Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29.292
Filter
1.
Biomed Pharmacother ; 179: 117423, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39260328

ABSTRACT

Hepatocellular carcinoma (HCC), a hypervascular tumor, is the most frequent primary malignant tumor of the liver. Angiogenesis inhibitors, such as endogenous angiogenesis inhibitors, are essential for HCC therapy and have generated significant interest owing to their safety, efficacy, and multitargeting attributes. Canstatin is an angiogenesis inhibitor derived from the basement membrane and exerts anti-tumor effects. However, the inhibitory effects and underlying mechanisms of action of canstatin on HCC remain unclear. Therefore, in this study, HepG2 and Huh7 cells were used to investigate the inhibitory effects of recombinant canstatin on HCC cells. Subsequently, the biosafety and inhibitory effects of recombinant canstatin on tumor growth were investigated in a xenograft animal model of liver cancer. Canstatin inhibited the growth of liver cancer cells by regulating their proliferation, apoptosis, and migration. Additionally, it suppressed the occurrence and progression of HCC by modulating the HIF-1α/VEGF signaling pathway. In mice, canstatin exerted no discernible harmful side effects and suppressed the growth of HCC subcutaneous xenograft tumors. Overall, our findings shed light on the molecular pathways underlying canstatin-induced HCC cell death that may help develop novel HCC treatments.


Subject(s)
Apoptosis , Carcinoma, Hepatocellular , Cell Proliferation , Disease Progression , Hypoxia-Inducible Factor 1, alpha Subunit , Liver Neoplasms , Mice, Nude , Signal Transduction , Vascular Endothelial Growth Factor A , Xenograft Model Antitumor Assays , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Animals , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Signal Transduction/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Vascular Endothelial Growth Factor A/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Mice , Recombinant Proteins/pharmacology , Hep G2 Cells , Cell Movement/drug effects , Mice, Inbred BALB C , Cell Line, Tumor , Angiogenesis Inhibitors/pharmacology , Male
2.
Cells ; 13(18)2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39329750

ABSTRACT

An essential aspect of harnessing the potential of pluripotent stem cells (PSCs) and their derivatives for regenerative medicine is the development of animal-free and chemically defined conditions for ex vivo cultivation. PSCs, including embryonic and induced PSCs (iPSCs), are in the early stages of clinical trials for various indications, including degenerative diseases and traumatic injury. A key step in the workflows generating these cells for more widespread clinical use is their safe and robust ex vivo cultivation. This entails optimization of cell culture media and substrates that are safe and consistent while maintaining robust functionality. Here, we describe the design of a human vitronectin (hVTN) variant with improved manufacturability in a bacterial expression system along with improved function in comparison to wild-type VTN and other previously characterized polypeptide fragments. In conjunction with an animal component-free media formulation, our hVTN fragment provides animal-free conditions for the enhanced expansion of iPSCs. This hVTN variant also supports the reprogramming of PBMCs into iPSCs. Furthermore, we show that these iPSCs can be efficiently differentiated into the three major germ layers and cortical neurons, thereby closing the loop on a completely defined animal-free workflow for cell types relevant for regenerative medicine.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells , Recombinant Proteins , Vitronectin , Vitronectin/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Recombinant Proteins/pharmacology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Proliferation , Cellular Reprogramming/genetics , Workflow , Mice
3.
ACS Appl Mater Interfaces ; 16(38): 50305-50320, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39255049

ABSTRACT

Excessive fibrotic scar formation during skin defect repair poses a formidable challenge, impeding the simultaneous acceleration of wound healing and prevention of scar formation and hindering the restoration of skin integrity and functionality. Drawing inspiration from the structural, compositional, and biological attributes of skin, we developed a hydrogel containing modified recombinant human collagen type III and thiolated hyaluronic acid to address the challenges of regenerating skin appendages and improving the recovery of skin functions after injury by reducing fibrotic scarring. The hydrogel displayed favorable biocompatibility, antioxidant properties, angiogenic potential, and fibroblast migration stimulation in vitro. In a rat full-layer defect model, it reduced inflammation, promoted microvascular formation, and significantly enhanced the wound healing speed and effectiveness. Additionally, by upregulating fibrosis-associated genes, such as TGFB1, it facilitated collagen accumulation and a beneficial balance between type I and type III collagen, potentially expediting skin regeneration and functional recovery. In conclusion, the utilization of rhCol III-HS demonstrated considerable potential as a wound dressing, offering a highly effective strategy for the restoration and rejuvenation of complete skin defects.


Subject(s)
Cicatrix , Collagen Type III , Hydrogels , Recombinant Proteins , Wound Healing , Wound Healing/drug effects , Collagen Type III/metabolism , Collagen Type III/genetics , Collagen Type III/chemistry , Animals , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Rats , Cicatrix/pathology , Cicatrix/drug therapy , Recombinant Proteins/pharmacology , Recombinant Proteins/chemistry , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/pathology , Male , Fibroblasts/drug effects , Fibroblasts/metabolism , Polysaccharides/chemistry , Polysaccharides/pharmacology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology
4.
Front Immunol ; 15: 1441908, 2024.
Article in English | MEDLINE | ID: mdl-39224597

ABSTRACT

Introduction: The antiviral activity of recombinant bovine interferon lambda 3 (bovIFN-λ3) against bovine viral diarrhea virus (BVDV) has been demonstrated in vitro in Madin-Darby bovine kidney cells (MDBK) and in vivo in cattle. However, anti-BVDV activity of bovIFN-λ3 has not been studied in bovine respiratory tract epithelial cells, supposedly a primary target of BVDV infection when entering the host by the oronasal route. Methods: Here we investigated the anti-BVDV activity of bovIFN-λ3 in bovine turbinate-derived primary epithelial cells (BTu) using BVDV infection and immunoperoxidase staining, TCID50, RT-qPCR, DNA and transcriptome sequencing, and transfection with plasmids containing the two subunits, IL-28Rα and IL-10Rß that constitute the bovIFN-λ3 receptor. Results: Our immunoperoxidase staining, RT-qPCR, and TCID50 results show that while BVDV was successfully cleared in MDBK cells treated with bovIFN-λ3 and bovIFN-α, only the latter, bovIFN-α, cleared BVDV in BTu cells. Preincubation of MDBK cells with bovIFN-λ3 before BVDV infection was needed to induce optimal antiviral state. Both cell types displayed intact type I and III IFN signaling pathways and expressed similar levels of IL-10Rß subunit of the type III IFN receptor. Sequencing of PCR amplicon of the IL-28Rα subunit revealed intact transmembrane domain and lack of single nucleotide polymorphisms (SNPs) in BTu cells. However, RT-qPCR and transcriptomic analyses showed a lower expression of IL-28Rα transcripts in BTu cells as compared to MDBK cells. Interestingly, transfection of BTu cells with a plasmid encoding IL-28Rα subunit, but not IL-10Rß subunit, established the bovIFN-λ3 sensitivity showing similar anti-BVDV activity to the response in MDBK cells. Conclusion: Our results demonstrate that the sensitivity of cells to bovIFN-λ3 depends not only on the quality but also of the quantity of the IL-28Rα subunit of the heterodimeric receptor. A reduction in IL-28Rα transcript expression was detected in BTu as compared to MDBK cells, despite the absence of spliced variants or SNPs. The establishment of bovIFN-λ3 induced anti-BVDV activity in BTu cells transfected with an IL-28Rα plasmid suggests that the level of expression of this receptor subunit is crucial for the specific antiviral activity of type III IFN in these cells.


Subject(s)
Interferon Lambda , Interferons , Turbinates , Animals , Cattle , Interferons/metabolism , Interferons/immunology , Turbinates/virology , Turbinates/immunology , Turbinates/metabolism , Antiviral Agents/pharmacology , Diarrhea Viruses, Bovine Viral/immunology , Diarrhea Viruses, Bovine Viral/physiology , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Epithelial Cells/virology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Interleukins/genetics , Interleukins/pharmacology , Interleukins/immunology , Interleukins/metabolism , Cell Line , Bovine Virus Diarrhea-Mucosal Disease/immunology , Bovine Virus Diarrhea-Mucosal Disease/virology , Recombinant Proteins/pharmacology , Interleukin-10 Receptor beta Subunit/genetics , Interleukin-10 Receptor beta Subunit/metabolism , Receptors, Cytokine
5.
Respir Res ; 25(1): 330, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227918

ABSTRACT

BACKGROUND: Vascular endothelial damage is involved in the development and exacerbation of ventilator-induced lung injury (VILI). Pulmonary endothelial glycocalyx and neutrophil extracellular traps (NETs) are endothelial protective and damaging factors, respectively; however, their dynamics in VILI and the effects of recombinant thrombomodulin and antithrombin on these dynamics remain unclear. We hypothesized that glycocalyx degradation and NETs are induced by VILI and suppressed by recombinant thrombomodulin, recombinant antithrombin, or their combination. METHODS: VILI was induced in male C57BL/6J mice by intraperitoneal lipopolysaccharide injection (20 mg/kg) and high tidal volume ventilation (20 mL/kg). In the intervention groups, recombinant thrombomodulin, recombinant antithrombin, or their combination was administered at the start of mechanical ventilation. Glycocalyx degradation was quantified by measuring serum syndecan-1, fluorescence-labeled lectin intensity, and glycocalyx-occupied area in the pulmonary vascular lumen. Double-stranded DNA in the bronchoalveolar fluid and fluorescent areas of citrullinated histone H3 and myeloperoxidase were quantified as NET formation. RESULTS: Serum syndecan-1 increased, and lectin fluorescence intensity decreased in VILI. Electron microscopy revealed decreases in glycocalyx-occupied areas within pulmonary microvessels in VILI. Double-stranded DNA levels in the bronchoalveolar lavage fluid and the fluorescent area of citrullinated histone H3 and myeloperoxidase in lung tissues increased in VILI. Recombinant thrombomodulin, recombinant antithrombin, and their combination reduced glycocalyx injury and NET marker levels. There was little difference in glycocalyx injury and NET makers between the intervention groups. CONCLUSION: VILI induced glycocalyx degradation and NET formation. Recombinant thrombomodulin and recombinant antithrombin attenuated glycocalyx degradation and NETs in our VILI model. The effect of their combination did not differ from that of either drug alone. Recombinant thrombomodulin and antithrombin have the potential to be therapeutic agents for biotrauma in VILI.


Subject(s)
Antithrombins , Endotoxemia , Extracellular Traps , Glycocalyx , Mice, Inbred C57BL , Recombinant Proteins , Thrombomodulin , Ventilator-Induced Lung Injury , Animals , Glycocalyx/metabolism , Glycocalyx/drug effects , Glycocalyx/pathology , Thrombomodulin/metabolism , Thrombomodulin/administration & dosage , Extracellular Traps/metabolism , Extracellular Traps/drug effects , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Mice , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology , Ventilator-Induced Lung Injury/drug therapy , Ventilator-Induced Lung Injury/prevention & control , Endotoxemia/metabolism , Endotoxemia/pathology , Endotoxemia/drug therapy , Endotoxemia/chemically induced , Antithrombins/pharmacology , Lung/metabolism , Lung/drug effects , Lung/pathology , Disease Models, Animal , Syndecan-1/metabolism
6.
Toxins (Basel) ; 16(9)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39330867

ABSTRACT

Hemolytic-uremic syndrome (HUS) is a rare complication of an infection with Shiga toxin (Stx)-producing Escherichia coli (STEC-HUS), characterized by severe acute kidney injury, thrombocytopenia and microangiopathic hemolytic anemia, and specific therapy is still lacking. Thrombomodulin (TM) is a multi-domain transmembrane endothelial cell protein and its N-terminal domain has been implicated in the pathophysiology of some cases of HUS. Indeed, the administration of recombinant human TM (rhTM) may have efficacy in HUS. We used a Stx-based murine model of HUS to characterize the role of the N-terminal domain of TM. We show that mice lacking that domain (TMLed (-/-)) are more sensitive to Stx, with enhanced HUS progression seen at 4 days and increased mortality at 7 days post-HUS induction. In spite of these changes, renal function was less affected in surviving Stx-challenged TMLed (-/-) mice compared to their wild-type counterparts TMLed (+/+) at 7 days. Contrary to few clinical case reports from Japan, the administration of rhTM (0.06 mg/kg) to wild-type mice (C57BL/6J) with HUS did not protect against disease progression. This overall promising, but also contradictory body of evidence, requires further systematic preclinical and clinical investigations to clarify the role of TM in HUS as a potential therapeutic strategy.


Subject(s)
Hemolytic-Uremic Syndrome , Recombinant Proteins , Thrombomodulin , Thrombomodulin/genetics , Thrombomodulin/therapeutic use , Animals , Hemolytic-Uremic Syndrome/drug therapy , Humans , Recombinant Proteins/therapeutic use , Recombinant Proteins/pharmacology , Shiga Toxin/toxicity , Mice , Disease Models, Animal , Protein Domains
7.
Int J Mol Sci ; 25(18)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39337581

ABSTRACT

Galectins are a class of lectins that are extensively expressed in all organisms. Galectins are involved in a range of functions, including early development, tissue regeneration, cancer and inflammation. It has been shown that galectin-8 is expressed in the villous and extravillous trophoblast (EVT) cells of the human placenta; however, its physiological role in pregnancy establishment has not been elucidated. Taking these factors into account, we investigated the functional role of galectin-8 in HTR-8/SVneo cells-a human EVT cell line-and human primary cytotrophoblast cells isolated from a first-trimester placenta. We analyzed the effects of recombinant human galectin-8 (rh galectin-8) on the adhesion, migration and invasion of HTR-8/SVneo cells. We used qPCR, cell-based ELISA (cELISA) and gelatin zymography to study the effects of galectin-8 on mediators of these processes, such as integrin subunits alpha-1 and beta-1 and matrix metalloproteinases (MMPs)-2 and -9, on the mRNA and protein levels. Further, we studied the effects of galectin-8 on primary cytotrophoblast cells' invasion. Galectin-8 stimulated the adhesion, migration and invasion of HTR-8/SVneo cells, as well as the invasion of primary cytotrophoblasts. In addition, the MMP-2 and -9 levels were increased, while the expression of integrins alpha-1 and beta-1 was not affected. Galectin-8 has the ability to positively affect EVTs' invasion, so it can be considered a significant factor in the trophoblast cell invasion process.


Subject(s)
Cell Adhesion , Cell Movement , Galectins , Matrix Metalloproteinase 2 , Trophoblasts , Humans , Trophoblasts/metabolism , Trophoblasts/cytology , Galectins/metabolism , Cell Movement/drug effects , Pregnancy , Female , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 2/genetics , Cell Line , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Placenta/metabolism , Placenta/cytology , Recombinant Proteins/pharmacology , Recombinant Proteins/metabolism
8.
Protein Expr Purif ; 223: 106562, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39094814

ABSTRACT

Previous studies have demonstrated the presence of chitinase in Bacillus velezensis through extensive genomic sequencing and experimental analyses. However, the detailed structure, functional roles, and antifungal activity of these chitinases remain poorly characterized. In this study, genomic screening identified three genes-chiA, chiB, and lpmo10-associated with chitinase degradation in B. velezensis S161. These genes encode chitinases ChiA and ChiB, and lytic polysaccharide monooxygenase LPMO10. Both ChiA and ChiB contain two CBM50 binding domains and one catalytic domain, whereas LPMO10 includes a signal peptide and a single catalytic domain. The chitinases ChiA, its truncated variant ChiA2, and ChiB were heterologously expressed in Escherichia coli. The purified enzymes efficiently degraded colloidal chitin and inhibited the spore germination of Penicillium digitatum. Notably, even after losing one CBM50 domain, the resultant enzyme, consisting of the remaining CBM50 domain and the catalytic domain, maintained its colloidal chitin hydrolysis and antifungal activity, indicating commendable stability. These results underscore the role of B. velezensis chitinases in suppressing plant pathogenic fungi and provide a solid foundation for developing and applying chitinase-based biocontrol strategies.


Subject(s)
Antifungal Agents , Bacillus , Chitinases , Penicillium , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Bacillus/enzymology , Bacillus/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/pharmacology , Chitin/chemistry , Chitinases/chemistry , Chitinases/pharmacology , Escherichia coli , Penicillium/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
9.
Int J Biol Macromol ; 278(Pt 1): 134632, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39128757

ABSTRACT

Recombinant high-density lipoprotein (rHDL) as anti-atherosclerosis (AS) vehicle has unique advantages including multiple anti-atherogenic functions and homing features to plaques. However, rHDL may be converted into dysfunctional forms due to complex treatment during preparation. Herein, oxidation-induced dysfunction of non-split HDL and rHDL was initially investigated. It was found that although both non-split HDL and rHDL showed oxidative dysfunction behavior, non-split HDL demonstrated superior oxidation defense compared to rHDL due to its intact composition and avoidance of overprocessing such as split and recombination. Unfortunately, in vivo oxidative stress could compromise the functionality of HDL. Therefore, surface engineering of non-split HDL and rHDL with cascade antioxidant enzyme analogues Ebselen and mitochondrial-targeted TPGS-Tempo was conducted to construct a dual-line defense HDL nano system (i.e., T@E-HDLs/rHDL), aiming to restore plaque redox balance and preserving the physiological function of HDL. Results indicated that both T@E-HDLs and rHDLs performed without distinction and exhibited greater resistance to oxidative stress damage as well as better functions than unmodified HDLs in macrophage foam cells. Overall, the modification of dual antioxidants strategy bridges the gap between non-split HDL and rHDL, and provides a promising resolution for the dilemmas of oxidative stress in plaques and HDL self dysfunction.


Subject(s)
Antioxidants , Lipoproteins, HDL , Oxidative Stress , Recombinant Proteins , Oxidative Stress/drug effects , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/pharmacology , Antioxidants/pharmacology , Recombinant Proteins/pharmacology , Animals , Humans , Mice , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Oxidation-Reduction/drug effects , Isoindoles/pharmacology , Organoselenium Compounds/pharmacology , Organoselenium Compounds/chemistry
10.
Domest Anim Endocrinol ; 89: 106880, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39163657

ABSTRACT

Porcine adrenocorticotrophic hormone (ACTH) has been considered valid for the ACTH stimulation test (ACTHST) in humans and dogs; however, its safety and efficacy for use in cats are unknown. Also, the equivalence between 5 µg/kg and 125 µg/cat dose of synthetic corticotropin (1-24 ACTH - cosyntropin/tetracosactide) is assumed for ACTHST in cats. This study evaluated the safety and effectiveness of different porcine recombinant ACTH doses for the ACTHST in healthy cats and its equivalence with tetracosactide. The study was divided into two arms. The first evaluated safety and equivalence of intravenous 1 µg/kg, 5 µg/kg, or 125 µg/cat porcine ACTH in seven healthy cats for the ACTHST evaluating basal and post-ACTH androstenedione, aldosterone, cortisol, and progesterone concentrations. In the second arm, the equivalence of the 125 µg/cat porcine ACTH dose was evaluated compared to results obtained using 125 µg/cat of tetracosactide in ten healthy cats regarding cortisol responses. In all tests, several cat-friendly strategies were adopted, and the ACTHST protocol involved basal and 60-minute post-ACTH blood sampling and intravenous ACTH injection. No adverse reactions were documented, and no tested cat showed any complications during the study. No porcine ACTH tested dose significantly increased androstenedione secretion. In contrast, all tested doses were able to increase progesterone concentration significantly (P < 0.05), and Δ-progesterone in response to 5 µg/kg or 125 µg/cat was considered equivalent (P > 0.99). The 125 µg/cat dose promoted greater responses for both cortisol and aldosterone, characterized by Δ-cortisol (P = 0.009) and Δ-aldosterone (P = 0.004). Despite equivalent Δ-cortisol results in response to 5 µg/kg or 125 µg/cat (P = 0.18); post-ACTH results of cortisol in response to 5 µg/kg only approximate statistical significance when compared with basal (P = 0.07). Porcine ACTH and tetracosactide significantly increased post-ACTH cortisol concentration (P < 0.0001) while the Δ-cortisol was slightly greater in response to the porcine ACTH (P = 0.006). These results suggest porcine ACTH could be an alternative source of corticotropin for the ACTHST in cats; however, maximum corticoadrenal stimulation seemed more reliable in response to a 125 µg/cat regarding cortisol and aldosterone.


Subject(s)
Adrenocorticotropic Hormone , Cosyntropin , Hydrocortisone , Animals , Cats/physiology , Adrenocorticotropic Hormone/pharmacology , Adrenocorticotropic Hormone/administration & dosage , Female , Male , Hydrocortisone/blood , Cosyntropin/pharmacology , Cosyntropin/administration & dosage , Swine , Recombinant Proteins/pharmacology , Aldosterone/blood , Progesterone/blood , Progesterone/pharmacology , Progesterone/administration & dosage , Androstenedione/blood , Androstenedione/pharmacology , Dose-Response Relationship, Drug
SELECTION OF CITATIONS
SEARCH DETAIL