Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.388
Filter
1.
Nat Commun ; 13(1): 160, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013289

ABSTRACT

Immune response and new tissue formation are important aspects of tissue repair. However, only a single aspect is generally considered in previous biomedical interventions, and the synergistic effect is unclear. Here, a dual-effect coating with immobilized immunomodulatory metal ions (e.g., Zn2+) and osteoinductive growth factors (e.g., BMP-2 peptide) is designed via mussel adhesion-mediated ion coordination and molecular clicking strategy. Compared to the bare TiO2 group, Zn2+ can increase M2 macrophage recruitment by up to 92.5% in vivo and upregulate the expression of M2 cytokine IL-10 by 84.5%; while the dual-effect of Zn2+ and BMP-2 peptide can increase M2 macrophages recruitment by up to 124.7% in vivo and upregulate the expression of M2 cytokine IL-10 by 171%. These benefits eventually significantly enhance bone-implant mechanical fixation (203.3 N) and new bone ingrowth (82.1%) compared to the bare TiO2 (98.6 N and 45.1%, respectively). Taken together, the dual-effect coating can be utilized to synergistically modulate the osteoimmune microenvironment at the bone-implant interface, enhancing bone regeneration for successful implantation.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone-Implant Interface/growth & development , Macrophages/drug effects , Titanium/pharmacology , Zinc/pharmacology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Biomarkers/metabolism , Bivalvia/chemistry , Cell Differentiation/drug effects , Femur/cytology , Femur/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/cytology , Macrophages/immunology , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/immunology , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/immunology , Osteogenesis/drug effects , Prostheses and Implants , Protein Precursors/pharmacology , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley
2.
Front Immunol ; 12: 616215, 2021.
Article in English | MEDLINE | ID: mdl-34447366

ABSTRACT

Tolerogenic vaccinations using beta-cell antigens are attractive for type 1 diabetes prevention, but clinical trials have been disappointing. This is probably due to the late timing of intervention, when multiple auto-antibodies are already present. We therefore devised a strategy to introduce the initiating antigen preproinsulin (PPI) during neonatal life, when autoimmunity is still silent and central tolerance mechanisms, which remain therapeutically unexploited, are more active. This strategy employs an oral administration of PPI-Fc, i.e. PPI fused with an IgG Fc to bind the intestinal neonatal Fc receptor (FcRn) that physiologically delivers maternal antibodies to the offspring during breastfeeding. Neonatal oral PPI-Fc vaccination did not prevent diabetes development in PPI T-cell receptor-transgenic G9C8.NOD mice. However, PPI-Fc was efficiently transferred through the intestinal epithelium in an Fc- and FcRn-dependent manner, was taken up by antigen presenting cells, and reached the spleen and thymus. Although not statistically significant, neonatal oral PPI-Fc vaccination delayed diabetes onset in polyclonal Ins2-/-.NOD mice that spontaneously develop accelerated diabetes. Thus, this strategy shows promise in terms of systemic and thymic antigen delivery via the intestinal FcRn pathway, but the current PPI-Fc formulation/regimen requires further improvements to achieve diabetes prevention.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/prevention & control , Histocompatibility Antigens Class I/immunology , Insulin/pharmacology , Protein Precursors/pharmacology , Receptors, Fc/immunology , Recombinant Fusion Proteins/pharmacology , Thymus Gland/immunology , Administration, Oral , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class I/genetics , Insulin/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Protein Precursors/genetics , Receptors, Fc/genetics , Recombinant Fusion Proteins/genetics
3.
Front Endocrinol (Lausanne) ; 12: 622647, 2021.
Article in English | MEDLINE | ID: mdl-33841327

ABSTRACT

Proinsulin is an abundant protein that is selectively expressed by pancreatic beta cells and has been a focus for development of antigen-specific immunotherapies for type 1 diabetes (T1D). In this study, we sought to comprehensively evaluate reactivity to preproinsulin by CD4 T cells originally isolated from pancreatic islets of organ donors having T1D. We analyzed 187 T cell receptor (TCR) clonotypes expressed by CD4 T cells obtained from six T1D donors and determined their response to 99 truncated preproinsulin peptide pools, in the presence of autologous B cells. We identified 14 TCR clonotypes from four out of the six donors that responded to preproinsulin peptides. Epitopes were found across all of proinsulin (insulin B-chain, C-peptide, and A-chain) including four hot spot regions containing peptides commonly targeted by TCR clonotypes derived from multiple T1D donors. Of importance, these hot spots overlap with peptide regions to which CD4 T cell responses have previously been detected in the peripheral blood of T1D patients. The 14 TCR clonotypes recognized proinsulin peptides presented by various HLA class II molecules, but there was a trend for dominant restriction with HLA-DQ, especially T1D risk alleles DQ8, DQ2, and DQ8-trans. The characteristics of the tri-molecular complex including proinsulin peptide, HLA-DQ molecule, and TCR derived from CD4 T cells in islets, provides an essential basis for developing antigen-specific biomarkers as well as immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Diabetes Mellitus, Type 1/metabolism , Insulin/pharmacology , Islets of Langerhans/drug effects , Protein Precursors/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Epitopes/metabolism , Humans , Islets of Langerhans/metabolism , Tissue Donors
4.
Peptides ; 140: 170532, 2021 06.
Article in English | MEDLINE | ID: mdl-33744371

ABSTRACT

OBJECTIVES: To analyse the peptidomics of mouse enteroendocrine cells (EECs) and human gastrointestinal (GI) tissue and identify novel gut derived peptides. METHODS: High resolution nano-flow liquid chromatography mass spectrometry (LC-MS/MS) was performed on (i) flow-cytometry purified NeuroD1 positive cells from mouse and homogenised human intestinal biopsies, (ii) supernatants from primary murine intestinal cultures, (iii) intestinal homogenates from mice fed high fat diet. Candidate bioactive peptides were selected on the basis of species conservation, high expression/biosynthesis in EECs and evidence of regulated secretionin vitro. Candidate novel gut-derived peptides were chronically administered to mice to assess effects on food intake and glucose tolerance. RESULTS: A large number of peptide fragments were identified from human and mouse, including known full-length gut hormones and enzymatic degradation products. EEC-specific peptides were largely from vesicular proteins, particularly prohormones, granins and processing enzymes, of which several exhibited regulated secretion in vitro. No regulated peptides were identified from previously unknown genes. High fat feeding particularly affected the distal colon, resulting in reduced peptide levels from GCG, PYY and INSL5. Of the two candidate novel peptides tested in vivo, a peptide from Chromogranin A (ChgA 435-462a) had no measurable effect, but a progastrin-derived peptide (Gast p59-79), modestly improved glucose tolerance in lean mice. CONCLUSION: LC-MS/MS peptidomic analysis of murine EECs and human GI tissue identified the spectrum of peptides produced by EECs, including a potential novel gut hormone, Gast p59-79, with minor effects on glucose tolerance.


Subject(s)
Enteroendocrine Cells/metabolism , Gastrins/pharmacology , Gastrointestinal Tract/metabolism , Glucose Tolerance Test/methods , Peptides/metabolism , Protein Precursors/pharmacology , Proteome/metabolism , Thinness/drug therapy , Animals , Cells, Cultured , Glucose/metabolism , Humans , Male , Mice , Models, Animal , Peptides/chemistry , Proteome/analysis , Thinness/metabolism
5.
Int J Mol Sci ; 23(1)2021 Dec 25.
Article in English | MEDLINE | ID: mdl-35008651

ABSTRACT

The prevalence of atopic dermatitis (AD), a disease characterized by severe pruritus, immune imbalance, and skin barrier dysfunction, is rapidly increasing worldwide. Deacetylasperulosidic acid (DAA) has anti-atopic activity in the three main cell types associated with AD: keratinocytes, mast cells, and eosinophils. Our study investigated the anti-atopic activity of DAA in 2,4-dinitrochlorobenzene-induced NC/Nga mice. DAA alleviated the symptoms of AD, including infiltration of inflammatory cells (mast cells and eosinophils), epidermal thickness, ear thickness, and scratching behavior. Furthermore, DAA reduced serum IgE, histamine, and IgG1/IgG2a ratio and modulated the levels of AD-related cytokines and chemokines, namely interleukin (IL)-1ß, IL-4, IL-6, IL-9, IL-10, IL-12, tumor necrosis factor-α, interferon-γ, thymic stromal lymphopoietin, thymus and activation-regulated chemokine, macrophage-derived chemokine, and regulated on activation the normal T cell expressed and secreted in the serum. DAA restored immune balance by regulating gene expression and secretion of Th1-, Th2-, Th9-, Th17-, and Th22-mediated inflammatory factors in the dorsal skin and splenocytes and restored skin barrier function by increasing the expression of the pro-filaggrin gene and barrier-related proteins filaggrin, involucrin, and loricrin. These results suggest DAA as a potential therapeutic agent that can alleviate the symptoms of AD by reducing pruritus, modulating immune imbalance, and restoring skin barrier function.


Subject(s)
Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/drug therapy , Dinitrochlorobenzene/adverse effects , Immunity/drug effects , Plant Extracts/pharmacology , Pruritus/drug therapy , Skin/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Chemokines/metabolism , Dermatitis, Atopic/metabolism , Filaggrin Proteins/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Male , Mast Cells/drug effects , Membrane Proteins/pharmacology , Mice , Protein Precursors/pharmacology , Pruritus/metabolism , Skin/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
6.
Food Funct ; 11(11): 10231-10241, 2020 Nov 18.
Article in English | MEDLINE | ID: mdl-33169751

ABSTRACT

The emergence of the plasmid-mediated colistin resistance mechanism (mcr-1) makes bacterial resistance to colistin increasingly serious. This mcr-1 mediated bacterial resistance to colicin is conferred primarily through modification of lipid A in lipopolysaccharides (LPS). In our previous research, antimicrobial peptide F1 was derived from Tibetan kefir and has been shown to effectively inhibit the growth of Gram-negative bacteria (E. coli), Gram-positive bacteria (Staphylococcus aureus), and other pathogenic bacteria. Based on this characteristic of antibacterial peptide F1, we speculated that it could inhibit the growth of the colicin-resistant E. coli SHP45 (mcr-1) and not easily produce drug resistance. Studies have shown that antimicrobial peptide F1 can destroy the liposome structure of the phospholipid bilayer by destroying the inner and outer membranes of bacteria, thereby significantly inhibiting the growth of E. coli SHP45 (mcr-1), but without depending on LPS. The results of this study confirmed our hypothesis, and we anticipate that antimicrobial peptide F1 will become a safe antibacterial agent that can assist in solving the problem of drug resistance caused by colistin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Enkephalin, Methionine/analogs & derivatives , Escherichia coli/drug effects , Protein Precursors/pharmacology , Colistin/pharmacology , Enkephalin, Methionine/pharmacology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Microbial Sensitivity Tests
7.
Science ; 369(6507)2020 08 28.
Article in English | MEDLINE | ID: mdl-32855309

ABSTRACT

Neuronal synapses undergo structural and functional changes throughout life, which are essential for nervous system physiology. However, these changes may also perturb the excitatory-inhibitory neurotransmission balance and trigger neuropsychiatric and neurological disorders. Molecular tools to restore this balance are highly desirable. Here, we designed and characterized CPTX, a synthetic synaptic organizer combining structural elements from cerebellin-1 and neuronal pentraxin-1. CPTX can interact with presynaptic neurexins and postsynaptic AMPA-type ionotropic glutamate receptors and induced the formation of excitatory synapses both in vitro and in vivo. CPTX restored synaptic functions, motor coordination, spatial and contextual memories, and locomotion in mouse models for cerebellar ataxia, Alzheimer's disease, and spinal cord injury, respectively. Thus, CPTX represents a prototype for structure-guided biologics that can efficiently repair or remodel neuronal circuits.


Subject(s)
C-Reactive Protein/pharmacology , Nerve Tissue Proteins/pharmacology , Neural Pathways/drug effects , Protein Precursors/pharmacology , Receptors, AMPA/metabolism , Recombinant Proteins/pharmacology , Synapses/drug effects , Alzheimer Disease/therapy , Animals , C-Reactive Protein/chemistry , C-Reactive Protein/therapeutic use , Cerebellar Ataxia/therapy , Disease Models, Animal , HEK293 Cells , Hippocampus , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/therapeutic use , Protein Domains , Protein Precursors/chemistry , Protein Precursors/therapeutic use , Receptors, Glutamate/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Spine/drug effects , Spine/physiology
8.
Biochem Biophys Res Commun ; 527(3): 744-750, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32439180

ABSTRACT

Proadrenomedullin N-terminal 20 peptide (PAMP) is a regulatory peptide that is found in various cell types. It is involved in many biological activities and is rich in basic and hydrophobic amino acids, a common feature of antimicrobial peptides (AMPs). In this study, the cell selectivity and antimicrobial mechanism of PAMP and its C-terminal peptide, PAMP(9-20), were investigated. PAMP and PAMP(9-20) displayed potent antimicrobial activity (minimum inhibitory concentration: 4-32 µM) against standard bacterial strains, but showed no hemolytic activity even at the highest tested concentration of 256 µM. PAMP(9-20) showed 2- to 4-fold increase in antimicrobial activity against gram-negative bacteria compared to PAMP. Cytoplasmic membrane depolarization, leakage of calcein dye from membrane mimic liposomes, SYTOX Green uptake, membrane permeabilization, and flow cytometry studies indicated that the major target of PAMP and PAMP(9-20) is not the microbial cell membrane. Interestingly, laser-scanning confocal microscopy demonstrated that FITC-labeled PAMP and PAMP(9-20) enter the cytoplasm of Escherichia coli similar to buforin-2, and gel retardation assay indicated that PAMP and PAMP(9-20) effectively bind to bacterial DNA. These results suggest that the intracellular target mechanism is responsible for the antimicrobial action of PAMP and PAMP(9-20). Collectively, PAMP and PAMP(9-20) could be considered promising candidates for the development of new antimicrobial agents.


Subject(s)
Adrenomedullin/pharmacology , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Peptide Fragments/pharmacology , Protein Precursors/pharmacology , Adrenomedullin/chemistry , Animals , Anti-Bacterial Agents/chemistry , Bacteria/metabolism , Bacterial Infections/drug therapy , Bacterial Infections/microbiology , Bacterial Outer Membrane/drug effects , Bacterial Outer Membrane/metabolism , DNA, Bacterial/metabolism , Hemolysis/drug effects , Microbial Sensitivity Tests , Peptide Fragments/chemistry , Protein Precursors/chemistry , Sheep
9.
Mol Biol Rep ; 47(5): 3949-3961, 2020 May.
Article in English | MEDLINE | ID: mdl-32385773

ABSTRACT

Soluble TNFα, a member of TNF superfamily attributes dual roles in apoptosis and cell proliferation whereas its precursor transmembrane TNFα (tmTNFα) has potential for tumor reduction without initiating proliferation. In this perspective, we recombinantly expressed functional tmTNFα and explored its potential in cell growth inhibition. While structural characterizations of purified tmTNFα revealed integrity of the protein, cell viability assays demonstrated significant antiproliferative effect on HepG2 (IC50: 36 nM) and HeLa (IC50: 23 nM) cells. Mechanistic insights into mode of cell death unveiled G1 arrest in HepG2 and G2/M arrest in HeLa cells accompanied with disruption of mitochondrial membrane potential and activation of executioner caspases. Subsequent, flow cytometry based assays resulted confirmatory evidence of apoptosis after treatment with the recombinant protein. Additionally, effect of the recombinant protein on 3D tumor spheroids was explored, which rendered reduction in tumor size due to cell death as evident from confocal microscopy studies. Effectiveness of the tmTNFα in 2D monolayer as well as in complex 3D spheroids demonstrate the therapeutic significance of the protein, featuring recombinant tmTNFα as an attractive option for cancer therapeutics in days to come.


Subject(s)
Protein Precursors/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , HeLa Cells , Hep G2 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factors/metabolism
10.
J Pharmacol Sci ; 143(2): 127-131, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32156464

ABSTRACT

The inhibition of retinal ischemia-induced damage by post-ischemic prothymosin alpha (ProTα) was not affected in toll-like receptor 4 knockout (TLR4-/-) mice but blocked by the pretreatment with antibody against F0/F1 ATPase α- or ß-subunit, novel candidate for ProTα-receptor. In addition to the previous observation of ProTα-induced ATP release from cells, the present study showed a ProTα-induced enhancement of ATP hydrolysis activity of recombinant ATP5A1/5B complex. As the protection of retinal function by post-ischemic ProTα was abolished by anti-P2Y12 antibody, the activation of F0/F1 ATPase and subsequent P2Y12 receptor system may play roles in beneficial actions by post-ischemic ProTα.


Subject(s)
Ischemia/metabolism , Ischemia/prevention & control , Protein Precursors/administration & dosage , Protein Precursors/pharmacology , Proton-Translocating ATPases/metabolism , Receptors, Purinergic P2Y12/metabolism , Retina , Thymosin/analogs & derivatives , Animals , Hydrolysis/drug effects , Male , Mice, Inbred C57BL , Mitochondrial Proton-Translocating ATPases/metabolism , Recombinant Proteins/metabolism , Thymosin/administration & dosage , Thymosin/pharmacology
11.
Peptides ; 126: 170265, 2020 04.
Article in English | MEDLINE | ID: mdl-31982448

ABSTRACT

Prothymosin alpha (ProTα)-mimetic hexapeptide (amino acid: NEVDQE, P6Q) inhibits cerebral or retinal ischemia-induced behavioral, electrophysiological and histological damage. P6Q also abolishes cerebral hemorrhage induced by ischemia with tissue plasminogen activator (tPA). In the present study we examined the beneficial effects of P6Q on other post-stroke prognostic psychology-related symptoms, which obstruct the motivation toward physical therapy. Intravenous (i.v.) administration with tPA (10 mg/kg) at 6 h after photochemically induced thrombosis (PIT) in mice resulted in bilateral central post-stroke pain in thermal and mechanical nociception tests and loss of social activity in the nest building test, both of which were significantly blocked by P6Q (30 mg/kg, i.v.) given at 5 h after PIT. P6Q (30 mg/kg, i.v.) also improved the memory-learning deficit in the step-through test and depression-like behavior in the tail suspension test when it was given 1 day after bilateral common carotid arteries occlusion (BCCAO) in mice. Thus, these studies suggest that P6Q could be a promising candidate to prevent negative prognostic psychological symptoms following focal and global ischemia.


Subject(s)
Brain Ischemia/drug therapy , Depression/drug therapy , Memory Disorders/drug therapy , Neuroprotective Agents/pharmacology , Pain/drug therapy , Protein Precursors/pharmacology , Stroke/drug therapy , Thymosin/analogs & derivatives , Animals , Brain Ischemia/chemically induced , Brain Ischemia/pathology , Brain Ischemia/psychology , Depression/etiology , Depression/pathology , Learning , Male , Memory Disorders/etiology , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Pain/etiology , Pain/pathology , Peptide Fragments/pharmacology , Stroke/chemically induced , Stroke/pathology , Stroke/psychology , Thymosin/pharmacology , Tissue Plasminogen Activator/toxicity
13.
Microb Pathog ; 137: 103763, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31574301

ABSTRACT

Procalcitonin and Mid-regional pro Adrenomedullin have been proposed for sepsis diagnosis, antibiotic therapy guidance and prognosis. A retrospective analysis of PCT and MR-proADM on 571 consecutive patients with sepsis diagnosis was performed. Median values were compared using the non-parametric Mann-Whitney's test. Receiver operating characteristic analysis was performed to define cutoff points for sepsis diagnosis. Pretest odds, posttest odds, and posttest probability have been calculated. Data were analyzed using Med-Calc 11.6.1.0 software. PCT resulted excellent in gram-negative, but less performant in gram-positive and fungal etiologies. MR-proADM values resulted homogenously distributed within the different microbial classes and increased significantly in septic shock. PCT highest PPV value was found to distinguish gram-negative from fungal sepsis and septic shock (>3. 57 ng/mL, PPV 0.96 and > 8.77 ng/mL, PPV 0.96, respectively). Good diagnostic accuracy was evidenced to discriminate gram-negative from gram-positive septic shock (>3.88 ng/mL PPV 0.89). Lower diagnostic accuracy was evidenced to discriminate gram-negative and gram-positive sepsis (>0.80 ng/mL, PPV 0.78) and gram-positive from fungal septic shock (>1.74 ng/mL PPV 0.75). The lowest PCT PPV (0.28) was found in gram-positive and fungal sepsis distinction. MR-proADM discriminating cut-offs were homogeneously distributed in Gram-negative and Gram-positive sepsis and were higher in septic shock, but not influenced by pathogen etiologies. MR-proADM cut-off values > 3.39 nmol/L in sepsis and >4.33 nmol/L in septic shock were associated with significant higher risk of 90-days mortality. In conclusion, PCT and MR-proADM combination represents an advantage for sepsis diagnosis and for 90-days mortality risk stratification.


Subject(s)
Adrenomedullin/pharmacology , Procalcitonin/pharmacology , Protein Precursors/pharmacology , Sepsis/diagnosis , Sepsis/drug therapy , Shock, Septic/diagnosis , Shock, Septic/drug therapy , Adrenomedullin/therapeutic use , Adult , Aged , Anti-Bacterial Agents/pharmacology , Bacteria/classification , Bacteria/pathogenicity , Drug Combinations , Female , Fungi/classification , Fungi/pathogenicity , Humans , Italy , Male , Middle Aged , Procalcitonin/therapeutic use , Prognosis , Protein Precursors/therapeutic use , ROC Curve , Retrospective Studies , Sepsis/microbiology , Sepsis/mortality , Shock, Septic/microbiology , Shock, Septic/mortality
14.
Bioresour Technol ; 291: 121882, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31377512

ABSTRACT

The aim of this study is to assess the effectiveness of protein-like precursors addition on promoting humification process during lignocellulose-like biomass composting through adding amino acids to compost. The humification indexes of R1 and R2 was significantly higher than that of CK (P < 0.05). The decreasing ratio of Maillard precursor concentration of R2 and R1 was higher than CK. Amino acids addition affected the bacteria community and environmental factors during composting. Variance partitioning analysis showed that humification process was strengthened with environmental factors, bacteria community, Maillard precursors. Structural equation model (SEM) analysis showed that amino acids had substantial impact on promoting humic acid (HA) formation. The combined application of protein-like wastes and lignocellulose-like wastes was suggested to improve carbon sequestration. This study lays a foundation for economically and effectively managing different types of straws by composting.


Subject(s)
Amino Acids/metabolism , Biomass , Composting , Lignin/metabolism , Protein Precursors/pharmacology , Humic Substances/analysis
15.
Neurochem Int ; 129: 104498, 2019 10.
Article in English | MEDLINE | ID: mdl-31278975

ABSTRACT

The present work aimed to explore the innovative hypothesis that different transcript/protein variants of a pro-neurotrophin may generate different biological outcomes in a cellular system. Nerve growth factor (NGF) is important in the development and progression of neurodegenerative and cancer conditions. Mature NGF (mNGF) originates from a precursor, proNGF, produced in mouse in two major variants, proNGF-A and proNGF-B. Different receptors bind mNGF and proNGF, generating neurotrophic or neurotoxic outcomes. It is known that dysregulation in the proNGF/mNGF ratio and in NGF-receptors expression affects brain homeostasis. To date, however, the specific roles of the two major proNGF variants remain unexplored. Here we attempted a first characterization of the possible differential effects of proNGF-A and proNGF-B on viability, differentiation and endogenous ngf gene expression in the PC12 cell line. We also investigated the differential involvement of NGF receptors in the actions of proNGF. We found that native mouse mNGF, proNGF-A and proNGF-B elicited different effects on PC12 cell survival and differentiation. Only mNGF and proNGF-A promoted neurotrophic responses when all NGF receptors are exposed at the cell surface. Tropomyosine receptor kinase A (TrkA) blockade inhibited cell differentiation, regardless of which NGF was added to culture media. Only proNGF-A exerted a pro-survival effect when TrkA was inhibited. Conversely, proNGF-B exerted differentiative effects when the p75 neurotrophin receptor (p75NTR) was antagonized. Stimulation with NGF variants differentially regulated the autocrine production of distinct proNgf mRNA. Overall, our findings suggest that mNGF and proNGF-A may elicit similar neurotrophic effects, not necessarily linked to activation of the same NGF-receptor, while the action of proNGF-B may be determined by the NGF-receptors balance. Thus, the proposed involvement of proNGF/NGF on the development and progression of neurodegenerative and tumor conditions may depend on the NGF-receptors balance, on specific NGF trancript expression and on the proNGF protein variant ratio.


Subject(s)
Nerve Growth Factor/pharmacology , PC12 Cells/drug effects , Protein Precursors/pharmacology , Animals , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Male , Mice , Nerve Growth Factors/biosynthesis , Nerve Growth Factors/genetics , Nerve Tissue Proteins , Protein Isoforms/pharmacology , Protein Precursors/biosynthesis , Protein Precursors/genetics , Proteome , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Receptor, trkA/antagonists & inhibitors , Receptors, Growth Factor , Receptors, Nerve Growth Factor/antagonists & inhibitors , Species Specificity
16.
Mater Sci Eng C Mater Biol Appl ; 103: 109762, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31349478

ABSTRACT

Early detection is the most effective mean of improving prognosis for many fatal diseases such as cancer. In this context, the Surface Enhanced Resonance Raman Scattering (SERRS) technique is being proposed as alternative to fluorescent methods in detection of biomarkers, because SERRS nanostructures are bright as fluorescent tags but more stable and clearly detectable using the narrow Raman "fingerprints" of a suitable reporter. Here we show that biocompatible SERRS active gold nanostructures, functionalized with an engineered PreS1 peptide (AuNP@PEG-PreS1), detect the presence of the SerpinB3 antigen overexpressed on liver tumor cells, a biomarker of the onset of liver cell carcinomatous transformation. A proper engineering of the targeting unit, linked to the nanostructure by a polymer chain, affords a sensitivity and specificity larger than 80%, at subnanomolar concentrations. Taking into account the high sensitivity of SERRS and that SB3 overexpression is an early event in liver cell carcinomatous transformation, AuNP@PEG-PreS1 nanostructures could be used in routine diagnostic activities, to improve the accuracy of HCC detection in particular in patients with chronic liver diseases.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Drug Delivery Systems , Gold , Hepatitis B Surface Antigens , Liver Neoplasms/drug therapy , Metal Nanoparticles , Peptides , Protein Precursors , Animals , Antigens, Neoplasm/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Gold/chemistry , Gold/pharmacology , Hep G2 Cells , Hepatitis B Surface Antigens/chemistry , Hepatitis B Surface Antigens/pharmacology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Transgenic , Neoplasm Proteins/metabolism , Peptides/chemistry , Peptides/pharmacology , Protein Precursors/chemistry , Protein Precursors/pharmacology , Serpins/metabolism , Spectrum Analysis, Raman
17.
Stem Cells ; 37(9): 1223-1237, 2019 09.
Article in English | MEDLINE | ID: mdl-31132299

ABSTRACT

The role of proNGF, the precursor of nerve growth factor (NGF), in the biology of adult neural stem cells (aNSCs) is still unclear. Here, we analyzed adult hippocampal neurogenesis in AD11 transgenic mice, in which the constitutive expression of anti-NGF antibody leads to an imbalance of proNGF over mature NGF. We found increased proliferation of progenitors but a reduced neurogenesis in the AD11 dentate gyrus (DG)-hippocampus (HP). Also in vitro, AD11 hippocampal neural stem cells (NSCs) proliferated more, but were unable to differentiate into morphologically mature neurons. By treating wild-type hippocampal progenitors with the uncleavable form of proNGF (proNGF-KR), we demonstrated that proNGF acts as mitogen on aNSCs at low concentration. The mitogenic effect of proNGF was specifically addressed to the radial glia-like (RGL) stem cells through the induction of cyclin D1 expression. These cells express high levels of p75NTR , as demonstrated by immunofluorescence analyses performed ex vivo on RGL cells isolated from freshly dissociated HP-DG or selected in vitro from NSCs by leukemia inhibitory factor. Clonogenic assay performed in the absence of mitogens showed that RGLs respond to proNGF-KR by reactivating their proliferation and thus leading to neurospheres formation. The mitogenic effect of proNGF was further exploited in the expansion of mouse-induced neural stem cells (iNSCs). Chronic exposure of iNSCs to proNGF-KR increased their proliferation. Altogether, we demonstrated that proNGF acts as mitogen on hippocampal and iNSCs. Stem Cells 2019;37:1223-1237.


Subject(s)
Dentate Gyrus/cytology , Hippocampus/cytology , Mitogens/pharmacology , Nerve Growth Factor/pharmacology , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Protein Precursors/pharmacology , Animals , Antibodies/genetics , Antibodies/immunology , Cell Proliferation/drug effects , Cells, Cultured , Leukemia Inhibitory Factor/pharmacology , Mice, Transgenic , Nerve Growth Factor/immunology , Nerve Growth Factor/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Protein Precursors/immunology , Protein Precursors/metabolism
18.
Skelet Muscle ; 9(1): 16, 2019 05 27.
Article in English | MEDLINE | ID: mdl-31133057

ABSTRACT

BACKGROUND: Growth differentiation factor 11 (GDF11) is a member of the transforming growth factor ß superfamily. The GDF11 propeptide, which is derived from the GDF11 precursor protein, blocks the activity of GDF11 and its homolog, myostatin, which are both potent inhibitors of muscle growth. Thus, treatment with GDF11 propeptide may be a potential therapeutic strategy for diseases associated with muscle atrophy like sarcopenia and the muscular dystrophies. Here, we evaluate the impact of GDF11 propeptide-Fc (GDF11PRO-Fc) gene delivery on skeletal muscle in normal and dystrophic adult mice. METHODS: A pull-down assay was used to obtain physical confirmation of a protein-protein interaction between GDF11PRO-Fc and GDF11 or myostatin. Next, differentiated C2C12 myotubes were treated with AAV6-GDF11PRO-Fc and challenged with GDF11 or myostatin to determine if GDF11PRO-Fc could block GDF11/myostatin-induced myotube atrophy. Localized expression of GDF11PRO-Fc was evaluated via a unilateral intramuscular injection of AAV9-GDF11PRO-Fc into the hindlimb of C57BL/6J mice. In mdx mice, intravenous injection of AAV9-GDF11PRO-Fc was used to achieve systemic expression. The impact of GDF11PRO-Fc on muscle mass, function, and pathological features were assessed. RESULTS: GDF11PRO-Fc was observed to bind both GDF11 and myostatin. In C2C12 myotubes, expression of GDF11PRO-Fc was able to mitigate GDF11/myostatin-induced atrophy. Following intramuscular injection in C57BL/6J mice, increased grip strength and localized muscle hypertrophy were observed in the injected hindlimb after 10 weeks. In mdx mice, systemic expression of GDF11PRO-Fc resulted in skeletal muscle hypertrophy without a significant change in cardiac mass after 12 weeks. In addition, grip strength and rotarod latency time were improved. Intramuscular fibrosis was also reduced in treated mdx mice; however, there was no change seen in central nucleation, membrane permeability to serum IgG or serum creatine kinase levels. CONCLUSIONS: GDF11PRO-Fc induces skeletal muscle hypertrophy and improvements in muscle strength via inhibition of GDF11/myostatin signaling. However, GDF11PRO-Fc does not significantly improve the dystrophic pathology in mdx mice.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Growth Differentiation Factors/antagonists & inhibitors , Muscular Dystrophy, Animal/drug therapy , Myostatin/antagonists & inhibitors , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Genetic Therapy , Genetic Vectors , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Myostatin/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Precursors/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology
19.
J Neurosci ; 39(23): 4489-4510, 2019 06 05.
Article in English | MEDLINE | ID: mdl-30936240

ABSTRACT

By virtue of their extensive axonal arborization and perisomatic synaptic targeting, cortical inhibitory parvalbumin (PV) cells strongly regulate principal cell output and plasticity and modulate experience-dependent refinement of cortical circuits during development. An interesting aspect of PV cell connectivity is its prolonged maturation time course, which is completed only by end of adolescence. The p75 neurotrophin receptor (p75NTR) regulates numerous cellular functions; however, its role on cortical circuit development and plasticity remains elusive, mainly because localizing p75NTR expression with cellular and temporal resolution has been challenging. By using RNAscope and a modified version of the proximity ligation assay, we found that p75NTR expression in PV cells decreases between the second and fourth postnatal week, at a time when PV cell synapse numbers increase dramatically. Conditional knockout of p75NTR in single PV neurons in vitro and in PV cell networks in vivo causes precocious formation of PV cell perisomatic innervation and perineural nets around PV cell somata, therefore suggesting that p75NTR expression modulates the timing of maturation of PV cell connectivity in the adolescent cortex. Remarkably, we found that PV cells still express p75NTR in adult mouse cortex of both sexes and that its activation is sufficient to destabilize PV cell connectivity and to restore cortical plasticity following monocular deprivation in vivo Together, our results show that p75NTR activation dynamically regulates PV cell connectivity, and represent a novel tool to foster brain plasticity in adults.SIGNIFICANCE STATEMENT In the cortex, inhibitory, GABA-releasing neurons control the output and plasticity of excitatory neurons. Within this diverse group, parvalbumin-expressing (PV) cells form the larger inhibitory system. PV cell connectivity develops slowly, reaching maturity only at the end of adolescence; however, the mechanisms controlling the timing of its maturation are not well understood. We discovered that the expression of the neurotrophin receptor p75NTR in PV cells inhibits the maturation of their connectivity in a cell-autonomous fashion, both in vitro and in vivo, and that p75NTR activation in adult PV cells promotes their remodeling and restores cortical plasticity. These results reveal a new p75NTR function in the regulation of the time course of PV cell maturation and in limiting cortical plasticity.


Subject(s)
Aging/physiology , Interneurons/physiology , Neuronal Plasticity/physiology , Receptors, Nerve Growth Factor/physiology , Sexual Maturation/physiology , Visual Cortex/growth & development , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Connectome , Evoked Potentials, Visual , Female , GABAergic Neurons/cytology , Gene Expression Regulation, Developmental , Interneurons/chemistry , Interneurons/ultrastructure , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Parvalbumins/analysis , Protein Precursors/pharmacology , Random Allocation , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Synapses/physiology , Vision, Monocular/physiology , Visual Cortex/cytology , Visual Cortex/metabolism
20.
Pain ; 160(6): 1383-1401, 2019 06.
Article in English | MEDLINE | ID: mdl-30720581

ABSTRACT

Generalized pain and fatigue are both hallmarks of fibromyalgia, a syndrome with an indefinite etiology. The treatment options for fibromyalgia are currently limited, probably because of its intricate pathophysiology. Thus, further basic and clinical research on this condition is currently needed. This study investigated the effects of nociceptin/orphanin FQ (N/OFQ) receptor (NOPr) ligands and the modulation of the NOP system in the preclinical mouse model of reserpine-induced fibromyalgia. The effects of administration of the natural agonist N/OFQ and the selective NOPr antagonists (UFP-101 and SB-612111) were evaluated in fibromyalgia-related symptoms in reserpine-treated mice. The expression of prepronociceptin/orphanin FQ and NOPr was assessed in central and peripheral sites at different time points after reserpine administration. Nociceptin/orphanin FQ displayed dual effects in the behavioral changes in the reserpine-elicited fibromyalgia model. The peptide NOPr antagonist UFP-101 produced analgesic and antifatigue effects, by preventing alterations in brain activity and skeletal muscle metabolism, secondary to fibromyalgia induction. The nonpeptide NOPr antagonist SB-612111 mirrored the favorable effects of UFP-101 in painful and fatigue alterations induced by reserpine. A time-related up- or downregulation of prepronociceptin/orphanin FQ and NOPr was observed in supraspinal, spinal, and peripheral sites of reserpine-treated mice. Our data shed new lights on the mechanisms underlying the fibromyalgia pathogenesis, supporting a role for N/OFQ-NOP receptor system in this syndrome.


Subject(s)
Analgesics/pharmacology , Fatigue/drug therapy , Fibromyalgia/drug therapy , Opioid Peptides/pharmacology , Animals , Disease Models, Animal , Female , Male , Mice , Narcotic Antagonists/pharmacology , Pain/drug therapy , Protein Precursors/pharmacology , Receptors, Opioid/drug effects , Nociceptin Receptor , Nociceptin
SELECTION OF CITATIONS
SEARCH DETAIL
...