Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.565
Filter
2.
ACS Biomater Sci Eng ; 10(7): 4400-4410, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38917429

ABSTRACT

Tympanic membrane perforation (TMP) is prevalent in clinical settings. Patients with TMPs often suffer from infections caused by Staphylococcus aureus and Pseudomonas aeruginosa, leading to middle ear and external ear canal infections, which hinder eardrum healing. The objective of this study is to fabricate an enzyme-responsive antibacterial electrospun scaffold using poly(lactic-co-glycolic acid) and hyaluronic acid for the treatment of infected TMPs. The properties of the scaffold were characterized, including morphology, wettability, mechanical properties, degradation properties, antimicrobial properties, and biocompatibility. The results indicated that the fabricated scaffold had a core-shell structure and exhibited excellent mechanical properties, hydrophobicity, degradability, and cytocompatibility. Furthermore, in vitro bacterial tests and ex vivo investigations on eardrum infections suggested that this scaffold possesses hyaluronidase-responsive antibacterial properties. It may rapidly release antibiotics when exposed to the enzyme released by S. aureus and P. aeruginosa. These findings suggest that the scaffold has great potential for repairing TMPs with infections.


Subject(s)
Anti-Bacterial Agents , Hyaluronic Acid , Hyaluronoglucosaminidase , Polylactic Acid-Polyglycolic Acid Copolymer , Pseudomonas aeruginosa , Staphylococcus aureus , Tissue Scaffolds , Tympanic Membrane , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/chemistry , Staphylococcus aureus/drug effects , Tissue Scaffolds/chemistry , Pseudomonas aeruginosa/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Animals , Humans , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacology , Lactic Acid/chemistry , Lactic Acid/pharmacology , Tympanic Membrane Perforation/drug therapy , Tympanic Membrane Perforation/therapy , Microbial Sensitivity Tests
3.
J Med Case Rep ; 18(1): 271, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38845030

ABSTRACT

OBJECTIVE: Extravasation of infused drugs is not a rare problem in medical practice. Acyclovir is a vesicant and an antiviral medication commonly used for young children. In the present study, we presented a neonate with soft tissue damage due to acyclovir extravasation. CASE REPORT: A female newborn (Iranian, Asian) with gestational age 37+2 weeks and breech presentation was born by Cesarean delivery from a mother with a recent history of Herpes simplex virus (HSV) infection (Yas Women's Hospital, Tehran, Iran). Intravenous administration of acyclovir was initiated through a peripheral catheter inserted on the dorsal side of the left hand. A few minutes after the second dose, the patient showed a diffused firm swelling, local discoloration, and induration in the dorsum of the hand. The peripheral catheter was removed immediately. Hyaluronidase was injected subcutaneously in five different regions around the catheterization site. Intermittent limb elevation and cold compression (for 10 minutes) were applied. Serial follow-ups and examinations were performed hourly to check limb inflammation, ischemia, and compartment syndrome. The limb swelling and discoloration significantly improved 4 hours after the second dose of hyaluronidase. CONCLUSION: Early diagnosis of acyclovir extravasation and immediate management could prevent severe complications in neonates. Further studies are needed to suggest a standard approach and treatment protocol for acyclovir extravasation.


Subject(s)
Acyclovir , Antiviral Agents , Extravasation of Diagnostic and Therapeutic Materials , Humans , Acyclovir/adverse effects , Acyclovir/administration & dosage , Acyclovir/therapeutic use , Female , Infant, Newborn , Antiviral Agents/adverse effects , Herpes Simplex/drug therapy , Hyaluronoglucosaminidase/administration & dosage
4.
Proc Natl Acad Sci U S A ; 121(24): e2320867121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38838015

ABSTRACT

O-GlcNAcase (OGA) is the only human enzyme that catalyzes the hydrolysis (deglycosylation) of O-linked beta-N-acetylglucosaminylation (O-GlcNAcylation) from numerous protein substrates. OGA has broad implications in many challenging diseases including cancer. However, its role in cell malignancy remains mostly unclear. Here, we report that a cancer-derived point mutation on the OGA's noncatalytic stalk domain aberrantly modulates OGA interactome and substrate deglycosylation toward a specific set of proteins. Interestingly, our quantitative proteomic studies uncovered that the OGA stalk domain mutant preferentially deglycosylated protein substrates with +2 proline in the sequence relative to the O-GlcNAcylation site. One of the most dysregulated substrates is PDZ and LIM domain protein 7 (PDLIM7), which is associated with the tumor suppressor p53. We found that the aberrantly deglycosylated PDLIM7 suppressed p53 gene expression and accelerated p53 protein degradation by promoting the complex formation with E3 ubiquitin ligase MDM2. Moreover, deglycosylated PDLIM7 significantly up-regulated the actin-rich membrane protrusions on the cell surface, augmenting the cancer cell motility and aggressiveness. These findings revealed an important but previously unappreciated role of OGA's stalk domain in protein substrate recognition and functional modulation during malignant cell progression.


Subject(s)
Cytoskeleton , LIM Domain Proteins , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Cytoskeleton/metabolism , Acetylglucosamine/metabolism , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/pathology , Cell Line, Tumor , Glycosylation , Hydrolysis , Mutation , Cell Movement , Antigens, Neoplasm , Hyaluronoglucosaminidase , Histone Acetyltransferases
5.
J Clin Immunol ; 44(7): 148, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896141

ABSTRACT

PURPOSE: Facilitated subcutaneous immunoglobulin (fSCIG; immune globulin infusion 10% [human] with recombinant human hyaluronidase [rHuPH20]) permits high-volume subcutaneous immunoglobulin (SCIG) infusion, shorter infusion times and reduced dosing frequency relative to conventional SCIG. It is initiated by gradually increasing infusion volumes over time (dose ramp-up) to achieve target dose level (TDL). Whether ramp-up strategies have tolerability or safety advantages over direct initiation at full TDL has not been evaluated clinically. METHODS: This phase 1 open-label study assessed tolerability and safety of fSCIG 10% with accelerated or no ramp-up compared with conventional ramp-up in healthy adults (NCT04578535). Participants were assigned to one of the three ramp-up arms to achieve TDLs of 0.4 or 1.0 g/kg/infusion. The primary endpoint was the proportion of infusions completed without interruption or infusion rate reduction owing to treatment-emergent adverse events (TEAEs). Safety was assessed as a secondary endpoint. RESULTS: Of 51 participants enrolled, 50 (98.0%) tolerated all fSCIG 10% infusions initiated (n = 174). Infusion rate was reduced in one participant owing to headache in the 0.4 g/kg/infusion conventional ramp-up arm. Study discontinuations were higher in the no ramp-up arm (70%) versus the conventional (0%) and accelerated (22%) arms at the 1.0 g/kg/infusion TDL. Safety outcomes did not substantially differ between treatment arms. CONCLUSION: The favorable tolerability and safety profiles of fSCIG 10% in healthy participants support initiating treatment with fSCIG 10% with accelerated ramp-up at TDLs up to 1.0 g/kg. Data support no ramp-up at TDLs close to 0.4 g/kg but additional data are needed for higher doses.


Subject(s)
Healthy Volunteers , Hyaluronoglucosaminidase , Infusions, Subcutaneous , Humans , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/adverse effects , Male , Female , Adult , Young Adult , Middle Aged , Immunoglobulins/administration & dosage , Immunoglobulins/adverse effects , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , Adolescent
6.
Sci Rep ; 14(1): 14958, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38942920

ABSTRACT

Cancer remains a formidable global health challenge, with metastasis being a key contributor to its lethality. Abundant high molecular mass hyaluronic acid, a major non-protein component of extracellular matrix, protects naked mole rats from cancer and reduces cancer incidence in mice. Hyaluronidase plays a critical role in degrading hyaluronic acid and is frequently overexpressed in metastatic cancer. Here we investigated the potential of targeting hyaluronidases to reduce metastasis. A high throughput screen identified delphinidin, a natural plant compound found in fruits and vegetables, as a potent hyaluronidase inhibitor. Delphinidin-mediated inhibition of hyaluronidase activity led to an increase in high molecular weight hyaluronic acid in cell culture and in mouse tissues, and reduced migration and invasion behavior of breast, prostate, and melanoma cancer cells. Moreover, delphinidin treatment suppressed melanoma metastasis in mice. Our study provides a proof of principle that inhibition of hyaluronidase activity suppresses cancer cell migration, invasion and metastasis. Furthermore, we identified a natural compound delphinidin as a potential anticancer therapeutic. Thus, we have identified a path for clinical translation of the cancer resistance mechanism identified in the naked mole rat.


Subject(s)
Anthocyanins , Cell Movement , Hyaluronoglucosaminidase , Neoplasm Metastasis , Animals , Female , Humans , Male , Mice , Anthocyanins/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Enzyme Inhibitors/pharmacology , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/antagonists & inhibitors , Hyaluronoglucosaminidase/metabolism , Mole Rats
7.
Nihon Yakurigaku Zasshi ; 159(4): 241-253, 2024.
Article in Japanese | MEDLINE | ID: mdl-38945908

ABSTRACT

Pertuzumab and trastuzumab are anti-HER2 humanized monoclonal antibodies with different mechanisms of action. Their combination is expected to suppress intracellular HER2 signaling additively or synergistically. Their combination is widely recommended worldwide and has been established as a standard of care for HER2-positive breast cancer. However, improvement is required because of the prolonged time of intravenous infusion. Vorhyaluronidase alfa (rHuPH20) depolymerizes hyaluronan in the subcutaneous connective tissue. It's reported to increase the permeability and absorption levels of drugs. PHESGO® combination for subcutaneous injection MA/IN (PHESGO®) is a fixed-dose combination of pertuzumab, trastuzumab, and rHuPH20. A confirmatory phase III study (FeDeriCa) was conducted following a dose-finding phase I study (BO30185). Patients with HER2-positive early breast cancer were randomly assigned to receive either intravenous infusion of pertuzumab and trastuzumab or subcutaneous injection of PHESGO®, in combination with chemotherapy, to compare the pharmacokinetics (PK), efficacy and safety. A phase II study (PHranceSCa) was also conducted to assess patients' preference and satisfaction. Based on these results, population PK analysis, and other data, PHESGO® obtained marketing approval in Japan in September 2023 with indications for "HER2-positive breast cancer" and "advanced or recurrent HER2-positive colorectal cancer that has progressed following cancer chemotherapy and is not amenable to curative resection". By reducing the administration time, PHESGO® is expected to contribute to various needs of patients and improvement of their daily lives. Since drug preparation is not required, it can provide convenience to healthcare professionals, leading to stress reduction of medical resources as well.


Subject(s)
Antibodies, Monoclonal, Humanized , Hyaluronoglucosaminidase , Trastuzumab , Humans , Trastuzumab/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Hyaluronoglucosaminidase/administration & dosage , Injections, Subcutaneous , Breast Neoplasms/drug therapy , Drug Combinations , Clinical Trials as Topic , Receptor, ErbB-2/metabolism , Female , Recombinant Proteins/administration & dosage , Cell Adhesion Molecules
8.
Thorac Cancer ; 15(18): 1397-1409, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38717936

ABSTRACT

BACKGROUND: Alterations in epigenetic factors are recognized as key contributors to the emergence of human cancer. The active and reversible alteration of N6-methyladenosine (m6A) RNA is crucial for controlling gene activity and determining cellular destiny. Even with these insights, the triggering of KIAA1429 (also called VIRMA) and its role in lung adenocarcinoma (LUAD) is mostly unclear. As a result, the objective of this study was to elucidate how KIAA1429 contributes to cancer development in LUAD. METHODS: This study utilized multiple methods for investigation, encompassing the in vitro functional examination of KIAA1429 in lung adenocarcinoma cells, transcriptome sequencing, methylation RNA immunoprecipitation sequencing (MeRIP-seq), as well as RNA stability tests to ascertain the half-life and stability of the target genes. RESULTS: The results indicated that modifying the expression of KIAA1429 regulated the proliferation and metastasis of LUAD. By employing transcriptome sequencing alongside MeRIP-seq analysis, the research pinpointed genes affected by m6A alterations triggered by KIAA1429. In a more detailed manner, it was discovered that KIAA1429 plays a regulatory role in the expression of ARHGAP30. Suppressing KIAA1429 results in reduced m6A levels in the mRNA of the target gene ARHGAP30, boosting its stability and expression, thus inhibiting tumor proliferation and metastasis. CONCLUSION: This study revealed the activation mechanism and pivotal function of KIAA1429 in LUAD tumor development, paving the way for molecular-based interventions for LUAD.


Subject(s)
Adenocarcinoma of Lung , Cell Proliferation , GTPase-Activating Proteins , Lung Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Mice , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Animals , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Hyaluronoglucosaminidase/genetics , Hyaluronoglucosaminidase/metabolism , Signal Transduction , RNA-Binding Proteins
9.
Arch Oral Biol ; 163: 105980, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38692246

ABSTRACT

OBJECTIVE: To determine the effect of hyaluronic acid (HA) degradation by hyaluronidase (HYAL) in inhibiting collagen fiber production by rat periodontal ligament cells (rPDLCs). DESIGN: Primary rPDLCs were isolated from the euthanized rats and used for in vitro experiments. The appropriate HYAL concentration was determined through CCK-8 testing for cytotoxicity detection and Alizarin red staining for mineralization detection. RT-qPCR and western blot assays were conducted to assess the effect of HYAL, with or without TGF-ß, on generation of collagen fiber constituents and expression of actin alpha 2, smooth muscle (ACTA2) of rPDLCs. RESULTS: Neither cell proliferation nor mineralization were significantly affected by treatment with 4 U/mL HYAL. HYAL (4 U/mL) alone downregulated type I collagen fiber (Col1a1 and Col1a2) and Acta2 mRNA expression; however, ACTA2 and COL1 protein levels were only downregulated by HYAL treatment after TGF-ß induction. CONCLUSIONS: Treatment of rPDLCs with HYAL can inhibit TGF-ß-induced collagen matrix formation and myofibroblast transformation.


Subject(s)
Cell Proliferation , Collagen , Fibroblasts , Hyaluronoglucosaminidase , Myofibroblasts , Periodontal Ligament , Transforming Growth Factor beta , Animals , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Hyaluronoglucosaminidase/pharmacology , Rats , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Transforming Growth Factor beta/metabolism , Collagen/metabolism , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Hyaluronic Acid/pharmacology , Cells, Cultured , Rats, Sprague-Dawley , Actins/metabolism , Blotting, Western , In Vitro Techniques , Collagen Type I/metabolism , Biomarkers/metabolism , Real-Time Polymerase Chain Reaction , Male , RNA, Messenger/metabolism
10.
Biomacromolecules ; 25(6): 3671-3684, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38720431

ABSTRACT

Reactive oxygen species (ROS) have emerged as a promising treatment option for antibacterial and biofilm eradication. However, their therapeutic efficacy is significantly hampered by the unique microenvironments of diabetic wounds. In this study, we designed and synthesized porphyrin-based Fe covalent organic frameworks (Fe-COF) through a Schiff base condensation reaction. Subsequently, Fe-COF were encapsulated with hyaluronic acid (HA) through electrostatic adsorption, resulting in a novel formulation named HA-Fe-COF for diabetic wound healing. HA-Fe-COF were engineered to respond to hyaluronidase in the infected wound, leading to the controlled release of Fe-COF. Those released Fe-COF served a dual role as photosensitizers, generating singlet oxygen and localized heating when exposed to dual light sources. Additionally, they acted as peroxidase-like nanozymes, facilitating the production of ROS through enzymatic reactions. This innovative approach enabled a synergistic therapeutic effect combining photodynamic, photothermal, and chemodynamic modalities. Furthermore, the sustained release of HA from HA-Fe-COF promoted angiogenesis, collagen deposition, and re-epithelialization during the diabetic wound healing process. This "all-in-one" strategy offers a novel approach for the development of antimicrobial and biofilm eradication strategies that minimize damage to healthy tissues in vivo.


Subject(s)
Hyaluronic Acid , Metal-Organic Frameworks , Porphyrins , Wound Healing , Wound Healing/drug effects , Animals , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology , Mice , Reactive Oxygen Species/metabolism , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Skin/drug effects , Humans , Wound Infection/drug therapy , Wound Infection/microbiology , Iron/chemistry , Photochemotherapy/methods , Hyaluronoglucosaminidase
11.
Chem Biol Interact ; 396: 111045, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38729283

ABSTRACT

Orbital connective tissue changes are contributors to the pathogenesis in thyroid eye disease (TED). Activated fibroblasts respond to immune stimuli with proliferation and increased hyaluronan (HA) production. Cyclosporin A (CsA) was reported to be beneficial in the treatment of TED. PDGF isoforms are increased in orbital tissue of TED patients and enhance HA production. We aimed to study the effect of CsA on HA production and hyaluronan synthase (HAS1, 2 and 3) and hyaluronidase (HYAL1 and 2) mRNA expressions in orbital fibroblasts (OFs). Measurements were performed in the presence or absence of CsA (10 µM) in unstimulated or PDGF-BB (10 ng/ml) stimulated OFs. The HA production of TED OFs (n = 7) and NON-TED OFs (n = 6) were measured by ELISA. The levels of mRNA expressions were examined using RT-PCR. The proliferation rate and metabolic activity were measured by BrdU incorporation and MTT assays, respectively. Treatment with CsA resulted in an average 42% decrease in HA production of OFs (p < 0.0001). CsA decreased the expression levels of HAS2, HAS3 and HYAL2 (p = 0.005, p = 0.005 and p = 0.002, respectively.) PDGF-BB increased HA production (p < 0.001) and HAS2 expression (p = 0.004). CsA could reduce the PDGF-BB-stimulated HA production (p < 0.001) and HAS2 expression (p = 0.005) below the untreated level. In addition, CsA treatment caused a decrease in proliferation potential (p = 0.002) and metabolic activity (p < 0.0001). These findings point to the fact that CsA affects HA metabolism via HAS2, HAS3 and HYAL2 inhibition in OFs. In addition to its well characterized immunosuppressant properties, CsA's beneficial effect in TED may be related to its direct inhibitory effect on basal and growth factor stimulated HA production.


Subject(s)
Becaplermin , Cell Proliferation , Cyclosporine , Fibroblasts , Glucuronosyltransferase , Graves Ophthalmopathy , Hyaluronan Synthases , Hyaluronic Acid , Hyaluronoglucosaminidase , Proto-Oncogene Proteins c-sis , Hyaluronic Acid/biosynthesis , Hyaluronic Acid/pharmacology , Humans , Becaplermin/metabolism , Becaplermin/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Hyaluronan Synthases/metabolism , Hyaluronan Synthases/genetics , Cyclosporine/pharmacology , Hyaluronoglucosaminidase/metabolism , Hyaluronoglucosaminidase/antagonists & inhibitors , Cell Proliferation/drug effects , Proto-Oncogene Proteins c-sis/metabolism , Glucuronosyltransferase/metabolism , Glucuronosyltransferase/genetics , Graves Ophthalmopathy/metabolism , Graves Ophthalmopathy/pathology , Graves Ophthalmopathy/drug therapy , Cells, Cultured , Orbit/metabolism , Orbit/drug effects , Orbit/pathology , RNA, Messenger/metabolism , RNA, Messenger/genetics , Cell Adhesion Molecules/metabolism , GPI-Linked Proteins
12.
Neurol Med Chir (Tokyo) ; 64(6): 222-229, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38719578

ABSTRACT

The detailed changes in disc properties after intradiscal injection of condoliase remain controversial. At 3 and 9 months after administration, radiographic changes in discs were investigated. A total of 41 patients (men, 25; median age, 46 years) who underwent regular follow-up magnetic resonance imaging at 3 and 9 months after administration without additional invasive therapy were retrospectively investigated. The intensity changes of the nucleus pulposus based on the Pfirrmann disc grading system, midsagittal disc height, and maximum protrusion length of herniation were assessed. In addition, disc height changes were compared between 24 patients aged <50 years (young group) and 17 patients aged ≥50 years (over 50s group). The overall mean disc heights were 9.1, 7.5, and 7.6 mm preoperatively, at 3 months, and at 9 months, respectively, with a significant reduction at 3 months (P < 0.001) and no significant changes thereafter. The mean maximum protrusion length of herniation significantly and gradually decreased. The overall proportions of Pfirrmann disc grades after administration were equivalent between 3 and 9 months. However, the recovery from Pfirrmann disc grades IV to III was confirmed in 8 of 17 cases (47.1%) between 3 and 9 months, whereas 6 of 20 cases (30.0%) showed a decline from III to IV. Patients in the young group with pretreatment disc height >11 mm had the greatest reduction in disc height than the over 50s group. In conclusion, the clinical outcomes in the over 50s group were comparable to those in the young group after injection of condoliase, whereas young patients with higher disc were more susceptible to disc height reduction.


Subject(s)
Intervertebral Disc Displacement , Lumbar Vertebrae , Humans , Intervertebral Disc Displacement/diagnostic imaging , Male , Middle Aged , Female , Adult , Lumbar Vertebrae/diagnostic imaging , Retrospective Studies , Aged , Intervertebral Disc/diagnostic imaging , Hyaluronoglucosaminidase/administration & dosage , Hyaluronoglucosaminidase/therapeutic use , Magnetic Resonance Imaging
13.
Int J Biol Macromol ; 271(Pt 1): 132626, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38795893

ABSTRACT

Immobilization of proteolytic enzymes onto nanocarriers is effective to improve drug diffusion in tumors through degrading the dense extracellular matrix (ECM). Herein, immobilization and release behaviors of hyaluronidase, bromelain, and collagenase (Coll) on mesoporous silica nanoparticles (MSNs) were explored. A series of cationic MSNs (CMSNs) with large and adjustable pore sizes were synthesized, and investigated together with two anionic MSNs of different pore sizes. CMSNs4.0 exhibited the highest enzyme loading capacity for hyaluronidase and bromelain, and CMSNs4.5 was the best for Coll. High electrostatic interaction, matched pore size, and large pore volume and surface area favor the immobilization. Changes of the enzyme conformations and surface charges with pH, existence of a space around the immobilized enzymes, and the depth of the pore structures, affect the release ratio and tunability. The optimal CMSNs-enzyme complexes exhibited deep and homogeneous penetration into pancreatic tumors, a tumor model with the densest ECM, with CMSNs4.5-Coll as the best. Upon loading with doxorubicin (DOX), the CMSNs-enzyme complexes induced high anti-tumor efficiencies. Conceivably, the DOX/CMSNs4.5-NH2-Coll nanodrug exhibited the most effective tumor therapy, with a tumor growth inhibition ratio of 86.1 %. The study provides excellent nanocarrier-enzyme complexes, and offers instructive theories for enhanced tumor penetration and therapy.


Subject(s)
Doxorubicin , Enzymes, Immobilized , Nanoparticles , Silicon Dioxide , Silicon Dioxide/chemistry , Enzymes, Immobilized/chemistry , Nanoparticles/chemistry , Porosity , Doxorubicin/chemistry , Doxorubicin/pharmacology , Animals , Humans , Mice , Drug Carriers/chemistry , Cell Line, Tumor , Hyaluronoglucosaminidase/chemistry , Hyaluronoglucosaminidase/metabolism , Drug Liberation , Collagenases/metabolism , Collagenases/chemistry , Bromelains/chemistry , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology
14.
Int J Biol Macromol ; 270(Pt 1): 132277, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735611

ABSTRACT

The high-glycemic microenvironment of diabetic wounds promotes bacterial proliferation, leading to persistent infections and delayed wound healing. This poses a significant threat to human health, necessitating the development of new nanodrug visualization platforms. In this study, we designed and synthesized cascade nano-systems modified with targeted peptide and hyaluronic acid for diabetic infection therapy. The nano-systems were able to target the site of infection using LL-37, and in the microenvironment of wound infection, the hyaluronic acid shell of the nano-systems was degraded by endogenous hyaluronidase. This precise degradation released a cascade of nano-enzymes on the surface of the bacteria, effectively destroying their cytoskeleton. Additionally, the metals in the nano-enzymes provided a photo-thermal effect, accelerating wound healing. The cascade nano-visualization platform demonstrated excellent bactericidal efficacy in both in vitro antimicrobial assays and in vivo diabetic infection models. In conclusion, this nano-system employs multiple approaches including targeting, enzyme-catalyzed therapy, photothermal therapy, and chemodynamic therapy to kill bacteria and promote healing. The Ag@Pt-Au-LYZ/HA-LL-37 formulation shows great potential for the treatment of diabetic wounds.


Subject(s)
Anti-Bacterial Agents , Bacterial Infections , Hyaluronic Acid , Wound Healing , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Wound Healing/drug effects , Bacterial Infections/drug therapy , Mice , Diabetes Mellitus, Experimental , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/chemistry , Hyaluronoglucosaminidase/metabolism , Cathelicidins , Humans , Diabetes Complications/drug therapy , Nanoparticles/chemistry
15.
Int Wound J ; 21(5): e14906, 2024 May.
Article in English | MEDLINE | ID: mdl-38745342

ABSTRACT

This study manufactured a 35 kDa hyaluronan fragment (HA35) by enzymatically degrading high-molecular-weight HA using hyaluronidase PH20 derived from bovine testis. The research then examined the therapeutic efficacy of locally administered, tissue-permeable HA35 in alleviating chronic wounds and their associated neuropathic pain. For 20 patients with nonhealing wounds and associated pain lasting over three months, 100 mg of HA35 was injected daily into the healthy skin surrounding the chronic wound for 10 days. Self-assessments before and after treatment indicated that HA35 significantly enhanced wound healing. This was evidenced by the formation of fresh granulation tissue on the wounds (p < 0.0001); reduced darkness, redness, dryness, and damage in the skin surrounding the wounds (p < 0.0001), and a decrease in wound size (p < 0.001). Remarkably, HA35 injections alleviated pain associated with chronic wounds within 24 hours (p < 0.0001). It can be concluded that the low-molecular-weight hyaluronan fragment HA35 potentially enhances the immune response and angiogenesis during wound healing.


Subject(s)
Hyaluronic Acid , Hyaluronoglucosaminidase , Wound Healing , Hyaluronic Acid/therapeutic use , Wound Healing/drug effects , Male , Humans , Middle Aged , Chronic Disease , Hyaluronoglucosaminidase/therapeutic use , Hyaluronoglucosaminidase/administration & dosage , Aged , Female , Adult , Treatment Outcome , Wounds and Injuries/drug therapy , Animals , Molecular Weight , Aged, 80 and over
16.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731830

ABSTRACT

Brevibacillus sp. JNUCC 41, characterized as a plant-growth-promoting rhizobacterium (PGPR), actively participates in lipid metabolism and biocontrol based on gene analysis. This study aimed to investigate the crucial secondary metabolites in biological metabolism; fermentation, extraction, and isolation were performed, revealing that methyl indole-3-acetate showed the best hyaluronidase (HAase) inhibitory activity (IC50: 343.9 µM). Molecular docking results further revealed that the compound forms hydrogen bonds with the residues Tyr-75 and Tyr-247 of HAase (binding energy: -6.4 kcal/mol). Molecular dynamics (MD) simulations demonstrated that the compound predominantly binds to HAase via hydrogen bonding (MM-PBSA binding energy: -24.9 kcal/mol) and exhibits good stability. The residues Tyr-247 and Tyr-202, pivotal for binding in docking, were also confirmed via MD simulations. This study suggests that methyl indole-3-acetate holds potential applications in anti-inflammatory and anti-aging treatments.


Subject(s)
Brevibacillus , Hyaluronoglucosaminidase , Molecular Docking Simulation , Molecular Dynamics Simulation , Hyaluronoglucosaminidase/antagonists & inhibitors , Hyaluronoglucosaminidase/metabolism , Brevibacillus/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Hydrogen Bonding , Genome, Bacterial
17.
Sci Rep ; 14(1): 11570, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38773189

ABSTRACT

Pre-clinical and clinical studies have shown that PEGPH20 depletes intratumoral hyaluronic acid (HA), which is linked to high interstitial fluid pressures and poor distribution of chemotherapies. 29 patients with metastatic advanced solid tumors received quantitative magnetic resonance imaging (qMRI) in 3 prospective clinical trials of PEGPH20: HALO-109-101 (NCT00834704), HALO-109-102 (NCT01170897), and HALO-109-201 (NCT01453153). Apparent Diffusion Coefficient of water (ADC), T1, ktrans, vp, ve, and iAUC maps were computed from qMRI acquired at baseline and ≥ 1 time point post-PEGPH20. Tumor ADC and T1 decreased, while iAUC, ktrans, vp, and ve increased, on day 1 post-PEGPH20 relative to baseline values. This is consistent with HA depletion leading to a decrease in tumor extracellular water content and an increase in perfusion, permeability, extracellular matrix space, and vascularity. Baseline parameter values predictive of pharmacodynamic responses were: ADC > 1.46 × 10-3 mm2/s (Balanced Accuracy (BA) = 72%, p < 0.01), T1 > 0.54 s (BA = 82%, p < 0.01), iAUC < 9.2 mM-s (BA = 76%, p < 0.05), ktrans < 0.07 min-1 (BA = 72%, p = 0.2), ve < 0.17 (BA = 68%, p < 0.01), and vp < 0.02 (BA = 60%, p < 0.01). A low ve at baseline was moderately predictive of response in any parameter (BA = 65.6%, p < 0.01 averaged across patients). These qMRI biomarkers are potentially useful for guiding patient pre-selection and post-treatment follow-up in future clinical studies of PEGPH20 and other tumor stroma-modifying anti-cancer therapies.


Subject(s)
Hyaluronic Acid , Hyaluronoglucosaminidase , Magnetic Resonance Imaging , Neoplasms , Adult , Aged , Female , Humans , Male , Middle Aged , Magnetic Resonance Imaging/methods , Neoplasms/drug therapy , Neoplasms/diagnostic imaging , Neoplasms/pathology , Polyethylene Glycols/therapeutic use , Prospective Studies
18.
Cell Rep ; 43(5): 114163, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38678556

ABSTRACT

Dysregulation of O-GlcNAcylation has emerged as a potential biomarker for several diseases, particularly cancer. The role of OGT (O-GlcNAc transferase) in maintaining O-GlcNAc homeostasis has been extensively studied; nevertheless, the regulation of OGA (O-GlcNAcase) in cancer remains elusive. Here, we demonstrated that the multifunctional protein RBM14 is a regulator of cellular O-GlcNAcylation. By investigating the correlation between elevated O-GlcNAcylation and increased RBM14 expression in lung cancer cells, we discovered that RBM14 promotes ubiquitin-dependent proteasomal degradation of OGA, ultimately mediating cellular O-GlcNAcylation levels. In addition, RBM14 itself is O-GlcNAcylated at serine 521, regulating its interaction with the E3 ligase TRIM33, consequently affecting OGA protein stability. Moreover, we demonstrated that mutation of serine 521 to alanine abrogated the oncogenic properties of RBM14. Collectively, our findings reveal a previously unknown mechanism for the regulation of OGA and suggest a potential therapeutic target for the treatment of cancers with dysregulated O-GlcNAcylation.


Subject(s)
Protein Stability , RNA-Binding Proteins , Humans , Acetylglucosamine/metabolism , Antigens, Neoplasm , beta-N-Acetylhexosaminidases/metabolism , Cell Line, Tumor , Glycosylation , HEK293 Cells , Histone Acetyltransferases , Hyaluronoglucosaminidase , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , N-Acetylglucosaminyltransferases/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Tripartite Motif Proteins/metabolism , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/metabolism
19.
Clin Transl Sci ; 17(4): e13788, 2024 04.
Article in English | MEDLINE | ID: mdl-38561908

ABSTRACT

Predicting the subcutaneous (SC) pharmacokinetics (PK) of antibodies in humans is challenging, with clinical data currently being the only reliable data source for modeling SC absorption and bioavailability. Recombinant human hyaluronidase PH20 (rHuPH20) is an enzyme that facilitates SC delivery of high-dose, high-volume therapeutics. Numerous monoclonal antibodies have been co-administered SC with rHuPH20 in a clinical setting, establishing an extensive PK database. The goal of this work is to demonstrate how aggregated clinical data can be leveraged in a universal modeling framework for characterizing SC antibody PK, resulting in parameterization that can be used in predictive simulations of new antibodies. Data for 10 individual antibodies co-administered SC with rHuPH20 were obtained from publicly available sources. PK modeling of each antibody was conducted using the same model structure, but uniquely parameterized. The model structure consisted of a two-compartment model to capture linear kinetics, plus a target-binding mechanism to accommodate nonlinear kinetics driven by antibody-target complex formation and elimination. The clinical PK profiles for all antibodies were accurately described using the universal modeling framework. The SC PK parameters of absorption and bioavailability were consistent across the range of antibody and target properties evaluated. SC administration with rHuPH20 yielded a 30% increase in absorption rate on average and similar or better bioavailability. These parameter values can serve as initial conditions for model-based PK predictions for new antibodies co-administered SC with rHuPH20 to enable evaluation of optimal SC dose and schedule regimens prior to and during clinical development.


Subject(s)
Antibodies, Monoclonal , Hyaluronoglucosaminidase , Humans , Injections, Subcutaneous , Biological Availability
SELECTION OF CITATIONS
SEARCH DETAIL
...