Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.289
Filter
1.
J Ethnopharmacol ; 336: 118711, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39181286

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: According to the theory of Qi and blood in Traditional Chinese Medicine (TCM), the combination of Qi-reinforcing herbs and blood-activating herbs has a synergistic effect in improving blood stasis syndrome, especially in tumor treatment. The classic "Radix Astragali - Salvia miltiorrhiza" duo exemplifies this principle, renowned for invigorating Qi and activating blood flow, employed widely in tumor therapies. Our prior research underscores the potent inhibition of pancreatic tumor xenografts by the combination of Formononetin (from Radix Astragali) and Salvianolic acid B (from Salvia miltiorrhiza) in vitro. However, it remains unclear whether this combination can inhibit the abnormal vascularization of pancreatic tumors to achieve its anti-cancer effect. AIM OF THE STUDY: Abnormal vasculature, known to facilitate tumor growth and metastasis. Strategies to normalize tumor-associated blood vessels provide a promising avenue for anti-tumor therapy. This study aimed to unravel the therapeutic potential of Formononetin combined with Salvianolic acid B (FcS) in modulating pancreatic cancer's impact on endothelial cells, illuminate the underlying mechanisms that govern this therapeutic interaction, thereby advancing strategies to normalize tumor vasculature and combat cancer progression. MATERIALS AND METHODS: A co-culture system involving Human Umbilical Vein Endothelial Cells (HUVECs) and PANC-1 cells was established to investigate the potential of targeting abnormal vasculature as a novel anti-tumor therapeutic strategy. We systematically compared HUVEC proliferation, migration, invasion, and lumenogenesis in both mono- and co-culture conditions with PANC-1 (H-P). Subsequently, FcS treatment of the H-P system was evaluated for its anti-angiogenic properties. Molecular docking was utilized to predict the interactions between Formononetin and Salvianolic acid B with RhoA, and the post-treatment expression of RhoA in HUVECs was assessed. Furthermore, we utilized shRhoA lentivirus to elucidate the role of RhoA in FcS-mediated effects on HUVECs. In vivo, a zebrafish xenograft tumor model was employed to assess FcS's anti-tumor potential, focusing on cancer cell proliferation, migration, apoptosis, and vascular development. RESULTS: FcS treatment demonstrated a significant, dose-dependent inhibition of PANC-1-induced alterations in HUVECs, including proliferation, migration, invasion, and tube formation capabilities. Molecular docking analyses indicated potential interactions between FcS and RhoA. Further, FcS treatment was found to downregulate RhoA expression and modulated the PI3K/AKT signaling pathway in PANC-1-induced HUVECs. Notably, the phenotypic inhibitory effects of FcS on HUVECs were attenuated by RhoA knockdown. In vivo zebrafish studies validated FcS's anti-tumor activity, inhibiting cancer cell proliferation, metastasis, and vascular sprouting, while promoting tumor cell apoptosis. CONCLUSIONS: This study underscores the promising potential of FcS in countering pancreatic cancer-induced endothelial alterations. FcS exhibits pronounced anti-abnormal vasculature effects, potentially achieved through downregulation of RhoA and inhibition of the PI3K/Akt signaling pathway, thereby presenting a novel therapeutic avenue for pancreatic cancer management.


Subject(s)
Benzofurans , Cell Movement , Human Umbilical Vein Endothelial Cells , Isoflavones , Pancreatic Neoplasms , rhoA GTP-Binding Protein , Isoflavones/pharmacology , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Animals , Benzofurans/pharmacology , rhoA GTP-Binding Protein/metabolism , Cell Line, Tumor , Human Umbilical Vein Endothelial Cells/drug effects , Cell Movement/drug effects , Neovascularization, Pathologic/drug therapy , Zebrafish , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Depsides
2.
Phytomedicine ; 134: 155990, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39243750

ABSTRACT

Diabetic foot ulcers (DFUs) represent a severe complication of diabetes mellitus. Ramulus Mori (Sangzhi) alkaloids (SZ-A), an approved oral medication for type 2 diabetes, have not been explored for their potential to enhance the processes involved in diabetic wound healing. This study aims to investigate SZ-A's role in diabetic wound healing mechanisms. The in vivo experimentation involves dividing the subjects into NC and SZ-A groups, with SZ-A dosed at 200 and 400 mg/kg, to assess the therapeutic efficacy of SZ-A. The results of the animal studies show that SZ-A intervention accelerates the processes of diabetic angiogenesis and wound healing in a manner dependent on its concentration. Additionally, a pathological model using advanced glycation end products (AGEs) in HUVECs demonstrates SZ-A's cytoprotective effect. In vitro, SZ-A intervention significantly increases cell proliferation, migration and tube formation, protecting HUVECs from oxidative stress injury induced by AGEs. Mechanistically, SZ-A exerts a protective effect on HUVECs from oxidative stress damage through the activation of the NRF2/HO-1/eNOS signaling pathway. The findings suggest that SZ-A exhibits considerable potential as a promising candidate for treating DFUs, which will aid in more effectively integrating plant-based therapies into clinical settings.


Subject(s)
Alkaloids , Diabetic Foot , Human Umbilical Vein Endothelial Cells , NF-E2-Related Factor 2 , Nitric Oxide Synthase Type III , Oxidative Stress , Wound Healing , Wound Healing/drug effects , NF-E2-Related Factor 2/metabolism , Animals , Humans , Alkaloids/pharmacology , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress/drug effects , Diabetic Foot/drug therapy , Human Umbilical Vein Endothelial Cells/drug effects , Male , Cell Proliferation/drug effects , Signal Transduction/drug effects , Rats, Sprague-Dawley , Diabetes Mellitus, Experimental/drug therapy , Glycation End Products, Advanced/metabolism , Cell Movement/drug effects , Rats
3.
Mol Biol Rep ; 51(1): 998, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39299971

ABSTRACT

BACKGROUND: Myricetin, a flavanol present in fruits, tea, and vegetables, has the potential to reduce chronic diseases like gastric cancer by promoting cell death and stopping cell growth. However, its limited bioactivity due to its short lifespan and poor solubility in water has been a challenge. The current research focuses on incorporating myricetin into alginate-cellulose hybrid nanocrystals to enhance its selective proapoptotic effects on human AGS gastric cancer cells. METHODS: MAC-NCs, myricetin-loaded alginate-cellulose hybrid nanocrystals, were synthesized using a combined co-precipitation/ultrasonic homogenization method and characterized through Dynamic Light Scattering (DLS), Fourier Transform Infrared Spectroscopy (FTIR), Field Emission Scanning Electron Microscope (FESEM), and Zeta-potential analyses. Their cytotoxic activity was tested on cancerous (AGS) and normal (Huvec) cells, revealing selective toxicity. Apoptotic markers, Caspase 8 and Caspase 9, gene expression was measured, and cell death type was confirmed using DAPI staining and flow cytometry on AGS cells. RESULTS: Synthesized MAC-NCs, measuring 40 nm, showed significant selective toxicity on human gastric cells (IC50 of 31.05 µg/mL) compared to normal endothelial cells (IC50 of 214.26 µg/mL). DAPI and annexin flow cytometry revealed increased apoptotic bodies in gastric cells, indicating apoptosis. However, the apoptosis was found to be independent of Caspase-8 and Caspase-9. CONCLUSION: The current study provides critical insights into the therapeutic potential of MAC-NCs for gastric cancer treatment. Based on the notable induction of apoptosis in the AGS cancer cell line, the synthesized MAC-NCs exhibit promising potential as a selective anti-gastric cancer agent. However, further in-vivo studies are necessary to confirm and quantify the nanoparticle's selective toxicity and pharmaceutical properties in future investigations.


Subject(s)
Alginates , Apoptosis , Cellulose , Flavonoids , Nanoparticles , Stomach Neoplasms , Humans , Alginates/chemistry , Alginates/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Apoptosis/drug effects , Nanoparticles/chemistry , Cell Line, Tumor , Cellulose/pharmacology , Cellulose/chemistry , Flavonoids/pharmacology , Caspase 9/metabolism , Caspase 9/genetics , Caspase 8/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Cell Survival/drug effects
4.
Phytomedicine ; 134: 155995, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39270591

ABSTRACT

BACKGROUND: Instead of completely suppressing blood vessels inside tumors, vascular normalization therapy is proposed to normalize and prune the abnormal vasculature in tumor microenvironment (TME) to acquire a normal and stable blood flow and perfusion. The theoretical basis for the use of "blood-activating and stasis-resolving" formulas in Traditional Chinese Medicine to treat cancer is highly consistent with the principle of vascular normalization therapy, suggesting the potential application of these traditional formulas in vascular normalization therapy. PURPOSE: To study the underlying mechanisms of a classical "blood-activating and stasis-resolving" formula, Taohong Siwu decoction (TSD), in enhancing the efficacy of chemotherapy for breast cancer treatment. STUDY DESIGN: HUVECs and transgenic zebrafish embryos were used as the major model in vitro. A 4T1 mouse breast cancer model was applied to study tumor vasculature normalization of TSD and the combination effects with DOX. RESULTS: Our data showed that TSD exhibited anti-angiogenic potential in HUVECs and transgenic zebrafish embryos. After 20 days treatment, TSD significantly normalized the tumor vasculature by remodeling vessel structure, reducing intratumoral hypoxia and vessel leakage, and promoting vessel maturation and blood perfusion in 4T1 breast tumor-bearing mice. Moreover, the anti-tumor efficacy of doxorubicin liposome in 4T1 breast tumors was significantly improved by TSD, including the suppression of tumor cell proliferation, angiogenesis, hypoxia, and the increase of cell apoptosis, which is likely through the vascular normalization induced by TSD. TSD also shifted the macrophage polarization from M2 to M1 phenotype in TME during the combination therapy, as evidenced by the reduced number of CD206+ macrophages and increased number of CD86+ macrophages. Additionally, TSD treatment protected against doxorubicin-induced cardiotoxicity in animals, as evidenced by the reduced cardiomyocytes apoptosis and improved heart function. CONCLUSION: This study demonstrated for the first time that TSD as a classical Chinese formula can enhance the drug efficacy and reduce the side effects of doxorubicin. These findings can support that TSD could be used as an adjuvant therapy in combination with conventional chemotherapy for the future breast cancer treatment.


Subject(s)
Doxorubicin , Drugs, Chinese Herbal , Human Umbilical Vein Endothelial Cells , Neovascularization, Pathologic , Zebrafish , Animals , Doxorubicin/pharmacology , Drugs, Chinese Herbal/pharmacology , Humans , Human Umbilical Vein Endothelial Cells/drug effects , Female , Mice , Neovascularization, Pathologic/drug therapy , Mice, Inbred BALB C , Animals, Genetically Modified , Tumor Microenvironment/drug effects , Breast Neoplasms/drug therapy , Cell Line, Tumor , Apoptosis/drug effects
5.
Sci Rep ; 14(1): 21852, 2024 09 19.
Article in English | MEDLINE | ID: mdl-39300121

ABSTRACT

5-Fluorouracil (5-FU) is used as a standard first-line drug for colorectal cancer malignancy (CRC), but it brings a series of side effects such as severe diarrhea and intestinal damage. Our previous study found that a large number of senescent cells increased while 5-Fu induced intestinal damage, and anti-senescence drugs can alleviate its side effects of inflammatory damage. Oleanolic acid (OA) is a common pentacyclic triterpenoid mainly derived from food fungi and medicinal plants, and studies have shown that it mainly possesses hepatoprotective, enzyme-lowering, anti-inflammatory, and anti-tumor effects. But its role in senescence is still unclear. In the present study, we demonstrated for the first time that OA ameliorated 5-Fu-induced human umbilical vein endothelial cells (HUVECs) and human normal intestinal epithelial cells (NCM460) in a 5-Fu-induced cellular senescence model by decreasing the activity of SA-ß-gal-positive cells, and the expression of senescence-associated proteins (p16), senescence-associated genes (p53 and p21), and senescence-associated secretory phenotypes (SASPs: IL-1ß, IL-6, IL-8, IFN-γ and TNF-α). Meanwhile, in this study, in a BALB/c mouse model, we demonstrated that 5-FU induced intestinal inflammatory response and injury, which was also found to be closely related to the increase of senescent cells, and that OA treatment was effective in ameliorating these adverse phenomena. Furthermore, our in vivo and in vitro studies showed that OA could alleviate senescence by inhibiting mTOR. In colon cancer cell models, OA also enhanced the ability of 5-FU to kill HCT116 cells and SW480 cells. Overall, this study demonstrates for the first time the potential role of OA in counteracting the side effects of 5-FU chemotherapy, providing a new option for the treatment of colorectal cancer to progressively achieve the goal of high efficacy and low toxicity of chemotherapy.


Subject(s)
Cellular Senescence , Fluorouracil , Human Umbilical Vein Endothelial Cells , Inflammation , Oleanolic Acid , Oleanolic Acid/pharmacology , Fluorouracil/adverse effects , Fluorouracil/pharmacology , Humans , Cellular Senescence/drug effects , Animals , Mice , Inflammation/drug therapy , Inflammation/pathology , Inflammation/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice, Inbred BALB C , Intestines/drug effects , Intestines/pathology , Male , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology
6.
Cell Mol Life Sci ; 81(1): 401, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39269632

ABSTRACT

Methylglyoxal (MGO), a reactive dicarbonyl metabolite of glucose, plays a prominent role in the pathogenesis of diabetes and vascular complications. Our previous studies have shown that MGO is associated with increased oxidative stress, inflammatory responses and apoptotic cell death in endothelial cells (ECs). Pyroptosis is a novel form of inflammatory caspase-1-dependent programmed cell death that is closely associated with the activation of the NOD-like receptor 3 (NLRP3) inflammasome. Recent studies have shown that sulforaphane (SFN) can inhibit pyroptosis, but the effects and underlying mechanisms by which SFN affects MGO-induced pyroptosis in endothelial cells have not been determined. Here, we found that SFN prevented MGO-induced pyroptosis by suppressing oxidative stress and inflammation in vitro and in vivo. Our results revealed that SFN dose-dependently prevented MGO-induced HUVEC pyroptosis, inhibited pyroptosis-associated biochemical changes, and attenuated MGO-induced morphological alterations in mitochondria. SFN pretreatment significantly suppressed MGO-induced ROS production and the inflammatory response by inhibiting the NLRP3 inflammasome (NLRP3, ASC, and caspase-1) signaling pathway by activating Nrf2/HO-1 signaling. Similar results were obtained in vivo, and we demonstrated that SFN prevented MGO-induced oxidative damage, inflammation and pyroptosis by reversing the MGO-induced downregulation of the NLRP3 signaling pathway through the upregulation of Nrf2. Additionally, an Nrf2 inhibitor (ML385) noticeably attenuated the protective effects of SFN on MGO-induced pyroptosis and ROS generation by inhibiting the Nrf2/HO-1 signaling pathway, and a ROS scavenger (NAC) and a permeability transition pore inhibitor (CsA) completely reversed these effects. Moreover, NLRP3 inhibitor (MCC950) and caspase-1 inhibitor (VX765) further reduced pyroptosis in endothelial cells that were pretreated with SFN. Collectively, these findings broaden our understanding of the mechanism by which SFN inhibits pyroptosis induced by MGO and suggests important implications for the potential use of SFN in the treatment of vascular diseases.


Subject(s)
Glucose , Human Umbilical Vein Endothelial Cells , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Oxidative Stress , Pyroptosis , Pyruvaldehyde , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/drug effects , Pyruvaldehyde/metabolism , Pyruvaldehyde/pharmacology , Humans , Oxidative Stress/drug effects , Inflammasomes/metabolism , Inflammasomes/drug effects , Animals , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Glucose/metabolism , Isothiocyanates/pharmacology , Mice , Sulfoxides/pharmacology , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Male , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mitochondria/metabolism , Mitochondria/drug effects
7.
Sci Rep ; 14(1): 20553, 2024 09 04.
Article in English | MEDLINE | ID: mdl-39232048

ABSTRACT

The plasma membrane protein caveolin-1 (CAV-1) regulates signaling by inhibiting a wide range of kinases and other enzymes. Our previous study demonstrated that the downregulation of CAV-1 in psoriatic epidermal cells contributes to inflammation by enhancing JAK/STAT signaling, cell proliferation, and chemokine production. Administration of the CAV-1 scaffolding domain (CSD) peptide suppressed imiquimod (IMQ)-induced psoriasis-like dermatitis. To identify an optimal therapeutic peptide derived from CAV-1, we have compared the efficacy of CSD and subregions of CSD that have been modified to make them water soluble. We refer to these modified peptides as sCSD, sA, sB, and sC. In IMQ-induced psoriasis-like dermatitis, while all four peptides showed major beneficial effects, sB caused the most significant improvements of skin phenotype and number of infiltrating cells, comparable or superior to the effects of sCSD. Phosphorylation of STAT3 was also inhibited by sB. Furthermore, sB suppressed angiogenesis both in vivo in the dermis of IMQ-induced psoriasis mice and in vitro by blocking the ability of conditioned media derived from CAV-1-silenced keratinocytes to inhibit tube formation by HUVEC. In conclusion, sB had similar or greater beneficial effects than sCSD not only by cytokine suppression but by angiogenesis inhibition adding to its ability to target psoriatic inflammation.


Subject(s)
Caveolin 1 , Cytokines , Imiquimod , Neovascularization, Pathologic , Psoriasis , STAT3 Transcription Factor , Psoriasis/drug therapy , Psoriasis/chemically induced , Psoriasis/pathology , Psoriasis/metabolism , Caveolin 1/metabolism , Animals , Mice , Cytokines/metabolism , Humans , STAT3 Transcription Factor/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Peptides/pharmacology , Peptides/chemistry , Skin/drug effects , Skin/metabolism , Skin/pathology , Keratinocytes/drug effects , Keratinocytes/metabolism , Disease Models, Animal , Water/chemistry , Solubility , Human Umbilical Vein Endothelial Cells/drug effects , Angiogenesis
8.
Biomed Pharmacother ; 179: 117418, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39265233

ABSTRACT

Fluoroquinolones, which are widely used antibiotics, have been linked to aortic disease, which prompted an FDA warning in 2018. Recent reports have challenged the perception that fluoroquinolones pose a significant risk for vascular diseases. This study aimed to investigate whether fluoroquinolones increase the risk of aortic diseases by focusing on the onset of aortic dissection. Levofloxacin (LVFX), a fluoroquinolone, was studied in vitro using cultured vascular cells and in vivo using a mouse model prone to aortic dissection. Risk of adverse drug events was analyzed using VigiBase, a global safety database, and a retrospective cohort analysis was conducted using the JMDC Claims database. LVFX resulted in endothelial cell injury and increased matrix metalloproteinases in vitro. However, in vivo studies showed no significant effect on elastin degradation or aortic dissection incidence. The effect of LVFX on endothelial injury was altered during the onset of dissection, exacerbating injury before onset but inhibiting it afterward. Safety database analysis showed no significant risk signals for aortic dissection associated with fluoroquinolones, which was supported by findings in the receipt database. Inconsistencies were observed in the in vitro and in vivo actions of fluoroquinolones and differences in their effects on aortic dissection and aneurysms. Despite cytotoxicity, the risk of aortic dissection was not significantly increased in clinical scenarios. Based on our findings, concerns regarding aortic diseases do not justify discontinuation of fluoroquinolone use. Further studies are needed to elucidate the conflicting actions of fluoroquinolones, taking into account background pathophysiology such as infection and inflammation.


Subject(s)
Aortic Dissection , Databases, Factual , Fluoroquinolones , Animals , Mice , Humans , Aortic Dissection/chemically induced , Fluoroquinolones/adverse effects , Male , Retrospective Studies , Levofloxacin/adverse effects , Mice, Inbred C57BL , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Female , Aortic Diseases/chemically induced , Human Umbilical Vein Endothelial Cells/drug effects , Aortic Aneurysm/chemically induced
9.
J Agric Food Chem ; 72(39): 21610-21623, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39292861

ABSTRACT

Fucoxanthin, a dietary carotenoid, is predominantly found in edible brown algae and is commonly consumed worldwide. Fucoxanthin has been shown to possess beneficial health activities such as antidiabetic, anti-inflammatory, antimutagenic, and antiobesity; however, the effects of fucoxanthin on VEGF-mediated angiogenesis and its possible binding with VEGF are unknown. Here, different lines of evidence supported the suppressive roles of fucoxanthin in VEGF-mediated angiogenesis. In human umbilical vein endothelial cells, fucoxanthin remarkedly suppressed VEGF-mediated cell proliferative, migration, and invasive abilities, as well as tube formation, without cytotoxicity. In addition, fucoxanthin inhibited the subintestinal vessel formation of zebrafish in vivo. In signaling cascades, fucoxanthin was proposed to interact with VEGF, thus attenuating VEGF's functions in activating the VEGF receptor and its related downstream signaling, i.e., phosphorylations of MEK and Erk. Fucoxanthin also significantly blocked VEGF-triggered ROS formation. Furthermore, the outcomes of applying fucoxanthin in cancer cells were identified, which included (i) inhibiting VEGF-mediated cell proliferation and migration and (ii) inhibiting NF-κB translocation via limiting MMP2 expression. These lines of investigations supported the antiangiogenic roles of fucoxanthin, as well as reviewing its signaling mechanisms, in blocking the VEGF-triggered responses. The results would benefit the potential development of fucoxanthin for the prevention and treatment of angiogenesis-related diseases.


Subject(s)
Cell Movement , Cell Proliferation , Human Umbilical Vein Endothelial Cells , Signal Transduction , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-2 , Xanthophylls , Zebrafish , Humans , Xanthophylls/pharmacology , Xanthophylls/chemistry , Signal Transduction/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Animals , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Cell Proliferation/drug effects , Cell Movement/drug effects , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry , Phaeophyceae/chemistry , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Angiogenesis
10.
Int Immunopharmacol ; 142(Pt B): 113060, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39321703

ABSTRACT

One component of the polycomb repressor complex 2 is histone methyltransferase zeste homolog 2 (EZH2), which is also called Enhancer of zeste homolog 2. It is considered a potential therapeutic target for inhibiting endothelial dysfunction.. Hence, directing efforts towards EZH2 to weaken endothelium damage and regulate vascular lesions proves to be a highly successful therapeutic approach for enhancing endothelial dysfunction. This study aimed to investigate the mechanism by which salidroside (SAL) improves hydrogen peroxide (H2O2)-induced endothelial dysfunction. The investigation involved the use of many techniques, including western blotting, real-time polymerase chain reaction (RT-PCR), a scratch test, molecular docking, and other methods. The experimental findings demonstrated that SAL has the ability to inhibit the impaired functioning of endothelial cells caused by H2O2 and decrease the levels of NF-κB p65, NLRP3, TNF-α, Beclin1, LC3, and P62 proteins. Additionally, there seems to be a targeting relationship between SAL and EZH2, and EZH2 knockdown can reproduce the protective effect of SAL on endothelial function. Overall, SAL inhibits H2O2-induced HUVEC dysfunction by regulating autophagy and inflammatory signaling pathways through EZH2.


Subject(s)
Autophagy , Enhancer of Zeste Homolog 2 Protein , Glucosides , Human Umbilical Vein Endothelial Cells , Hydrogen Peroxide , Phenols , Humans , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Hydrogen Peroxide/metabolism , Autophagy/drug effects , Glucosides/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Phenols/pharmacology , Inflammation/drug therapy , Signal Transduction/drug effects , Cells, Cultured , Molecular Docking Simulation , Anti-Inflammatory Agents/pharmacology
11.
Lipids Health Dis ; 23(1): 279, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227809

ABSTRACT

BACKGROUND: NOD-like receptor protein 3 (NLRP3) inflammasome activation is indispensable for atherogenesis. Mitophagy has emerged as a potential strategy to counteract NLRP3 inflammasome activation triggered by impaired mitochondria. Our previous research has indicated that dihydromyricetin, a natural flavonoid, can mitigate NLRP3-mediated endothelial inflammation, suggesting its potential to treat atherosclerosis. However, the precise underlying mechanisms remain elusive. This study sought to investigate whether dihydromyricetin modulates endothelial mitophagy and inhibits NLRP3 inflammasome activation to alleviate atherogenesis, along with the specific mechanisms involved. METHODS: Apolipoprotein E-deficient mice on a high-fat diet were administered daily oral gavages of dihydromyricetin for 14 weeks. Blood samples were procured to determine the serum lipid profiles and quantify proinflammatory cytokine concentrations. Aortas were harvested to evaluate atherosclerotic plaque formation and NLRP3 inflammasome activation. Concurrently, in human umbilical vein endothelial cells, Western blotting, flow cytometry, and quantitative real-time PCR were employed to elucidate the mechanistic role of mitophagy in the modulation of NLRP3 inflammasome activation by dihydromyricetin. RESULTS: Dihydromyricetin administration significantly attenuated NLRP3 inflammasome activation and vascular inflammation in mice on a high-fat diet, thereby exerting a pronounced inhibitory effect on atherogenesis. Both in vivo and in vitro, dihydromyricetin treatment markedly enhanced mitophagy. This enhancement in mitophagy ameliorated the mitochondrial damage instigated by saturated fatty acids, thereby inhibiting the activation and nuclear translocation of NF-κB. Consequently, concomitant reductions in the transcript levels of NLRP3 and interleukin-1ß (IL-1ß), alongside decreased activation of NLRP3 inflammasome and IL-1ß secretion, were discerned. Notably, the inhibitory effects of dihydromyricetin on the activation of NF-κB and subsequently the NLRP3 inflammasome were determined to be, at least in part, contingent upon its capacity to promote mitophagy. CONCLUSION: This study suggested that dihydromyricetin may function as a modulator to promote mitophagy, which in turn mitigates NF-κB activity and subsequent NLRP3 inflammasome activation, thereby conferring protection against atherosclerosis.


Subject(s)
Atherosclerosis , Diet, High-Fat , Flavonols , Human Umbilical Vein Endothelial Cells , Inflammasomes , Mitophagy , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Mitophagy/drug effects , Animals , Flavonols/pharmacology , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Atherosclerosis/pathology , Atherosclerosis/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Mice , Humans , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Diet, High-Fat/adverse effects , Male , Mice, Inbred C57BL , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Mitochondria/drug effects , Mitochondria/metabolism
12.
Int J Biol Macromol ; 279(Pt 3): 135329, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39236943

ABSTRACT

Open wounds present a significant challenge in healthcare, requiring careful management to prevent infection and promote wound healing. Advanced wound dressings are critical need to enhance their hemostatic capabilities, antimicrobial properties, and ability to support angiogenesis and sustained moisture for optimal healing. This study introduces a flexible hemostatic dressing designed for open wounds, integrating chitosan (CS) for hemostasis and biocompatibility, silk fibroin (SF) for mechanical strength, and montmorillonite (MMT) for enhanced drug transport. The CSSF@MMT dressings showed promising mechanical strength and swift hemostasis. The CIP-loaded CSSF@MMT demonstrated sustained release for up to one week, exhibiting antibacterial properties against both Gram-positive and Gram-negative bacteria. In vitro cell migration assay demonstrated that erythropoietin-loaded CSSF@MMT dressings promoted the proliferation and migration of endothelial cells. Similarly, the chick embryo chorioallantoic membrane study indicated the same dressings exhibited a significant increase in vascular regeneration. This research suggests that the CSSF@MMT sponge dressing, incorporated with CIP and erythropoietin, holds promise in effectively halting bleeding, creating a protective environment, diminishing inflammation, and fostering wound tissue regeneration. This potential makes it a significant advancement in open wound care, potentially lowering the need for limb amputation and decreasing wound care burden worldwide.


Subject(s)
Bandages , Bentonite , Chitosan , Fibroins , Hemostasis , Neovascularization, Physiologic , Wound Healing , Chitosan/chemistry , Chitosan/pharmacology , Fibroins/chemistry , Fibroins/pharmacology , Wound Healing/drug effects , Animals , Bentonite/chemistry , Bentonite/pharmacology , Humans , Neovascularization, Physiologic/drug effects , Hemostasis/drug effects , Cell Movement/drug effects , Chick Embryo , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Cell Proliferation/drug effects , Angiogenesis
13.
Int Immunopharmacol ; 142(Pt B): 113177, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39298820

ABSTRACT

BACKGROUND: Angiotensin 1-7 (Ang1-7) is the classical end product of angiotensin II, which has the effects of dilating blood vessels, protecting endothelial cells, anti-hypertension, improving cardiac function, and inhibiting atherosclerosis. We hypothesize that Ang1-7 inhibits human umbilical vein endothelial cells (HUVEC) ferroptosis through NF-κB/P53 signal pathway, and reduces extracorporeal membrane oxygenation (ECMO) vascular injury. METHODS: Cultured HUVEC were seeded into 15 wells and randomly divided into five groups: the control group and four experimental groups (erastin, erastin + Ang1-7, erastin + Ang1-7 + Betulinic acid, erastin + Betulinic acid). After stimulation, cell viability, lactate dehydrogenase (LDH), malondialdehyde (MDA), and superoxide dismutase (SOD) activity were measured. The effects of Ang1-7 on HUVEC microstructure, antioxidant enzymes (ferritin heavy chain 1 (FTH1), cystine/glutamic acid reverse transport solute carrier family 7 members 11 (SLC7A11 or XCT), superoxide dismutase-2 (SOD-2) and glutathione peroxidase 4 (GPX4)), NF-κB, P-NF-κB, P53, and P-P53). RESULTS: Erastin stimulation promoted HUVEC lipid peroxidation, decreased antioxidant enzyme expression, increased P-NF-κB, P53, and P-P53 expressions, and damaged HUVEC mitochondrial structure. Ang1-7 alleviated the effect of erastin on HUVEC, which was destroyed by Betulinic acid. CONCLUSION: Angiotensin1-7 pretreatment inhibited vascular endothelial cells' ferroptosis and alleviated ECMO vessel injury through NF-κB /P53 signal pathway.


Subject(s)
Angiotensin I , Extracorporeal Membrane Oxygenation , Ferroptosis , Human Umbilical Vein Endothelial Cells , Peptide Fragments , Humans , Ferroptosis/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Angiotensin I/pharmacology , Angiotensin I/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Vascular System Injuries/drug therapy , Vascular System Injuries/metabolism , Tumor Suppressor Protein p53/metabolism , Superoxide Dismutase/metabolism , Cells, Cultured , Cell Survival/drug effects
14.
Am J Physiol Cell Physiol ; 327(4): C1150-C1161, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39250819

ABSTRACT

In the era of immunotherapy, lenvatinib (LEN) still holds an important position in the sequential treatment of advanced hepatocellular carcinoma (HCC). However, the sustained therapeutic effect of LEN is not sufficient, and there is a need to address the development of resistance. Neuropilin-1 (NRP1) is known to act as a coreceptor for epidermal growth factor receptor (EGFR), Met, and vascular endothelial growth factor receptor 2 (VEGFR2), which have been reported to be involved in LEN resistance. In this study, we used cell culture and in vivo xenograft models to evaluate the contribution of NRP1 in the acquisition of LEN resistance in HCC as well as the potential of NRP1 as a therapeutic target. LEN resistance increased EGF/EGFR and hepatocyte growth factor (HGF)/Met signaling in liver cancer cells and VEGFA/VEGFR2 and HGF/Met signaling in vascular endothelial cells, thereby promoting cell proliferation, cell migration, and angiogenesis. We found that activation of NRP1 is essential for the enhancement of these signaling. In addition, NRP1 inhibition combined with LEN therapy synergistically improved the antitumor effects against LEN-resistant HCC, indicating that NRP1 is an attractive therapeutic target.NEW & NOTEWORTHY We demonstrated that neuropilin-1 (NRP1) was an essential coreceptor mediating the activation of multiple signaling pathways in the acquisition of resistance to lenvatinib (LEN) in HCC. The addition of NRP1 inhibition to LEN had a synergistic antitumor effect on LEN-resistant HCC in culture and in vivo xenograft models.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Drug Resistance, Neoplasm , Liver Neoplasms , Neovascularization, Pathologic , Neuropilin-1 , Phenylurea Compounds , Quinolines , Xenograft Model Antitumor Assays , Quinolines/pharmacology , Neuropilin-1/metabolism , Neuropilin-1/genetics , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/genetics , Phenylurea Compounds/pharmacology , Humans , Animals , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Drug Resistance, Neoplasm/drug effects , Cell Proliferation/drug effects , Signal Transduction/drug effects , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Mice, Nude , Mice , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Hepatocyte Growth Factor/metabolism , Angiogenesis Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Mice, Inbred BALB C , Hep G2 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Angiogenesis
15.
Int J Mol Sci ; 25(17)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39273200

ABSTRACT

Thrombosis is a key process that determines acute coronary syndrome and ischemic stroke and is the leading cause of morbidity and mortality in the world, together with cancer. Platelet adhesion and subsequent activation and aggregation are critical processes that cause thrombus formation after endothelial damage. To date, high hopes are associated with compounds of natural origin, which show anticoagulant action without undesirable effects and can be proposed as supportive therapies. We investigated the effect of the new combination of four natural compounds, escin-bromelain-ginkgo biloba-sage miltiorrhiza (EBGS), on the initial process of the coagulation cascade, which is the adhesion of platelets to activated vascular endothelium. Our results demonstrated that EBGS pretreatment of endothelial cells reduces platelet adhesion even in the presence of the monocyte-lymphocyte population. Our data indicate that EBGS exerts its effects by inhibiting the transcription of adhesion molecules, including P-selectin, platelet membrane glycoprotein GP1b, integrins αV and ß3, and reducing the secretion of the pro-inflammatory cytokines interleukin 6, interleukin 8, and the metalloproteinases MMP-2 and MMP-9. Furthermore, we demonstrated that EBGS inhibited the expression of focal adhesion kinase (FAK), strictly involved in platelet adhesion, and whose activity is correlated with that of integrin ß3. The results shown in this manuscript suggest a possible inhibitory role of the new combination EBGS in the reduction in platelet adhesion to activated endothelium, thus possibly preventing coagulation cascade initiation.


Subject(s)
Endothelium, Vascular , Platelet Adhesiveness , Signal Transduction , Tumor Necrosis Factor-alpha , Humans , Platelet Adhesiveness/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Blood Platelets/metabolism , Blood Platelets/drug effects , Salvia miltiorrhiza/chemistry , Focal Adhesion Kinase 1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Plant Extracts/pharmacology
16.
Int J Mol Sci ; 25(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39273228

ABSTRACT

Vascular aging is an important factor leading to cardiovascular diseases such as hypertension and atherosclerosis. Hyperlipidemia or fat accumulation may play an important role in vascular aging and cardiovascular disease. Isopropyl 3-(3,4-dihydroxyphenyl)-2-hydroxypropanoate (IDHP) has biological activity and can exert cardiovascular protection, which may be related to ferroptosis. However, the exact mechanism remains undefined. We hypothesized that IDHP may have a protective effect on blood vessels by regulating vascular aging caused by hyperlipidemia or vascular wall fat accumulation. The aim of this study is to investigate the protective effect and mechanism of IDHP on palmitic acid-induced human umbilical vein endothelial cells (HUVEC) based on senescence and ferroptosis. We found that IDHP could delay vascular aging, reduce the degree of ferrous ion accumulation and lipid peroxidation, and protect vascular cells from injury. These effects may be achieved by attenuating excessive reactive oxygen species (ROS) and ferroptosis signaling pathways generated in vascular endothelial cells. In short, our study identified IDHP as one of the antioxidant agents to slow down lipotoxicity-induced vascular senescence through the ROS/ferroptosis pathway. IDHP has new medicinal value and provides a new therapeutic idea for delaying vascular aging in patients with dyslipidemia.


Subject(s)
Cellular Senescence , Ferroptosis , Human Umbilical Vein Endothelial Cells , Palmitic Acid , Reactive Oxygen Species , Signal Transduction , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Reactive Oxygen Species/metabolism , Palmitic Acid/pharmacology , Cellular Senescence/drug effects , Ferroptosis/drug effects , Signal Transduction/drug effects , Lipid Peroxidation/drug effects , Antioxidants/pharmacology
17.
Exp Physiol ; 109(9): 1593-1603, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39092897

ABSTRACT

The purpose of this study was to determine the effect of circulating microvesicles isolated from chronic electronic (e-)cigarette users on cultured human umbilical vein endothelial cell (HUVEC) expression of nuclear factor-κB (NF-κB), cellular cytokine release, phosphorylation of endothelial nitric oxide synthase (eNOS) and NO production. The HUVECs were treated with microvesicles isolated via flow cytometry from nine non-tobacco users (five male and four female; 22 ± 2 years of age) and 10 e-cigarette users (six male and four female; 22 ± 2 years of age). Microvesicles from e-cigarette users induced significantly greater release of interleukin-6 (183.4 ± 23.6 vs. 150.6 ± 15.4 pg/mL; P = 0.002) and interleukin-8 (160.0 ± 31.6 vs. 129.4 ± 11.2 pg/mL; P = 0.01), in addition to expression of p-NF-κB p65 (Ser536) (18.8 ± 3.4 vs. 15.6 ± 1.5 a.u.; P = 0.02) from HUVECs compared with microvesicles from non-tobacco users. Nuclear factor-κB p65 was not significantly different between microvesicles from the non-tobacco users and from the e-cigarette users (87.6 ± 8.7 vs. 90.4 ± 24.6 a.u.; P = 0.701). Neither total eNOS (71.4 ± 21.8 vs. 80.4 ± 24.5 a.u.; P = 0.413) nor p-eNOS (Thr495) (229.2 ± 26.5 vs. 222.1 ± 22.7 a.u.; P = 0.542) was significantly different between microvesicle-treated HUVECs from non-tobacco users and e-cigarette users. However, p-eNOS (Ser1177) (28.9 ± 6.2 vs. 45.8 ± 9.0 a.u.; P < 0.001) expression was significantly lower from e-cigarette users compared with non-tobacco users. Nitric oxide production was significantly lower (8.2 ± 0.6 vs. 9.7 ± 0.9 µmol/L; P = 0.001) in HUVECs treated with microvesicles from e-cigarette users compared with microvesicles from non-tobacco users. This study demonstrated increased NF-κB activation and inflammatory cytokine production, in addition to diminished eNOS activity and NO production resulting from e-cigarette use. HIGHLIGHTS: What is the central question of this study? Circulating microvesicles contribute to cardiovascular health and disease via their effects on the vascular endothelium. The impact of electronic (e-)cigarette use on circulating microvesicle phenotype is not well understood. What is the main finding and its importance? Circulating microvesicles from e-cigarette users increase endothelial cell inflammation and impair endothelial nitric oxide production. Endothelial inflammation and diminished nitric oxide bioavailability are central factors underlying endothelial dysfunction and, in turn, cardiovascular disease risk. Deleterious changes in the functional phenotype of circulating microvesicles might contribute to the reported adverse effects of e-cigarette use on cardiovascular health.


Subject(s)
Cell-Derived Microparticles , Electronic Nicotine Delivery Systems , Human Umbilical Vein Endothelial Cells , Inflammation , NF-kappa B , Nitric Oxide Synthase Type III , Nitric Oxide , Humans , Nitric Oxide/metabolism , Male , Female , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Cell-Derived Microparticles/metabolism , Nitric Oxide Synthase Type III/metabolism , Young Adult , Inflammation/metabolism , NF-kappa B/metabolism , Vaping/adverse effects , Vaping/metabolism , Adult , Interleukin-6/metabolism , Cytokines/metabolism , Interleukin-8/metabolism , Cells, Cultured , Phosphorylation
18.
Int J Nanomedicine ; 19: 8661-8679, 2024.
Article in English | MEDLINE | ID: mdl-39193530

ABSTRACT

Purpose: Hydroxyapatite-based nanoparticles have found diverse applications in drug delivery, gene carriers, diagnostics, bioimaging and tissue engineering, owing to their ability to easily enter the bloodstream and target specific sites. However, there is limited understanding of the potential adverse effects and molecular mechanisms of these nanoparticles with varying geometries upon their entry into the bloodstream. Here, we used two commercially available hydroxyapatite nanoparticles (HANPs) with different geometries (less than 100 nm in size each) to investigate this issue. Methods: First, the particle size, Zeta potential, and surface morphology of nano-hydroxyapatite were characterized. Subsequently, the effects of 2~2000 µM nano-hydroxyapatite on the proliferation, migration, cell cycle distribution, and apoptosis levels of umbilical vein endothelial cells were evaluated. Additionally, the impact of nanoparticles of various shapes on the differential expression of genes was investigated using transcriptome sequencing. Additionally, we investigated the in vivo biocompatibility of HANPs through gavage administration of nanohydroxyapatite in mice. Results: Our results demonstrate that while rod-shaped HANPs promote proliferation in Human Umbilical Vein Endothelial Cell (HUVEC) monolayers at 200 µM, sphere-shaped HANPs exhibit significant toxicity to these monolayers at the same concentration, inducing apoptosis/necrosis and S-phase cell cycle arrest through inflammation. Additionally, sphere-shaped HANPs enhance SULT1A3 levels relative to rod-shaped HANPs, facilitating chemical carcinogenesis-DNA adduct signaling pathways in HUVEC monolayers. In vivo experiments have shown that while HANPs can influence the number of blood cells and comprehensive metabolic indicators in blood, they do not exhibit significant toxicity. Conclusion: In conclusion, this study has demonstrated that the geometry and surface area of HANPs significantly affect VEC survival status and proliferation. These findings hold significant implications for the optimization of biomaterials in cell engineering applications.


Subject(s)
Apoptosis , Cell Proliferation , Durapatite , Human Umbilical Vein Endothelial Cells , Nanoparticles , Particle Size , Durapatite/chemistry , Durapatite/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Nanoparticles/chemistry , Animals , Cell Proliferation/drug effects , Apoptosis/drug effects , Mice , Cell Movement/drug effects
19.
ACS Nano ; 18(36): 24909-24928, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39208278

ABSTRACT

This research introduces a vascular phenotypic and proteomic analysis (VPT) platform designed to perform high-throughput experiments on vascular development. The VPT platform utilizes an open-channel configuration that facilitates angiogenesis by precise alignment of endothelial cells, allowing for a 3D morphological examination and protein analysis. We study the effects of antiangiogenic agents─bevacizumab, ramucirumab, cabozantinib, regorafenib, wortmannin, chloroquine, and paclitaxel─on cytoskeletal integrity and angiogenic sprouting, observing an approximately 50% reduction in sprouting at higher drug concentrations. Precise LC-MS/MS analyses reveal global protein expression changes in response to four of these drugs, providing insights into the signaling pathways related to the cell cycle, cytoskeleton, cellular senescence, and angiogenesis. Our findings emphasize the intricate relationship between cytoskeletal alterations and angiogenic responses, underlining the significance of integrating morphological and proteomic data for a comprehensive understanding of angiogenesis. The VPT platform not only advances our understanding of drug impacts on vascular biology but also offers a versatile tool for analyzing proteome and morphological features across various models beyond blood vessels.


Subject(s)
Angiogenesis Inhibitors , Human Umbilical Vein Endothelial Cells , Proteomics , Humans , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Phenotype , Neovascularization, Physiologic/drug effects
20.
Eur J Pharm Biopharm ; 203: 114448, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39134098

ABSTRACT

Diabetic ulcers present a formidable obstacle in diabetes management, typically leading to high mortality and amputation rates. To overcome traditional monotherapy drawbacks, We developed a novel microneedle strategy for combined antimicrobial action: ingeniously integrating quercetin with Platelet-derived Growth Factor-BB(PDGF-BB) and Sucrose Octasulfate(SOS) into the microneedle system(QPS MN). This method allows to penetrate through biofilms, administering quercetin nanocrystals and PDGF-BB deep into the tissue to combat microbial infection, mitigate inflammation, and promote angiogenesis. The accompanying backing material contains SOS, which absorbs wound exudate and forms a dressing that provides a moist environment for wound healing In an in vitro wound-scratch assay demonstrated that co-cultivating Human Umbilical Vein Endothelial Cells(HUVEC) with QPS MN for 48 h (90.3 ± 2.51 %) significantly enhanced cell migration compared to the control group (20.2 ± 1.41 %). Moreover, treatment of streptozotocin-induced diabetic wounds in rats with QPS MN for 14 days resulted in a wound healing rate of 96.56 ± 3.44 %, far surpassing the healing rate of only 40.34 ± 7.26 % observed in the untreated control group. Furthermore, the QPS MN treated wounds exhibited a notable increase in skin appendages and neovascularisation, indicating promising potential for achieving complete wound healing. These results suggest that QPS MN may offer substantial therapeutic benefits for addressing diabetic wounds.


Subject(s)
Anti-Inflammatory Agents , Diabetes Mellitus, Experimental , Human Umbilical Vein Endothelial Cells , Needles , Wound Healing , Wound Healing/drug effects , Animals , Humans , Rats , Human Umbilical Vein Endothelial Cells/drug effects , Diabetes Mellitus, Experimental/drug therapy , Male , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Becaplermin/administration & dosage , Becaplermin/pharmacology , Quercetin/administration & dosage , Quercetin/pharmacology , Anti-Infective Agents/pharmacology , Anti-Infective Agents/administration & dosage , Neovascularization, Physiologic/drug effects , Nanoparticles/chemistry , Angiogenesis Inducing Agents/administration & dosage , Angiogenesis Inducing Agents/pharmacology , Rats, Sprague-Dawley , Cell Movement/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL