Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 151
Filter
1.
Nanoscale ; 16(18): 8843-8850, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38644775

ABSTRACT

Extensive modifications have been made to the synthesis protocol for porous silica particles to improve the shape, size and yield percentage, but problems associated with improvement in biodegradability and decrease in chances to induce side effects still remain a concern. To circumvent these limitations, a facile modification strategy has been employed through in situ carbonization of porous silica particles. Herein, carbon particles were integrated within porous silica core-shell particles (Si-P-CNPs) during the synthesis process and found to preserve the ordered structural morphology. Curcumin was used as a model drug for loading in prepared Si-P-CNPs whereas lung cancer cells were used as a model system to study the in vitro fate. These Si-P-CNPs showed improved drug loading, drug effectivity, biodegradability and avoidance of interaction with transforming growth factor ß1 (TGF-ß1) indicating the possibility of reducing the chances of lung fibrosis and thereby enhancing the safety profile over conventional porous silica particles.


Subject(s)
Carbon , Curcumin , Drug Carriers , Silicon Dioxide , Transforming Growth Factor beta1 , Silicon Dioxide/chemistry , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/chemistry , Humans , Porosity , Drug Carriers/chemistry , Carbon/chemistry , Curcumin/chemistry , Curcumin/pharmacology , A549 Cells , Cell Line, Tumor , Fibrosis , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
2.
Biophys J ; 122(1): 82-89, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36419349

ABSTRACT

Thermally stable or resilient proteins are usually stabilized at intermediate states during thermal stress to prevent irreversible denaturation. However, the mechanism by which their conformations are stabilized to resist high temperature remains elusive. Herein, we investigate the conformational and thermal stability of transforming growth factor-ß1 (TGF-ß1), a key signaling molecule in numerous biological pathways. We report that the TGF-ß1 molecule is thermally resilient as it gradually denatures during thermal treatment when the temperature increases to 90°C-100°C but recovers native folding when the temperature decreases. Using this protein as a model, further studies show the maintenance of its bioactive functional properties after thermal stress, as demonstrated by differentiation induction of NIH/3T3 fibroblasts and human mesenchymal stem cells into myofibroblasts and chondrocytes, respectively. Molecular dynamic simulations revealed that although the protein's secondary structure is unstable under thermal stress, its conformation is partially stabilized by newly formed turns. Given the importance and/or prevalence of TGF-ß1 in biological processes, potential therapeutics, and the human diet, our findings encourage consideration of its thermostability for biomedical applications and nutrition.


Subject(s)
Myofibroblasts , Transforming Growth Factor beta1 , Humans , Cell Differentiation , Fibroblasts/metabolism , Protein Conformation , Signal Transduction , Transforming Growth Factor beta1/chemistry
3.
Carbohydr Polym ; 278: 118924, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34973742

ABSTRACT

This work aims to use carboxymethyl cellulose (CMC) as main structural and functional component of 3D printed scaffolds for healing of diabetic wounds. Differently from previous inks involving small contents in CMC, herein sterile (steam-heated) concentrated CMC solely dispersions (10-20%w/v) were screened regarding printability and fidelity properties. CMC (15%w/v)-citric acid inks showed excellent self-healing rheological properties and stability during storage. CMC scaffolds loaded with platelet rich plasma (PRP) sustained the release of relevant growth factors. CMC scaffolds both with and without PRP promoted angiogenesis in ovo, stem cell migration in vitro, and wound healing in a diabetic model in vivo. Transparent CMC scaffolds allowed direct monitoring of bilateral full-thickness wounds created in rat dorsum. CMC scaffolds facilitated re-epithelialization, granulation, and angiogenesis in full-thickness skin defects, and the performance was improved when combined with PRP. Overall, CMC is pointed out as outstanding component of active dressings for diabetic wounds.


Subject(s)
Carboxymethylcellulose Sodium/chemistry , Drug Delivery Systems , Intercellular Signaling Peptides and Proteins/pharmacology , Printing, Three-Dimensional , Tissue Scaffolds/chemistry , Wound Healing/drug effects , Animals , Diabetes Mellitus, Type 1 , Intercellular Signaling Peptides and Proteins/chemistry , Male , Particle Size , Platelet-Rich Plasma/chemistry , Rats , Rats, Sprague-Dawley , Transforming Growth Factor beta1/chemistry , Vascular Endothelial Growth Factors/chemistry
4.
Int J Mol Sci ; 23(2)2022 Jan 15.
Article in English | MEDLINE | ID: mdl-35055112

ABSTRACT

In 3D bioprinting for cartilage regeneration, bioinks that support chondrogenic development are of key importance. Growth factors covalently bound in non-printable hydrogels have been shown to effectively promote chondrogenesis. However, studies that investigate the functionality of tethered growth factors within 3D printable bioinks are still lacking. Therefore, in this study, we established a dual-stage crosslinked hyaluronic acid-based bioink that enabled covalent tethering of transforming growth factor-beta 1 (TGF-ß1). Bone marrow-derived mesenchymal stromal cells (MSCs) were cultured over three weeks in vitro, and chondrogenic differentiation of MSCs within bioink constructs with tethered TGF-ß1 was markedly enhanced, as compared to constructs with non-covalently incorporated TGF-ß1. This was substantiated with regard to early TGF-ß1 signaling, chondrogenic gene expression, qualitative and quantitative ECM deposition and distribution, and resulting construct stiffness. Furthermore, it was successfully demonstrated, in a comparative analysis of cast and printed bioinks, that covalently tethered TGF-ß1 maintained its functionality after 3D printing. Taken together, the presented ink composition enabled the generation of high-quality cartilaginous tissues without the need for continuous exogenous growth factor supply and, thus, bears great potential for future investigation towards cartilage regeneration. Furthermore, growth factor tethering within bioinks, potentially leading to superior tissue development, may also be explored for other biofabrication applications.


Subject(s)
Bioprinting/methods , Cartilage, Articular/cytology , Hyaluronic Acid/chemistry , Mesenchymal Stem Cells/cytology , Transforming Growth Factor beta1/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Hydrogels , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds , Transforming Growth Factor beta1/chemistry
5.
ACS Appl Mater Interfaces ; 14(1): 307-323, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-34968038

ABSTRACT

Mesangial cell (MC)-mediated glomerulonephritis is a frequent cause of end-stage renal disease, with immune inflammatory damage and fibrosis as its basic pathological processes. However, the treatment of glomerulonephritis remains challenging owing to limited drug accumulation and serious side effects. Hence, the specific codelivery of "anti-inflammatory/antifibrosis" drugs to the glomerular MC region is expected to yield better therapeutic effects. In this study, liposome-nanoparticle hybrids (Au-LNHy) were formed by coating the surface of gold nanoparticles with a phospholipid bilayer; the Au-LNHys formed were comodified with PEG and α8 integrin antibodies to obtain gold nanoparticle immunoliposomes (Au-ILs). Next, the Au-ILs were loaded with dexamethasone and TGFß1 siRNA to obtain DXMS/siRNA@Au-ILs. Our results showed that the functionalized nanoparticles had a core-shell structure, a uniform and suitable particle size, low cytotoxicity, and good MC entry, and lysosomal escape abilities. The nanoparticles were found to exhibit enhanced retention in glomerular MCs due to anti-α8 integrin antibody mediation. In vivo and in vitro pharmacodynamic studies showed the enhanced efficacy of DXMS/siRNA@Au-ILs modified with α8 integrin antibodies in the treatment of glomerulonephritis. In addition, DXMS/siRNA@Au-ILs were capable of effectively reducing the expression levels of TNF-α, TGF-ß1, and other cytokines, thereby improving pathological inflammatory and fibrotic conditions in the kidney, and significantly mediating the dual regulation of inflammation and fibrosis. In summary, our results demonstrated that effectively targeting the MCs of the glomerulus for drug delivery can inhibit local inflammation and fibrosis and produce better therapeutic effects, providing a new strategy and promising therapeutic approach for the development of targeted therapies for glomerular diseases.


Subject(s)
Dexamethasone/therapeutic use , Glomerulonephritis/drug therapy , Gold/therapeutic use , Metal Nanoparticles/chemistry , RNA, Small Interfering/therapeutic use , Transforming Growth Factor beta1/chemistry , Animals , Cells, Cultured , Dexamethasone/chemistry , Gold/chemistry , Humans , Liposomes/chemistry , Male , Materials Testing , Mice , Mice, Inbred Strains , RNA, Small Interfering/chemistry
6.
ACS Appl Bio Mater ; 4(1): 545-558, 2021 01 18.
Article in English | MEDLINE | ID: mdl-34458689

ABSTRACT

The bypass graft is the mainstream of surgical intervention to treat vascular diseases. Ideal bypass materials, yet to be developed, require mechanical properties, availability, clinically feasible manufacturing logistics, and bioactivities with precise physicochemical cues defined to guide cell activities for arterial regeneration. Such needs instigated our fabrication of vascular grafts, which consist of coaxial, nanostructured fibers exhibiting a polycaprolactone (PCL) core and a photoclickable, 4-arm thiolated polyethylene glycol-norbornene (PEG-NB) sheath. The graft strength and bioactivity were modulated by the PCL concentration and the peptides (RGD, transforming growth factor ß-1 or TGF-ß1) conjugated to thiol-ene of PEG-NB, respectively. Structural, physical, and mechanical characterizations demonstrated that the fibrous grafts mimicked the key features of the native extracellular matrix, including a crosslinked fiber network for structural stability, viscoelasticity emulating arteries, hydration property, and high porosity for cell infiltration. Meanwhile, these grafts displayed strength and toughness exceeding or meeting surgical criteria. Furthermore, the grafts with higher PCL concentration (3 vs 1.8%) showed thicker fibers, lower porosity and pore size, and increased elastic and storage moduli. Graft bioactivity was determined by the mesenchymal stem cell (MSC) behaviors on the grafts and arterial regeneration in vivo using interposition grafting. Results showed that the cell adhesion and proliferation increased with the RGD density (25 vs 5 mM). After 1 week implantation, all peptide-functionalized PCL/PEG-NB grafts with or without MSC preseeding, as opposed to PCL grafts, showed expeditious endothelial lining, abundant vascular cell infiltration, and matrix production. Compared to RGD grafts, RGD/TGF-ß1 grafts enhanced MSC differentiation into smooth muscle cells in vitro and developed thicker smooth muscle cell layers in vivo. Overall, the versatile porous vascular grafts offer superior properties and tunability for future translation.


Subject(s)
Ligands , Polymers/chemistry , Regeneration , Animals , Cell Adhesion/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Elastic Modulus , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Norbornanes/chemistry , Oligopeptides/chemistry , Peptides/chemistry , Polyesters/chemistry , Polyethylene Glycols/chemistry , Polymers/pharmacology , Rats , Rats, Sprague-Dawley , Regeneration/drug effects , Transforming Growth Factor beta1/chemistry
7.
Biochem Pharmacol ; 192: 114697, 2021 10.
Article in English | MEDLINE | ID: mdl-34302795

ABSTRACT

The TGF-ß1 cytokine is a key mediator of many biological processes. Complex regulatory mechanisms are in place that allow one single molecule to exert so many distinct indispensable activities. The complexity of TGF-ß1 biology is further illustrated by the opposing dual roles it plays during cancer progression. Risks of toxicities combined with lack of convincing therapeutical efficacy explain at least in part why therapies targeting TGF-ß1 have lagged behind in past decades. However, recent successes of immunostimulatory antibodies for the immunotherapy of cancer and findings that TGF-ß1 activity associates with resistance to immunotherapeutic drugs have revived the field. In this review, we discuss the biology of TGF-ß1 with a special focus on its roles in regulating immune responses in the context of cancer. We describe the various therapeutic approaches available to inhibit TGF-ß signalling, and more recent findings that allow selective targeting of specific sources of TGF-ß activity, which may prove relevant to increase the efficacy and reduce the toxicity of cancer immunotherapy.


Subject(s)
Immunity, Cellular/immunology , Immunosuppression Therapy/methods , Immunotherapy/methods , Neoplasms/immunology , Transforming Growth Factor beta1/immunology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/immunology , Humans , Neoplasms/therapy , Protein Structure, Secondary , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/chemistry
8.
Biomed Res Int ; 2021: 6631848, 2021.
Article in English | MEDLINE | ID: mdl-33869629

ABSTRACT

Oral submucous fibrosis (OSMF) is considered a premalignant condition characterized by aggressive fibrosis of the submucosal tissues of the oral cavity reflecting its malignant transformation potential. Activation of transforming growth factor beta (TGF-ß) signaling has been reported to lead increased collagen production and fibrosis. Recently, significant upregulation of TGF-ß1 has been reported in OSMF as compared to normal tissues. Therefore, inhibition of the TGF-ß1 may pave for the development of therapeutics of OSMF. Based on the structure-assisted drug designing, we found "silmitasertib" as potent inhibitor of TGF-ß1. We suggest that this molecule can be validated and implemented for the treatment of OSMF.


Subject(s)
Drug Repositioning , Naphthyridines/therapeutic use , Oral Submucous Fibrosis/drug therapy , Phenazines/therapeutic use , Small Molecule Libraries/therapeutic use , Transforming Growth Factor beta1/antagonists & inhibitors , Humans , Hydrogen Bonding , Molecular Docking Simulation , Naphthyridines/chemistry , Naphthyridines/pharmacology , Oral Submucous Fibrosis/pathology , Phenazines/chemistry , Phenazines/pharmacology , Protein Binding/drug effects , Protein Structure, Secondary , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Time Factors , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/metabolism
9.
Front Immunol ; 12: 586220, 2021.
Article in English | MEDLINE | ID: mdl-33763059

ABSTRACT

Type 1 diabetes (T1D) is a disorder of impaired glucoregulation due to lymphocyte-driven pancreatic autoimmunity. Mobilizing dendritic cells (DC) in vivo to acquire tolerogenic activity is an attractive therapeutic approach as it results in multiple and overlapping immunosuppressive mechanisms. Delivery of agents that can achieve this, in the form of micro/nanoparticles, has successfully prevented a number of autoimmune conditions in vivo. Most of these formulations, however, do not establish multiple layers of immunoregulation. all-trans retinoic acid (RA) together with transforming growth factor beta 1 (TGFß1), in contrast, has been shown to promote such mechanisms. When delivered in separate nanoparticle vehicles, they successfully prevent the progression of early-onset T1D autoimmunity in vivo. Herein, we show that the approach can be simplified into a single microparticle formulation of RA + TGFß1 with surface decoration with the T1D-relevant insulin autoantigen. We show that the onset of hyperglycemia is prevented when administered into non-obese diabetic mice that are at the mid-stage of active islet-selective autoimmunity. Unexpectedly, the preventive effects do not seem to be mediated by increased numbers of regulatory T-lymphocytes inside the pancreatic lymph nodes, at least following acute administration of microparticles. Instead, we observed a mild increase in the frequency of regulatory B-lymphocytes inside the mesenteric lymph nodes. These data suggest additional and potentially-novel mechanisms that RA and TGFß1 could be modulating to prevent progression of mid-stage autoimmunity to overt T1D. Our data further strengthen the rationale to develop RA+TGFß1-based micro/nanoparticle "vaccines" as possible treatments of pre-symptomatic and new-onset T1D autoimmunity.


Subject(s)
Autoantigens/immunology , Autoimmunity/drug effects , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Transforming Growth Factor beta1/pharmacology , Tretinoin/pharmacology , Animals , Dendritic Cells , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/drug therapy , Drug Compounding , Female , Insulin/metabolism , Lymphocyte Count , Mice , Mice, Inbred NOD , Pancreas/metabolism , Pancreas/pathology , Severity of Illness Index , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/chemistry , Tretinoin/chemistry
10.
Respirology ; 26(3): 255-263, 2021 03.
Article in English | MEDLINE | ID: mdl-33025706

ABSTRACT

BACKGROUND AND OBJECTIVE: Lipid metabolism dysregulation has been implicated in the pathogenesis of IPF; however, the roles of most lipid metabolites in lung fibrosis remain unexplored. Therefore, we aimed to identify changes in lipid metabolites in the lung tissues of IPF patients and determine their roles in pulmonary fibrosis. METHODS: Free fatty acids in the lung tissues of IPF patients and controls were quantified using a metabolomic approach. The roles of free fatty acids in fibroblasts or epithelial cells treated with TGF-ß1 were evaluated using fibrotic markers. The antifibrotic role of stearic acid was also assessed in a bleomycin-induced lung fibrosis mouse model. Protein levels in cell lysates or tissues were measured by western blotting. RESULTS: The levels of stearic acid were lower in IPF lung tissues than in control lung tissues. Stearic acid significantly reduced TGF-ß1-induced α-SMA and collagen type 1 expression in MRC-5 cells. Furthermore, stearic acid decreased the levels of p-Smad2/3 and ROS in MRC-5 cells treated with TGF-ß1 and disrupted TGF-ß1-induced EMT in Beas-2B cells. Stearic acid reduced the levels of bleomycin-induced hydroxyproline in a mouse model. CONCLUSION: Changes in the free fatty acid profile, including low levels of stearic acid, were observed in IPF patients. Stearic acid may exert antifibrotic activity by regulating profibrotic signalling.


Subject(s)
Bleomycin/pharmacology , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis , Lung/physiology , Stearic Acids/chemistry , Transforming Growth Factor beta1/chemistry , Animals , Bleomycin/chemistry , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Mice , Signal Transduction/physiology , Transforming Growth Factor beta1/metabolism
11.
J Mol Recognit ; 34(4): e2881, 2021 04.
Article in English | MEDLINE | ID: mdl-33137847

ABSTRACT

Transforming growth factor-ß1 (TGF-ß1) signaling pathway has been implicated in the fibroblast activation of hypertrophic scarring (HS). Previously, we proposed a new biotherapeutic strategy to combat HS by disrupting the intermolecular interaction of TGF-ß1 with its cognate type-II receptor (TßR-II). Here, we further demonstrate that the binding site of TGF-ß1 to TßR-II is not overlapped with the conformational wrist epitope and linear knuckle epitope that are traditionally recognized as the functional binding sites of bone morphogenetic protein-2 (BMP-2) to its type-II receptor (BMPR-II), which can thus be regarded as a new functional site we called elbow epitope. Structural, energetic, and dynamic investigations reveal that the elbow epitope consists of two sequentially discontinuous, spatially vicinal segments Loop30-34 and Turn90-95 ; they cannot work effectively to independently interact with TßR-II. Rational redesign of the epitope is performed using an integrated in silio-in vitro method based on crystal and modeled structure data. In the procedure, the two epitope segments are split from the interface of TGF-ß1-TßR-II complex and then connected with each other in a head-to-tail manner by adding a flexible poly-(Gly)n linker between them, thus resulting in a series of combined peptides. We found that the peptide affinity reaches maximum at n = 2, which shares a consistent binding mode with the elbow epitope at native complex interface. The linker of either too long (n > 2) or too short (n < 2) cannot properly place the gap space between the two segments, thus impairing the binding compatibility of designed peptides with TßR-II active site.


Subject(s)
Epitopes/chemistry , Epitopes/metabolism , Peptide Fragments/metabolism , Receptor, Transforming Growth Factor-beta Type II/metabolism , Transforming Growth Factor beta1/immunology , Binding Sites , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein Receptors, Type II/chemistry , Bone Morphogenetic Protein Receptors, Type II/immunology , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cicatrix, Hypertrophic/therapy , Fluorescence Polarization , Humans , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Peptide Fragments/immunology , Receptor, Transforming Growth Factor-beta Type II/chemistry , Receptor, Transforming Growth Factor-beta Type II/immunology , Thermodynamics , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/metabolism
12.
Front Immunol ; 11: 574447, 2020.
Article in English | MEDLINE | ID: mdl-33193362

ABSTRACT

Current monotherapeutic agents fail to restore tolerance to self-antigens in autoimmune individuals without systemic immunosuppression. We hypothesized that a combinatorial drug formulation delivered by a poly-lactic-co-glycolic acid (PLGA) dual-sized microparticle (dMP) system would facilitate tunable drug delivery to elicit immune tolerance. Specifically, we utilized 30 µm MPs to provide local sustained release of granulocyte-macrophage colony-stimulating factor (GM-CSF) and transforming growth factor ß1 (TGF-ß1) along with 1 µm MPs to facilitate phagocytic uptake of encapsulated antigen and 1α,25(OH)2 Vitamin D3 (VD3) followed by tolerogenic antigen presentation. We previously demonstrated the dMP system ameliorated type 1 diabetes (T1D) and experimental autoimmune encephalomyelitis (EAE) in murine models. Here, we investigated the system's capacity to impact human cell activity in vitro to advance clinical translation. dMP treatment directly reduced T cell proliferation and inflammatory cytokine production. dMP delivery to monocytes and monocyte-derived dendritic cells (DCs) increased their expression of surface and intracellular anti-inflammatory mediators. In co-culture, dMP-treated DCs (dMP-DCs) reduced allogeneic T cell receptor (TCR) signaling and proliferation, while increasing PD-1 expression, IL-10 production, and regulatory T cell (Treg) frequency. To model antigen-specific activation and downstream function, we co-cultured TCR-engineered autoreactive T cell "avatars," with dMP-DCs or control DCs followed by ß-cell line (ßlox5) target cells. For G6PC2-specific CD8+ avatars (clone 32), dMP-DC exposure reduced Granzyme B and dampened cytotoxicity. GAD65-reactive CD4+ avatars (clone 4.13) exhibited an anergic/exhausted phenotype with dMP-DC presence. Collectively, these data suggest this dMP formulation conditions human antigen presenting cells toward a tolerogenic phenotype, inducing regulatory and suppressive T cell responses.


Subject(s)
Dendritic Cells/drug effects , Diabetes Mellitus, Type 1/immunology , Immune Tolerance/drug effects , T-Lymphocytes/immunology , Antigen Presentation/drug effects , Autoantigens/immunology , Calcitriol/chemistry , Calcitriol/pharmacology , Dendritic Cells/immunology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunomodulation , Lymphocyte Activation , Monocytes/drug effects , Particle Size , Phenotype , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/pharmacology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/drug effects , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/pharmacology
13.
Int J Biol Macromol ; 165(Pt A): 1392-1401, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33045298

ABSTRACT

Transforming growth factor ß1 (TGFß1) is a polyfunctional cytokine with important roles in growth, differentiation and immune function in various animals. In this study, PCR, bioinformatics, real-time quantitative PCR, prokaryotic expression, protein purification and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-TOF-MS) were applied to investigate the structural features and function of TGFß1-b in crucian carp. The complete coding sequence (CDS) of TGFß1-b was 1134 bp in length and was submitted to GenBank (ID: MH473141). TGFß1-b encoded a putative protein of 377 amino acids and included a signal peptide consisting of 22 amino acids. TGFß1-b was relatively conservative in fish and distant from mammals in terms of evolutionary relationship. TGFß1-b was found to be expressed in various tissues, with the highest expression in the kidney. The expressions of TGFß1-b in muscle, heart and liver were increased with the addition of Rhodopseudomonas palustris, Bacillus subtilis and Enterococcus faecium at 30 days (p < 0.01). While, the expressions of SMAD2, SMAD3 and SMAD7 were also up-regulated with the addition of R. palustris at 20 days (p < 0.01). The expression of TGFß1-b could be affected by time and group factors (p < 0.05). Moreover, the expression vector TGFß1-b-pDE2 was successfully constructed. Prokaryotic expression indicated that a 43 kDa target protein was obtained after induction with 1.5 mM isopropyl-beta-D-thiogalactopyranoside (IPTG) for 3.5 h at 37 °C for 200 r/h. The activities of alkaline phosphatase and lysozyme in injection TGFß1-b protein group (ITg) and feeding broken bacterial liquid group (BTg) were significantly increased at 24 h (p < 0.01). And the activities of superoxide dismutase in ITg were significantly increased at 36 h (p < 0.01). Besides, the expressions of heat shock protein 30 and heat shock protein 47 in ITg and BTg were significantly increased (p < 0.01). Whereas, the expression of interleukin-11 was significantly reduced (p < 0.01). These results indicated that TGFß1-b protein might play a role in immunity of crucian carp.


Subject(s)
Carps/genetics , Phylogeny , Transforming Growth Factor beta1/genetics , Amino Acid Sequence/genetics , Animals , Cloning, Molecular , Gene Expression Regulation/genetics , Immunity, Innate/genetics , Sequence Alignment , Transforming Growth Factor beta1/chemistry
14.
Syst Biol Reprod Med ; 66(4): 267-280, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32735465

ABSTRACT

The prevalence of infertility is increasing and worrisome. About 10 to 30% of infertility is classified as idiopathic or unexplained infertility (UI).TGF-ß is multifunctional and immunoregulatry cytokine which regulates both implantation and adhesion of trophoblasts to the extracellular matrix during pregnancy. The aim of the current study was to investigate the association between two polymorphisms rs1800470 (C29T) and rs1800471 (G74C) of the TGF-ß1 gene in Iranian patients with unexplained infertility. A total of 250 UI patients and 484 healthy individuals with no history of infertility were included in the study. The amplification and sequencing of target DNA fragments were done using PCR and automated sequencing methods, respectively. The effects of these polymorphisms on both TGF-ß1 structure and function of mRNA and protein were analyzed using new in-silico tools. The frequency distribution of the alleles, genotypes, and haplotypes of both rs1800470 and rs1800471 polymorphisms had a statistically significant difference between subjects and controls. CC genotype of TGF-ß1 rs1800470 (29C→T) increase the risk of UI in male UI patients. Moreover, C alleles of TGF-ß1 rs1800471 was associated with increased risk of UI in female UI patients. Couples, subgroup analysis revealed a significant association between TGF-ß1 polymorphisms (rs1800470, rs1800471) and the risk of UI in male, female, and all UI patients. The frequency of TG and CG haplotypes were statistically different in both UI and healthy subjects group (P < 0.05). RS1800471 polymorphisms changed the secondary structure of TGF-ß1 mRNA and resulted in the removal of one mRNA arm and creation of two new arms. Taken together, the results of the current study suggest that TGF-ß1 functional polymorphisms may play an important role in the susceptibility to UI in Iranian population. According to in silico analysis, polymorphisms in TGF-ß1 can reduce mRNA half-life and, therefore, reduced TGF-ß1 expression. .


Subject(s)
Infertility/genetics , Polymorphism, Single Nucleotide , Transforming Growth Factor beta1/genetics , Adult , Alleles , Case-Control Studies , Computer Simulation , Female , Gene Frequency , Genetic Association Studies , Genotype , Haplotypes , Humans , Iran , Male , Sequence Analysis, DNA , Structure-Activity Relationship , Transforming Growth Factor beta1/chemistry
15.
J Mater Sci Mater Med ; 31(8): 62, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32696084

ABSTRACT

The aim of this study was to evaluate the effect of biofunctionalization with two TGF-ß1 inhibitor peptides, P17 and P144, on osseointegration of CP-Ti dental implants. A total of 36 implants (VEGA, Klockner®) with 3.5 × 8 mm internal connection were used in this study, divided in three groups: (1) control group (n = 12), (2) implants which surfaces were biofunctionalized with P17 peptide inhibitor (n = 12), (3) implants with surfaces biofunctionalized by P144 peptide (n = 12). Three implants, one from each group, were inserted in both hemimandibles of 6 beagle dogs, 2 months after tooth extraction. Two animals were sacrificed at 2, 4 and 8 weeks post implant insertion, respectively. The samples were analyzed by Backscattering Scanning Electron Microscopy (BS-SEM) and histological analysis. Histomorphometric analysis of bone to implant contact (BIC), peri-implant bone fraction (BF) and interthread bone (IB) were carried out. Bone formation around implants measured by quantitative analysis, BS-SEM, was significantly higher in the P17-biofunctionalized implants, 4 and 8 weeks after the implantation. Histomorphometric analysis of BIC, BF and IB showed higher values in the P17-biofunctionalized group at initial stages of healing (2 weeks) and early osseointegration both at 4 and 8 weeks. For P144 biofunctionalized implants, the histomorphometric values obtained are also higher than control group. Accordingly, better results in the experimental groups were proven both by the quantitative and the qualitative analysis. Surface biofunctionalization with TGF-ß1 inhibitor peptides, P17 and P144, resulted in better quantitative and qualitative parameters relative to implant osseointegration.


Subject(s)
Coated Materials, Biocompatible/pharmacology , Dental Implants , Osseointegration/drug effects , Peptide Fragments/chemistry , Peptides/chemistry , Receptors, Transforming Growth Factor beta/chemistry , Animals , Coated Materials, Biocompatible/chemistry , Dental Implantation, Endosseous , Dental Prosthesis Design , Dogs , Implants, Experimental , Male , Mandible/pathology , Mandible/surgery , Materials Testing , Osteogenesis/drug effects , Surface Properties/drug effects , Transforming Growth Factor beta1/chemistry
16.
Org Lett ; 22(15): 5726-5730, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32525688

ABSTRACT

Aspongdopamines A and B (1 and 2), unusual adducts composed of N-acetyldopamine and adenine were isolated from the insect Aspongopus chinensis. Compounds 1 and 2 are positional isomers both isolated as racemates. Chiral separation assisted by 14-step total synthesis and computation including vibrational circular dichroism calculations allowed us to unambiguously assign the absolute configurations of eight stereoisomers. Renal fibrosis inhibition of the stereoisomers was evaluated in TGF-ß1-induced rat kidney epithelial cells.


Subject(s)
Adenine/chemical synthesis , Biological Products/pharmacology , Dopamine/analogs & derivatives , Insecta/drug effects , Transforming Growth Factor beta1/chemistry , Adenine/chemistry , Animals , Circular Dichroism , Dopamine/chemical synthesis , Dopamine/chemistry , Molecular Structure , Rats , Stereoisomerism , Transforming Growth Factor beta1/metabolism
17.
Blood Transfus ; 18(3): 208-216, 2020 05.
Article in English | MEDLINE | ID: mdl-32281925

ABSTRACT

BACKGROUND: There are many advantages to using cord blood (CB) as a source of therapeutic platelet and plasma derivatives for regenerative medicine. These include availability, universal use, young donor source, and virally safe biological material, rich in tissue regenerative factors. MATERIALS AND METHODS: We aimed to validate a bioprocess design for the production of cord blood-derived platelet concentrates (CBPC) in a public Cord Blood Bank (CBB). CBPC was defined as a product of 10±5 mL, 1,000±200×109/L total platelets, free of erythrocytes and leukocytes. A total of 300 CB units were centrifuged in two steps to enrich for platelets, in compliance with Good Manufacturing Practice. The samples were tested for the degree of platelet activation present, and the levels of growth factor were analysed to evaluate their potential function. CBPC were then activated after thawing with 10% calcium gluconate to generate platelet gels (CBPG) to treat patients with diabetic foot ulcers. RESULTS: After processing, 84% of the products fulfilled the acceptance criteria. Final products contained 1,017±149×106 platelets/mL in 10±3mL of plasma. Platelet recovery was 50±9%. The methods described here ensure depletion of white and red blood cells down to a residual concentration of 0.2±0.1×106/mL and 0.03±0.02×106/mL, respectively. Platelets showed low levels of activation during processing, but were significantly activated after thawing, as indicated by an increase in CD62p expression. The growth factors EGF, VEGF, bFGF, PDGF AB/BB and TGF-ß1 were at concentrations of 1,706±123 pg/mL; 1,602±227 pg/mL; 314±26 pg/mL; 30±1.5 ng/mL; 24±2 ng/mL (mean±standard error of mean), respectively. For clinical evaluation, a total of 21 CBPG were applied in 3 patients, with no reported adverse events and improvement of ulcers in all of them. DISCUSSION: We designed and validated a highly reproducible, closed system method to manufacture high quality CBPC suitable for clinical applications using CB units not suitable for transplantation in a public CBB.


Subject(s)
Blood Banks , Fetal Blood/chemistry , Platelet-Derived Growth Factor/chemistry , Platelet-Rich Plasma/chemistry , Transforming Growth Factor beta1/chemistry , Blood Platelets , Diabetic Foot/drug therapy , Humans
18.
Mater Sci Eng C Mater Biol Appl ; 111: 110757, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32279738

ABSTRACT

Creating a microenvironment with low inflammation and favorable for the chondrogenic differentiation of endogenous stem cells plays an essential role in cartilage repairing. In the present study, we design a novel ginsenoside Rb1/TGF-ß1 loaded silk fibroin-gelatin porous scaffold (GSTR) with the function of attenuating inflammation and promoting chondrogenesis. The scaffold has porous microstructure, proper mechanical strength, degradation rate and sustained release of Rb1 and TGF-ß1. Rat bone marrow-derived mesenchymal stem cells (rBMSCs) seeded into GSTR scaffolds are homogeneously distributed and display a higher proliferation rate than non-loaded scaffolds (GS). GSTR scaffolds promote the chondrogenic differentiation of rBMSCs and suppress the expression of inflammation genes. Under the stimulation of IL-1ß, the inflammation level of the chondrocytes seeded in GSTR scaffolds is also significantly down-regulated. Moreover, GSTR scaffolds implanted into the osteochondral defects in rats effectively promote the regeneration of hyaline cartilage 12 weeks after surgery when compared with other groups. It is demonstrated that this scaffold loaded with Rb1 and TGF-ß1 can synergistically create a microenvironment favorable for cartilage regeneration by promoting the chondrogenesis and suppressing the inflammation levels in vivo. These results prove it has a great potential to develop this Rb1/TGF-ß1 releasing scaffold into a novel and promising therapeutic for cartilage repair.


Subject(s)
Cartilage/physiology , Fibroins/chemistry , Gelatin/chemistry , Ginsenosides/chemistry , Regeneration , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/chemistry , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/therapeutic use , Cell Differentiation/drug effects , Cell Survival/drug effects , Chondrogenesis/drug effects , Compressive Strength , Interleukin-1beta/metabolism , Joint Diseases/pathology , Joint Diseases/therapy , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Porosity , Rats , Rats, Sprague-Dawley , Regeneration/drug effects
19.
Biofabrication ; 12(2): 025030, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32106097

ABSTRACT

Due to the increasing aging population and the high probability of sport injury among young people nowadays, it is of great demand to repair/regenerate diseased/defected osteochondral tissue. Given that osteochondral tissue mainly consists of a subchondral layer and a cartilage layer which are structurally heterogeneous and mechanically distinct, developing a biomimetic bi-phasic scaffold with excellent bonding strength to regenerate osteochondral tissue is highly desirable. Three-dimensional (3D) printing is advantageous in producing scaffolds with customized shape, designed structure/composition gradients and hence can be used to produce heterogeneous scaffolds for osteochondral tissue regeneration. In this study, bi-layered osteochondral scaffolds were developed through cryogenic 3D printing, in which osteogenic peptide/ß-tricalcium phosphate/poly(lactic-co-glycolic acid) water-in-oil composite emulsions were printed into hierarchically porous subchondral layer while poly(D,L-lactic acid-co-trimethylene carbonate) water-in-oil emulsions were printed into thermal-responsive cartilage frame on top of the subchondral layer. The cartilage frame was further filled/dispensed with transforming growth factor-ß1 loaded collagen I hydrogel to form the cartilage module. Although the continuously constructed osteochondral scaffolds had distinct microscopic morphologies and varied mechanical properties at the subchondral zone and cartilage zone at 37 °C, respectively, the two layers were closely bonded together, showing excellent shear strength and peeling strength. Rat bone marrow derived mesenchymal stem cells (rBMSCs) exhibited high viability and proliferation at both subchondral- and cartilage layer. Moreover, gradient rBMSC osteogenic/chondrogenic differentiation was obtained in the osteochondral scaffolds. This proof-of-concept study provides a facile way to produce integrated osteochondral scaffolds for concurrently directing rBMSC osteogenic/chondrogenic differentiation at different regions.


Subject(s)
Peptides/metabolism , Printing, Three-Dimensional , Regeneration , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Transforming Growth Factor beta1/metabolism , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Calcium Phosphates/chemistry , Cartilage/physiology , Cell Differentiation/drug effects , Cell Survival/drug effects , Chondrogenesis/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Peptides/chemistry , Peptides/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rats , Regeneration/drug effects , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/pharmacology
20.
Cell ; 180(3): 490-501.e16, 2020 02 06.
Article in English | MEDLINE | ID: mdl-31955848

ABSTRACT

Integrin αvß8 binds with exquisite specificity to latent transforming growth factor-ß (L-TGF-ß). This binding is essential for activating L-TGF-ß presented by a variety of cell types. Inhibiting αvß8-mediated TGF-ß activation blocks immunosuppressive regulatory T cell differentiation, which is a potential therapeutic strategy in cancer. Using cryo-electron microscopy, structure-guided mutagenesis, and cell-based assays, we reveal the binding interactions between the entire αvß8 ectodomain and its intact natural ligand, L-TGF-ß, as well as two different inhibitory antibody fragments to understand the structural underpinnings of αvß8 binding specificity and TGF-ß activation. Our studies reveal a mechanism of TGF-ß activation where mature TGF-ß signals within the confines of L-TGF-ß and the release and diffusion of TGF-ß are not required. The structural details of this mechanism provide a rational basis for therapeutic strategies to inhibit αvß8-mediated L-TGF-ß activation.


Subject(s)
Cryoelectron Microscopy/methods , Integrins/chemistry , Integrins/metabolism , Latent TGF-beta Binding Proteins/chemistry , Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta1/chemistry , Transforming Growth Factor beta1/metabolism , Animals , Antibodies/immunology , Binding Sites , Bronchi/cytology , CHO Cells , Cricetulus , Female , Humans , Immunoglobulin Fab Fragments/immunology , Integrins/immunology , Lymphocyte Activation , Male , Mink , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...