Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Front Microbiol ; 15: 1324403, 2024.
Article in English | MEDLINE | ID: mdl-38903788

ABSTRACT

Microbiome research has gained much attention in recent years as the importance of gut microbiota in regulating host health becomes increasingly evident. However, the impact of radiation on the microbiota in the murine bone marrow transplantation model is still poorly understood. In this paper, we present key findings from our study on how radiation, followed by bone marrow transplantation with or without T cell depletion, impacts the microbiota in the ileum and caecum. Our findings show that radiation has different effects on the microbiota of the two intestinal regions, with the caecum showing increased interindividual variation, suggesting an impaired ability of the host to regulate microbial symbionts, consistent with the Anna Karenina principle. Additionally, we observed changes in the ileum composition, including an increase in bacterial taxa that are important modulators of host health, such as Akkermansia and Faecalibaculum. In contrast, radiation in the caecum was associated with an increased abundance of several common commensal taxa in the gut, including Lachnospiraceae and Bacteroides. Finally, we found that high doses of radiation had more substantial effects on the caecal microbiota of the T-cell-depleted group than that of the non-T-cell-depleted group. Overall, our results contribute to a better understanding of the complex relationship between radiation and the gut microbiota in the context of bone marrow transplantation and highlight the importance of considering different intestinal regions when studying microbiome responses to environmental stressors.

2.
J Colloid Interface Sci ; 664: 389-399, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38479275

ABSTRACT

Electrochemical water electrolysis is a promising method for sustainable hydrogen production while transiting towards hydrogen economy. Among many, the Anion Exchange Membrane (AEM) based water electrolyzer is an emerging yet potentially affordable technology on maturity for producing large-scale hydrogen accommodating the usage of Non-Platinum Group Metal (non-PGM) based inexpensive electrocatalysts. Herein, we demonstrate the excellent performance of a bifunctional Nickel Copper Phosphide-Nickel sulphide (NCP-NS) electrocatalyst with a unique tensile nanostructure obtained via a facile, controlled ambient galvanic displacement route. An AEM electrolyzer with a larger active area of 10 cm2 stacked with the symmetric NCP-NS electrodes and a membrane demonstrates scalability with a requirement of a mere 1.66 V to reach a current density of 10 mA cm-2. The nickel-copper phosphide boosts the kinetics of charge transfer between the electrode and electrolyte interface, while a unique combination of a few nickel sulphide phases present in the catalyst provides sufficiently appropriate active sites for the overall water electrolysis. For the first time, we report a room temperature performance of âˆ¼ 230 mA cm-2 at 2 V for a non-PGM-based bifunctional electrocatalyst with exceptional durability for over 300 h of operation in an AEM water electrolyser with a retention rate of 95 %-97 % at a current density range of 80-800 mA cm-2.

3.
Endocrine ; 85(2): 626-637, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38424350

ABSTRACT

AIM: Much focus of immunotherapy for type 1 diabetes (T1D) has been devoted on selectively boosting regulatory T (Treg) cells using low dose IL-2 due to their constitutive expression of IL-2Rα, CD25. However, several clinical trials using a low dose of IL-2 only showed a limited improvement of metabolic control. It can therefore be hypothesized that further decreasing IL-2 dosage may increase the selective responsiveness of Treg cells. METHODS: We induced experimental T1D using multiple low dose streptozotocin (STZ) injections and treated the mice with an ultra-low dose IL-2 (uIL-2, approximately 7-fold lower than low dose). Immune response was studied using multicolor flow cytometry. RESULTS: We found that uIL-2 did not protect STZ mice from developing hyperglycemia. It did neither increase Treg cell proportions, nor did it correct the phenotypic shift of Treg cells seen in T1D. It only partially decreased the proportion of IFN-γ+ T cells. Likewise, uIL-2 also did not protect the dysfunction of regulatory B (Breg) cells. Strikingly, when administered in combination with an anti-inflammatory cytokine IL-35, uIL-2 abrogated IL-35's protective effect. Low dose IL-2, on the other hand, protected half of the STZ mice from developing hyperglycemia. No difference was found in the Treg and Breg response, and it only tended to decrease CD80 expression in macrophages and dendritic cells. CONCLUSION: In conclusion, further decreasing IL-2 dosage may not be a suitable approach for T1D therapy, and the limited success suggests that an alternative low dose IL-2 therapy strategy or other immunotherapies should be considered.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Interleukin-2 , T-Lymphocytes, Regulatory , Animals , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , T-Lymphocytes, Regulatory/drug effects , Mice , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/immunology , Interleukins , Male , Mice, Inbred C57BL , Immunotherapy/methods
4.
Front Immunol ; 14: 1219598, 2023.
Article in English | MEDLINE | ID: mdl-37483613

ABSTRACT

The incidence of Diabetes Mellitus is increasing globally. Individuals who have been burdened with diabetes for many years often develop complications as a result of hyperglycemia. More and more research is being conducted highlighting inflammation as an important factor in disease progression. In all kinds of diabetes, hyperglycemia leads to activation of alternative glucose metabolic pathways, resulting in problematic by-products including reactive oxygen species and advanced glycation end products. This review takes a look into the pathogenesis of three specific diabetic complications; retinopathy, nephropathy and neuropathy as well as their current treatment options. By considering recent research papers investigating the effects of immunotherapy on relevant conditions in animal models, multiple strategies are suggested for future treatment and prevention of diabetic complications with an emphasis on molecular targets associated with the inflammation.


Subject(s)
Diabetes Complications , Diabetes Mellitus , Hyperglycemia , Animals , Prospective Studies , Inflammation , Immunotherapy
5.
Water Sci Technol ; 87(1): 193-217, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36640032

ABSTRACT

In this work, heavy metals were removed simultaneously using wheat bran as an adsorbent. For batch experiments, the Box-Behnken design of response surface methodology was used and the effect of dye on metal removal was analysed. It has been observed that the presence of dye has reduced the removal of each metal in the range of 100-20% with no appreciable reduction in dye adsorption. The optimum pH, temperature, and adsorbent dose were found to be 7.59, 33.23 °C, and 2.90 g/L, respectively, for 79.70% chromium, 99.9% cadmium and 87.27% copper removal. It was found that Langmuir isotherm fits well with the experimental data (RMSE value up to 0.033). The maximum adsorption capacity obtained for copper, chromium, cadmium and dye were 2.17 mg/g, 1.76 mg/g, 1.52 mg/g and 3.215 mg/g, respectively. The continuous study was performed for parameters, i.e. bed height (0.15-0.45 m), flow rate (5-15 mL/min) and initial metal concentration (100-500 mg/L). In continuous study, dye acted as an interfering species and as a result breakthrough and exhaustion time decreased. The modelling and simulation of continuous adsorption process were performed. A dynamic mathematical model was developed for continuous fixed bed adsorption column to compare the breakthrough curve with experimental results.


Subject(s)
Metals, Heavy , Water Pollutants, Chemical , Wastewater , Copper , Cadmium , Hydrogen-Ion Concentration , Chromium , Adsorption , Kinetics
6.
Biomolecules ; 12(10)2022 10 16.
Article in English | MEDLINE | ID: mdl-36291702

ABSTRACT

In type 1 diabetes, dysfunctional glucose regulation occurs due to the death of insulin-producing beta-cells in the pancreatic islets. Initiation of this process is caused by the inheritance of an adaptive immune system that is predisposed to responding to beta-cell antigens, most notably to insulin itself, coupled with unknown environmental insults priming the autoimmune reaction. While autoimmunity is a primary driver in beta-cell death, there is growing evidence that cellular stress participates in the loss of beta-cells. In the beta-cell fragility model, partial loss of islet mass requires compensatory upregulation of insulin production in the remaining islets, driving a cellular stress capable of triggering apoptosis in the remaining cells. The Glis3-Manf axis has been identified as being pivotal to the relative fragility or robustness of stressed islets, potentially operating in both type 1 and type 2 diabetes. Here, we have used an AAV-based gene delivery system to enhance the expression of the anti-apoptotic protein Manf in the beta-cells of NOD mice. Gene delivery substantially lowered the rate of diabetes development in treated mice. Manf-treated mice demonstrated minimal insulitis and superior preservation of insulin production. Our results demonstrating the therapeutic potential of Manf delivery to enhance beta-cell robustness and avert clinical diabetes.


Subject(s)
Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Islets of Langerhans , Mice , Animals , Mice, Inbred NOD , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/prevention & control , Islets of Langerhans/metabolism , Insulin/genetics , Gene Transfer Techniques , Glucose , Apoptosis Regulatory Proteins/genetics , Nerve Growth Factors
7.
Eur J Med Chem ; 242: 114642, 2022 Nov 15.
Article in English | MEDLINE | ID: mdl-35987021

ABSTRACT

The Secretin/Secretin receptor (SCTR) axis is well-known for its important role in water/salt homeostasis and blood pressure control. Recent studies revealed that absence of Secretin could lead to hypertension in animals and the administration of external Secretin leads to a sharp drop in blood pressure. Therefore, Secretin receptor has emerged as a crucial drug target of interest. In this report, using structure based drug design strategy, we have identified a small compound-based Secretin receptor modulator (i.e. purmorphamine or KSD179019). The virtual docking of KSD179019 with SCTR crystal structure and homology models revealed similar binding interactions. Based on active pharmacophores of KSD179019, several derivatives were designed and sythesized. SAR studies revealed that KSD179019 is the most effective SCTR modulator and chosen for further biological evaluation, including drug like properties and anti-hypertensive effect. KSD179019 not only has a similar blood pressure lowering effect as SCT peptide, but more importantly, it has a much longer half-life (∼8 h) and can be taken orally. Preliminary preclinical studies revealed extended bioavailability and low toxicity of this compound.


Subject(s)
Antihypertensive Agents , Secretin , Animals , Antihypertensive Agents/pharmacology , Morpholines , Peptides , Purines , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone , Secretin/metabolism , Water
8.
Int J Mol Sci ; 23(14)2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35887317

ABSTRACT

Macrophages play an important role in the early development of type 1 diabetes (T1D). Based on the phenotype, macrophages can be classified into pro-inflammatory (M1) and anti-inflammatory (M2) macrophages. Despite intensive research in the field of macrophages and T1D, the kinetic response of M1/M2 ratio has not been studied in T1D. Thus, herein, we studied the M1 and M2 macrophages in the early development of T1D using the multiple low dose streptozotocin (MLDSTZ) mouse model. We determined the proportions of M1 and M2 macrophages in thymic glands, pancreatic lymph nodes and spleens on days 3, 7 and 10 after the first injection of STZ. In addition, we investigated the effect of IL-35 in vivo on the M1/M2 ratio and IL-35+ plasmacytoid dendritic cells in diabetic mice and in vitro on the sorted macrophages. Our results revealed that the M1/M2 ratio is higher in STZ-treated mice but this was lowered upon the treatment with IL-35. Furthermore, IL-35 treated mice had lower blood glucose levels and a higher proportion of IL-35+ cells among pDCs. Macrophages treated with IL-35 in vitro also had a higher proportion of M2 macrophages. Together, our data indicate that, under diabetic conditions, pro-inflammatory macrophages increased, but IL-35 treatment decreased the pro-inflammatory macrophages and increased anti-inflammatory macrophages, further suggesting that IL-35 prevents hyperglycemia by maintaining the anti-inflammatory phenotype of macrophages and other immune cells. Thus, IL-35 should be further investigated for the treatment of T1D and other autoimmune disorders.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Hyperglycemia , Animals , Diabetes Mellitus, Type 1/pathology , Interleukins , Macrophages , Mice , Streptozocin
10.
J Exp Med ; 219(7)2022 07 04.
Article in English | MEDLINE | ID: mdl-35699942

ABSTRACT

Interleukin 2 (IL-2) is a key homeostatic cytokine, with therapeutic applications in both immunogenic and tolerogenic immune modulation. Clinical use has been hampered by pleiotropic functionality and widespread receptor expression, with unexpected adverse events. Here, we developed a novel mouse strain to divert IL-2 production, allowing identification of contextual outcomes. Network analysis identified priority access for Tregs and a competitive fitness cost of IL-2 production among both Tregs and conventional CD4 T cells. CD8 T and NK cells, by contrast, exhibited a preference for autocrine IL-2 production. IL-2 sourced from dendritic cells amplified Tregs, whereas IL-2 produced by B cells induced two context-dependent circuits: dramatic expansion of CD8+ Tregs and ILC2 cells, the latter driving a downstream, IL-5-mediated, eosinophilic circuit. The source-specific effects demonstrate the contextual influence of IL-2 function and potentially explain adverse effects observed during clinical trials. Targeted IL-2 production therefore has the potential to amplify or quench particular circuits in the IL-2 network, based on clinical desirability.


Subject(s)
Interleukin-2 , Killer Cells, Natural , T-Lymphocytes, Regulatory , Animals , Immunity, Innate , Interleukin-2/biosynthesis , Interleukin-2/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
11.
Nat Immunol ; 23(6): 878-891, 2022 06.
Article in English | MEDLINE | ID: mdl-35618831

ABSTRACT

The ability of immune-modulating biologics to prevent and reverse pathology has transformed recent clinical practice. Full utility in the neuroinflammation space, however, requires identification of both effective targets for local immune modulation and a delivery system capable of crossing the blood-brain barrier. The recent identification and characterization of a small population of regulatory T (Treg) cells resident in the brain presents one such potential therapeutic target. Here, we identified brain interleukin 2 (IL-2) levels as a limiting factor for brain-resident Treg cells. We developed a gene-delivery approach for astrocytes, with a small-molecule on-switch to allow temporal control, and enhanced production in reactive astrocytes to spatially direct delivery to inflammatory sites. Mice with brain-specific IL-2 delivery were protected in traumatic brain injury, stroke and multiple sclerosis models, without impacting the peripheral immune system. These results validate brain-specific IL-2 gene delivery as effective protection against neuroinflammation, and provide a versatile platform for delivery of diverse biologics to neuroinflammatory patients.


Subject(s)
Astrocytes , Biological Products , Animals , Brain , Humans , Interleukin-2/genetics , Interleukins , Mice , Neuroinflammatory Diseases , T-Lymphocytes, Regulatory
12.
J Allergy Clin Immunol ; 149(1): 275-291, 2022 01.
Article in English | MEDLINE | ID: mdl-34111449

ABSTRACT

BACKGROUND: P17, a peptide isolated from Tetramorium bicarinatum ant venom, is known to induce an alternative phenotype of human monocyte-derived macrophages via activation of an unknown G protein-coupled receptor (GPCR). OBJECTIVE: We sought to investigate the mechanism of action and the immunomodulatory effects of P17 mediated through MRGPRX2 (Mas-related G protein-coupled receptor X2). METHODS: To identify the GPCR for P17, we screened 314 GPCRs. Upon identification of MRGPRX2, a battery of in silico, in vitro, ex vivo, and in vivo assays along with the receptor mutation studies were performed. In particular, to investigate the immunomodulatory actions, we used ß-hexosaminidase release assay, cytokine releases, quantification of mRNA expression, cell migration and differentiation assays, immunohistochemical labeling, hematoxylin and eosin, and immunofluorescence staining. RESULTS: P17 activated MRGPRX2 in a dose-dependent manner in ß-arrestin recruitment assay. In LAD2 cells, P17 induced calcium and ß-hexosaminidase release. Quercetin- and short hairpin RNA-mediated knockdown of MRGPRX2 reduced P17-evoked ß-hexosaminidase release. In silico and in vitro mutagenesis studies showed that residue Lys8 of P17 formed a cation-π interaction with the Phe172 of MRGPRX2 and [Ala8]P17 lost its activity partially. P17 activated LAD2 cells to recruit THP-1 and human monocytes in Transwell migration assay, whereas MRGPRX2-impaired LAD2 cells cannot. In addition, P17-treated LAD2 cells stimulated differentiation of THP-1 and human monocytes, as indicated by the enhanced expression of macrophage markers cluster of differentiation 11b and TNF-α by quantitative RT-PCR. Immunohistochemical and immunofluorescent staining suggested monocyte recruitment in mice ears injected with P17. CONCLUSIONS: Our data provide novel structural information regarding the interaction of P17 with MRGPRX2 and intracellular pathways for its immunomodulatory action.


Subject(s)
Peptides/pharmacology , Receptors, G-Protein-Coupled/metabolism , Animals , Binding Sites , Capillary Permeability/drug effects , Cell Differentiation/drug effects , Cell Line , Chemotaxis/drug effects , Cricetulus , Cytokines/metabolism , Edema/immunology , Edema/metabolism , Evans Blue/metabolism , Gene Silencing , Humans , Male , Mast Cells/drug effects , Mice, Inbred C57BL , Models, Molecular , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Receptors, G-Protein-Coupled/genetics
13.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34884797

ABSTRACT

The anti-inflammatory role of regulatory B cells (Breg cells) has been associated with IL-35 based on studies of experimental autoimmune uveitis and encephalitis. The role of Breg cells and IL-35+ Breg cells for type 1 diabetes (T1D) remains to be investigated. We studied PBMCs from T1D subjects and healthy controls (HC) and found lowered proportions of Breg cells and IL-35+ Breg cells in T1D. To elucidate the role of Breg cells, the lymphoid organs of two mouse models of T1D were examined. Lower proportions of Breg cells and IL-35+ Breg cells were found in the animal models of T1D compared with control mice. In addition, the systemic administration of recombinant mouse IL-35 prevented hyperglycemia after multiple low dose streptozotocin (MLDSTZ) injections and increased the proportions of Breg cells and IL-35+ Breg cells. A higher proportion of IFN-γ+ cells among Breg cells were found in the PBMCs of the T1D subjects. In the MLDSTZ mice, IL-35 administration decreased the proportions of IFN-γ+ cells among the Breg cells. Our data illustrate that Breg cells may play an important role in the development of T1D and that IL-35 treatment prevents the development of hyperglycemia by maintaining the phenotype of the Breg cells under an experimental T1D condition.


Subject(s)
Anti-Inflammatory Agents/pharmacology , B-Lymphocytes, Regulatory/immunology , Diabetes Mellitus, Type 1/prevention & control , Hyperglycemia/prevention & control , Interleukins/pharmacology , Adult , Animals , Anti-Inflammatory Agents/blood , Cells, Cultured , Disease Models, Animal , Female , Humans , Hyperglycemia/chemically induced , Interferon-gamma/blood , Interleukins/blood , Lymphocyte Count , Male , Mice , Mice, Inbred NOD , Streptozocin/toxicity
14.
Biomark Med ; 15(17): 1693-1708, 2021 12.
Article in English | MEDLINE | ID: mdl-34743546

ABSTRACT

Purpose: Neurodegenerative disorders are a global health burden with costly and invasive diagnoses relying on brain imaging technology or CSF-based biomarkers. Therefore, considerable efforts to identify blood-biomarkers for Alzheimer's (AD) and Parkinson's diseases (PD) are ongoing. Objectives: This review evaluates the blood biomarkers for AD and PD for their diagnostic value. Methods: This study systematically reviewed articles published between July 1984 and February 2021. Among 1266 papers, we selected 42 studies for a systematic review and 23 studies for meta-analysis. Results & conclusion: Our analysis highlights P-tau181, T-tau and Nfl as promising blood biomarkers for AD diagnosis. Nfl levels were consistently raised in 16 AD and three PD cohorts. P-tau181 and T-tau were also significantly increased in 12 and eight AD cohorts, respectively.


Subject(s)
Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Biomarkers/blood , Parkinson Disease/blood , Parkinson Disease/diagnosis , Case-Control Studies , Humans , Longitudinal Studies , Publication Bias , Publications , Risk
15.
Sci Immunol ; 6(61)2021 07 23.
Article in English | MEDLINE | ID: mdl-34301799

ABSTRACT

Regulatory T cells (Tregs) are indispensable for the control of immune homeostasis and have clinical potential as a cell therapy for treating autoimmunity. Tregs can lose expression of the lineage-defining Foxp3 transcription factor and acquire effector T cell (Teff) characteristics, a process referred to as Treg plasticity. The extent and reversibility of such plasticity during immune responses remain unknown. Here, using a murine genetic fate-mapping system, we show that Treg stability is maintained even during exposure to a complex microbial/antigenic environment. Furthermore, we demonstrate that the observed plasticity of Tregs after adoptive transfer into a lymphopenic environment is a property limited to only a subset of the Treg population, with the nonconverting majority of Tregs being resistant to plasticity upon secondary stability challenge. The unstable Treg fraction is a complex mixture of phenotypically distinct Tregs, enriched for naïve and neuropilin-1-negative Tregs, and includes peripherally induced Tregs and recent thymic emigrant Tregs These results suggest that a "purging" process can be used to purify stable Tregs that are capable of robust fate retention, with potential implications for improving cell transfer therapy.


Subject(s)
T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Cytokines/blood , Feces/chemistry , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gastrointestinal Microbiome/genetics , Male , Mice, Transgenic , Neuropilin-1/immunology
16.
J Mol Biol ; 433(7): 166843, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33539880

ABSTRACT

Kisspeptin receptor (Kiss1R) is an important receptor that plays central regulatory roles in reproduction by regulating hormone release in the hypothalamus. We hypothesize that the formation of heterocomplexes between Kiss1R and other hypothalamus G protein-coupled receptors (GPCRs) affects their cellular signaling. Through screening of potential interactions between Kiss1R and hypothalamus GPCRs, we identified G protein-coupled estrogen receptor (GPER) as one interaction partner of Kiss1R. Based on the recognised function of kisspeptin and estrogen in regulating the reproductive system, we investigated the Kiss1R/GPER heterocomplex in more detail and revealed that complex formation significantly reduced Kiss1R-mediated signaling. GPER did not directly antagonize Kiss1R conformational changes upon ligand binding, but it rather reduced the cell surface expression of Kiss1R. These results therefore demonstrate a regulatory mechanism of hypothalamic hormone receptors via receptor cooperation in the reproductive system and modulation of receptor sensitivity.


Subject(s)
Hypothalamus/metabolism , Multiprotein Complexes/genetics , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1/genetics , Animals , Hormones/biosynthesis , Hormones/genetics , Humans , Multiprotein Complexes/ultrastructure , Protein Binding/genetics , Receptors, Cell Surface/genetics , Receptors, Estrogen/ultrastructure , Receptors, G-Protein-Coupled/ultrastructure , Receptors, Kisspeptin-1/ultrastructure , Signal Transduction/genetics
17.
Microb Cell Fact ; 20(1): 26, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33509221

ABSTRACT

Engine oil used in automobiles is a threat to soil and water due to the recalcitrant properties of its hydrocarbons. It pollutes surrounding environment which affects both flora and fauna. Microbes can degrade hydrocarbons containing engine oil and utilize it as a substrate for their growth. Our results demonstrated that cell-free broth of Bacillus velezensis KLP2016 (Gram + ve, endospore forming; Accession number KY214239) recorded an emulsification index (E24%) from 52.3% to 65.7% against different organic solvents, such as benzene, pentane, cyclohexane, xylene, n-hexane, toluene and engine oil. The surface tension of the cell-free broth of B. velezensis grown in Luria-Bertani broth at 35 °C decreased from 55 to 40 mN m-1at critical micelle concentration 17.2 µg/mL. The active biosurfactant molecule of cell-free broth of Bacillus velezensis KLP2016 was purified by Dietheylaminoethyl-cellulose and size exclusion chromatography, followed by HPLC (RT = 1.130), UV-vis spectrophotometry (210 nm) and thin layer chromatography (Rf = 0.90). The molecular weight of purified biosurfactant was found to be ~ 1.0 kDa, based on Electron Spray Ionization-MS. A concentration of 1980 × 10-2 parts per million of CO2 was trapped in a KOH solution after 15 days of incubation in Luria-Bertani broth containing 1% engine oil. Our results suggest that bacterium Bacillus velezensis KLP2016 may promise a new dimension to solving the engine oil pollution problem in near future.


Subject(s)
Bacillus/metabolism , Lipopeptides/isolation & purification , Petroleum Pollution , Surface-Active Agents/isolation & purification , Bacillus/growth & development , Biodegradation, Environmental , Carbon Dioxide/chemistry , Chromatography, Gel , Emulsions , Gas Chromatography-Mass Spectrometry , Hydrocarbons/analysis , Micelles , Reference Standards , Surface Tension
18.
J Neurochem ; 157(6): 1850-1860, 2021 06.
Article in English | MEDLINE | ID: mdl-33078390

ABSTRACT

Pituitary adenylate cyclase-activating peptide (PACAP) receptor (PAC1R) is a class B Gprotein-coupled receptor (GPCR) that is widely expressed in the human body and is involved in neuronal differentiation. As class B GPCRs are known to form heterocomplexes with family members, we hypothesized that PAC1R mediates neuronal differentiation through interaction with a class B GPCR. We used the BRET assay to identify potential interactions between PAC1R and 11 class B GPCRs. Gastric inhibitory polypeptide receptor (GIPR) and secretin receptor were identified as putative binding partners of PAC1R. The effect of heterocomplex formation by PAC1R on receptor activation was evaluated with the cyclic (c)AMP, luciferase reporter, and calcium signaling assays; and the effects on receptor internalization and subcellular localization were examined by confocal microscopy. The results suggested he PAC1R/GIPR heterocomplex suppressed signaling events downstream of PAC1R, including cAMP production, serum response element and calcium signaling, and ß-arrestin recruitment. Protein-protein interaction was analyzed in silico, and induction of neuronal differentiation by the PAC1R heterocomplex was assessed in SH-SY5Y neuronal cells by measure the morphological changes and marker genes expression by real-time quantitative PCR and western blot. Over-expression of GIPR suppressed PACAP/PAC1R-mediated neuronal differentiation and the differentiation markers expression in SH-SY5Y cells. GIPR regulates neuronal differentiation through heterocomplex formation with PAC1R.


Subject(s)
Cell Differentiation/physiology , Neurons/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/chemistry , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , HEK293 Cells , Humans , Protein Binding/physiology , Protein Structure, Secondary , Receptors, Gastrointestinal Hormone/genetics , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics
19.
Matrix Biol ; 93: 25-42, 2020 11.
Article in English | MEDLINE | ID: mdl-32534153

ABSTRACT

Heparanase, the sole heparan sulfate degrading endoglycosidase, regulates multiple biological activities that enhance tumor growth, angiogenesis and metastasis. Much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth and metastasis. We have utilized mice over-expressing (Hpa-tg) heparanase to reveal the role of host heparanase in tumor initiation, growth and metastasis. While in wild type mice tumor development in response to DMBA carcinogenesis was restricted to the mammary gland, Hpa-tg mice developed tumors also in their lungs and liver, associating with reduced survival of the tumor-bearing mice. Consistently, xenograft tumors (lymphoma, melanoma, lung carcinoma, pancreatic carcinoma) transplanted in Hpa-tg mice exhibited accelerated tumor growth and shorter survival of the tumor-bearing mice compared with wild type mice. Hpa-tg mice were also more prone to the development of metastases following intravenous or subcutaneous injection of tumor cells. In some models, the growth advantage was associated with infiltration of heparanase-high host cells into the tumors. However, in other models, heparanase-high host cells were not detected in the primary tumor, implying that the growth advantage in Hpa-tg mice is due to systemic factors. Indeed, we found that plasma from Hpa-tg mice enhanced tumor cell migration and invasion attributed to increased levels of pro-tumorigenic factors (i.e., RANKL, SPARC, MIP-2) in the plasma of Hpa-Tg vs. wild type mice. Furthermore, tumor aggressiveness and short survival time were demonstrated in wild type mice transplanted with bone marrow derived from Hpa-tg but not wild type mice. These results were attributed, among other factors, to upregulation of pro-tumorigenic (i.e., IL35+) and downregulation of anti-tumorigenic (i.e., IFN-γ+) T-cell subpopulations in the spleen, lymph nodes and blood of Hpa-tg vs. wild type mice and their increased infiltration into the primary tumor. Collectively, our results emphasize the significance of host heparanase in mediating the pro-tumorigenic and pro-metastatic interactions between the tumor cells and the host tumor microenvironment, immune cells and systemic factors.


Subject(s)
Glucuronidase/genetics , Glucuronidase/metabolism , Neoplasm Metastasis/pathology , Neoplasms/pathology , Up-Regulation , Animals , Anthracenes/adverse effects , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Transgenic , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , Neoplasm Transplantation , Neoplasms/chemically induced , Neoplasms/genetics , Neoplasms/metabolism , Piperidines/adverse effects , Tumor Microenvironment
20.
Molecules ; 25(5)2020 Feb 25.
Article in English | MEDLINE | ID: mdl-32106575

ABSTRACT

Anaphylactoid shock is a fatal hypersensitivity response caused by non-IgE mediated mast cell activation. These reactions are mediated by a family of G protein-coupled receptors (GPCRs) known as Mas related GPCRX2 (MRGPRX2). Several US FDA approved drugs which are used in day to day life have been reported to cause anaphylactoid shock. Surprisingly, no therapeutic drugs are available which can directly target MRGPRX2 for treatment of anaphylactoid shock. Genistein is a non-steroidal polyphenol known for its diverse physiological and pharmacological activities. In recent studies, Genistein has been reported for its anti-inflammatory activity on mast cells. However, the effects and mechanistic pathways of Genistein on anaphylactoid reaction remain unknown. In the present study, we designed a battery of in-vitro, in-silico and in-vivo experiments to evaluate the anti-anaphylactoid activity of Genistein in order to understand the possible molecular mechanisms of its action. The in-vitro results demonstrated the inhibitory activity of Genistein on MRGPRX2 activation. Further, a mouse model of anaphylactoid shock was used to evaluate the inhibitory activity of Genistein on blood vessel leakage and hind paw edema. Taken together, our findings have demonstrated a therapeutic potential of Genistein as a lead compound in the treatment of anaphylactoid shock via MRGPRX2.


Subject(s)
Anaphylaxis/drug therapy , Genistein/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Protective Agents/pharmacology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Neuropeptide/antagonists & inhibitors , Anaphylaxis/chemically induced , Anaphylaxis/genetics , Anaphylaxis/pathology , Animals , Cell Degranulation/drug effects , Disease Models, Animal , Drug Hypersensitivity/drug therapy , Drug Hypersensitivity/genetics , Genistein/chemistry , Humans , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/pathology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Protective Agents/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, Neuropeptide/genetics , p-Methoxy-N-methylphenethylamine/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL