Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
Mol Cell ; 84(10): 1995-2005.e7, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38614096

ABSTRACT

Cytokines regulate immune responses by binding to cell surface receptors, including the common subunit beta (ßc), which mediates signaling for GM-CSF, IL-3, and IL-5. Despite known roles in inflammation, the structural basis of IL-5 receptor activation remains unclear. We present the cryo-EM structure of the human IL-5 ternary receptor complex, revealing architectural principles for IL-5, GM-CSF, and IL-3. In mammalian cell culture, single-molecule imaging confirms hexameric IL-5 complex formation on cell surfaces. Engineered chimeric receptors show that IL-5 signaling, as well as IL-3 and GM-CSF, can occur through receptor heterodimerization, obviating the need for higher-order assemblies of ßc dimers. These findings provide insights into IL-5 and ßc receptor family signaling mechanisms, aiding in the development of therapies for diseases involving deranged ßc signaling.


Subject(s)
Cryoelectron Microscopy , Granulocyte-Macrophage Colony-Stimulating Factor , Interleukin-3 , Protein Multimerization , Signal Transduction , Humans , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interleukin-3/metabolism , Interleukin-3/chemistry , Interleukin-3/genetics , HEK293 Cells , Protein Binding , Models, Molecular , Interleukin-5/metabolism , Cytokine Receptor Common beta Subunit/metabolism , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/chemistry , Single Molecule Imaging , Structure-Activity Relationship , Binding Sites , Receptors, Interleukin-5/metabolism , Receptors, Interleukin-5/genetics , Receptors, Interleukin-5/chemistry
2.
Amino Acids ; 54(4): 601-613, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34453584

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a cytokine and a white blood cell growth factor that has found usage as a therapeutic protein. During analysis of different fermentation batches of GM-CSF recombinantly expressed in E. coli, a covalent modification was identified on the protein by intact mass spectrometry. The modification gave a mass shift of + 70 Da and peptide mapping analysis demonstrated that it located to the protein N-terminus and lysine side chains. The chemical composition of C4H6O was found to be the best candidate by peptide fragmentation using tandem mass spectrometry. The modification likely contains a carbonyl group, since the mass of the modification increased by 2 Da by reduction with borane pyridine complex and it reacted with 2,4-dinitrophenylhydrazine. On the basis of chemical and tandem mass spectrometry fragmentation behavior, the modification could be attributed to crotonaldehyde, a reactive compound formed during lipid peroxidation. A low recorded oxygen pressure in the reactor during protein expression could be linked to the formation of this compound. This study shows the importance of maintaining full control over all reaction parameters during recombinant protein production.


Subject(s)
Escherichia coli , Granulocyte-Macrophage Colony-Stimulating Factor , Escherichia coli/genetics , Escherichia coli/metabolism , Fermentation , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mass Spectrometry , Recombinant Proteins/chemistry
3.
Clin Sci (Lond) ; 135(22): 2559-2573, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34778899

ABSTRACT

Granulocyte macrophage colony stimulating factor (GM-CSF) is a key participant in, and a clinical target for, the treatment of inflammatory diseases including rheumatoid arthritis (RA). Therapeutic inhibition of GM-CSF signalling using monoclonal antibodies to the α-subunit of the GM-CSF receptor (GMCSFRα) has shown clear benefit in patients with RA, giant cell arteritis (GCAs) and some efficacy in severe SARS-CoV-2 infection. However, GM-CSF autoantibodies are associated with the development of pulmonary alveolar proteinosis (PAP), a rare lung disease characterised by alveolar macrophage (AM) dysfunction and the accumulation of surfactant lipids. We assessed how the anti-GMCSFRα approach might impact surfactant turnover in the airway. Female C57BL/6J mice received a mouse-GMCSFRα blocking antibody (CAM-3003) twice per week for up to 24 weeks. A parallel, comparator cohort of the mouse PAP model, GM-CSF receptor ß subunit (GMCSFRß) knock-out (KO), was maintained up to 16 weeks. We assessed lung tissue histopathology alongside lung phosphatidylcholine (PC) metabolism using stable isotope lipidomics. GMCSFRß KO mice reproduced the histopathological and biochemical features of PAP, accumulating surfactant PC in both broncho-alveolar lavage fluid (BALF) and lavaged lung tissue. The incorporation pattern of methyl-D9-choline showed impaired catabolism and not enhanced synthesis. In contrast, chronic supra-pharmacological CAM-3003 exposure (100 mg/kg) over 24 weeks did not elicit a histopathological PAP phenotype despite some changes in lung PC catabolism. Lack of significant impairment of AM catabolic function supports clinical observations that therapeutic antibodies to this pathway have not been associated with PAP in clinical trials.


Subject(s)
Arthritis, Rheumatoid/metabolism , COVID-19/therapy , Pulmonary Alveolar Proteinosis/immunology , Pulmonary Surfactants/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Arthritis, Rheumatoid/therapy , Autoantibodies/chemistry , Bronchoalveolar Lavage Fluid , COVID-19/immunology , Choline/analogs & derivatives , Female , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Inflammation , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pulmonary Alveolar Proteinosis/genetics , SARS-CoV-2/immunology , Surface-Active Agents
4.
Biochem J ; 478(8): 1571-1583, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33734311

ABSTRACT

The α1,6-fucosyltransferase, FUT8, is the sole enzyme catalyzing the core-fucosylation of N-glycoproteins in mammalian systems. Previous studies using free N-glycans as acceptor substrates indicated that a terminal ß1,2-GlcNAc moiety on the Man-α1,3-Man arm of N-glycan substrates is required for efficient FUT8-catalyzed core-fucosylation. In contrast, we recently demonstrated that, in a proper protein context, FUT8 could also fucosylate Man5GlcNAc2 without a GlcNAc at the non-reducing end. We describe here a further study of the substrate specificity of FUT8 using a range of N-glycans containing different aglycones. We found that FUT8 could fucosylate most of high-mannose and complex-type N-glycans, including highly branched N-glycans from chicken ovalbumin, when the aglycone moiety is modified with a 9-fluorenylmethyloxycarbonyl (Fmoc) moiety or in a suitable peptide/protein context, even if they lack the terminal GlcNAc moiety on the Man-α1,3-Man arm. FUT8 could also fucosylate paucimannose structures when they are on glycoprotein substrates. Such core-fucosylated paucimannosylation is a prominent feature of lysosomal proteins of human neutrophils and several types of cancers. We also found that sialylation of N-glycans significantly reduced their activity as a substrate of FUT8. Kinetic analysis demonstrated that Fmoc aglycone modification could either improve the turnover rate or decrease the KM value depending on the nature of the substrates, thus significantly enhancing the overall efficiency of FUT8 catalyzed fucosylation. Our results indicate that an appropriate aglycone context of N-glycans could significantly broaden the acceptor substrate specificity of FUT8 beyond what has previously been thought.


Subject(s)
Erythropoietin/metabolism , Fucose/metabolism , Fucosyltransferases/metabolism , Glycoproteins/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mannose/metabolism , Polysaccharides/metabolism , Animals , Carbohydrate Sequence , Chickens , Erythropoietin/chemistry , Erythropoietin/genetics , Fluorenes/chemistry , Fucose/chemistry , Fucosyltransferases/chemistry , Fucosyltransferases/genetics , Gene Expression , Glycoproteins/chemistry , Glycoproteins/genetics , Glycosylation , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , HEK293 Cells , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV-1/genetics , HIV-1/metabolism , Humans , Kinetics , Mannose/chemistry , Ovalbumin/chemistry , Ovalbumin/genetics , Ovalbumin/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Polysaccharides/chemistry , Substrate Specificity
5.
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
6.
Biochemistry ; 59(38): 3541-3553, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32897051

ABSTRACT

Granulocyte macrophage colony stimulating factor (GMCSF) is an immunomodulatory cytokine that is harnessed as a therapeutic. GMCSF is known to interact with other clinically important molecules, such as heparin, suggesting that endogenous and administered GMCSF has the potential to modulate orthogonal treatment outcomes. Thus, molecular level characterization of GMCSF and its interactions with biologically active compounds is critical to understanding these mechanisms and predicting clinical consequences. Here, we dissect the biophysical factors that facilitate the GMCSF-heparin interaction, previously shown to be pH-dependent, using nuclear magnetic resonance spectroscopy, surface plasmon resonance, and molecular dynamics simulations. We find that the affinity of GMCSF for heparin increases not only with a transition to acidic pH but also with an increase in heparin chain length. Changes in local flexibility, including a disruption of the N-terminal helix at acidic pH, also accompany the binding of heparin to GMCSF. We use molecular dynamics simulations to propose a mechanism in which a positive binding pocket that is not fully solvent accessible at neutral pH becomes more accessible at acidic pH, facilitating the binding of heparin to the protein.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Heparin/metabolism , Animals , Binding Sites , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Heparin/chemistry , Humans , Hydrogen-Ion Concentration , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Surface Plasmon Resonance , Swine
7.
Acc Chem Res ; 53(9): 1749-1760, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32786230

ABSTRACT

A major function of the immune system is to detect threat from foreign invaders, tissue damage, or cancer and to mount a counter response that resolves the threat, restores homeostasis, and supplies immunological memory to prevent a second assault. Our increasing understanding of the immune system has opened up numerous avenues for modulating immune responses against infections, cancer, and autoimmunity. However, agents used for immunomodulation have been traditionally administered systemically via bolus injection, leading to unintended consequences by disrupting homeostasis at nontarget sites. Consequently, systemic hyperactivation and hypoactivation can result from bolus administration of immune-activators and immunosuppressants, respectively. Macroscale biomaterial scaffolds can instead be placed at the intended target site to provide both localized, controlled release of immunomodulatory agents and control over local immune cell trafficking and function, potentially maximizing therapeutic efficacy and limiting systemic exposure. These scaffolds have found utility in the area of cancer immunotherapy, especially in situ cancer vaccination where controlled release of factors such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and the local presentation of tumor antigen and danger signals lead to the recruitment of immature dendritic cells and facilitate their activation and antigen presentation. These cells eventually migrate into secondary lymphoid organs where they prime tumor specific T cells for downstream tumor clearance. Scaffolds can also be used in adoptive T cell therapy to generate large numbers of potent antigen specific T cells or chimeric antigen receptor (CAR) T cells in vitro for subsequent delivery to patients. Macroscale biomaterial scaffolds have also found utility beyond cancer immunotherapy and have been developed to promote immune tolerance by regulatory T cell induction and to expedite tissue regeneration. The design of these macroscale biomaterial scaffolds considers their biocompatibility, biodegradability, mode of delivery, porosity, and kinetics of therapeutic cargo release. Consequently, the numerous approaches that have been developed to fabricate biomaterial scaffolds are aimed at tuning these parameters to achieve the desired therapeutic outcome. This Account will discuss the use of biomaterial scaffolds as niches for immunomodulation and will focus on (1) approaches that have been used to fabricate various biomaterial systems being employed as niches for immunomodulation and (2) how these biomaterial systems have been used to modulate immune responses, specifically in area of cancer immunotherapy, where we will discuss the role of macroscale biomaterial scaffolds for in situ vaccination and in vitro T cell expansion. We will also briefly discuss the utility of biomaterial scaffolds beyond cancer, drawing examples from tolerance and tissue regeneration.


Subject(s)
Biocompatible Materials/chemistry , Immunologic Factors/therapeutic use , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/therapeutic use , Gels/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Immunologic Factors/chemistry , Immunotherapy , Nanoparticles/chemistry , Neoplasms/drug therapy , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Receptors, Chimeric Antigen/metabolism , Stem Cell Transplantation , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Bioconjug Chem ; 31(5): 1551-1561, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32319752

ABSTRACT

Human granulocyte-macrophage colony-stimulating factor (hGMCSF) is crucial in the immune system as it stimulates survival, proliferation, differentiation, and functional activation of myeloid hematopoietic cells. hGMCSF is integral to approved therapies, including monoclonal antibodies against checkpoint inhibitors, chimeric antigen receptors, and prevention of chemotherapy-induced neutropenia. Recombinant hGMCSF can be purified from Escherichia. coli; however, it forms inclusion bodies that require solubilization and refolding. Alternatively, this manuscript describes its fusion with an elastin-like polypeptide (ELP). Previously reported as purification tags and solubility enhancers, ELPs are recombinant polypeptides that undergo reversible temperature-dependent phase separation. This report is the first to show that fusion to an ELP enables direct purification of hGMCSF fusions from the soluble fraction of bacterial lysate. Surprisingly, these ELP-fusions assemble stable, small, spherical nanoparticles that maintain pro-mitotic activity of hGMCSF. These nanoparticles exhibit ELP-mediated phase separation; however, nanoparticle assembly significantly increases the entropic and enthalpic cost of phase separation compared to ELP alone. The attachment of a high molecular weight ELP to a difficult-to-express protein, like hGMCSF, appears to be a useful strategy to stabilize bioactive, protein-based nanoparticles, which may have broad applications in medicine and biology.


Subject(s)
Elastin/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Nanoparticles , Entropy , Humans , Temperature
9.
Int J Pharm ; 567: 118421, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31176849

ABSTRACT

Intratumoral injection of biocompatible gels is increasingly used for the sustained delivery of drugs and vaccines to enhance the anti-cancer immune response. Granulocyte-macrophage colony stimulating factor (GM-CSF) has become an attractive adjuvant thanks to its ability to boost the antitumor immune response by inducing proliferation, maturation and migration of the dendritic-cells (DCs) and the differentiation of lymphocytes. Killed Mycobacteria, such as Heat-killed Mycobacterium tuberculosis (HKMT) have been used in several studies as TLR-2 agonist to increase maturation of DCs. In this study, we designed a mucoadhesive thermosensitive formulation for the local delivery of GM-CSF and HKMT in order to enhance DCs activation and improve the local antitumor immune response. This formulation was selected based on its elastic and mucoadhesive properties obtained thanks to rheological studies. More importantly, intratumoral residence time of the labelled gel and protein were evidenced by means of MRI and non invasive in vivo optical imaging. Then, the efficacy of the combination of immunomodulators loaded thermogel was demonstated in vitro and in vivo. The selected thermogel exhibits rheological properties which confer a good elasticity and increased residence time of the immunostimulatory agents in the tumor, thus increasing the recruitment of DCs and T cytotoxic CD8+ lymphocytes.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Hydrogels/administration & dosage , Immunologic Factors/administration & dosage , Mycobacterium tuberculosis , Neoplasms/drug therapy , Adhesiveness , Animals , Bone Marrow/drug effects , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Dendritic Cells/drug effects , Drug Delivery Systems , Female , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Hydrogels/chemistry , Immunologic Factors/chemistry , Mice , Mice, Inbred BALB C , Mucins/chemistry , NIH 3T3 Cells , Neoplasms/immunology , Neoplasms/pathology , Optical Imaging , Poloxamer/administration & dosage , Poloxamer/chemistry , Rheology
10.
Biophys J ; 117(2): 214-228, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31235180

ABSTRACT

Understanding how polypeptides can efficiently and reproducibly attain a self-entangled conformation is a compelling biophysical challenge that might shed new light on our general knowledge of protein folding. Complex lassos, namely self-entangled protein structures characterized by a covalent loop sealed by a cysteine bridge, represent an ideal test system in the framework of entangled folding. Indeed, because cysteine bridges form in oxidizing conditions, they can be used as on/off switches of the structure topology to investigate the role played by the backbone entanglement in the process. In this work, we have used molecular dynamics to simulate the folding of a complex lasso glycoprotein, granulocyte-macrophage colony-stimulating factor, modeling both reducing and oxidizing conditions. Together with a well-established Go-like description, we have employed the elastic folder model, a coarse-grained, minimalistic representation of the polypeptide chain driven by a structure-based angular potential. The purpose of this study is to assess the kinetically optimal pathways in relation to the formation of the native topology. To this end, we have implemented an evolutionary strategy that tunes the elastic folder model potentials to maximize the folding probability within the early stages of the dynamics. The resulting protein model is capable of folding with high success rate, avoiding the kinetic traps that hamper the efficient folding in the other tested models. Employing specifically designed topological descriptors, we could observe that the selected folding routes avoid the topological bottleneck by locking the cysteine bridge after the topology is formed. These results provide valuable insights on the selection of mechanisms in self-entangled protein folding while, at the same time, the proposed methodology can complement the usage of established minimalistic models and draw useful guidelines for more detailed simulations.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Protein Folding , Algorithms , Molecular Dynamics Simulation
11.
Protein Expr Purif ; 160: 66-72, 2019 08.
Article in English | MEDLINE | ID: mdl-30998976

ABSTRACT

The most common approaches to improve soluble expression of heterologous proteins are applications of molecular chaperones such as DnaK, DnaJ, GrpE, GroEL and GroES. The aim of present study was to enhance soluble expression of granulocyte-macrophage colony-stimulating factor (GM-CSF) in Escherichia coli by different approaches including modification of cultivation and induction conditions, and thermally, genetically and chemically enhancement of expression of cellular chaperones. To genetically enhance amount of molecular chaperones, co-expression of pET28-GM-CSF and pKJE7 plasmids was performed. The soluble expressed protein was affinity purified and subjected to endotoxin removal. Co-expression with molecular chaperones significantly increased soluble expression of GM-CSF. Addition of chemical chaperones and osmolytes like NaCl (0.5 M), sucrose (0.5 M), sorbitol (0.5 M) and MgCl2 (1 mM) to growing media could improve solubility of GM-CSF. Biological activity of purified GM-CSF was confirmed based on its proliferative effect on HL-60 cell lines. The approach developed in the present study can be applied to improve soluble expression of other recombinant protein proteins.


Subject(s)
Escherichia coli Proteins/genetics , Escherichia coli/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Molecular Chaperones/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Humans , Molecular Chaperones/metabolism , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Solubility
12.
Protein Expr Purif ; 159: 69-74, 2019 07.
Article in English | MEDLINE | ID: mdl-30917920

ABSTRACT

Human granulocyte-macrophage colony stimulating factor (hGM-CSF) is a hematopoietic growth factor. It is widely employed as a therapeutic agent targeting neutropenia in cancer patients undergoing chemotherapy and in patients with AIDS or after bone marrow transplantation. In this study, we constructed the recombinant baculoviruses for the expression of recombinant hGM-CSF (rhGM-CSF) with two small affinity tags (His-tag and Strep-tag) at the N or C-terminus. Compared to N-tagged rhGM-CSF, C-tagged rhGM-CSF was highly recovered from silkworm hemolymph. The purified rhGM-CSF proteins migrated as a diffuse band and were confirmed to hold N-glycosylations. A comparable activity was achieved when commercial hGM-CSF was tested as a control. Considering the high price of hGM-CSF in the market, our results and strategies using silkworm-baculovirus system can become a great reference for mass production of the active rhGM-CSF at a lower cost.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Animals , Baculoviridae/genetics , Base Sequence , Bombyx/genetics , Cell Extracts/chemistry , Cell Line , Chromatography, Affinity , Gene Expression , Glycosylation , Humans , Hydrogen-Ion Concentration , Protein Stability , Virus Diseases
13.
Biomed Mater ; 14(3): 035003, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30690433

ABSTRACT

In recent years, applications of biopolymers such as hyaluronic acid (HA) for wound dressing have attracted more attention. However, the poor mechanical properties of HA-based wound dressings limit their clinical applications. Incorporation of reinforcing agents such as nanocrystalline cellulose (CNC) in HA-based wound dressings can improve their mechanical properties. In addition, controlled delivery of growth factors to the wound site using nanoparticles can significantly improve the healing process. In this study, we focus on development and characterization of a novel CNC reinforced HA-based composite containing chitosan nanoparticles loaded with GM-CSF (CNC-HA/GM-CSF-Chi-NPs composite) as an effective wound dressing. CNC-HA/GM-CSF-Chi-NPs composite showed some physicochemical characteristics such as appropriate mechanical properties, high swelling capacity (swelling ratio: 2622.1% ± 35.2%) and controlled release of GM-CSF up to 48 h which make it an excellent candidate for wound dressing. In vivo investigation showed that, after 13 d, the wounds covered with CNC-HA/GM-CSF-Chi-NPs composite could reach to nearly full wound closure and complete re-epithelialization compared to the normal saline treated wounds which exhibited nearly 70% of wound size reduction. Furthermore, the CNC-HA/GM-CSF-Chi-NPs composite treated wounds exhibited significantly lower inflammatory reaction, enhanced re-epithelialization and improved granulation tissue formation compared with CNC-HA/Chi-NPs composite treated wound; it might be due to positive effects of GM-CSF on the wound healing process. Our results suggest that CNC-HA/GM-CSF-Chi-NPs composite can be potentially applied in clinical practice for wound treatment.


Subject(s)
Cellulose/chemistry , Chitosan/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Hyaluronic Acid/chemistry , Nanoparticles/chemistry , Wound Healing , Animals , Bandages , Humans , Inflammation , Male , Materials Testing , Nanocomposites , Rats , Rats, Wistar , Re-Epithelialization , Spectroscopy, Fourier Transform Infrared , Stress, Mechanical , Tensile Strength
14.
Adv Healthc Mater ; 7(10): e1701469, 2018 05.
Article in English | MEDLINE | ID: mdl-29441705

ABSTRACT

A covalently crosslinked methacrylated (MA)-alginate cryogel vaccine has been previously shown to generate a potent response against murine melanoma, but is not mechanically robust and requires a large 16G needle for delivery. Here, covalent and ionic crosslinking of cryogels are combined with the hypothesis that this will result in a tough MA-alginate cryogel with improved injectability. All tough cryogels can be injected through a smaller, 18G needle without sustaining any damage, while covalently crosslinked-only cryogels break after injection. Cytosine-phosphodiester-guanine (CpG)-delivering tough cryogels effectively activate dendritic cells (DCs). Granulocyte macrophage colony-stimulating factor releasing tough cryogels recruit four times more DCs than blank gels by day 7 in vivo. The tough cryogel vaccine induces strong antigen-specific cytotoxic T-lymphocyte and humoral responses. These vaccines prevent tumor formation in 80% of mice inoculated with HER2/neu-overexpressing DD breast cancer cells. The MA-alginate tough cryogels provide a promising minimally invasive delivery platform for cancer vaccinations.


Subject(s)
Alginates/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/pharmacology , Cryogels/pharmacology , Mammary Neoplasms, Experimental/therapy , Alginates/chemistry , Animals , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/chemistry , Cryogels/chemistry , Female , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/pharmacology
15.
J Control Release ; 259: 195-202, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28389408

ABSTRACT

Using recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) as a model drug, the present study demonstrated a practically feasible process to produce polymeric microspheres for sustained-release delivery of protein drugs with preserved integrity. This process is featured with pre-loading proteins into polysaccharide fine particles via a self-standing aqueous-aqueous "emulsion", prior to microencapsulation into the microspheres. The protein drug, rhGM-CSF, was partitioned thermodynamically into a dextran dispersed phase of the aqueous-aqueous emulsion, followed by lyophilization and removal of the polyethylene glycol (PEG) continuous phase (using an organic solvent not penetrating into dextran matrix). The harvested dextran particles were then suspended in a dichloromethane solution of polylatic-co-glyclic acids (PLGA) and emulsified in a polyvinyl alcohol (PVA) and NaCl solution of small volume to form embryonic microspheres. The emulsion was then transferred into a NaCl solution of large volume to extract the organic solvent and harden the embryonic microspheres. The obtained rhGM-CSF microspheres showed a satisfied release profile with the day-to-day variation within 9 folds over the multi-weeks long release period. At the same time, the integrity (defined freedom of aggregates measured by SEC-HPLC) and bioactivity (defined by TF-1 cell proliferation) of the proteins were well preserved. The present formulation process ensured good reproducibility and over 89% protein encapsulation efficiency, and practically feasible to adapt to scaled productions.


Subject(s)
Delayed-Action Preparations/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Microspheres , Animals , Cell Line , Cell Survival/drug effects , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Dextrans/chemistry , Drug Liberation , Granulocyte-Macrophage Colony-Stimulating Factor/blood , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacokinetics , Humans , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Rats, Wistar
16.
Bioengineered ; 8(2): 120-123, 2017 Mar 04.
Article in English | MEDLINE | ID: mdl-27459024

ABSTRACT

Most of the cancer treatment strategies from chemotherapy to radiotherapy render cancer cells apoptotic and these apoptotic cancer cells accumulate at the tumor sites. The accumulation of apoptotic cancer cells often result in inflammation and autoimmune responses causing serious health implications. Macrophages, which are effective immune combatants, can help in the clearance of these deleterious occupants. Granulocyte macrophage colony stimulating factor (GM-CSF) is a key cytokine, modulator of immune system and responsible for growth and differentiation of granulocytes and macrophages. In this regard, supply of recombinant GM-CSF can enhance the capability of macrophages for clearance of apoptotic cancer cells. However, delivery of the cytokine in vivo can suffer from certain disadvantages like faster depletion, less stability and low targeting efficiency. We believe that the stability and sustained release of GM-CSF can be improved through its encapsulation inside appropriately designed nanoparticles.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Immunotherapy , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasms/therapy , Apoptosis/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Macrophages/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
17.
Nat Commun ; 7: 13228, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27819269

ABSTRACT

Subversion of the host immune system by viruses is often mediated by molecular decoys that sequester host proteins pivotal to mounting effective immune responses. The widespread mammalian pathogen parapox Orf virus deploys GIF, a member of the poxvirus immune evasion superfamily, to antagonize GM-CSF (granulocyte macrophage colony-stimulating factor) and IL-2 (interleukin-2), two pleiotropic cytokines of the mammalian immune system. However, structural and mechanistic insights into the unprecedented functional duality of GIF have remained elusive. Here we reveal that GIF employs a dimeric binding platform that sequesters two copies of its target cytokines with high affinity and slow dissociation kinetics to yield distinct complexes featuring mutually exclusive interaction footprints. We illustrate how GIF serves as a competitive decoy receptor by leveraging binding hotspots underlying the cognate receptor interactions of GM-CSF and IL-2, without sharing any structural similarity with the cytokine receptors. Our findings contribute to the tracing of novel molecular mimicry mechanisms employed by pathogenic viruses.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-2/immunology , Parapoxvirus/immunology , Viral Proteins/immunology , Crystallography, X-Ray , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Interleukin-2/chemistry , Interleukin-2/metabolism , Models, Molecular , Multiprotein Complexes/chemistry , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Parapoxvirus/metabolism , Poxviridae Infections/immunology , Poxviridae Infections/metabolism , Poxviridae Infections/virology , Protein Binding , Viral Proteins/chemistry , Viral Proteins/metabolism
18.
Biochem Biophys Res Commun ; 479(2): 266-271, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27638310

ABSTRACT

Recombinant human GM-CSF (rhGM-CSF) from yeast has been clinically applied to immunosuppressed patients. The production of suspension-cultured rice-cell-derived rhGM-CSF (rrhGM-CSF), which has a longer blood clearance time and the same bioactivity as yeast-derived rhGM-CSF, and the analysis of its N-glycans have been reported recently. However, there are no previous reports of the O-glycosylation of rhGM-CSF from plant cells, and so this study investigated O-glycans, O-glycosylation sites, and their structural role in rrhGM-CSF. Monosaccharide analysis revealed the presence of O-glycans comprising arabinose and galactose. Eight O-glycans comprising four arabinose residues with zero to seven galactose residues along with their relative quantities were analyzed. Analysis of pronase-digested glycopeptides indicated that the O-glycans are partially attached to Ser 5, Ser 7, Ser 9, or Thr 10 residues, and glycan heterogeneity was confirmed at each site. Pro-to-hydroxyproline conversions occurred at Pro 2, Pro 6, and Pro 8 residues. The preparation of deglycosylated rrhGM-CSFs revealed that deglycosylation greatly affects their α-helix structures. These findings indicate that O-glycans of rrhGM-CSF are essential for maintaining its structural stability and result in an extended in vivo half-life, but without affecting its biological function. This is the first report on the O-glycosylation of rhGM-CSF derived from plant cells.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Oryza/metabolism , Polysaccharides/chemistry , Arabinose/chemistry , Cells, Cultured , Chromatography, High Pressure Liquid , Circular Dichroism , Galactose/chemistry , Glycopeptides/chemistry , Glycosylation , Humans , Monosaccharides/chemistry , Protein Domains , Protein Structure, Secondary , Recombinant Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
19.
Structure ; 24(8): 1271-1281, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27396825

ABSTRACT

The GM-CSF, IL-3, and IL-5 receptors constitute the ßc family, playing important roles in inflammation, autoimmunity, and cancer. Typical of heterodimeric type I cytokine receptors, signaling requires recruitment of the shared subunit to the initial cytokine:α subunit binary complex through an affinity conversion mechanism. This critical process is poorly understood due to the paucity of crystal structures of both binary and ternary receptor complexes for the same cytokine. We have now solved the structure of the binary GM-CSF:GMRα complex at 2.8-Å resolution and compared it with the structure of the ternary complex, revealing distinct conformational changes. Guided by these differences we performed mutational and functional studies that, importantly, show GMRα interactions playing a major role in receptor signaling while ßc interactions control high-affinity binding. These results support the notion that conformational changes underlie the mechanism of GM-CSF receptor activation and also suggest how related type I cytokine receptors signal.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Protein Subunits/chemistry , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Amino Acid Motifs , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
20.
Protein Expr Purif ; 121: 52-60, 2016 May.
Article in English | MEDLINE | ID: mdl-26792558

ABSTRACT

Super magnetic nanoparticle NiFe2O4 with high magnetization, physical and chemical stability was introduced as a core particle which exhibits high thermal stability (>97%) during the harsh coating process. Instead of multi-stage process for coating, the magnetic nanoparticles was mineralized via one step coating by a cheap, safe, stable and recyclable alumina sol-gel lattice (from bohemite source) saturated by nickel ions. The TEM, SEM, VSM and XRD imaging and BET analysis confirmed the structural potential of NiFe2O4@NiAl2O4 core-shell magnetic nanoparticles for selective and sensitive purification of His-tagged protein, in one step. The functionality and validity of the nickel magnetic nanoparticles were attested by purification of three different bioactive His-tagged recombinant fusion proteins including hIGF-1, GM-CSF and bFGF. The bonding capacity of the nickel magnetics nanoparticles was studied by Bradford assay and was equal to 250 ± 84 µg Protein/mg MNP base on protein size. Since the metal ion leakage is the most toxicity source for purification by nickel magnetic nanoparticles, therefor the nickel leakage in purified final protein was determined by atomic absorption spectroscopy and biological activity of final purified protein was confirmed in comparison with reference. Also, in vitro cytotoxicity of nickel magnetic nanoparticles and trace metal ions were investigated by MTS assay analysis. The results confirmed that the synthesized nickel magnetic nanoparticles did not show metal ion toxicity and not affected on protein folding.


Subject(s)
Ferric Compounds/chemistry , Magnetite Nanoparticles/chemistry , Nickel/chemistry , Recombinant Fusion Proteins/isolation & purification , Aluminum/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/chemistry , Granulocyte-Macrophage Colony-Stimulating Factor/isolation & purification , Histidine/chemistry , Humans , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/isolation & purification , Phase Transition , Recombinant Fusion Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...