Agents/chemistry , Base Sequence , Cathelicidins/chemistry , Phylogeny , Sequence Alignment
11.
J Pept Sci ; 26(8): e3269, 2020 Aug.
Article En | MEDLINE | ID: mdl-32558003

The inflammatory response plays important roles in acne vulgaris and pain pathogenesis. In previous study, Esc-1GN with anti-inflammatory, antimicrobial, and lipopolysacchride (LPS) binding activity was identified from the skin of the frog Hylarana guentheri. Here, we report its therapeutic potentials for acne vulgaris and inflammatory pain. Esc-1GN destroyed the cell membrane of Propionibacteria acnes in the membrane permeability assays. In addition, bacterial agglutination test suggested that Esc-1GN triggered the agglutination of P. acnes, which was affected by LPS and Ca2+ . Meanwhile, in vivo anti-P. acnes and anti-inflammatory effects of Esc-1GN were confirmed by reducing the counts of P. acnes in mice ear, relieving P. acnes-induced mice ear swelling, decreasing mRNA expression and the production of pro-inflammatory cytokines, and attenuating the infiltration of inflammatory cells. Moreover, Esc-1GN also displayed antinociceptive effect in mice induced by acetic acid and formalin. Therefore, Esc-1GN is a promising candidate drug for treatment of acne vulgaris and inflammatory pain.


Acne Vulgaris/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/drug therapy , Pain/drug therapy , Pore Forming Cytotoxic Proteins/metabolism , Propionibacterium acnes/drug effects , Acne Vulgaris/microbiology , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Mice , Microbial Sensitivity Tests , Pore Forming Cytotoxic Proteins/chemical synthesis , Pore Forming Cytotoxic Proteins/chemistry
12.
Chem Biol Interact ; 327: 109179, 2020 Aug 25.
Article En | MEDLINE | ID: mdl-32534990

Excessive osteoclast leads to the imbalance in bone reconstruction and results in osteolytic diseases, such as osteoporosis and rheumatic arthritis. Integrin αvß3 abundantly expresses on osteoclast and plays a critical role in the formation and function of osteoclast, therefore, blockage of αvß3 has become an attractive therapeutic option for osteolytic diseases. In this study, we find that Tablysin-15, a RGD motif containing disintegrin, concentration-dependently suppresses RANKL-induced osteoclastogenesis, F-actin ring formation and bone resorption without affecting the cell viabilities. Tablysin-15 binds to integrin αvß3 and inhibits the activation of FAK-associated signaling pathways. Tablysin-15 also suppresses the activation of NF-кB, MAPK, and Akt-NFATc1 signaling pathways, which are crucial transcription factors during osteoclast differentiation. Moreover, Tablysin-15 decreases the osteoclastogenesis marker gene expression, including MMP-9, TRAP, CTSK, and c-Src. Finally, Tablysin-15 significantly inhibits LPS-induced bone loss in a mouse model. Taken together, our results indicate that Tablysin-15 significantly suppresses osteoclastogenesis in vitro and in vivo, thus it might be a excellent candidate for treating osteolytic-related diseases.


Bone Density Conservation Agents/pharmacology , Bone Resorption/prevention & control , Insect Proteins/pharmacology , Osteogenesis/drug effects , Salivary Proteins and Peptides/pharmacology , Animals , Bone Density Conservation Agents/toxicity , Bone Resorption/chemically induced , Femur/drug effects , Femur/pathology , Insect Proteins/toxicity , Integrin alphaVbeta3/metabolism , Lipopolysaccharides , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , RANK Ligand/metabolism , RAW 264.7 Cells , Salivary Proteins and Peptides/toxicity , Transcription Factor RelA/metabolism , Up-Regulation/drug effects
13.
Life Sci ; 254: 117753, 2020 Aug 01.
Article En | MEDLINE | ID: mdl-32387419

AIMS: This study aimed to explore the antioxidant properties and neuroprotective effects of Esc-1GN. MAIN METHODS: FRAP assay and ABTS, DPPH, and NO radicals radical scavenging assays were performed to investigated the Antioxidant activities of Esc-1GN in vitro. Hydrogen peroxide (H2O2)-induced cell damage model was used to determine the neuroprotective effects of Esc-1GN. Carrageenan-injected inflamed paw model was performed to analysis the antioxidant and anti-inflammatory properties of Esc-1GN in vivo. KEY FINDINGS: Esc-1GN scavenged the ABTS, DPPH, and NO radicals and reduced Fe3+ in a concentration-dependent manner. Moreover, Esc-1GN exhibited neuroprotective activity by decreasing malondialdehyde and reactive oxygen species accumulation, restoring endogenous antioxidant enzyme activity, and inhibiting H2O2-induced cell cycle arrest and apoptosis in PC12 cells. Esc-1GN significantly reversed the dysregulation of MAPK, AKT and NF-κB signaling caused by H2O2. In vivo, Esc-1GN decreased MDA, COX-2, NO, TNF-α, IL-6, and Il-1ß levels and increased SOD, CAT activity and GSH level in carrageenan-injected inflamed paw tissues. SIGNIFICANCE: These findings suggest that Esc-1GN might serve as a potential antioxidant agent with therapeutic potential in human neurodegenerative diseases.


Antioxidants/pharmacology , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , MAP Kinase Signaling System , Mice
14.
Curr Cancer Drug Targets ; 20(4): 306-315, 2020.
Article En | MEDLINE | ID: mdl-31893992

BACKGROUND: Integrins are crucial anti-cancer therapy targets. We previously showed that tablysin-15 is an integrin antagonist with its Arg-Gly-Asp motif in a novel structural context. OBJECTIVE: Here we investigated the anti-cancer effects and mechanisms of action of tablysin-15 in melanoma cells. METHODS: Cell adhesion, competitive binding, cell viability, and ATP chemiluminescence assays were used to analyze the binding of tablysin-15 to αvß3 integrin and its phenotypic effects. Wound healing, transwells, and zymography were performed to detect motility and matrix metalloproteinase- 2/-9 activities. PARP and caspase-3 cleavage were used as apoptosis assays, while LDH release and flow cytometry were used for necrosis and cell cycle analysis. The expression of mRNAs and proteins of target molecules was measured by qRT-PCR and western blotting, respectively. RESULTS: Tablysin-15 dose-dependently inhibited the proliferation, migration, and invasion of M21 cells through integrin αvß3. The proliferation inhibition caused by tablysin-15 was attributable to G0/G1 phase arrest rather than apoptosis or necrosis. Furthermore, tablysin-15 suppressed MMP-2/- 9 activities and the mRNA expression of MMP-2/-9 and COX-2 but was upregulated TIMP-1 in M21 cells. Meanwhile, tablysin-15 suppressed the expression of cyclin D1/E and CDK 2/6, the phosphorylation of FAK, Akt, and ERK, and nuclear translocation of NF-κB, while increasing the expression of the CDK inhibitor p21waf1/C1. Taken together, tablysin-15 might inhibit melanoma cell metastasis and proliferation by competing with αvß3 integrin, thereby blocking FAK-associated signaling pathways and nuclear translocation of NF-κB. CONCLUSION: Tablysin-15 has reliable anti-cancer effects against M21 melanoma cells, suggesting tablysin-15 is a promising anti-tumor drug.


Insect Proteins/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/drug therapy , NF-kappa B/metabolism , Salivary Proteins and Peptides/pharmacology , Skin Neoplasms/drug therapy , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Disintegrins/chemistry , Disintegrins/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Focal Adhesion Kinase 1/metabolism , Humans , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Melanoma/metabolism , Melanoma/pathology , Oligopeptides/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
15.
Int J Biol Macromol ; 129: 1155-1167, 2019 May 15.
Article En | MEDLINE | ID: mdl-30660566

αvß3 integrin expressed on cancer cell surfaces is associated with important cancer hallmarks including survival and metastasis and is thus a potential anticancer drug target. Tablysin-15 contains the RGD motif and is a high-affinity αvß3 integrin antagonist. The aim of this study was to investigate the antitumor effect and mechanism of action of tablysin-15 against αvß3 integrin high-expressing breast cancer cell lines in vitro and in vivo. Tablysin-15 dose-dependently inhibited the proliferation, migration, and invasion of two breast cancer cell lines via the αvß3 integrin in vitro. Proliferation inhibition was attributable to G0/G1 phase cell cycle arrest rather than apoptosis or necrosis. Furthermore, tablysin-15 downregulated the activity and mRNA expression of MMP-2/-9, VEGF, and COX-2 but upregulated TIMP-1/-2 mRNA in both cell lines. Further, tablysin-15 suppressed the expression of CDK2, CDK6, cyclin D1, and cyclin E, the phosphorylation of FAK, Akt, GSK-3ß, and ERK, and the nuclear translocation of NF-κB while increasing the expression of the CDK inhibitor p21waf1/C1. Lastly, tablysin-15 provided effective antitumor protection in vivo. Thus, tablysin-15 inhibits the metastasis and proliferation of breast cancer cells through binding αvß3 integrin and blocking FAK-associated signaling pathways as well as nuclear translocation of NF-κB.


Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Disintegrins/chemistry , Disintegrins/pharmacology , Oligopeptides/chemistry , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin alphaVbeta3/metabolism , Mice , Neoplasm Invasiveness , Resting Phase, Cell Cycle/drug effects , Xenograft Model Antitumor Assays
...