Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29.209
Filter
1.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1498-1508, 2024 May 25.
Article in Chinese | MEDLINE | ID: mdl-38783811

ABSTRACT

To investigate the role of recombinant mussel mucin in wound healing, we aimed to prepare this mucin using Pichia pastoris as the host microbe. Our method involved constructing a genetically engineered strain of P. pastoris that expressed a fusion protein consisting of Mfp-3 and preCol-P peptide segments of mussel. After fermentation and purification, we obtained a pure recombinant mussel mucin product. We then conducted experiments to evaluate its effect at both the cellular and animal levels. At the cellular level, we examined its impact on the proliferation and migration of mouse fibroblast L929. At the animal level, we assessed its ability to promote wound healing after full-layer skin resection in rats. Our results showed that the recombinant mussel mucin protein has a content of 90.28% and a purity of 96.49%. The content of 3,4-dihydroxyphenylalanine (DOPA) was 0.73 wt%, and the endotoxin content was less than 0.5 EU/mg. Importantly, the recombinant mussel mucin protein significantly promoted both the migration and proliferation of mouse fibroblast, as well as the wound healing in rat skin. In conclusion, our findings demonstrate that recombinant mussel mucin has the potential to promote wound healing and can be considered a promising medical biomaterial.


Subject(s)
Wound Healing , Animals , Wound Healing/drug effects , Rats , Mice , Mucins/metabolism , Mucins/genetics , Bivalvia , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Fibroblasts/metabolism , Fibroblasts/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/pharmacology , Male , Rats, Sprague-Dawley , Saccharomycetales
2.
Int Immunopharmacol ; 134: 112188, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38728880

ABSTRACT

Neuroinflammation is one of the extensive secondary injury processes that aggravate metabolic and cellular dysfunction and tissue loss following spinal cord injury (SCI). Thus, an anti-inflammatory strategy is crucial for modulating structural and functional restoration during the stage of acute and chronic SCI. Recombinant fibroblast growth factor 4 (rFGF4) has eliminated its mitogenic activity and demonstrated a metabolic regulator for alleviating hyperglycemia in type 2 diabetes and liver injury in non-alcoholic steatohepatitis. However, it remains to be explored whether or not rFGF4 has a neuroprotective effect for restoring neurological disorders, such as SCI. Here, we identified that rFGF4 could polarize microglia/macrophages into the restorative M2 subtype, thus exerting an anti-inflammatory effect to promote neurological functional recovery and nerve fiber regeneration after SCI. Importantly, these effects by rFGF4 were related to triggering PI3K/AKT/GSK3ß and attenuating TLR4/NF-κB signaling axes. Conversely, gene silencing of the PI3K/AKT/GSK3ß signaling or pharmacological reactivation of the TLR4/NF-κB axis aggravated inflammatory reaction. Thus, our findings highlight rFGF4 as a potentially therapeutic regulator for repairing SCI, and its outstanding effect is associated with regulating macrophage/microglial polarization.


Subject(s)
Glycogen Synthase Kinase 3 beta , Macrophages , Microglia , NF-kappa B , Nerve Regeneration , Recovery of Function , Spinal Cord Injuries , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/drug therapy , Animals , Microglia/drug effects , Microglia/metabolism , Macrophages/drug effects , Macrophages/immunology , Nerve Regeneration/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , NF-kappa B/metabolism , Recombinant Proteins/therapeutic use , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Mice , Male , Axons/metabolism , Axons/drug effects , Axons/pathology , Proto-Oncogene Proteins c-akt/metabolism , Mice, Inbred C57BL , Rats, Sprague-Dawley , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phenotype , Rats , Humans , Disease Models, Animal , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology
3.
Mol Med Rep ; 30(1)2024 Jul.
Article in English | MEDLINE | ID: mdl-38757335

ABSTRACT

Thrombin, which plays a crucial role in hemostasis, is also implicated in cancer progression. In the present study, the effects of the thrombin­targeting recombinant tyrosine­sulfated madanin­1 on cancer cell behavior and signaling pathways compared with madanin­1 wild­type (WT) were investigated. Recombinant madanin­1 2 sulfation (madanin­1 2S) and madanin­1 WT proteins were generated using Escherichia coli. SKOV3 and MDA­MB­231 cells were treated with purified recombinant proteins with or without thrombin stimulation. Migration and invasion of cells were analyzed by wound healing assay and Transwell assay, respectively. Thrombin markedly increased cell migration and invasion in both SKOV3 and MDA­MB­231 cells, which were significantly suppressed by madanin­1 2S (P<0.05). Madanin­1 2S also significantly suppressed thrombin­induced expression of phosphorylated (p)­Akt and p­extracellular signal­regulated kinase in both cell lines (P<0.05), whereas madanin­1 WT had no effect on the expression levels of these proteins in MDA­MB­231 cells. Furthermore, madanin­1 2S significantly reversed the effects of thrombin on E­cadherin, N­cadherin and vimentin expression in MDA­MB­231 cells (P<0.05), whereas madanin­1 WT did not show any effect. In conclusion, madanin­1 2S suppressed the migration and invasion of cancer cells more effectively than madanin­1 WT. It is hypothesized that inhibiting thrombin via the sulfated form of madanin­1 may be a potential candidate for enhanced cancer therapy; however, further in vivo validation is required.


Subject(s)
Cell Movement , Recombinant Proteins , Thrombin , Humans , Cell Movement/drug effects , Thrombin/pharmacology , Cell Line, Tumor , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Tyrosine/metabolism , Tyrosine/pharmacology , Cadherins/metabolism , Cadherins/genetics
4.
Anticancer Res ; 44(6): 2359-2367, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821601

ABSTRACT

BACKGROUND/AIM: The alkylating agent trabectedin, which binds the minor groove of DNA, is second-line therapy for soft-tissue sarcoma but has only moderate efficacy. The aim of the present study was to determine the synergistic efficacy of recombinant methioninase (rMETase) and trabectedin on fibrosarcoma cells in vitro, compared with normal fibroblasts. MATERIALS AND METHODS: HT1080 human fibrosarcoma cells expressing green fluorescent protein (GFP) in the nucleus and red fluorescent protein (RFP) in the cytoplasm and Hs27 normal human fibroblasts, were used. Each cell line was cultured in vitro and divided into four groups: no-treatment control; trabectedin treated; rMETase treated; and trabectedin plus rMETase treated. The dual-color HT1080 cells were used to quantitate nuclear fragmentation in each treatment group. RESULTS: The combination of rMETase and trabectedin was highly synergistic to decrease HT1080 cell viability. In contrast, there was no synergy on Hs27 cells. Moreover, nuclear fragmentation occurred synergistically with the combination of trabectedin and rMETase on dual-color HT1080 cells. CONCLUSION: The combination treatment of trabectedin plus rMETase was highly synergistic on fibrosarcoma cells in vitro suggesting that the combination can improve the outcome of trabectedin alone in future clinical studies. The lack of synergy of rMETase and trabectedin on normal fibroblasts suggests the combination is not toxic to normal cells. Synergy of the two drugs may be due to the high rate of nuclear fragmentation on treated HT1080 cells, and the late-S/G2 cell-cycle block of cancer cells by rMETase, which is a target for trabectedin. The results of the present study suggest the future clinical potential of the combination of rMETase and trabectedin for soft-tissue sarcoma.


Subject(s)
Carbon-Sulfur Lyases , Cell Survival , Dioxoles , Drug Synergism , Fibroblasts , Fibrosarcoma , Tetrahydroisoquinolines , Trabectedin , Humans , Fibrosarcoma/drug therapy , Fibrosarcoma/pathology , Fibrosarcoma/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Trabectedin/pharmacology , Carbon-Sulfur Lyases/pharmacology , Carbon-Sulfur Lyases/administration & dosage , Tetrahydroisoquinolines/pharmacology , Dioxoles/pharmacology , Cell Survival/drug effects , Recombinant Proteins/pharmacology , Cell Line, Tumor , Antineoplastic Agents, Alkylating/pharmacology , Cell Nucleus/metabolism , Cell Nucleus/drug effects
5.
Cell Mol Biol (Noisy-le-grand) ; 70(5): 243-247, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38814208

ABSTRACT

Burns are the most severe type of trauma, and the resulting ischemia and hypoxia damage can promote the dysfunction and even failure of tissues and organs throughout the body, endangering patients' life safety. Recombinant human growth hormone (rhGH) has the functions of promoting protein synthesis to reverse negative nitrogen balance, accelerating wound healing, and improving immune function, which is widely used in the treatment of burns. However, the exact mechanism and pathway of rhGH's action is not yet fully understood. In this study, we observed the wound repair effect of recombinant human growth hormone (rhGH) on burned mice and further analyzed the mechanism of action, which can provide more comprehensive reference opinions for clinical practice. First, by establishing a burn mouse model and and intervening with different doses of rhGH, we found that the wound healing capacity of mice was significantly enhanced and the inflammatory and oxidative stress responses were obviously alleviated, confirming the excellent promotion of wound repair and anti-inflammatory and antioxidant effects of rhGH. Subsequently, we found that the expression of p-ERK1/2/ERK1/2, EGF, TGF-ß, and VEGF proteins was elevated in the traumatic tissues of mice after rhGH intervention, suggesting that the pathway of action of rhGH might be related to the activation of ERK pathway to promote the regeneration of traumatic capillaries.


Subject(s)
Burns , Human Growth Hormone , MAP Kinase Signaling System , Neovascularization, Physiologic , Recombinant Proteins , Wound Healing , Animals , Burns/drug therapy , Burns/pathology , Wound Healing/drug effects , MAP Kinase Signaling System/drug effects , Recombinant Proteins/pharmacology , Mice , Human Growth Hormone/pharmacology , Humans , Neovascularization, Physiologic/drug effects , Male , Oxidative Stress/drug effects , Disease Models, Animal , Vascular Endothelial Growth Factor A/metabolism , Transforming Growth Factor beta/metabolism , Mice, Inbred C57BL , Epidermal Growth Factor/pharmacology , Angiogenesis
6.
Stem Cell Res Ther ; 15(1): 144, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38764077

ABSTRACT

BACKGROUND: The aim of this study was to evaluate potential synergistic effects of a single, local application of human umbilical cord MSC-derived sEVs in combination with a low dose of recombinant human rhBMP-2 to promote the regeneration of a metaphyseal femoral defect in an osteoporotic rat model. METHODS: 6 weeks after induction of osteoporosis by bilateral ventral ovariectomy and administration of a special diet, a total of 64 rats underwent a distal femoral metaphyseal osteotomy using a manual Gigli wire saw. Defects were stabilized with an adapted Y-shaped mini-locking plate and were subsequently treated with alginate only, or alginate loaded with hUC-MSC-sEVs (2 × 109), rhBMP-2 (1.5 µg), or a combination of sEVs and rhBMP-2 (n = 16 for each group). 6 weeks post-surgery, femora were evaluated by µCT, descriptive histology, and biomechanical testing. RESULTS: Native radiographs and µCT analysis confirmed superior bony union with callus formation after treatment with hUC-MSC-sEVs in combination with a low dose of rhBMP-2. This finding was further substantiated by histology, showing robust defect consolidation 6 weeks after treatment. Torsion testing of the explanted femora revealed increased stiffness after application of both, rhBMP-2 alone, or in combination with sEVs, whereas torque was only significantly increased after treatment with rhBMP-2 together with sEVs. CONCLUSION: The present study demonstrates that the co-application of hUC-MSC-sEVs can improve the efficacy of rhBMP-2 to promote the regeneration of osteoporotic bone defects.


Subject(s)
Bone Morphogenetic Protein 2 , Extracellular Vesicles , Femur , Osteoporosis , Recombinant Proteins , Umbilical Cord , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/genetics , Osteoporosis/pathology , Rats , Female , Humans , Femur/pathology , Femur/drug effects , Femur/diagnostic imaging , Umbilical Cord/cytology , Extracellular Vesicles/metabolism , Bone Regeneration/drug effects , Rats, Sprague-Dawley , Transforming Growth Factor beta/pharmacology , Disease Models, Animal , X-Ray Microtomography , Mesenchymal Stem Cells/metabolism
7.
Chem Biol Drug Des ; 103(5): e14533, 2024 May.
Article in English | MEDLINE | ID: mdl-38684373

ABSTRACT

Hirudin is one of the specific inhibitors of thrombin, which has been confirmed to have strong bioactivities, including inhibiting tumors. However, the function and mechanism of hirudin and protease-activated receptor 1 (PAR-1) in diffuse large B-cell lymphoma (DLBCL) have not been clear. Detecting the expression PAR-1 in DLBCL tissues and cells by RT-qPCR and IHC. Transfected sh-NC, sh-PAR-1, or pcDNA3.1-PAR-1 in DLBCL cells or processed DLBCL cells through added thrombin, Vorapaxar, Recombinant hirudin (RH), or Na2S2O4 and co-culture with EA.hy926. And built DLBCL mice observed tumor growth. Detecting the expression of related genes by RT-qPCR, Western blot, IHC, and immunofluorescence, measured the cellular hypoxia with Hypoxyprobe-1 Kit, and estimated the cell inflammatory factors, proliferation, migration, invasion, and apoptosis by ELISA, CCK-8, flow cytometry, wound-healing and Transwell. Co-immunoprecipitation and pull-down measurement were used to verify the relationship. PAR-1 was highly expressed in DLBCL tissues and cells, especially in SUDHL2. Na2S2O4 induced SUDHL2 hypoxia, and PAR-1 did not influence thrombin-activated hypoxia. PAR-1 could promote SUDHL2 proliferation, migration, and invasion, and it was unrelated to cellular hypoxia. PAR-1 promoted proliferation, migration, and angiogenesis of EA.hy926 or SUDHL2 through up-regulation vascular endothelial growth factor (VEGF). RH inhibited tumor growth, cell proliferation, and migration, promoted apoptosis of DLBCL, and inhibited angiogenesis by down-regulating PAR-1-VEGF. RH inhibits proliferation, migration, and angiogenesis of DLBCL cells by down-regulating PAR-1-VEGF.


Subject(s)
Apoptosis , Cell Proliferation , Hirudins , Lymphoma, Large B-Cell, Diffuse , Neovascularization, Pathologic , Receptor, PAR-1 , Recombinant Proteins , Vascular Endothelial Growth Factor A , Humans , Hirudins/pharmacology , Receptor, PAR-1/metabolism , Receptor, PAR-1/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Animals , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Mice , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Apoptosis/drug effects , Recombinant Proteins/pharmacology , Recombinant Proteins/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Angiogenesis
8.
Clin Appl Thromb Hemost ; 30: 10760296241247558, 2024.
Article in English | MEDLINE | ID: mdl-38656136

ABSTRACT

INTRODUCTION: Andexanet alfa (AA) - zhzo, recombinant coagulation factor Xa, is an approved antidote for oral Xa inhibitors (apixaban and rivaroxaban). Unfractionated heparin (UFH) is commonly used for therapeutic, interventional, and surgical indications. Protamine sulfate (PrSO4) is frequently used to neutralize UFH. This study aimed to investigate the comparative neutralization profiles of AA and PrSO4 for heparins of bovine, ovine, and porcine origin. MATERIALS AND METHODS: The neutralization effect of PrSO4 at 25 µg/ml and AA at 100 µg/ml was studied on an approximate surgical/interventional concentration of heparin by supplementing whole blood with each of the heparins at 25 µg/ml. For the clotting profile (activated partial thromboplastin time: aPTT), amidolytic (anti-Xa and anti-IIa), and thrombin generation assay each of the heparin were supplemented from -10-0.62 µg/ml. RESULTS: In the whole blood ACT studies, all three heparins produced strong anti-coagulant effects (400-450 seconds) compared to saline (130-150 seconds). Both AA and PrSO4 almost fully neutralized the anti-coagulant effects of heparins (140-160 seconds). Both antidotes completely reversed the anticoagulant effects of all three heparins in the aPTT and thrombin generation assay. However, PrSO4 was more effective in neutralizing the anti-Xa, and anti-IIa effects than AA, which only partially neutralized these effects. CONCLUSION: Andexanet alfa at 100 µg/ml effectively neutralizes the therapeutic and surgical/interventional concentrations of heparins in in-vitro settings. While differences in the anti-Xa, and anti-IIa effects between heparins were noted, anti-coagulant effect of these agents in the aPTT assay were comparable. A similar neutralization profile was observed in the ACT and thrombin generation assays by both agents.


Subject(s)
Anticoagulants , Factor Xa , Heparin , Protamines , Recombinant Proteins , Animals , Cattle , Protamines/pharmacology , Heparin/pharmacology , Sheep , Factor Xa/metabolism , Recombinant Proteins/pharmacology , Anticoagulants/pharmacology , Swine , Humans , Blood Coagulation/drug effects , Factor Xa Inhibitors/pharmacology
9.
Cell Commun Signal ; 22(1): 236, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38650003

ABSTRACT

BACKGROUND: The preservation of retinal ganglion cells (RGCs) and the facilitation of axon regeneration are crucial considerations in the management of various vision-threatening disorders. Therefore, we investigate the efficacy of interleukin-4 (IL-4), a potential therapeutic agent, in promoting neuroprotection and axon regeneration of retinal ganglion cells (RGCs) as identified through whole transcriptome sequencing in an in vitro axon growth model. METHODS: A low concentration of staurosporine (STS) was employed to induce in vitro axon growth. Whole transcriptome sequencing was utilized to identify key target factors involved in the molecular mechanism underlying axon growth. The efficacy of recombinant IL-4 protein on promoting RGC axon growth was validated through in vitro experiments. The protective effect of recombinant IL-4 protein on somas of RGCs was assessed using RBPMS-specific immunofluorescent staining in mouse models with optic nerve crush (ONC) and N-methyl-D-aspartic acid (NMDA) injury. The protective effect on RGC axons was evaluated by anterograde labeling of cholera toxin subunit B (CTB), while the promotion of RGC axon regeneration was assessed through both anterograde labeling of CTB and immunofluorescent staining for growth associated protein-43 (GAP43). RESULTS: Whole-transcriptome sequencing of staurosporine-treated 661 W cells revealed a significant upregulation in intracellular IL-4 transcription levels during the process of axon regeneration. In vitro experiments demonstrated that recombinant IL-4 protein effectively stimulated axon outgrowth. Subsequent immunostaining with RBPMS revealed a significantly higher survival rate of RGCs in the rIL-4 group compared to the vehicle group in both NMDA and ONC injury models. Axonal tracing with CTB confirmed that recombinant IL-4 protein preserved long-distance projection of RGC axons, and there was a notably higher number of surviving axons in the rIL-4 group compared to the vehicle group following NMDA-induced injury. Moreover, intravitreal delivery of recombinant IL-4 protein substantially facilitated RGC axon regeneration after ONC injury. CONCLUSION: The recombinant IL-4 protein exhibits the potential to enhance the survival rate of RGCs, protect RGC axons against NMDA-induced injury, and facilitate axon regeneration following ONC. This study provides an experimental foundation for further investigation and development of therapeutic agents aimed at protecting the optic nerve and promoting axon regeneration.


Subject(s)
Axons , Interleukin-4 , Nerve Regeneration , Retinal Ganglion Cells , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Animals , Interleukin-4/pharmacology , Axons/drug effects , Axons/metabolism , Nerve Regeneration/drug effects , Mice , Mice, Inbred C57BL , Optic Nerve Injuries/pathology , Optic Nerve Injuries/drug therapy , N-Methylaspartate/pharmacology , Staurosporine/pharmacology , Neuroprotective Agents/pharmacology , Recombinant Proteins/pharmacology
10.
Int J Biol Macromol ; 266(Pt 2): 131276, 2024 May.
Article in English | MEDLINE | ID: mdl-38561117

ABSTRACT

Skin aging, a complex physiological progression marked by collagen degradation, poses substantial challenges in dermatology. Recombinant collagen emerges as a potential option for skin revitalization, yet its application is constrained by difficulties in forming hydrogels. We have for the first time developed a highly bioactive Tetrakis(hydroxymethyl) phosphonium chloride (THPC)-crosslinked recombinant collagen hydrogel implant for aging skin rejuvenation. THPC demonstrated superior crosslinking efficiency compared to traditional agents such as EDC/NHS and BDDE, achieving complete recombinant collagen crosslinking at minimal concentrations and effectively inducing hydrogel formation. THPC's four reactive hydroxymethyl groups facilitate robust crosslinking with triple helical recombinant collagen, producing hydrogels with enhanced mechanical strength, excellent injectability, increased stability, and greater durability. Moreover, the hydrogel exhibited remarkable biocompatibility and bioactivity, significantly promoting the proliferation, adhesion, and migration of human foreskin fibroblast-1. In photoaged mice skin models, the THPC-crosslinked collagen hydrogel implant notably improved dermal density, skin elasticity, and reduced transepidermal water loss, creating a conducive environment for fibroblast activity and healthy collagen regeneration. Additionally, it elevated superoxide dismutase (SOD) activity and displayed substantial anti-calcification properties. The THPC-crosslinked recombinant collagen hydrogel implant presents an innovative methodology in combating skin aging, offering significant promise in dermatology and tissue engineering.


Subject(s)
Collagen , Hydrogels , Rejuvenation , Skin Aging , Hydrogels/chemistry , Animals , Collagen/chemistry , Skin Aging/drug effects , Mice , Humans , Cross-Linking Reagents/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Fibroblasts/drug effects , Skin/drug effects , Cell Proliferation/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology
11.
Res Vet Sci ; 173: 105274, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38669867

ABSTRACT

A systematic review and meta-analysis were conducted to assess the impact of recombinant bovine somatotropin (rbST) on lactation performance, feed efficiency, and blood metabolites in dairy cows. In the systematic review, articles were selected based on the following criteria: (1) Data focusing on the influence of bovine somatotropin doses on milk production; (2) Submission of original data; (3) Articles published in journals; and (4) Articles in English or Portuguese. The analysis of variance was used with a completely randomized design and mixed models methodology. Polynomial regression was applied to significant fixed effects (rbST dose). The use of rbST resulted in increased milk yield and 4% fat-corrected milk yield, while fat, protein, and lactose contents remained unaffected. Dry matter and metabolizable energy intakes, as well as milk/feed efficiency, exhibited a linear increase, but body condition score (BCS) was negatively impacted. The administration of rbST led to higher blood concentrations of triglycerides and insulin. Cows treated with rbST showed a 23% increase in non-esterified fatty acid (NEFA) concentrations compared to non-treated cows. Additionally, growth factors IGF-1 and IGF-2 displayed a linear increase with rbST treatment. In summary, rbST administration increased milk yield and fat-corrected milk yield without affecting milk components. However, despite increasing intake, it resulted in BCS losses and alterations in blood parameters such as NEFA, IGF-1, and IGF-2.


Subject(s)
Growth Hormone , Lactation , Animals , Lactation/drug effects , Cattle/physiology , Female , Growth Hormone/blood , Growth Hormone/pharmacology , Growth Hormone/administration & dosage , Milk/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/administration & dosage , Animal Feed/analysis
12.
Int J Mol Sci ; 25(8)2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38673749

ABSTRACT

The anticancer potential of Levilactobacillus brevis KU15176 against the stomach cancer cell line AGS has been reported previously. In this study, we aimed to analyze the genome of L. brevis KU15176 and identify key genes that may have potential anticancer properties. Among potential anticancer molecules, the role of arginine deiminase (ADI) in conferring an antiproliferative functionality was confirmed. In vitro assay against AGS cell line confirmed that recombinant ADI from L. brevis KU15176 (ADI_br, 5 µg/mL), overexpressed in E. coli BL21 (DE3), exerted an inhibitory effect on AGS cell growth, resulting in a 65.32% reduction in cell viability. Moreover, the expression of apoptosis-related genes, such as bax, bad, caspase-7, and caspase-3, as well as the activity of caspase-9 in ADI_br-treated AGS cells, was higher than those in untreated (culture medium-only) cells. The cell-scattering behavior of ADI_br-treated cells showed characteristics of apoptosis. Flow cytometry analyses of AGS cells treated with ADI_br for 24 and 28 h revealed apoptotic rates of 11.87 and 24.09, respectively, indicating the progression of apoptosis in AGS cells after ADI_br treatment. This study highlights the potential of ADI_br as an effective enzyme for anticancer applications.


Subject(s)
Apoptosis , Cell Proliferation , Hydrolases , Levilactobacillus brevis , Stomach Neoplasms , Humans , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Hydrolases/metabolism , Hydrolases/genetics , Hydrolases/pharmacology , Levilactobacillus brevis/genetics , Levilactobacillus brevis/enzymology , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
13.
ACS Biomater Sci Eng ; 10(5): 3188-3202, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38592024

ABSTRACT

Chronic wound repair is a clinical treatment challenge. The development of multifunctional hydrogels is of great significance in the key aspects of treating chronic wounds, including reducing oxidative stress, promoting angiogenesis, and improving the natural remodeling of extracellular matrix and immune regulation. In this study, we prepared a composite hydrogel, sodium alginate (SA)@MnO2/recombinant humanized collagen III (RHC)/mesenchymal stem cells (MSCs), composed of SA, MnO2 nanoparticles, RHC, and MSCs. The hydrogel has high mechanical properties and good biocompatibility. In vitro, SA@MnO2/RHC/MSCs hydrogel effectively enhanced the formation of intricate tubular structures and angiogenesis and showed synergistic effects on cell proliferation and migration. In vivo, the SA@MnO2/RHC/MSCs hydrogel enhanced diabetes wound healing, rapid re-epithelization, favorable collagen deposition, and abundant wound angiogenesis. These findings demonstrated that the combined effects of SA, MnO2, RHC, and MSCs synergistically accelerate healing, resulting in a reduced healing time. These observed healing effects demonstrated the potential of this multifunctional hydrogel to transform chronic wound care and improve patient outcomes.


Subject(s)
Hydrogels , Manganese Compounds , Mesenchymal Stem Cells , Oxides , Wound Healing , Wound Healing/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Animals , Manganese Compounds/chemistry , Manganese Compounds/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Humans , Oxides/chemistry , Oxides/pharmacology , Diabetes Mellitus, Experimental , Cell Proliferation/drug effects , Collagen/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Alginates/chemistry , Alginates/pharmacology , Male , Mice
14.
Stem Cell Res Ther ; 15(1): 124, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38679735

ABSTRACT

BACKGROUND: Recombinant human bone morphogenetic protein 2 (rhBMP-2) and human bone marrow mesenchymal stromal cells (hBM-MSCs) have been thoroughly studied for research and translational bone regeneration purposes. rhBMP-2 induces bone formation in vivo, and hBM-MSCs are its target, bone-forming cells. In this article, we studied how rhBMP-2 drives the multilineage differentiation of hBM-MSCs both in vivo and in vitro. METHODS: rhBMP-2 and hBM-MSCs were tested in an in vivo subcutaneous implantation model to assess their ability to form mature bone and undergo multilineage differentiation. Then, the hBM-MSCs were treated in vitro with rhBMP-2 for short-term or long-term cell-culture periods, alone or in combination with osteogenic, adipogenic or chondrogenic media, aiming to determine the role of rhBMP-2 in these differentiation processes. RESULTS: The data indicate that hBM-MSCs respond to rhBMP-2 in the short term but fail to differentiate in long-term culture conditions; these cells overexpress the rhBMP-2 target genes DKK1, HEY-1 and SOST osteogenesis inhibitors. However, in combination with other differentiation signals, rhBMP-2 acts as a potentiator of multilineage differentiation, not only of osteogenesis but also of adipogenesis and chondrogenesis, both in vitro and in vivo. CONCLUSIONS: Altogether, our data indicate that rhBMP-2 alone is unable to induce in vitro osteogenic terminal differentiation of hBM-MSCs, but synergizes with other signals to potentiate multiple differentiation phenotypes. Therefore, rhBMP-2 triggers on hBM-MSCs different specific phenotype differentiation depending on the signalling environment.


Subject(s)
Bone Morphogenetic Protein 2 , Cell Differentiation , Mesenchymal Stem Cells , Osteogenesis , Recombinant Proteins , Humans , Adipogenesis/drug effects , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Chondrogenesis/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
15.
Int J Biol Macromol ; 268(Pt 1): 131723, 2024 May.
Article in English | MEDLINE | ID: mdl-38649072

ABSTRACT

Endometrial injury poses a significant challenge in tissue regeneration, with type III collagen (COL III) playing a pivotal role in maintaining endometrial integrity and facilitating repair. Our study explored the utility of recombinant human type III collagen (RHC) as an intervention for endometrial damage. To address the challenges associated with the inherent instability and rapid degradation of COL III in vivo, we developed an RHC-HA hydrogel by conjugating RHC with hyaluronic acid (HA), thus ensuring a more stable and sustained delivery. Our findings suggested that the RHC-HA hydrogel significantly promoted endometrial regeneration and restored fertility. The hydrogel facilitated prolonged retention of RHC in the uterus, leading to a substantial improvement in the repair process. The synergistic interaction between RHC and HA greatly enhances cell proliferation and adhesion, surpassing the efficacy of HA or RHC alone. Additionally, the RHC-HA hydrogel demonstrated notable anti-fibrotic effects, which are crucial for preventing abnormalities during endometrial healing. These findings suggested that the RHC-HA hydrogel presented a therapeutic strategy in the treatment of uterine endometrial injuries, which may improve female reproductive health.


Subject(s)
Collagen Type III , Endometrium , Extracellular Matrix , Hyaluronic Acid , Hydrogels , Recombinant Proteins , Regeneration , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Female , Endometrium/drug effects , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Recombinant Proteins/pharmacology , Recombinant Proteins/administration & dosage , Animals , Collagen Type III/metabolism , Extracellular Matrix/drug effects , Regeneration/drug effects , Cell Proliferation/drug effects , Biomimetic Materials/pharmacology , Biomimetic Materials/chemistry , Rats , Cell Adhesion/drug effects
16.
J Physiol ; 602(9): 1939-1951, 2024 May.
Article in English | MEDLINE | ID: mdl-38606903

ABSTRACT

Recombinant human proteoglycan 4 (rhPRG4) is a macromolecular mucin-like glycoprotein that is classically studied as a lubricant within eyes and joints. Given that endogenously produced PRG4 is present within atherosclerotic lesions and genetic PRG4 deficiency increases atherosclerosis susceptibility in mice, in the current study we investigated the anti-atherogenic potential of chronic rhPRG4 treatment. Female low-density lipoprotein receptor knockout mice were fed an atherogenic Western-type diet for 6 weeks and injected three times per week intraperitoneally with 0.5 mg rhPRG4 or PBS as control. Treatment with rhPRG4 was associated with a small decrease in plasma-free cholesterol levels, without a change in cholesteryl ester levels. A marked increase in the number of peritoneal foam cells was detected in response to the peritoneal rhPRG4 administration, which could be attributed to elevated peritoneal leukocyte MSR1 expression levels. However, rhPRG4-treated mice exhibited significantly smaller aortic root lesions of 278 ± 21 × 103 µm2 compared with 339 ± 15 × 103 µm2 in the aortic root of control mice. The overall decreased atherosclerosis susceptibility coincided with a shift in the monocyte and macrophage polarization states towards the patrolling and anti-inflammatory M2-like phenotypes, respectively. Furthermore, rhPRG4 treatment significantly reduced macrophage gene expression levels as well as plasma protein levels of the pro-inflammatory/pro-atherogenic cytokine TNF-alpha. In conclusion, we have shown that peritoneal administration and subsequent systemic exposure to rhPRG4 beneficially impacts the inflammatory state and reduces atherosclerosis susceptibility in mice. Our findings highlight that PRG4 is not only a lubricant but also acts as an anti-inflammatory agent. KEY POINTS: Endogenously produced proteoglycan 4 is found in atherosclerotic lesions and its genetic deficiency in mice is associated with enhanced atherosclerosis susceptibility. In this study we investigated the anti-atherogenic potential of chronic treatment with recombinant human PRG4 in hypercholesterolaemic female low-density lipoprotein receptor knockout mice. We show that recombinant human PRG4 stimulates macrophage foam cell formation, but also dampens the pro-inflammatory state of monocyte/macrophages, eventually leading to a significant reduction in plasma TNF-alpha levels and a lowered atherosclerosis susceptibility. Our findings highlight that peritoneal recombinant human PRG4 treatment can execute effects both locally and systemically and suggest that it will be of interest to study whether rhPRG4 treatment is also able to inhibit the progression and/or induce regression of previously established atherosclerotic lesions.


Subject(s)
Atherosclerosis , Inflammation , Mice, Knockout , Proteoglycans , Receptors, LDL , Recombinant Proteins , Animals , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Female , Proteoglycans/pharmacology , Proteoglycans/metabolism , Proteoglycans/genetics , Receptors, LDL/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/administration & dosage , Mice , Humans , Inflammation/drug therapy , Inflammation/metabolism , Mice, Inbred C57BL , Aorta/metabolism , Aorta/drug effects , Aorta/pathology , Macrophages/metabolism , Macrophages/drug effects , Foam Cells/metabolism , Foam Cells/drug effects
17.
Int J Biol Macromol ; 267(Pt 2): 131610, 2024 May.
Article in English | MEDLINE | ID: mdl-38621565

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is a neurotrophic protein that promotes neuronal survival, increases neurotransmitter synthesis, and has potential therapeutic effects in neurodegenerative and psychiatric diseases, but its drug development has been limited by the fact that recombinant proteins of BDNF are unstable and do not penetrate the blood-brain barrier (BBB). In this study, we fused a TAT membrane-penetrating peptide with BDNF to express a recombinant protein (TBDNF), which was then PEG-modified to P-TBDNF. Protein characterization showed that P-TBDNF significantly improved the stability of the recombinant protein and possessed the ability to penetrate the BBB, and in cellular experiments, P-TBDNF prevented MPTP-induced nerve cell oxidative stress damage, apoptosis and inflammatory response, and its mechanism of action was closely related to the activation of tyrosine kinase B (TrkB) receptor and inhibition of microglia activation. In animal experiments, P-TBDNF improved motor and cognitive deficits in MPTP mice and inhibited pathological changes in Parkinson's disease (PD). In conclusion, this paper is expected to reveal the mechanism of action of P-TBDNF in inhibiting neurotoxicity, provide a new way for treating PD, and lay the foundation for the future development of recombinant P-TBDNF.


Subject(s)
Brain-Derived Neurotrophic Factor , Neuroprotective Agents , Animals , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/pharmacology , Brain-Derived Neurotrophic Factor/genetics , Mice , Neuroprotective Agents/pharmacology , Recombinant Proteins/pharmacology , Blood-Brain Barrier/metabolism , Male , Oxidative Stress/drug effects , Humans , Apoptosis/drug effects , Receptor, trkB/metabolism , Neurons/metabolism , Neurons/drug effects , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Mice, Inbred C57BL
18.
J Agric Food Chem ; 72(18): 10439-10450, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38676695

ABSTRACT

Trypsin inhibitors derived from plants have various pharmacological activities and promising clinical applications. In our previous study, a Bowman-Birk-type major trypsin inhibitor from foxtail millet bran (FMB-BBTI) was extracted with antiatherosclerotic activity. Currently, we found that FMB-BBTI possesses a prominent anticolorectal cancer (anti-CRC) activity. Further, a recombinant FMB-BBTI (rFMB-BBTI) was successfully expressed in a soluble manner in host strain Escherichia coli. BL21 (DE3) was induced by isopropyl-ß-d-thiogalactoside (0.1 mM) at 37 °C for 3.5 h by the pET28a vector system. Fortunately, a purity greater than 93% of rFMB-BBTI with anti-CRC activity was purified by nickel-nitrilotriacetic acid affinity chromatography. Subsequently, we found that rFMB-BBTI displays a strikingly anti-CRC effect, characterized by the inhibition of cell proliferation and clone formation ability, cell cycle arrest at the G2/M phase, and induction of cell apoptosis. It is interesting that the rFMB-BBTI treatment had no obvious effect on normal colorectal cells in the same concentration range. Importantly, the anti-CRC activity of rFMB-BBTI was further confirmed in the xenografted nude mice model. Taken together, our study highlights the anti-CRC activity of rFMB-BBTI in vitro and in vivo, uncovering the clinical potential of rFMB-BBTI as a targeted agent for CRC in the future.


Subject(s)
Colorectal Neoplasms , Plant Extracts , Plant Proteins , Setaria Plant , Trypsin Inhibitors , Animals , Humans , Male , Mice , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Gene Expression , Mice, Inbred BALB C , Mice, Nude , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Proteins/genetics , Plant Proteins/isolation & purification , Plant Proteins/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Setaria Plant/genetics , Setaria Plant/chemistry , Trypsin Inhibitors/pharmacology , Trypsin Inhibitors/isolation & purification , Trypsin Inhibitors/chemistry
19.
Int J Biol Macromol ; 270(Pt 1): 131886, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677696

ABSTRACT

Type V collagen is an essential component of the extracellular matrix (ECM), and its remodeling releases specific protein fragments that can specifically inhibit endothelial cell responses such as proliferation, migration, and invasion. In this study, we have successfully constructed two engineered strains of Pichia pastoris capable of producing recombinant collagen through a new genetic engineering approach. Through high-density fermentation, the expression of 1605 protein and 1610 protein could reach 2.72 g/L and 4.36 g/L. With the increase of repetition times, the yield also increased. Bioactivity analysis showed that recombinant collagen could block the angiogenic effect of FGF-2 on endothelial cells by eliminating FGF-2-induced endothelial cell migration and invasion. Collectively, the recombinant proteins we successfully expressed have a wide range of potential for inhibiting angiogenesis in the biomaterials and biomedical fields.


Subject(s)
Recombinant Proteins , Recombinant Proteins/pharmacology , Recombinant Proteins/genetics , Humans , Collagen/chemistry , Collagen/pharmacology , Cell Movement/drug effects , Repetitive Sequences, Amino Acid , Amino Acid Sequence , Human Umbilical Vein Endothelial Cells/drug effects , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 2/chemistry , Gene Expression , Fermentation , Saccharomycetales/genetics , Saccharomycetales/metabolism
20.
Biomed Pharmacother ; 175: 116661, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38678965

ABSTRACT

Alzheimer's disease (AD) is a global medical challenge. Studies have shown that neurotoxicity caused by pathological aggregation of ß-amyloid (Aß) is an important factor leading to AD. Therefore, inhibiting the pathological aggregation of Aß is the key to treating AD. The recombinant human HspB5-ACD structural domain protein (AHspB5) prepared by our group in the previous period has been shown to have anti-amyloid aggregation effects, but its inability to penetrate biological membranes has limited its development. In this study, we prepared a recombinant fusion protein (T-AHspB5) of TAT and AHspB5. In vitro experiments showed that T-AHspB5 inhibited the formation of Aß1-42 protofibrils and had the ability to penetrate the blood-brain barrier; in cellular experiments, T-AHspB5 prevented Aß1-42-induced oxidative stress damage, apoptosis, and inflammatory responses in neuronal cells, and its mechanism of action was related to microglia activation and mitochondria-dependent apoptotic pathway. In animal experiments, T-AHspB5 improved memory and cognitive dysfunction and inhibited pathological changes of AD in APP/PS1 mice. In conclusion, this paper is expected to reveal the intervention mechanism and biological effect of T-AHspB5 on pathological aggregation of Aß1-42, provide a new pathway for the treatment of AD, and lay the foundation for the future development and application of T-AHspB5.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Amyloid beta-Peptides/metabolism , Animals , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Apoptosis/drug effects , Oxidative Stress/drug effects , Peptide Fragments/pharmacology , Peptide Fragments/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Mice, Transgenic , alpha-Crystallin B Chain/metabolism , Recombinant Fusion Proteins/pharmacology , Male , Recombinant Proteins/pharmacology , Protein Domains , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...