Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.387.202
Filter
1.
Biomaterials ; 312: 122714, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39079462

ABSTRACT

Osteosarcoma, a malignant bone tumor often characterized by high hedgehog signaling activity, residual tumor cells, and substantial bone defects, poses significant challenges to both treatment response and postsurgical recovery. Here, we developed a nanocomposite hydrogel for the sustained co-delivery of bioactive magnesium ions, anti-PD-L1 antibody (αPD-L1), and hedgehog pathway antagonist vismodegib, to eradicate residual tumor cells while promoting bone regeneration post-surgery. In a mouse model of tibia osteosarcoma, this hydrogel-mediated combination therapy led to remarkable tumor growth inhibition and hence increased animal survival by enhancing the activity of tumor-suppressed CD8+ T cells. Meanwhile, the implanted hydrogel improved the microenvironment of osteogenesis through long-term sustained release of Mg2+, facilitating bone defect repair by upregulating the expression of osteogenic genes. After 21 days, the expression levels of ALP, COL1, RUNX2, and BGLAP in the Vis-αPD-L1-Gel group were approximately 4.1, 5.1, 5.5, and 3.4 times higher than those of the control, respectively. We believe that this hydrogel-based combination therapy offers a potentially valuable strategy for treating osteosarcoma and addressing the tumor-related complex bone diseases.


Subject(s)
Bone Neoplasms , Hydrogels , Immunotherapy , Nanocomposites , Osteosarcoma , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/therapy , Animals , Hydrogels/chemistry , Nanocomposites/chemistry , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Mice , Immunotherapy/methods , Cell Line, Tumor , Bone Regeneration/drug effects , Humans , Osteogenesis/drug effects , B7-H1 Antigen/metabolism , Mice, Inbred BALB C , Magnesium/chemistry
2.
Biomaterials ; 312: 122720, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39084098

ABSTRACT

Mesenchymal stem cells (MSCs) are expected to be useful therapeutics in osteoarthritis (OA), the most common joint disorder characterized by cartilage degradation. However, evidence is limited with regard to cartilage repair in clinical trials because of the uncontrolled differentiation and weak cartilage-targeting ability of MSCs after injection. To overcome these drawbacks, here we synthesized CuO@MSN nanoparticles (NPs) to deliver Sox9 plasmid DNA (favoring chondrogenesis) and recombinant protein Bmp7 (inhibiting hypertrophy). After taking up CuO@MSN/Sox9/Bmp7 (CSB NPs), the expressions of chondrogenic markers were enhanced while hypertrophic markers were decreased in response to these CSB-engineered MSCs. Moreover, a cartilage-targeted peptide (designated as peptide W) was conjugated onto the surface of MSCs via a click chemistry reaction, thereby prolonging the residence time of MSCs in both the knee joint cavity of mice and human-derived cartilage. In a surgery-induced OA mouse model, the NP and peptide dual-modified W-CSB-MSCs showed an enhancing therapeutic effect on cartilage repair in knee joints compared with other engineered MSCs after intra-articular injection. Most importantly, W-CSB-MSCs accelerated cartilage regeneration in damaged cartilage explants derived from OA patients. Thus, this new peptide and NPs dual engineering strategy shows potential for clinical applications to boost cartilage repair in OA using MSC therapy.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Nanoparticles , Osteoarthritis , Peptides , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Osteoarthritis/therapy , Osteoarthritis/pathology , Nanoparticles/chemistry , Humans , Cell Differentiation/drug effects , Peptides/chemistry , Mesenchymal Stem Cell Transplantation/methods , Chondrogenesis/drug effects , Mice , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Cartilage, Articular/pathology , Cartilage, Articular/drug effects , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/pharmacology , Tissue Engineering/methods , Regeneration/drug effects
3.
Biomaterials ; 312: 122711, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39088911

ABSTRACT

The unsuitable deformation stimulus, harsh urine environment, and lack of a regenerative microenvironment (RME) prevent scaffold-based urethral repair and ultimately lead to irreversible urethral scarring. The researchers clarify the optimal elastic modulus of the urethral scaffolds for urethral repair and design a multilayered PVA hydrogel scaffold for urethral scar-free healing. The inner layer of the scaffold has self-healing properties, which ensures that the wound effectively resists harsh urine erosion, even when subjected to sutures. In addition, the scaffold's outer layer has an extracellular matrix-like structure that synergizes with adipose-derived stem cells to create a favorable RME. In vivo experiments confirm successful urethral scar-free healing using the PVA multilayered hydrogel scaffold. Further mechanistic study shows that the PVA multilayer hydrogel effectively resists the urine-induced inflammatory response and accelerates the transition of urethral wound healing to the proliferative phase by regulating macrophage polarization, thus providing favorable conditions for urethral scar-free healing. This study provides mechanical criteria for the fabrication of urethral tissue-engineered scaffolds, as well as important insights into their design.


Subject(s)
Elastic Modulus , Hydrogels , Tissue Scaffolds , Urethra , Wound Healing , Tissue Scaffolds/chemistry , Animals , Hydrogels/chemistry , Tissue Engineering/methods , Mice , Regeneration , Cicatrix/pathology , Male , Cellular Microenvironment , Rats, Sprague-Dawley , Stem Cells/cytology
4.
Biomaterials ; 312: 122719, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39088912

ABSTRACT

Acute myeloid leukemia (AML) is a deadly form of leukemia with ineffective traditional treatment and frequent chemoresistance-associated relapse. Personalized drug screening holds promise in identifying optimal regimen, nevertheless, primary AML cells undergo spontaneous apoptosis during cultures, invalidating the drug screening results. Here, we reconstitute a 3D osteogenic niche (3DON) mimicking that in bone marrow to support primary AML cell survival and phenotype maintenance in cultures. Specifically, 3DON derived from osteogenically differentiated mesenchymal stem cells (MSC) from healthy and AML donors are co-cultured with primary AML cells. The AML cells under the AML_3DON niche showed enhanced viability, reduced apoptosis and maintained CD33+ CD34-phenotype, associating with elevated secretion of anti-apoptotic cytokines in the AML_3DON niche. Moreover, AML cells under the AML_3DON niche exhibited low sensitivity to two FDA-approved chemotherapeutic drugs, further suggesting the physiological resemblance of the AML_3DON niche. Most interestingly, AML cells co-cultured with the healthy_3DON niche are highly sensitive to the same sample drugs. This study demonstrates the differential responses of AML cells towards leukemic and healthy bone marrow niches, suggesting the impact of native cancer cell niche in drug screening, and the potential of re-engineering healthy bone marrow niche in AML patients as chemotherapeutic adjuvants overcoming chemoresistance, respectively.


Subject(s)
Cell Survival , Leukemia, Myeloid, Acute , Mesenchymal Stem Cells , Phenotype , Tumor Microenvironment , Humans , Leukemia, Myeloid, Acute/pathology , Tumor Microenvironment/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Cell Survival/drug effects , Coculture Techniques/methods , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Marrow/pathology , Bone Marrow/drug effects , Stem Cell Niche/drug effects , Bone Marrow Cells/cytology , Male , Cell Differentiation/drug effects , Female
5.
Biomaterials ; 312: 122732, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39088913

ABSTRACT

Fully restoring the lost population of cardiomyocytes and heart function remains the greatest challenge in cardiac repair post myocardial infarction. In this study, a pioneered highly ROS-eliminating hydrogel was designed to enhance miR-19a/b induced cardiomyocyte proliferation by lowering the oxidative stress and continuously releasing miR-19a/b in infarcted myocardium in situ. In vivo lineage tracing revealed that ∼20.47 % of adult cardiomyocytes at the injected sites underwent cell division in MI mice. In MI pig the infarcted size was significantly reduced from 40 % to 18 %, and thereby marked improvement of cardiac function and increased muscle mass. Most importantly, our treatment solved the challenge of animal death--all the treated pigs managed to live until their hearts were harvested at day 50. Therefore, our strategy provides clinical conversion advantages and safety for healing damaged hearts and restoring heart function post MI, which will be a powerful tool to battle cardiovascular diseases in patients.


Subject(s)
Cell Proliferation , MicroRNAs , Myocardial Infarction , Myocytes, Cardiac , Oxidative Stress , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Oxidative Stress/drug effects , Mice , Swine , Hydrogels/chemistry , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism
6.
Biomaterials ; 312: 122740, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39096839

ABSTRACT

Metastasis stands as the primary contributor to mortality associated with tumors. Chemotherapy and immunotherapy are frequently utilized in the management of metastatic solid tumors. Nevertheless, these therapeutic modalities are linked to serious adverse effects and limited effectiveness in preventing metastasis. Here, we report a novel therapeutic strategy named starvation-immunotherapy, wherein an immune checkpoint inhibitor is combined with an ultra-long-acting L-asparaginase that is a fusion protein comprising L-asparaginase (ASNase) and an elastin-like polypeptide (ELP), termed ASNase-ELP. ASNase-ELP's thermosensitivity enables it to generate an in-situ depot following an intratumoral injection, yielding increased dose tolerance, improved pharmacokinetics, sustained release, optimized biodistribution, and augmented tumor retention compared to free ASNase. As a result, in murine models of oral cancer, melanoma, and cervical cancer, the antitumor efficacy of ASNase-ELP by selectively and sustainably depleting L-asparagine essential for tumor cell survival was substantially superior to that of ASNase or Cisplatin, a first-line anti-solid tumor medicine, without any observable adverse effects. Furthermore, the combination of ASNase-ELP and an immune checkpoint inhibitor was more effective than either therapy alone in impeding melanoma metastasis. Overall, the synergistic strategy of starvation-immunotherapy holds excellent promise in reshaping the therapeutic landscape of refractory metastatic tumors and offering a new alternative for next-generation oncology treatments.


Subject(s)
Asparaginase , Immune Checkpoint Inhibitors , Immunotherapy , Animals , Asparaginase/therapeutic use , Asparaginase/pharmacology , Asparaginase/chemistry , Immunotherapy/methods , Female , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Mice , Humans , Cell Line, Tumor , Drug Synergism , Elastin/chemistry , Elastin/metabolism , Neoplasm Metastasis , Mice, Inbred C57BL , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Tissue Distribution
7.
Biomaterials ; 312: 122712, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39098305

ABSTRACT

Immunosuppressive tumor microenvironment (ITM) severely limited the efficacy of immunotherapy against triple-negative breast cancer (TNBC). Herein, Apt-LPR, a light-activatable photodynamic therapy (PDT)/RNAi immune synergy-enhancer was constructed by co-loading miR-34a and photosensitizers in cationic liposomes (in phase III clinical trial). Interestingly, the introduction of tumor-specific aptamers creates a special "Liposome-Aptamer-Target" interface, where the aptamers are initially in a "lying down" state but transform to "standing up" after target binding. The interfacing mechanism was elaborately revealed by computational and practical experiments. This unique interface endowed Apt-LPR with neutralized surface potential of cationic liposomes to reduce non-specific cytotoxicity, enhanced DNase resistance to protect aptamers, and preserved target-binding ability for selective drug delivery. Upon near-infrared irradiation, the generated reactive oxygen species would oxidize unsaturated phospholipids to destabilize both liposomes and lysosomes, realizing stepwise lysosomal escape of miR-34a for tumor cell apoptosis and downregulation of PD-L1 to suppress immune escape. Together, tumor-associated antigens released from PDT-damaged mitochondria and endoplasmic reticulum could activate the suppressive immune cells to establish an "immune hot" milieu. The collaborative immune-enhancing strategy effectively aroused systemic antitumor immunity and inhibited primary and distal tumor progression as well as lung metastasis in 4T1 xenografted mouse models. The photo-controlled drug release and specific tumor-targeting capabilities of Apt-LPR were also visualized in MDA-MB-231 xenografted zebrafish models. Therefore, this photoswitchable PDT/RNAi immune stimulator offered a powerful approach to reprogramming ITM and reinforcing cancer immunotherapy efficacy.


Subject(s)
Liposomes , MicroRNAs , Photochemotherapy , Photosensitizing Agents , Triple Negative Breast Neoplasms , Tumor Microenvironment , Animals , Humans , Liposomes/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Photochemotherapy/methods , Tumor Microenvironment/drug effects , Cell Line, Tumor , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Female , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Mice , Aptamers, Nucleotide/chemistry , Delayed-Action Preparations/chemistry , RNA Interference , Zebrafish
8.
Biomaterials ; 312: 122746, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39106816

ABSTRACT

Postoperative radiotherapy remains the gold standard for malignant glioma treatment. Clinical limitations, including tumor growth between surgery and radiotherapy and the emergence of radioresistance, reduce treatment effectiveness and result in local disease progression. This study aimed to develop a local drug delivery system to inhibit tumor growth before radiotherapy and enhance the subsequent anticancer effects of limited-dose radiotherapy. We developed a compound of carboplatin-loaded hydrogel (CPH) incorporated with carboplatin-loaded calcium carbonate (CPCC) to enable two-stage (peritumoral and intracellular) release of carboplatin to initially inhibit tumor growth and to synergize with limited-dose radiation (10 Gy in a single fraction) to eliminate malignant glioma (ALTS1C1 cells) in a C57BL/6 mouse subcutaneous tumor model. The doses of carboplatin in CPH and CPCC treatments were 150 µL (carboplatin concentration of 5 mg/mL) and 15 mg (carboplatin concentration of 4.1 µg/mg), respectively. Mice receiving the combination of CPH-CPCC treatment and limited-dose radiation exhibited significantly reduced tumor growth volume compared to those receiving double-dose radiation alone. Furthermore, combining CPH-CPCC treatment with limited-dose radiation resulted in significantly longer progression-free survival than combining CPH treatment with limited-dose radiation. Local CPH-CPCC delivery synergized effectively with limited-dose radiation to eliminate mouse glioma, offering a promising solution for overcoming clinical limitations.


Subject(s)
Calcium Carbonate , Carboplatin , Glioma , Hydrogels , Mice, Inbred C57BL , Animals , Glioma/pathology , Glioma/drug therapy , Glioma/radiotherapy , Carboplatin/administration & dosage , Carboplatin/therapeutic use , Carboplatin/pharmacology , Hydrogels/chemistry , Cell Line, Tumor , Calcium Carbonate/chemistry , Mice , Drug Delivery Systems/methods , Drug Liberation , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy
9.
Biomaterials ; 312: 122721, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39106817

ABSTRACT

Silver nanoparticles (AgNPs) are a potential antiviral agent due to their ability to disrupt the viral particle or alter the virus metabolism inside the host cell. In vitro, AgNPs exhibit antiviral activity against the most common human respiratory viruses. However, their capacity to modulate immune responses during respiratory viral infections has yet to be explored. This study demonstrates that administering AgNPs directly into the lungs prior to infection can reduce viral loads and therefore virus-induced cytokines in mice infected with influenza virus or murine pneumonia virus. The prophylactic effect was diminished in mice with depleted lymphoid cells. We showed that AgNPs-treatment resulted in the recruitment and activation of lymphocytes in the lungs, particularly natural killer (NK) cells. Mechanistically, AgNPs enhanced the ability of alveolar macrophages to promote both NK cell migration and IFN-γ production. By contrast, following infection, in mice treated with AgNPs, NK cells exhibited decreased activation, indicating that these nanoparticles can regulate the potentially deleterious activation of these cells. Overall, the data suggest that AgNPs may possess prophylactic antiviral properties by recruiting and controlling the activation of lymphoid cells through interaction with alveolar macrophages.


Subject(s)
Killer Cells, Natural , Lung , Metal Nanoparticles , Orthomyxoviridae Infections , Silver , Animals , Silver/chemistry , Silver/pharmacology , Metal Nanoparticles/chemistry , Lung/virology , Lung/pathology , Lung/drug effects , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Mice , Killer Cells, Natural/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Mice, Inbred C57BL , Lymphocytes/drug effects , Lymphocytes/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Female , Lymphocyte Activation/drug effects
10.
Biomaterials ; 312: 122733, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39106819

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) demonstrates unique characteristics in anticancer therapies as it selectively induces apoptosis in cancer cells. However, most cancer cells are TRAIL-resistant. Odanacatib (ODN), a cathepsin K inhibitor, is considered a novel sensitizer for cancer treatment. Combination therapy between TRAIL and sensitizers is considered a potent platform that improves TRAIL-based anticancer therapies beyond TRAIL monotherapy. Herein, we developed ODN loaded poly(lactic-co-glycolic) nanoparticles conjugated to GST-TRAIL (TRAIL-ODN-PLGA-NPs) to target and treat TRAIL-resistant cancer. TRAIL-ODN-PLGA-NPs demonstrated a significant increase in cellular uptake via death receptors (DR5 and DR4) on surface of cancer cells. TRAIL-ODN-PLGA-NPs exposure destroyed more TRAIL-resistant cells compared to a single treatment with free drugs. The released ODN decreased the Raptor protein, thereby increasing damage to mitochondria by elevating reactive oxygen species (ROS) generation. Additionally, Bim protein stabilization improved TRAIL-resistant cell sensitization to TRAIL-induced apoptosis. The in vivo biodistribution study revealed that TRAIL-ODN-PLGA-NPs demonstrated high location and retention in tumor sites via the intravenous route. Furthermore, TRAIL-ODN-PLGA-NPs significantly inhibited xenograft tumor models of TRAIL-resistant Caki-1 and TRAIL-sensitive MDA-MB-231 cells.The inhibition was associated with apoptosis activation, Raptor protein stabilizing Bim protein downregulation, Bax accumulation, and mitochondrial ROS generation elevation. Additionally, TRAIL-ODN-PLGA-NPs affected the tumor microenvironment by increasing tumor necrosis factor-α and reducing interleukin-6. In conclusion, we evealed that our formulation demonstrated synergistic effects against TRAIL compared with the combination of free drug in vitro and in vivo models. Therefore, TRAIL-ODN-PLGA-NPs may be a novel candidate for TRAIL-induced apoptosis in cancer treatment.


Subject(s)
Antineoplastic Agents , Biphenyl Compounds , Drug Resistance, Neoplasm , Mice, Nude , Nanoparticles , Polylactic Acid-Polyglycolic Acid Copolymer , TNF-Related Apoptosis-Inducing Ligand , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Nanoparticles/chemistry , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Biphenyl Compounds/therapeutic use , Biphenyl Compounds/pharmacology , Biphenyl Compounds/chemistry , Drug Resistance, Neoplasm/drug effects , Apoptosis/drug effects , Mice , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Female , Tissue Distribution
11.
Biomaterials ; 312: 122742, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39106821

ABSTRACT

Hypertrophic scar (HS) tends to raised above skin level with high inflammatory microenvironment and excessive proliferation of myofibroblasts. The HS therapy remains challenging due to dense scar tissue which makes it hard to penetrate, and the side effects resulting from intralesional corticosteroid injection which is the mainstay treatment in clinic. Herein, bilayer microneedle patches combined with dexamethasone and colchicine (DC-MNs) with differential dual-release pattern is designed. Two drugs loaded in commercially available materials HA and PLGA, respectively. Specifically, after administration, outer layer rapidly releases the anti-inflammatory drug dexamethasone, which inhibits macrophage polarization to pro-inflammatory phenotype in scar tissue. Subsequently, inner layer degrades sustainedly, releasing antimicrotubular agent colchicine, which suppresses the overproliferation of myofibroblasts with extremely narrow therapeutic window, and inhibits the overexpression of collagen, as well as promotes the regular arrangement of collagen. Only applied once, DC-MNs directly delivered drugs to the scar tissue. Compared to traditional treatment regimen, DC-MNs significantly suppressed HS at lower dosage and frequency by differential dual-release design. Therefore, this study put forward the idea of integrated DC-MNs accompany the development of HS, providing a non-invasive, self-applicable, more efficient and secure strategy for treatment of HS.


Subject(s)
Anti-Inflammatory Agents , Cicatrix, Hypertrophic , Colchicine , Dexamethasone , Myofibroblasts , Needles , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/pathology , Animals , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Dexamethasone/pharmacology , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Colchicine/pharmacology , Colchicine/administration & dosage , Mice , Drug Delivery Systems , Humans , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
12.
Biomaterials ; 312: 122749, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39121725

ABSTRACT

The prevalence of Alzheimer's disease (AD) is increasing globally due to population aging. However, effective clinical treatment strategies for AD still remain elusive. The mechanisms underlying AD onset and the interplay between its pathological factors have so far been unclear. Evidence indicates that AD progression is ultimately driven by neuronal loss, which in turn is caused by neuroapoptosis and neuroinflammation. Therefore, the inhibition of neuroapoptosis and neuroinflammation could be a useful anti-AD strategy. Nonetheless, the delivery of active drug agents into the brain parenchyma is hindered by the blood-brain barrier (BBB). To address this challenge, we fabricated a black phosphorus nanosheet (BP)-based methylene blue (MB) delivery system (BP-MB) for AD therapy. After confirming the successful preparation of BP-MB, we proved that its BBB-crossing ability was enhanced under near-infrared light irradiation. In vitro pharmacodynamics analysis revealed that BP and MB could synergistically scavenge excessive reactive oxygen species (ROS) in okadaic acid (OA)-treated PC12 cells and lipopolysaccharide (LPS)-treated BV2 cells, thus efficiently reversing neuroapoptosis and neuroinflammation. To study in vivo pharmacodynamics, we established a mouse model of AD mice, and behavioral tests confirmed that BP-MB treatment could successfully improve cognitive function in these animals. Notably, the results of pathological evaluation were consistent with those of the in vitro assays. The findings demonstrated that BP-MB could scavenge excessive ROS and inhibit Tau hyperphosphorylation, thereby alleviating downstream neuroapoptosis and regulating the polarization of microglia from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. Overall, this study highlights the therapeutic potential of a smart nanomedicine with the capability of reversing neuroapoptosis and neuroinflammation for AD treatment.


Subject(s)
Alzheimer Disease , Apoptosis , Blood-Brain Barrier , Methylene Blue , Nanomedicine , Neuroinflammatory Diseases , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Apoptosis/drug effects , PC12 Cells , Neuroinflammatory Diseases/drug therapy , Rats , Mice , Nanomedicine/methods , Methylene Blue/pharmacology , Methylene Blue/therapeutic use , Male , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL
13.
Biomaterials ; 312: 122747, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39142219

ABSTRACT

Directly administering medication to inflamed intestinal sites for treating ulcerative colitis (UC), poses significant challenges like retention time, absorption variability, side effects, drug stability, and non-specific delivery. Recent advancements in therapy to treat colitis aim to improve local drug availability that is enema therapy at the site of inflammation, thereby reducing systemic adverse effects. Nevertheless, a key limitation lies in enemas' inability to sustain medication in the colon due to rapid peristaltic movement, diarrhea, and poor local adherence. Therefore, in this work, we have developed site-specific thiolated mucoadhesive anionic nanoliposomes to overcome the limitations of conventional enema therapy. The thiolated delivery system allows prolonged residence of the delivery system at the inflamed site in the colon, confirmed by the adhesion potential of thiolated nanoliposomes using in-vitro and in-vivo models. To further provide therapeutic efficacy thiolated nanoliposomes were loaded with gallic acid (GA), a natural compound known for its antibacterial, antioxidant, and potent anti-inflammatory properties. Consequently, Gallic Acid-loaded Thiolated 2,6 DALP DMPG (GATh@APDL) demonstrates the potential for targeted adhesion to the inflamed colon, facilitated by their small size 100 nm and anionic nature. Therapeutic studies indicate that this formulation offers protective effects by mitigating colonic inflammation, downregulating the expression of NF-κB, HIF-1α, and MMP-9, and demonstrating superior efficacy compared to the free GA enema. The encapsulated GA inhibits the NF-κB expression, leading to enhanced expression of MUC2 protein, thereby promoting mucosal healing in the colon. Furthermore, GATh@APDL effectively reduces neutrophil infiltration and regulates immune cell quantification in colonic lamina propria. Our findings suggest that GATh@APDL holds promise for alleviating UC and addressing the limitations of conventional enema therapy.


Subject(s)
Colitis, Ulcerative , Liposomes , Sulfhydryl Compounds , Colitis, Ulcerative/drug therapy , Liposomes/chemistry , Animals , Sulfhydryl Compounds/chemistry , Humans , Nanoparticles/chemistry , Mice , Colon/pathology , Colon/drug effects , Colon/metabolism , Male , Drug Delivery Systems
14.
Biomaterials ; 312: 122731, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39153324

ABSTRACT

Tumor-associated inflammation drives cancer progression and therapy resistance, often linked to the infiltration of monocyte-derived tumor-associated macrophages (TAMs), which are associated with poor prognosis in various cancers. To advance immunotherapies, testing on immunocompetent pre-clinical models of human tissue is crucial. We have developed an in vitro model of microvascular networks with tumor spheroids or patient tissues to assess monocyte trafficking into tumors and evaluate immunotherapies targeting the human tumor microenvironment. Our findings demonstrate that macrophages in vascularized breast and lung tumor models can enhance monocyte recruitment via CCL7 and CCL2, mediated by CSF-1R. Additionally, a multispecific antibody targeting CSF-1R, CCR2, and neutralizing TGF-ß (CSF1R/CCR2/TGF-ß Ab) repolarizes TAMs towards an anti-tumoral M1-like phenotype, reduces monocyte chemoattractant protein secretion, and blocks monocyte migration. This antibody also inhibits monocyte recruitment in patient-specific vascularized tumor models. In summary, this vascularized tumor model recapitulates the monocyte recruitment cascade, enabling functional testing of innovative therapeutic antibodies targeting TAMs in the tumor microenvironment.


Subject(s)
Monocytes , Receptor, Macrophage Colony-Stimulating Factor , Receptors, CCR2 , Tumor Microenvironment , Humans , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , Monocytes/metabolism , Monocytes/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Female , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Mice , Cell Movement/drug effects , Neoplasms/immunology , Neoplasms/pathology
15.
J Biomed Opt ; 30(Suppl 1): S13702, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39034960

ABSTRACT

Significance: Near-infrared autofluorescence (NIRAF) utilizes the natural autofluorescence of parathyroid glands (PGs) to improve their identification during thyroid surgeries, reducing the risk of inadvertent removal and subsequent complications such as hypoparathyroidism. This study evaluates NIRAF's effectiveness in real-world surgical settings, highlighting its potential to enhance surgical outcomes and patient safety. Aim: We evaluate the effectiveness of NIRAF in detecting PGs during thyroidectomy and central neck dissection and investigate autofluorescence characteristics in both fresh and paraffin-embedded tissues. Approach: We included 101 patients diagnosed with papillary thyroid cancer who underwent surgeries in 2022 and 2023. We assessed NIRAF's ability to locate PGs, confirmed via parathyroid hormone assays, and involved both junior and senior surgeons. We measured the accuracy, speed, and agreement levels of each method and analyzed autofluorescence persistence and variation over 10 years, alongside the expression of calcium-sensing receptor (CaSR) and vitamin D. Results: NIRAF demonstrated a sensitivity of 89.5% and a negative predictive value of 89.1%. However, its specificity and positive predictive value (PPV) were 61.2% and 62.3%, respectively, which are considered lower. The kappa statistic indicated moderate to substantial agreement (kappa = 0.478; P < 0.001 ). Senior surgeons achieved high specificity (86.2%) and PPV (85.3%), with substantial agreement (kappa = 0.847; P < 0.001 ). In contrast, junior surgeons displayed the lowest kappa statistic among the groups, indicating minimal agreement (kappa = 0.381; P < 0.001 ). Common errors in NIRAF included interference from brown fat and eschar. In addition, paraffin-embedded samples retained stable autofluorescence over 10 years, showing no significant correlation with CaSR and vitamin D levels. Conclusions: NIRAF is useful for PG identification in thyroid and neck surgeries, enhancing efficiency and reducing inadvertent PG removals. The stability of autofluorescence in paraffin samples suggests its long-term viability, with false positives providing insights for further improvements in NIRAF technology.


Subject(s)
Optical Imaging , Parathyroid Glands , Spectroscopy, Near-Infrared , Thyroidectomy , Humans , Parathyroid Glands/surgery , Parathyroid Glands/metabolism , Male , Female , Middle Aged , Optical Imaging/methods , Adult , Spectroscopy, Near-Infrared/methods , Paraffin Embedding/methods , Aged , Thyroid Cancer, Papillary/surgery , Thyroid Cancer, Papillary/pathology , Thyroid Cancer, Papillary/metabolism , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/analysis
16.
Can J Urol ; 31(4): 11921-11930, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39217515

ABSTRACT

INTRODUCTION: Renal cell carcinoma is as the most prevalent form of kidney cancer, with the clear cell subtype comprising approximately 75% of cases. The identification of predictive and prognostic biomarkers has emerged as a crucial area of research within the field. Despite advancements in treatment, metastatic renal cell carcinoma presents formidable challenges, with survival rates heavily dependent upon the optimal choice of treatment. MATERIALS AND METHODS: This review summarizes the current literature regarding the prognostic and predictive value of biomarkers in patients with renal cell carcinoma. We conducted a comprehensive literature search to identify studies that reference biomarkers of interest in this domain. We selected studies based on their relevance, publication date, and the quality of the research. Data from these selected papers were compiled and analyzed to provide an overview of the current understanding and advancements in the field. The findings were then synthesized into a concise discussion highlighting the state of biomarker research in renal cell carcinoma today. RESULTS AND CONCLUSIONS: While various nucleic acid and protein biomarkers have shown promise in other malignancies, their application in renal cell carcinoma remains limited by the lack of validated predictors. This review aims to highlight the pressing need for robust predictive and prognostic biomarkers in renal cell carcinoma to guide clinicians in tailoring optimal therapeutic strategies. The discussion encompasses the limitations of existing markers and underscores the significance of the most recent advancements within the field. Despite these strides, the clinical application of renal cell carcinoma biomarkers requires further study and validation.


Subject(s)
Biomarkers, Tumor , Carcinoma, Renal Cell , Kidney Neoplasms , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Humans , Kidney Neoplasms/pathology , Biomarkers, Tumor/blood , Prognosis , Predictive Value of Tests
17.
Can J Urol ; 31(4): 11955-11962, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39217520

ABSTRACT

INTRODUCTION: Most men diagnosed with very-low and low-risk prostate cancer are candidates for active surveillance; however, there is still a misclassification risk. We examined whether PI-RADS category 4 or 5 combined with ISUP 1 on prostate biopsy predicts upgrading and/or adverse pathology at radical prostatectomy. MATERIALS AND METHODS: A total of 127 patients had ISUP 1 cancer on biopsy after multiparametric MRI (mpMRI) and then underwent radical prostatectomy. We then evaluated them for ISUP upgrading and/or adverse pathology on radical prostatectomy. RESULTS: Eight-nine patients (70%) were diagnosed with PI-RADS 4 or 5 lesions. ISUP upgrading was significantly higher among patients with PI-RADS 4-5 lesions (84%) compared to patients with equivocal or non-suspicious mpMRI findings (26%, p < 0.001). Both PI-RADS 4-5 lesions (OR 24.3, 95% CI 7.3, 80.5, p < 0.001) and stage T2 on DRE (OR 5.9, 95% CI 1.2, 29.4, p = 0.03) were independent predictors of upgrading on multivariate logistic regression analysis. Men with PI-RADS 4-5 lesions also had significantly more extra-prostatic extension (51% vs. 3%, p < 0.001) and positive surgical margins (16% vs. 3%. p = 0.03). The only independent predictor of adverse pathology was PI-RADS 4-5 (OR 21.7, 95% CI 4.8, 99, p < 0.001). CONCLUSION: PI-RADS 4 or 5 lesions on mpMRI were strong independent predictors of upgrading and adverse pathology. Incorporating mpMRI findings when selecting patients for active surveillance must be further evaluated in future studies.


Subject(s)
Multiparametric Magnetic Resonance Imaging , Prostatectomy , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Prostatic Neoplasms/diagnostic imaging , Prostatectomy/methods , Middle Aged , Aged , Predictive Value of Tests , Neoplasm Grading , Prostate/pathology , Prostate/diagnostic imaging , Retrospective Studies , Biopsy , Neoplasm Staging , Magnetic Resonance Imaging , Watchful Waiting , Risk Assessment
18.
Clinics (Sao Paulo) ; 79: 100477, 2024.
Article in English | MEDLINE | ID: mdl-39217675

ABSTRACT

OBJECTIVES: To determine the prevalence and association of HPV and Herpesviruses in saliva and tissue samples of patients with orofacial tumors. METHODS: Biopsies of tumors were done, and saliva samples were collected from patients with orofacial tumors for the determination of viruses using nested multiplex PCR. Independent variables were sex, age, comorbidities, tumor stage, and length of stay. Outcome variables were the presence or absence of herpesviruses and HPV. Descriptive summaries and inferential statistics were done. RESULTS: A hundred patients were included in the study. Prevalence of herpesviruses and HPV were 17.6 % and 57.0 % in tumors, and 48.3 % and 60.0 % in the saliva of patients respectively. Herpesviruses detected included EBV (21.3 %), HHV-7 (11.2 %), CMV (6.7 %), HSV-1 (5.1 %), HSV-2 (1.1 %), VZV (1.1 %), and Kaposi sarcoma virus (0.6 %). The most prevalent HPV genotypes were HPV-42 (29 %), HPV-43 (22.7 %), HPV-52 (22.2 %), HPV-39 (18.8 %), and HPV-18 (9.1 %). The odds of EBV being detected in malignant orofacial tumors were 2 times that of benign orofacial tumors. HPV DNA in the saliva of patients with orofacial tumors was 69.7 %, compared to 18.2 % of the control sample (p < 0.001). The median length of stay for all participants was 6.5 days, those associated with viruses stayed longer. CONCLUSION: There was a high prevalence of Herpesviruses and HPV in saliva and tumor samples of patients with orofacial tumors, signalling some potential for more work to be done in this area.


Subject(s)
Herpesviridae , Papillomaviridae , Saliva , Humans , Female , Saliva/virology , Male , Middle Aged , Herpesviridae/isolation & purification , Herpesviridae/genetics , Adult , Papillomaviridae/isolation & purification , Papillomaviridae/genetics , Aged , Biopsy , Young Adult , Papillomavirus Infections/virology , Papillomavirus Infections/epidemiology , Herpesviridae Infections/virology , Herpesviridae Infections/epidemiology , Prevalence , DNA, Viral/analysis , Mouth Neoplasms/virology , Mouth Neoplasms/pathology , Adolescent , Brazil/epidemiology , Aged, 80 and over , Multiplex Polymerase Chain Reaction , Human Papillomavirus Viruses
19.
Biochemistry (Mosc) ; 89(7): 1239-1250, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39218021

ABSTRACT

Huntington's disease (HD) is an incurable hereditary disease caused by expansion of the CAG repeats in the HTT gene encoding the mutant huntingtin protein (mHTT). Despite numerous studies in cellular and animal models, the mechanisms underlying the biological role of mHTT and its toxicity to striatal neurons have not yet been established and no effective therapy for HD patients has been developed so far. We produced and characterized a new line of dermal fibroblasts (HDDF, Huntington's disease dermal fibroblasts) from a patient with a confirmed HD diagnosis. We also studied the growth characteristics of HDDF cells, stained them for canonical markers, karyotyped these cells, and investigated their phenotype. HDDF cells was successfully reprogrammed into induced striatal neurons via transdifferentiation. The new fibroblast line can be used as a cell model to study the biological role of mHTT and manifestations of HD pathogenesis in both fibroblasts and induced neuronal cells obtained from them by reprogramming techniques.


Subject(s)
Fibroblasts , Huntington Disease , Huntington Disease/pathology , Huntington Disease/metabolism , Huntington Disease/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Neurons/metabolism , Neurons/pathology , Cell Line , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Cell Transdifferentiation , Male
20.
Biochemistry (Mosc) ; 89(7): 1300-1312, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39218026

ABSTRACT

To date, the molecular mechanisms of the common neurodegenerative disorder Parkinson's disease (PD) are unknown and, as a result, there is no neuroprotective therapy that may stop or slow down the process of neuronal cell death. The aim of the current study was to evaluate the prospects of using the mTOR molecule as a potential target for PD therapy due to the dose-dependent effect of mTOR kinase activity inhibition on cellular parameters associated with, PD pathogenesis. The study used peripheral blood monocyte-derived macrophages and SH-SY5Y neuroblastoma cell line. As a result, we have for the first time showed that inhibition of mTOR by Torin1 only at a concentration of 100 nM affects the level of the lysosomal enzyme glucocerebrosidase (GCase), encoded by the GBA1 gene. Mutations in GBA1 are considered a high-risk factor for PD development. This concentration led a decrease in pathological phosphorylated alpha-synuclein (Ser129), an increase in its stable tetrameric form with no changes in the lysosomal enzyme activities and concentrations of lysosphingolipids. Our findings suggest that inhibition of the mTOR protein kinase could be a promising approach for developing therapies for PD, particularly for GBA1-associated PD.


Subject(s)
Lysosomes , Macrophages , Parkinson Disease , TOR Serine-Threonine Kinases , alpha-Synuclein , Humans , alpha-Synuclein/metabolism , Lysosomes/metabolism , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Cell Line, Tumor , Macrophages/metabolism , Macrophages/drug effects , Neuroblastoma/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Dose-Response Relationship, Drug , Glucosylceramidase/metabolism , Glucosylceramidase/antagonists & inhibitors , Naphthyridines
SELECTION OF CITATIONS
SEARCH DETAIL