Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.614
Filter
1.
Int J Nanomedicine ; 19: 9145-9160, 2024.
Article in English | MEDLINE | ID: mdl-39258005

ABSTRACT

Background: Triple negative breast cancer (TNBC) is one of the worst prognosis types of breast cancer that urgently needs effective therapy methods. However, cancer is a complicated disease that usually requires multiple treatment modalities. Methods: A tumor microenvironment (TME)-responsive PFC/TRIM37@Fe-TA@HA (abbreviated as PTFTH) nanoplatform was constructed by coating Fe3+ and tannic acid (TA) on the surface of TRIM37-siRNA loaded phase-transition perfluorocarbon (PFC) nanodroplets and further modifying them with hyaluronic acid (HA) to achieve tumor-specific mild photothermal/gene/ferroptosis synergistic therapy (MPTT/GT/ Ferroptosis) in vitro. Once internalized into tumor cells through CD44 receptor-mediated active targeting, the HA shell of PTFTH would be preliminarily disassembled by hyaluronidase (HAase) to expose the Fe-TA metal-phenolic networks (MPNs), which would further degrade in response to an acidic lysosomal environment, leading to HAase/pH dual-responsive release of Fe3+ and PFC/TRIM37. Results: PTFTH showed good biocompatibility in vitro. On the one hand, the released Fe3+ could deplete the overexpressed glutathione (GSH) through redox reactions and produce Fe2+, which in turn converts endogenous H2O2 into highly cytotoxic hydroxyl radicals (•OH) for chemodynamic therapy (CDT). On the other hand, the local hyperthermia generated by PTFTH under 808 nm laser irradiation could not only improve CDT efficacy through accelerating the Fe2+-mediated Fenton reaction, but also enhance TRIM37-siRNA delivery for gene therapy (GT). The consumption of GSH and accumulation of •OH synergistically augmented intracellular oxidative stress, resulting in substantial tumor cell ferroptosis. Moreover, PTFTH possessed outstanding contrast enhanced ultrasound (CEUS), photoacoustic imaging (PAI) and magnetic resonance imaging (MRI) ability. Conclusion: This PTFTH based multiple-mode therapeutic strategy has successfully achieved a synergistic anticancer effect in vitro and has the potential to be translated into clinical application for tumor therapy in future.


Subject(s)
Ferroptosis , Glutathione , Hyaluronic Acid , Nanoparticles , Photothermal Therapy , RNA, Small Interfering , Tannins , Triple Negative Breast Neoplasms , Tumor Microenvironment , Humans , Ferroptosis/drug effects , Glutathione/metabolism , Glutathione/chemistry , Tumor Microenvironment/drug effects , Cell Line, Tumor , Tannins/chemistry , Tannins/pharmacology , Nanoparticles/chemistry , Hyaluronic Acid/chemistry , Female , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/genetics , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , RNA, Small Interfering/genetics , Photothermal Therapy/methods , Fluorocarbons/chemistry , Fluorocarbons/pharmacology , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Genetic Therapy/methods , Combined Modality Therapy/methods , Animals , Iron/chemistry , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/metabolism
2.
Clin Epigenetics ; 16(1): 130, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285429

ABSTRACT

BACKGROUND: Coronary heart disease (CHD) and stroke have become the leading cause of premature mortality and morbidity worldwide. Therefore, sensitive and accurate biomarkers for early detection of CHD and stroke are urgently needed for effective prevention and treatment. We aim to investigate the association between blood-based HYAL2 methylation and the risk of CHD and stroke in Chinese population. METHODS: In a prospective nested case-control study comprising 171 CHD cases, 139 stroke cases, who developed the diseases after recruitment and 356 controls who remained healthy during the 2.5 years of follow-up time, the methylation level of HYAL2 in the peripheral blood was quantified using mass spectrometry, and the association was calculated by logistic regression adjusted for covariant. RESULTS: Significant association between HYAL2 methylation in the peripheral blood and increased risk of preclinical CHD and stroke were identified [odds ratios (ORs) per - 10% methylation: 1.35-1.64, p ≤ 0.045 for HYAL2_CpG_1, HYAL2_CpG_2 and HYAL2_CpG_3 in CHD; ORs per - 10% methylation: 0.76-1.64, p ≤ 0.033 for HYAL2_CpG_2 and HYAL2_CpG_4 in stroke]. The association in CHD was further enhanced by female gender, younger age (< 70 years old), without the history of hypertension and cancer. The combination of four HYAL2 methylation sites showed an effective discrimination of CHD and stroke cases without hypertension from controls [area under curve (AUC) = 0.78 and 0.75, respectively]. CONCLUSIONS: This study presents a strong association of altered HYAL2 methylation in peripheral blood with preclinical CHD and stroke, providing a novel biomarker for risk assessment and early detection of cardiovascular diseases.


Subject(s)
Biomarkers , Coronary Disease , DNA Methylation , Hyaluronoglucosaminidase , Stroke , Humans , Male , Female , Middle Aged , DNA Methylation/genetics , Case-Control Studies , Stroke/genetics , Stroke/blood , Prospective Studies , Coronary Disease/genetics , Coronary Disease/blood , Coronary Disease/diagnosis , Aged , Biomarkers/blood , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/blood , China , GPI-Linked Proteins/genetics , GPI-Linked Proteins/blood , Early Diagnosis , Cell Adhesion Molecules
3.
BMC Oral Health ; 24(1): 1018, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39215323

ABSTRACT

BACKGROUND: Limiting postoperative edema, pain, trismus, and infection is crucial for smooth healing. This prospective, controlled clinical trial investigated and compared the effectiveness of dexamethasone and hyaluronidase in relieving these complications. METHODS: In groups Ia and IIa, 8 mg of dexamethasone and 150 IU of hyaluronidase were administered following the removal of impacted teeth, respectively. The contralateral sides (groups Ib and IIb) were determined as control groups. Edema, pain, trismus, and infection were clinically evaluated on the 1st, 2nd, 3rd, and 7th postoperative days. RESULTS: 60 patients were enrolled in the study. Hyaluronidase provided significantly more edema relief than dexamethasone on the 1st, 2nd, 3rd, and 7th postoperative days (P = 0.031, 0.002, 0.000, and 0.009, respectively). No statistical difference was found between dexamethasone and hyaluronidase in VAS and rescue analgesic intake amount values for all time points. Hyaluronidase was more effective in reducing trismus than dexamethasone on the 2nd and 3rd postoperative days (P = 0.029, 0.024, respectively). Neither of the agents significantly increased the postoperative infection rate. CONCLUSIONS: Hyaluronidase can be selected when postoperative excessive edema and trismus are anticipated. Dexamethasone may be a cost-effective option if postoperative pain control is merely targeted. TRIAL REGISTRATION: This trial was registered in the Clinical Trials Protocol Registration and Results System (ClinicalTrials.gov identifier number: NCT05466604) on 20/07/2022.


Subject(s)
Dexamethasone , Edema , Hyaluronoglucosaminidase , Molar, Third , Pain, Postoperative , Tooth, Impacted , Trismus , Humans , Dexamethasone/therapeutic use , Hyaluronoglucosaminidase/therapeutic use , Trismus/prevention & control , Edema/prevention & control , Pain, Postoperative/prevention & control , Pain, Postoperative/drug therapy , Molar, Third/surgery , Tooth, Impacted/surgery , Male , Female , Prospective Studies , Adult , Young Adult , Tooth Extraction/adverse effects , Anti-Inflammatory Agents/therapeutic use , Postoperative Complications/prevention & control , Pain Measurement , Adolescent , Surgical Wound Infection/prevention & control
5.
J Control Release ; 374: 369-383, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39089505

ABSTRACT

Hyaluronan (HA) is a glycosaminoglycan that forms a gel-like barrier in the subcutaneous (SC) space, limiting bulk fluid flow and the dispersion of SC-administered therapeutics. Recombinant human hyaluronidase PH20 (rHuPH20) facilitates the rapid delivery of co-administered therapeutics by depolymerizing HA in the SC space. Administration of rHuPH20 can induce the formation of anti-rHuPH20 antibodies, or anti-drug antibodies (ADAs), with the potential to bind endogenous PH20 hyaluronidase in the adult testes and epididymis. Using a variety of relevant animal models and multiple dose regimens of rHuPH20 across the full spectrum of animal development, we demonstrated that rHuPH20 administration resulted in the formation of ADAs. Although these ADAs can bind both the recombinant rHuPH20 enzyme and recombinant versions of animal model-specific hyaluronidases, they had no impact on fertility parameters (as measured by sperm concentration and motility, litter size, and litter viability) or fetal development. We present the result of our nonclinical studies in order of the developmental lifecycle, beginning with adults. Toxicology studies that extend beyond the standard package are also presented. These studies demonstrate the favorable safety profile of rHuPH20 and ADAs in nonclinical models. Additionally, we identified substantial safety margins for clinically relevant doses of rHuPH20.


Subject(s)
Hyaluronoglucosaminidase , Recombinant Proteins , Hyaluronoglucosaminidase/administration & dosage , Animals , Humans , Recombinant Proteins/administration & dosage , Antibodies/administration & dosage , Male , Female , Hyaluronic Acid/chemistry , Cell Adhesion Molecules
6.
Eur J Pharm Biopharm ; 203: 114474, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39191305

ABSTRACT

The stratum corneum of the skin presents the initial barrier to transdermal penetration. The dense structure of the extracellular matrix (ECM) further impedes local drug dispersion. Hyaluronidase (HAase) is a key component for the degradation of glycosidic bonding sites in hyaluronic acid (HA) within the ECM to overcome this barrier and enhance drug dispersion. HAase activity is optimal at 37-45 °C and in the pH range 4.5-5.5. Numerous FDA-approved formulations are available for the clinical treatment of extravasation and other diseases. HAase combined with various new nanoformulations can markedly improve intradermal dispersion. By degrading HA to create tiny channels that reduce the ECM density, these small nanoformulations then use these channels to deliver drugs to deeper layers of the skin. This deep penetration may increase local drug concentration or facilitate penetration into the blood or lymphatic circulation. Based on the generalization of 114 studies from 2010 to 2024, this article summarizes the most recent strategies to overcome the HAase-based ECM barrier for local drug delivery, discusses opportunities and challenges in clinical applications, and provides references for the future development of HAase. In the future, HAase-assisted topical administration is necessary to achieve systemic effects and to standardize HAase application protocols.


Subject(s)
Administration, Cutaneous , Drug Delivery Systems , Extracellular Matrix , Hyaluronic Acid , Hyaluronoglucosaminidase , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/metabolism , Extracellular Matrix/metabolism , Humans , Drug Delivery Systems/methods , Hyaluronic Acid/chemistry , Hyaluronic Acid/administration & dosage , Animals , Skin Absorption/drug effects , Skin/metabolism , Nanoparticles/chemistry
7.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167473, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39173890

ABSTRACT

BACKGROUND: Excessive activation of cardiac fibroblasts (CFs) significantly contributes to adverse cardiac remodeling post-myocardial infarction (MI). CEMIP, initially recognized as an enzyme involved in hyaluronic acid (HA) degradation, has also been implicated in the activation of pulmonary fibroblasts. Nevertheless, the role and mechanism of CEMIP in adverse cardiac remodeling following MI remain largely unexplored. MATERIALS AND METHODS: RNA sequencing (RNA-seq) was performed on cardiac tissue harvested from the infarct/peri-infarct region of mice 28 days post-MI. RNA-seq was conducted on primary cardiac fibroblasts (CFs) transfected with adenovirus overexpressing CEMIP. Adeno-associated virus serotype 9 (AAV9) was engineered for in vivo CEMIP knockdown to elucidate its impact on cardiac remodeling. Immunoprecipitation coupled with mass spectrometry (IP-MS) and co-immunoprecipitation (co-IP) were employed to elucidate the mechanism by which CEMIP affected cardiac remodeling. KEY FINDINGS: RNA-seq of fibrotic heart tissue at day 28 post-MI revealed a significant upregulation of CEMIP. In vitro, CEMIP facilitated the activation of cardiac fibroblasts. In vivo, knockdown of CEMIP markedly reduced cardiac fibrosis and improved cardiac function post-MI. IP-MS and co-immunoprecipitation (co-IP) confirmed that CEMIP interacted with TSP4 through the G8 domain. Further experiments confirmed that CEMIP promoted TSP4 degradation in lysosomes in an ACTN4-dependent manner, thereby activating the FAK signaling pathway. SIGNIFICANCE: Our findings suggest that CEMIP significantly contributes to cardiac remodeling post-MI, which might be a novel approach for treating cardiac fibrosis following MI.


Subject(s)
Myocardial Infarction , Thrombospondins , Ventricular Remodeling , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Mice , Ventricular Remodeling/genetics , Male , Thrombospondins/genetics , Thrombospondins/metabolism , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Signal Transduction , Gene Knockdown Techniques , Mice, Inbred C57BL , Humans , Fibrosis , Myocardium/metabolism , Myocardium/pathology , Disease Models, Animal
8.
Molecules ; 29(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125044

ABSTRACT

Eleutherococcus divaricatus (Siebold and Zucc.) S. Y. Hu. has been used in Traditional Chinese Medicine (TCM) due to its anticancer, immunostimulant, and anti-inflammatory activities. However, its mechanism of action and chemical composition are still insufficiently understood and require more advanced research, especially for cases in which anti-inflammatory properties are beneficial. The aim of this study was to evaluate the impact of E. divaricatus root extracts and fractions on proinflammatory serum hyaluronidase and tyrosinase in children diagnosed with acute lymphoblastic leukemia. Antioxidant and anti-melanoma activities were also examined and correlated with metabolomic data. For the first time, we discovered that the ethyl acetate fraction significantly inhibits hyaluronidase activity, with mean group values of 55.82% and 63.8% for aescin used as a control. However, interestingly, the fraction showed no activity against human tyrosinase, and in A375 melanoma cells treated with a doxorubicin fraction, doxorubicin activity decreased. This fraction exhibited the most potent antioxidant activity, which can be attributed to high contents of polyphenols, especially caffeic acid (24 mg/g). The findings suggest an important role of the ethyl acetate fraction in hyaluronidase inhibition, which may additionally indicate its anti-inflammatory property. The results suggest that this fraction can be used in inflammatory-related diseases, although with precautions in cases of patients undergoing chemotherapy.


Subject(s)
Acetates , Antioxidants , Eleutherococcus , Hyaluronoglucosaminidase , Melanoma , Monophenol Monooxygenase , Plant Extracts , Plant Roots , Hyaluronoglucosaminidase/antagonists & inhibitors , Hyaluronoglucosaminidase/metabolism , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/metabolism , Humans , Antioxidants/pharmacology , Antioxidants/chemistry , Plant Roots/chemistry , Plant Extracts/chemistry , Plant Extracts/pharmacology , Melanoma/drug therapy , Melanoma/metabolism , Acetates/chemistry , Eleutherococcus/chemistry , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry
9.
J Biochem Mol Toxicol ; 38(9): e23829, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39215765

ABSTRACT

KIAA1429 is an important 'writer' of the N6-methyladenine (m6A) modification, which is involved in tumour progression. This study was conducted to explore the mechanism of action of KIAA1429 in colon adenocarcinoma (COAD). KIAA1429-silenced COAD cell and xenograft tumour models were constructed, and the function of KIAA1429 was explored through a series of in vivo and in vitro assays. The downstream mechanisms of KIAA1429 were explored using transcriptome sequencing. Dimethyloxalylglycine (DMOG), an activator of HIF-1α, was used for feedback verification. The expression of KIAA1429 in COAD tumour tissues and cells was elevated, and KIAA1429 exhibited differential expression at different stages of the tumour. Silencing of KIAA1429 inhibited the proliferation, migration, and invasion of HT29 and HCT116 cells. The expression levels of NLRP3, GSDMD and Caspase-1 were decreased in KIAA1429-silenced HT29 cells, indicating the pyroptotic activity was inhibited. Additionally, KIAA1429 silencing inhibited the growth of tumour xenograft. Transcriptome sequencing and reverse transcription quantitative polymerase chain reaction revealed that after KIAA1429 silencing, the expression of AKR1C1, AKR1C2, AKR1C3 and RDH8 was elevated, and the expression of VIRMA, GINS1, VBP1 and ARF3 was decreased. In HT29 cells, KIAA1429 silencing blocked the HIF-1 signalling pathway, accompanied by the decrease in AKT1 and HIF-1α protein levels. The activation of HIF-1 signalling pathway, mediated by DMOG, reversed the antitumour role of KIAA1429 silencing. KIAA1429 silencing inhibits COAD development by blocking the HIF-1 signalling pathway.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Methyltransferases , Signal Transduction , Humans , Colonic Neoplasms/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Animals , Mice , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Methyltransferases/metabolism , Methyltransferases/genetics , HT29 Cells , Mice, Nude , Gene Silencing , Male , HCT116 Cells , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice, Inbred BALB C , Female , Disease Progression , Xenograft Model Antitumor Assays , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1/genetics , Hyaluronoglucosaminidase
10.
Toxins (Basel) ; 16(8)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39195737

ABSTRACT

Ananteris is a scorpion genus that inhabits dry and seasonal areas of South and Central America. It is located in a distinctive morpho-group of Buthids, the 'Ananteris group', which also includes species distributed in the Old World. Because of the lack of information on venom composition, the study of Ananteris species could have biological and medical relevance. We conducted a venomics analysis of Ananteris platnicki, a tiny scorpion that inhabits Panama and Costa Rica, which shows the presence of putative toxins targeting ion channels, as well as proteins with similarity to hyaluronidases, proteinases, phospholipases A2, members of the CAP-domain family, and hemocyanins, among others. Venom proteolytic and hyaluronidase activities were corroborated. The determination of the primary sequences carried out by mass spectrometry evidences that several peptides are similar to the toxins present in venoms from Old World scorpion genera such as Mesobuthus, Lychas, and Isometrus, but others present in Tityus and Centruroides toxins. Even when this venom displays the characteristic protein families found in all Buthids, with a predominance of putative Na+-channel toxins and proteinases, some identified partial sequences are not common in venoms of the New World species, suggesting its differentiation into a distinctive group separated from other Buthids.


Subject(s)
Scorpion Venoms , Scorpions , Costa Rica , Panama , Animals , Scorpion Venoms/chemistry , Amino Acid Sequence , Hyaluronoglucosaminidase/metabolism , Molecular Sequence Data
11.
ACS Nano ; 18(34): 23497-23507, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39146387

ABSTRACT

Colorectal cancer (CRC) is a major global health concern, and the development of effective treatment strategies is crucial. Enzyme prodrug therapy (EPT) shows promise in combating tumors but faces challenges in achieving sustained expression of therapeutic enzymes and optimal biological distribution. To address these issues, a fungi-triggered in situ chemotherapeutics generator (named as SC@CS@5-FC) was constructed via oral delivery of a prodrug (5-fluorocytosine, 5-FC) for the treatment of orthotopic colorectal tumor. When SC@CS@5-FC targets the tumor through tropism by Saccharomyces cerevisiae (SC), the chemotherapeutic generator could be degraded under abundant hyaluronidase (HAase) in the tumor microenvironment by an enzyme-responsive gate to release prodrug (5-FC). And nontoxic 5-FC was catalyzed to toxic chemotherapy drug 5-fluorouracil (5-FU) by cytosine deaminase (CD) of SC. Meanwhile, SC and zinc-coordinated chitosan nanoparticles could be used as immune adjuvants to activate antigen-presenting cells and further enhance the therapeutic effect. Our results demonstrated that SC@CS@5-FC could effectively inhibit tumor growth and prolong mouse survival in an orthotopic colorectal cancer model. This work utilizes living SC as a dynamotor and positioning system for the chemotherapeutic generator SC@CS@5-FC, providing a strategy for oral enzyme prodrug therapy for the treatment of orthotopic colorectal.


Subject(s)
Colorectal Neoplasms , Flucytosine , Fluorouracil , Immunotherapy , Prodrugs , Saccharomyces cerevisiae , Prodrugs/chemistry , Prodrugs/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Animals , Mice , Humans , Flucytosine/pharmacology , Flucytosine/chemistry , Administration, Oral , Fluorouracil/pharmacology , Fluorouracil/chemistry , Fluorouracil/administration & dosage , Cytosine Deaminase/metabolism , Chitosan/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Hyaluronoglucosaminidase/metabolism , Mice, Inbred BALB C , Nanoparticles/chemistry , Drug Screening Assays, Antitumor
12.
Mol Immunol ; 174: 69-76, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39216237

ABSTRACT

Immune checkpoint inhibitor-based cancer immunotherapy has shown promise as a potential treatment in the clinic. It has been reported that anti-PD-L1 combined with cisplatin treatment can improve the antitumor effect. However, the therapeutic outcome is limited due to the abundance of tumor stroma in pancreatic cancer (PC), which prevented the penetration of cisplatin and anti-PD-L1 into tumor regions, thus impeding the effectiveness in the treatment of PC. In this study, a nanocarrier-mediated codelivery system of hyaluronidase and cisplatin was constructed, which can degrade the stroma and promote cisplatin and anti-PD-L1 to penetrate the tumor stroma into the deep tumor, so as to suppress PC effectively. When combined the cisplatin nanocarrier system BPEI-SS-Pt/HAase@CaP (BSP/H@CaP) with an immune checkpoint inhibitor to overcome the poor therapeutic outcome of PC, the results indicated that the therapeutic effect of BSP/H@CaP combined with anti-PD-L1 was better than that of BSP/H@CaP and single anti-PD-L1 group. Because the stroma is degrading, a higher amount of BPEI-SS-Pt and anti-PD-L1 can enter the tumor stroma and reach the inner depths of the tumor for immune stimulation, leading to a synergistically augmented chemotherapy and immunotherapy for PC. The above combination therapy is useful for clinical translation to overcome the treatment resistance of matrix-rich PC.


Subject(s)
B7-H1 Antigen , Cisplatin , Immune Checkpoint Inhibitors , Pancreatic Neoplasms , Cisplatin/pharmacology , Cisplatin/administration & dosage , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/administration & dosage , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Animals , Humans , Mice , Cell Line, Tumor , Drug Carriers/chemistry , Nanoparticles , Immunotherapy/methods , Hyaluronoglucosaminidase , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Female , Mice, Inbred BALB C
13.
Int J Rheum Dis ; 27(8): e15282, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39091178

ABSTRACT

OBJECTIVE: To investigate the impact of IGJ on the proliferation, inflammation, and motility of rheumatoid arthritis (RA) fibroblast-like synoviocytes and elucidate the underlying mechanism. METHODS: The expression of IGJ RA fibroblast-like synoviocytes was assessed using immunoblot and qPCR. Cell growth was evaluated using CCK-8 and FCM assays. The effects on inflammatory response were determined by ELISA and immunoblot assays. Cell motility was assessed using transwell and immunoblot assays. The mechanism was further confirmed using immunoblot assays. RESULTS: IGJ expression was found to be elevated in fibroid synovial cells of RA. IGJ ablation inhibited the growth of MH7A cells and suppressed the inflammatory response. Knockdown of IGJ also blocked cell motility. Mechanically, the knockdown of IGJ suppressed the NF-κB axis in MH7A cells. CONCLUSION: IGJ suppresses RA in fibroblast-like synoviocytes via NF-κB pathway.


Subject(s)
Arthritis, Rheumatoid , Cell Movement , Cell Proliferation , Fibroblasts , NF-kappa B , Signal Transduction , Synoviocytes , Humans , Synoviocytes/metabolism , Synoviocytes/pathology , Synoviocytes/drug effects , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/immunology , NF-kappa B/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Cells, Cultured , Cell Line , Hyaluronoglucosaminidase
14.
Biomed Pharmacother ; 178: 117261, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39106708

ABSTRACT

BACKGROUND: Long-term anti-angiogenesis leads to pruned vasculature, densely deposited extracellular matrix (ECM), and consequently reduced chemotherapy delivery in esophagogastric cancer (EGC). To address this issue, we evaluated the efficacy of adding a hyaluronidase or a NO-donor to the regimen of chemotherapy and anti-angiogenic drugs. METHODS: A patient-derived EGC xenograft model was developed. Grafted mice were randomly assigned to four experimental groups and one control group. The experimental groups received DC101, a murine angiogenesis inhibitor, and nab-paclitaxel (NPTX), with the addition of hyaluronidase (PEGPH20), or NO-donor (nitroglycerine, NTG), or their combination, respectively. We compared tumor growth during 17 days of treatment. We performed immunohistochemistry for ECM components hyaluronan (HA) and collagen, CD31 for endothelial cells, and γH2AX for DNA damage. The positively stained areas were quantified, and vessel diameters were measured using QuPath software. RESULTS: Prolonged DC101 treatment induced deposition of HA (p<0.01) and collagen (p<0.01). HA was effectively degraded by PEGPH20 (p<0.001), but not by NTG as expected. Both PEGPH20 (p<0.05) and NTG (p<0.01) dilated vessels collapsed in response to long-term DC101 treatment. However, only PEGPH20 (rather than NTG) was found to significantly inhibit tumor growth (p<0.05) in combination with NPTX and DC101. CONCLUSIONS: These findings suggest that the mechanical barrier of HA is the major reason responsible for the resistance developed during prolonged anti-angiogenesis in EGC. Incorporating PEGPH20 into the existing treatment regimen is promising to improve outcomes for patients with EGC.


Subject(s)
Albumins , Angiogenesis Inhibitors , Esophageal Neoplasms , Hyaluronoglucosaminidase , Neovascularization, Pathologic , Paclitaxel , Stomach Neoplasms , Xenograft Model Antitumor Assays , Animals , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use , Hyaluronoglucosaminidase/administration & dosage , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Humans , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/administration & dosage , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Albumins/pharmacology , Albumins/administration & dosage , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Mice , Hyaluronic Acid/pharmacology , Mice, Nude , Female , Angiogenesis , Antibodies, Monoclonal
15.
J Clin Immunol ; 44(8): 181, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39158670

ABSTRACT

Facilitated subcutaneous immunoglobulin (fSCIG) 10% is an immunoglobulin replacement therapy that utilizes recombinant human hyaluronidase (rHuPH20) to enhance immunoglobulin dispersion and absorption, allowing for longer treatment intervals similar to intravenous immunoglobulin (up to once monthly). fSCIG 10% is indicated in the USA for treating adults and children aged ≥ 2 years with primary immunodeficiency diseases (PIDs). This prospective, non-interventional, open-label, multicenter, post-authorization safety study (NCT02593188) was conducted in the USA from November 2015 to October 2021 to assess the long-term safety of fSCIG 10% in routine clinical practice. Patients with PIDs aged ≥ 16 years who were prescribed and/or had started fSCIG 10% treatment were enrolled. In total, 253 patients were enrolled and included (full analysis set). Participants received fSCIG 10% treatment for a median (interquartile range) of 10.0 (3.5-11.8) months, with the majority of infusions administered every 4 weeks (54.4% [1197/2201 infusions]) and at home (62.6% [1395/2230 infusions]). Overall, 98.5% of infusions were administered without rate reduction, interruption, or discontinuation due to adverse events (AEs). Treatment-related, non-serious AEs were experienced by 52 patients (20.6%, 284 events). Two patients (0.8%) each experienced one treatment-related serious AE (aseptic meningitis and deep vein thrombosis). Development of antibodies against rHuPH20 was uncommon; 14/196 patients (7.1%) tested positive for binding antibodies (titer ≥ 1:160) with no neutralizing antibodies detected. There was no relationship between anti-rHuPH20 antibody positivity and the occurrence of treatment-related serious or non-serious AEs. Long-term, repeated self-administration of fSCIG 10% was well tolerated in US clinical practice by patients with PIDs.


Subject(s)
Hyaluronoglucosaminidase , Humans , Male , Female , United States , Adult , Adolescent , Prospective Studies , Hyaluronoglucosaminidase/therapeutic use , Hyaluronoglucosaminidase/administration & dosage , Primary Immunodeficiency Diseases/drug therapy , Middle Aged , Infusions, Subcutaneous , Child , Young Adult , Immunoglobulins/administration & dosage , Immunoglobulins/adverse effects , Immunoglobulins/therapeutic use , Injections, Subcutaneous , Treatment Outcome , Aged , Child, Preschool , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/therapy
16.
J Nanobiotechnology ; 22(1): 484, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138477

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a progressive and debilitating inflammatory disease of the gastrointestinal tract (GIT). Despite recent advances, precise treatment and noninvasive monitoring remain challenging. METHODS: Herein, we developed orally-administered, colitis-targeting and hyaluronic acid (HA)-modified, core-shell curcumin (Cur)- and cerium oxide (CeO2)-loaded nanoprobes (Cur@PC-HA/CeO2 NPs) for computed tomography (CT) imaging-guided treatment and monitoring of IBD in living mice. RESULTS: Following oral administration, high-molecular-weight HA maintains integrity with little absorption in the upper GIT, and then actively accumulates at local colitis sites owing to its colitis-targeting ability, leading to specific CT enhancement lasting for 24 h. The retained NPs are further degraded by hyaluronidase in the colon to release Cur and CeO2, thereby exerting anti-inflammatory and antioxidant effects. Combined with the ability of NPs to regulate intestinal flora, the oral NPs result in substantial relief in symptoms. Following multiple treatments, the gradually decreasing range of the colon with high CT attenuation correlates with the change in the clinical biomarkers, indicating the feasibility of treatment response and remission. CONCLUSION: This study provides a proof-of-concept for the design of a novel theranostic integration strategy for concomitant IBD treatment and the real-time monitoring of treatment responses.


Subject(s)
Cerium , Curcumin , Hyaluronic Acid , Inflammatory Bowel Diseases , Nanoparticles , Theranostic Nanomedicine , Animals , Inflammatory Bowel Diseases/drug therapy , Mice , Cerium/chemistry , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/therapeutic use , Theranostic Nanomedicine/methods , Administration, Oral , Nanoparticles/chemistry , Hyaluronic Acid/chemistry , Hyaluronoglucosaminidase/metabolism , Tomography, X-Ray Computed , Mice, Inbred C57BL , Colon/diagnostic imaging , Colon/pathology , Colon/metabolism , Humans , Colitis/drug therapy
17.
PLoS One ; 19(8): e0308370, 2024.
Article in English | MEDLINE | ID: mdl-39121049

ABSTRACT

Human hyaluronidase 1 (HYAL1) and PH20 play vital roles in degrading hyaluronic acids through the substrate-assisted double displacement mechanism. While HYAL1, a lysosomal enzyme, functions optimally under acidic conditions, PH20, a sperm surface hyaluronidase, displays a broader pH range, from acidic to neutral. Our objective was to extend HYAL1's pH range towards neutral pH by introducing repulsive charge-charge interactions involving the catalytic Glu131, increasing its pKa as the proton donor. Substituting individual acidic residues in the ß3-loop (S77D), ß3'-ß3″ hairpin (T86D and P87E), and at Ala132 (A132D and A132E) enabled HYAL1 to demonstrate enzyme activity at pH 7, with the mutants S77D, P87E, and A132E showing the highest activity in the substrate gel assay. However, double and triple substitutions, including S77D/T86D/A132E as found in the PH20 configuration, did not result in enhanced activity compared to single substitutions. Conversely, PH20 mutants with non-acidic substitutions, such as D94S in the ß3-loop and D103T in the ß3'-ß3″ hairpin, significantly reduced activity within the pH range of 4 to 7. However, the PH20 mutant E149A, reciprocally substituted compared to A132E in HYAL1, exhibited activity similar to PH20 wild-type (WT) at pH 7. In a turbidimetric assay, HYAL1 mutants with single acidic substitutions exhibited activity similar to that of PH20 WT at pH 7. These results suggest that substituting acidic residues near Glu131 results in HYAL1 activity at neutral pH through electrostatic repulsion. This study highlights the significance of charge-charge interactions in both HYAL1 and PH20 in regulating the pH-dependent activity of hyaluronidases.


Subject(s)
Hyaluronoglucosaminidase , Humans , Amino Acid Substitution , Catalytic Domain , Cell Adhesion Molecules , Glutamic Acid/metabolism , Glutamic Acid/chemistry , Hyaluronic Acid/metabolism , Hyaluronic Acid/chemistry , Hyaluronoglucosaminidase/chemistry , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/genetics , Hydrogen-Ion Concentration , Models, Molecular , Mutation
18.
Plant Foods Hum Nutr ; 79(3): 693-699, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39001986

ABSTRACT

Apple pomace is the residue left after apples are squeezed. The majority of pomace produced worldwide is produced by the apple manufacturing industry, however, most of the pomace produced by the industry is discarded. Apple pomace contains functional ingredients, such as polyphenols and triterpenoids, and exerts several beneficial effects on human health; however, studies on its cosmetic effects on the skin are lacking. Therefore, herein, we investigated the effects of apple pomace extract (APE) on human skin fibroblasts (HSFs) in vitro. When HSFs were cultured with the extract for 72 h, the number of HSFs increased at concentrations of 10 and 20 µg/mL. Transcriptome analysis and reverse transcription-quantitative PCR results revealed that the extract upregulated the expression of hyaluronan synthase (HAS) 1, HAS2, and HAS3 and downregulated the expression of HYAL1, a gene encoding the hyaluronan-degrading enzyme, in HSFs. Additionally, enzyme-linked immunosorbent assay revealed increased amounts of factors related to skin extracellular matrix, such as type I collagen and hyaluronic acid, secreted in the culture supernatant. The western blotting results suggested that the extract induced extracellular signal-regulated kinase and protein kinase B phosphorylation in HSFs. Additionally, several GO_Terms related to mitosis were detected in the Gene Ontology analysis. This is the first study to show that APE induces the proliferation of HSFs and production of factors related to skin anti-aging.


Subject(s)
Cell Proliferation , Collagen Type I , Fibroblasts , Hyaluronan Synthases , Hyaluronic Acid , Malus , Plant Extracts , Skin , Humans , Malus/chemistry , Fibroblasts/drug effects , Fibroblasts/metabolism , Hyaluronic Acid/metabolism , Cell Proliferation/drug effects , Plant Extracts/pharmacology , Hyaluronan Synthases/metabolism , Hyaluronan Synthases/genetics , Skin/cytology , Skin/drug effects , Skin/metabolism , Collagen Type I/metabolism , Collagen Type I/genetics , Cells, Cultured , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/genetics , Phosphorylation , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , Glucuronosyltransferase/metabolism , Glucuronosyltransferase/genetics
19.
J Pediatr Urol ; 20(5): 898-903, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39079875

ABSTRACT

INTRODUCTION: Phimosis is defined as the inability to retract the foreskin, preventing partial or complete exposure of the glans. OBJECTIVES: To compare the efficacy of topical treatment with betamethasone alone and in combination with hyaluronidase, evaluate systemic absorption of cortisol, and identify factors that predispose the success of topical treatment of phimosis in children aged 3-10 years. METHODS: This randomized double-blinded clinical trial involved 152 participants (3-10 years old) with phimosis. The children were divided into two groups: betamethasone associated with hyaluronidase (betamethasone valerate 2.5 mg + hyaluronidase 150 UTR; Group A) and betamethasone (betamethasone valerate 2.5 mg; Group B). Parents were instructed on how to use the ointment (twice a day, after hygiene, for 60 days) and on collecting salivary cortisol measurements at 11pm and 9am, before and after treatment. Participants were evaluated after 30 and 60 days. Fisher's exact test and paired t-test were used to analyze the data. RESULTS: Ninety children (69.77%) were successfully treated with the proposed treatment, with the success rate for Group A being 75.38% versus 64.06% for Group B, p = 0.18). Systemic absorption evaluated by salivary cortisol did not show differences after the intervention (p > 0.05), indicating that there was no systemic absorption when using ointments with or without hyaluronidase. The factors of age (OR = 0.98 - CI: 0.97-1.00), adherence (OR = 1.49 - CI: 0.53-4.16), balanoposthitis (OR = 1.85 - CI: 0.47-7.19), and previous use of corticosteroids (OR = 1.21 - CI: 0.53-2.72) also did not show influenced results CONCLUSION: Topical therapy for true phimosis with betamethasone 0.2% + hyaluronidase, despite showing no differences when compared with betamethasone 0.2% alone, for a period of up to 60 days, proved to be safe, effective, and with good results. The variables analyzed could not predict the expected clinical response. REBEC: RBR-76bhgyb.


Subject(s)
Administration, Topical , Betamethasone , Drug Therapy, Combination , Glucocorticoids , Hyaluronoglucosaminidase , Phimosis , Humans , Hyaluronoglucosaminidase/administration & dosage , Double-Blind Method , Male , Phimosis/drug therapy , Child, Preschool , Child , Treatment Outcome , Glucocorticoids/administration & dosage , Betamethasone/administration & dosage , Betamethasone/analogs & derivatives , Hydrocortisone/administration & dosage , Hydrocortisone/analogs & derivatives
20.
Arch Biochem Biophys ; 759: 110098, 2024 09.
Article in English | MEDLINE | ID: mdl-39009271

ABSTRACT

Mouse transmembrane protein 2 (mTMEM2) has been identified as a hyaluronidase, which has extracellularly G8 and GG domains and PbH1 repeats; however, our previously study showed that human TMEM2 (hTMEM2) is not a catalytic hyaluronidase due to the absence of the critical amino acid residues (His248/Ala303) in the GG domain. Naked mole-rats (NMRs) accumulate abundant high-molecular weight hyaluronan (HA) in their tissues, suggesting decreased HA degradation. Therefore, we aimed to evaluate the HA-degrading activity of NMR TMEM2 (nmrTMEM2) and compare it with those of mTMEM2 and hTMEM2. The amino acid residues of nmrTMEM2 (Asn247/Val302) are similar to Asn248/Phe303 of hTMEM2, and nmrTMEM2-expressing HEK293T cells showed negligible activity. We confirmed the significance of these amino acid residues using an inactive chimeric TMEM2 with the human GG domain, which acquired catalytic activity when Asn248/Phe303 was substituted with His248/Ala303. Semi-quantitative comparison of the activities of the membrane-fractions derived from m/h/nmrTMEM2-expressing HEK293T cells revealed that at least 20- and 14-fold higher amounts of nmr/hTMEM2 were required to degrade HA to the same extent as by mTMEM2. Thus, unlike mTMEM2, nmrTMEM2 is not a physiological hyaluronidase. The inability of nmrTMEM2 to degrade HA might partially account for the high-molecular-weight HA accumulation in NMR tissues.


Subject(s)
Hyaluronic Acid , Hyaluronoglucosaminidase , Membrane Proteins , Mole Rats , Humans , Hyaluronic Acid/metabolism , Animals , HEK293 Cells , Mole Rats/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/chemistry , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/chemistry , Amino Acid Sequence , Mice , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL