Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Publication year range
1.
Lab Invest ; 98(12): 1538-1548, 2018 12.
Article in English | MEDLINE | ID: mdl-30206309

ABSTRACT

Human ß-defensins contain an oncolytic motif that binds to tumor cell membranes and mediate permeabilization, rapid induction of cytolysis, and apoptosis. Previous studies have indicated that a fragment of the mature human ß-defensin-1 (HBD1) peptide (DF) has antitumor properties. While targeted drug treatments using fusion proteins have been shown to increase drug efficacy, this phenomenon has not been studied for this defensin. Thus, in this study, we designed and prepared a fusion protein containing this HBD1 fragment and an epidermal growth factor receptor (EGFR)-targeting oligopeptide (Ec) as well as lidamycin (LDM), an extremely potent cytotoxic antitumor antibiotic, which consists of an apoprotein (LDP) and a highly active enediyne (AE). The fusion protein (Ec-LDP-DF) and its enediyne-integrated fusion protein (Ec-LDP(AE)-DF) were then purified and used to treat lung carcinoma cells in culture as well as lung carcinoma xenograft mouse models. The multifunctional fusion protein Ec-LDP-DF was shown to effectively bind to EGFR-expressing tumor cells. Furthermore, the enediyne-energized Ec-LDP(AE)-DF analog exhibited extremely potent cytotoxicity in NSCLC cell lines and an IC50 less than 10-10 mol/L. Ec-LDP(AE)-DF also significantly inhibited the growth of human carcinoma A549 and H460 xenografts in athymic mice at well-tolerated doses. Treatment resulted in cell cycle arrest and apoptosis in a dose-dependent manner. EGF-stimulated EGFR phosphorylation was also abolished by Ec-LDP(AE)-DF. In summary, our understanding of the role of defensins in cancer development and progression is continually expanding, and Ec-LDP(AE)-DF is a promising cancer cell-targeting agent for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , beta-Defensins/administration & dosage , A549 Cells , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Humans , Mice, Inbred BALB C , Mice, Nude , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Xenograft Model Antitumor Assays
2.
J Drugs Dermatol ; 17(4): 426-441, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29601620

ABSTRACT

BACKGROUND/OBJECTIVES: Anti-aging strategies utilizing stem cells are in the forefront. Alpha and beta defensins are natural immune peptides that have been shown to activate an LGR6-positive stem cell locus in the hair follicle, identified as the source of most new epidermal cells during acute wound healing. We investigated the ability of biomimetic alpha and beta defensin molecules, supplemented with supportive cosmetic ingredients, formulated into three skin care products, at improving the structure and function of aging skin. METHODS: A participant- and investigator -blinded, placebo-controlled, multi-center trial was performed in outpatient settings. Forty-four healthy female subjects, aged 41-71 years, skin types I-V, completed the study with 2/3 receiving full formula and 1/3 receiving the placebo formula. A skin care regimen of 3 products (serum, cream, and mask) containing alpha-defensin 5 and beta-defensin 3, and other cosmetic ingredients, was applied to the face, post-auricular, and neck skin two times per day for 12 weeks in those receiving full formula, whereas the placebo group received the identically packaged regimen without the active ingredients. Methods of evaluation included histopathology and immunohistochemistry (7 subjects), clinical evaluation of pores, superficial and deep wrinkles based on Griffiths scale, and high-resolution photography (all subjects). In addition, a subset of 15 patients were evaluated with the QuantifiCare system (3-dimensional imaging and skin care scores for evenness, pores, oiliness) and Cortex measurements (high-resolution skin ultrasound, TEWL, elasticity, color, and hydration). Data points for evaluation included baseline, 6 weeks, and 12 weeks. All patients used the same sunscreen and cleanser, which was provided to them. RESULTS: The full formula regimen caused a significantly (P equals 0.027) increased thickness of the epidermis as seen in histology, not seen in the placebo group, with no signs of inflammation. No excessive cell proliferation was detected in either group as measured by Ki67-immunohistochemistry. Reduction in visible pores, superficial wrinkles, oiliness, pigmentation, and improvement of skin evenness, were statistically significant. A trend for improvement was also observed in skin elasticity, TEWL, and hydration; these did not achieve statistical significance. Ultrasound and histopathology demonstrated increases in dermal thickness in individual patients, without statistical significance. Comprehensive improvement in all 5 parameters, including visible pores, hyperpigmentation, superficial and deep wrinkles, and epidermal thickness, was statistically significant when the subset of participants assigned for histology in full formula group was compared with the placebo group participants. CONCLUSIONS: A 3-product skin care regimen containing alpha and beta defensins globally improves the visual appearance and structure of aging skin without irritation, dryness, or inflammation. Specifically, this regimen increases epidermal thickness, reduces appearance of pores, reduces wrinkles, and reduces melanin. This skin care regimen stimulates rejuvenation without evidence of increase of a marker of carcinogenic stimulation. This data is consistent with the hypothesis that a defensin-containing skin care regimen activates the body's own dormant stem cells to generate healthy new epidermal cells.

J Drugs Dermatol. 2018;17(4):426-441.

THIS ARTICLE HAD BEEN MADE AVAILABLE FREE OF CHARGE. PLEASE SCROLL DOWN TO ACCESS THE FULL TEXT OF THIS ARTICLE WITHOUT LOGGING IN. NO PURCHASE NECESSARY. PLEASE CONTACT THE PUBLISHER WITH ANY QUESTIONS.


Subject(s)
Dermatologic Agents/administration & dosage , Skin Aging/drug effects , Skin Aging/pathology , Ultrasonography, Interventional/methods , alpha-Defensins/administration & dosage , beta-Defensins/administration & dosage , Adult , Aged , Cosmetics/administration & dosage , Double-Blind Method , Epidermis/diagnostic imaging , Epidermis/drug effects , Epidermis/pathology , Female , Humans , Imaging, Three-Dimensional/methods , Middle Aged , Photography , Skin Care/methods
3.
BMC Microbiol ; 17(1): 43, 2017 Feb 23.
Article in English | MEDLINE | ID: mdl-28231771

ABSTRACT

BACKGROUND: Avian ß-defensins (AvBD) possess broad-spectrum antimicrobial, LPS neutralizing and chemotactic properties. AvBD-12 is a chemoattractant for avian immune cells and mammalian dendritic cells (JAWSII) - a unique feature that is relevant to the applications of AvBDs as chemotherapeutic agents in mammalian hosts. To identify the structural components essential to various biological functions, we have designed and evaluated seven AvBD analogues. RESULTS: In the first group of analogues, the three conserved disulfide bridges were eliminated by replacing cysteines with alanine and serine residues, peptide hydrophobicity and charge were increased by changing negatively charged amino acid residues to hydrophobic (AvBD-12A1) or positively charged residues (AvBD-12A2 and AvBD-12A3). All three analogues in this group showed improved antimicrobial activity, though AvBD-12A3, with a net positive charge of +9, hydrophobicity of 40% and a predicted CCR2 binding domain, was the most potent antimicrobial peptide. AvBD-12A3 also retained more than 50% of wild type chemotactic activity. In the second group of analogues (AvBD-12A4 to AvBD-12A6), one to three disulfide bridges were removed via substitution of cysteines with isosteric amino acids. Their antimicrobial activity was compromised and chemotactic activity abolished. The third type of analogue was a hybrid that had the backbone of AvBD-12 and positively charged amino acid residues AvBD-6. The antimicrobial and chemotactic activities of the hybrid resembled that of AvBD-6 and AvBD-12, respectively. CONCLUSIONS: While the net positive charge and charge distribution have a dominating effect on the antimicrobial potency of AvBDs, the three conserved disulfide bridges are essential to the chemotactic property and the maximum antimicrobial activity. Analogue AvBD-12A3 with a high net positive charge, a moderate degree of hydrophobicity and a CCR2-binding domain can serve as a template for the design of novel antimicrobial peptides with chemotactic property and salt resistance.


Subject(s)
Birds/immunology , Disulfides/chemistry , Hydrophobic and Hydrophilic Interactions , beta-Defensins/chemical synthesis , beta-Defensins/pharmacology , Alanine/chemistry , Amino Acid Sequence , Animals , Anti-Infective Agents/chemical synthesis , Anti-Infective Agents/pharmacology , Bacteria/drug effects , Bacteria/growth & development , Bacterial Load , Cell Line/drug effects , Chemotaxis , Chickens , Colony Count, Microbial/methods , Cysteine/chemistry , Dendritic Cells/immunology , Drug Combinations , Macrophages/drug effects , Microbial Sensitivity Tests/methods , Microscopy, Electron, Scanning , Peptides/chemical synthesis , Protein Conformation , Serine/chemistry , Sodium Chloride/administration & dosage , Sodium Chloride/pharmacology , beta-Defensins/administration & dosage
4.
Transgenic Res ; 24(2): 353-64, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25417183

ABSTRACT

We describe the use of plant-made ß-defensins as effective antimicrobial substances for controlling salmonellosis, a deadly infection caused by Salmonella typhimurium (referred to further as S. typhi). Human ß-defensin-1 (hBD-1) and -2 (hBD-2) were expressed under the control of strong constitutive promoters in tobacco plants, and bio-active ß-defensins were successfully extracted. In the in vitro studies, enriched recombinant plant-derived human ß-defensin-1 (phBD-1) and -2 (phBD-2) obtained from both T1 and T2 transgenic plants showed significant antimicrobial activity against Escherichia coli and S. typhi when used individually and in various combinations. The 2:1 peptide combination of phBD-1:phBD-2 with peptides isolated from T1-and T2-generation plants reduced the growth of S. typhi by 96 and 85 %, respectively. In vivo studies employing the mouse model (Balb/c) of Salmonella infection clearly demonstrated that the administration of plant-derived defensins individually and in different combinations enhanced the mean survival time of Salmonella-infected animals. When treatment consisted of the 2:1 phBD-1:phBD-2 combination, approximately 50 % of the infected mice were still alive at 206 h post-inoculation; the lowest number of viable S. typhi was observed in the liver and spleen of infected animals. We conclude that plant-made recombinant ß-defensins (phBD-1 and phBD-2) are promising antimicrobial substances and have the potential to become additional tools against salmonellosis, particularly when used in combination.


Subject(s)
Plants, Genetically Modified/metabolism , Salmonella Infections/drug therapy , beta-Defensins/biosynthesis , Amino Acid Sequence , Animals , Escherichia coli/genetics , Humans , Mice , Plants, Genetically Modified/genetics , RNA, Messenger/biosynthesis , Salmonella/drug effects , Salmonella/pathogenicity , Salmonella Infections/microbiology , Nicotiana/genetics , Nicotiana/metabolism , beta-Defensins/administration & dosage , beta-Defensins/genetics
5.
J Surg Res ; 199(2): 458-69, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26119274

ABSTRACT

BACKGROUND: The infection of orthopedic implantation devices with Staphylococcus has been a serious concern within the biomaterial community. Treatments are not always successful because of antibiotic-resistant bacteria biofilm infection. Recent studies have shown that combination of antibiotics with low-frequency ultrasound (US) can enhance the bactericidal activity effectively against the formation of biofilms in vitro pilot study. Meanwhile, microbubbles evolved as targeted drug-delivery agents can provide nuclei for inertial cavitation and lower the threshold for US-induced cavitation. Human ß-defensin 3 (HBD-3) is a cationic antimicrobial peptide considered particularly promising for future bactericidal employment and has effect on antibiotic-resistant Staphylococcus biofilms. But the effect has not been reported when combined with US-targeted microbubble destruction (UTMD) in vivo. METHODS: In this study, we evaluated the effect of HBD-3 combined with UTMD on two tested Staphylococcus by the spread plate method, crystal violet staining, confocal laser scanning microscopy, scanning electron microscopy, and real-time polymerase chain reaction. RESULTS: In the study, we found that the biofilm densities, the percentage of live cells, and the viable counts of two tested Staphylococcus that recovered from the biofilm on the titanium surface in mice were significantly decreased in the group of the HBD-3 combined with UTMD, compared with those of other groups. Furthermore, in the experiment, we found out that UTMD could enhance HBD-3 activity, which inhibits the biofilm-associated genes expression of icaAD and the methicillin-resistance genes expression of MecA by promoting the icaR expression simultaneously. CONCLUSIONS: The combination of HBD-3 with UTMD can play a significant role on the elimination of the antibiotic-resistant Staphylococcus biofilms in vivo.


Subject(s)
Biofilms , Microbubbles , Prosthesis-Related Infections/drug therapy , Ultrasonic Therapy , beta-Defensins/administration & dosage , Animals , Bacterial Proteins/metabolism , Drug Evaluation, Preclinical , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Microscopy, Confocal , Microscopy, Electron, Scanning , Penicillin-Binding Proteins , Phenotype , Staphylococcus aureus , Staphylococcus epidermidis , Titanium
6.
Avian Dis ; 59(4): 518-24, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26629626

ABSTRACT

Inclusion body hepatitis (IBH) is one of the major infectious diseases adversely affecting the poultry industry of the United States and Canada. Currently, no effective and safe vaccine is available for the control of IBH virus (IBHV) infection in chickens. However, based on the excellent safety and immunogenic profiles of experimental veterinary vaccines developed with the use of new generation adjuvants, we hypothesized that characterization of vaccine formulations containing inactivated IBHV or its capsid protein hexon as antigens, along with poly[di(sodium carboxylatoethylphenoxy)phosphazene] (PCEP) and avian beta defensin 2 (ABD2) as vaccine adjuvants, will be helpful in development of an effective and safe vaccine formulation for IBH. Our data demonstrated that experimental administration of vaccine formulations containing inactivated IBHV and a mixture of PCEP with or without ABD2 as an adjuvant induced significantly higher antibody responses compared with other vaccine formulations, while hexon protein-based vaccine formulations showed relatively lower levels of antibody responses. Thus, a vaccine formulation containing inactivated IBHV with PCEP or a mixture of PCEP and ABD2 (with a reduced dosage of PCEP) as an adjuvant may serve as a potential vaccine candidate. However, in order to overcome the risks associated with whole virus inactivated vaccines, characterization of additional viral capsid proteins, including fiber protein and penton of IBHV along with hexon protein in combination with more new generation adjuvants, will be helpful in further improvements of vaccines against IBHV infection.


Subject(s)
Adenoviridae Infections/prevention & control , Adenovirus Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Chickens , Fowl adenovirus A/immunology , Hepatitis, Animal/prevention & control , Poultry Diseases/prevention & control , Viral Hepatitis Vaccines/immunology , Adenoviridae Infections/virology , Adenovirus Vaccines/administration & dosage , Animals , Capsid Proteins/administration & dosage , Capsid Proteins/immunology , Hepatitis Viruses/immunology , Hepatitis, Animal/virology , Immunity, Innate , Phenylpropionates/administration & dosage , Phenylpropionates/immunology , Polymers/administration & dosage , Poultry Diseases/virology , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology , Viral Hepatitis Vaccines/administration & dosage , beta-Defensins/administration & dosage , beta-Defensins/immunology
7.
Hell J Nucl Med ; 18(3): 233-7, 2015.
Article in English | MEDLINE | ID: mdl-26574693

ABSTRACT

OBJECTIVE: Human beta-defensin-3 (HBD-3) is an antimicrobial peptide which is up-regulated during inflammation. Based on the previously demonstrated capacity of technetium-99m ((99m)Tc) labelled HBD-3 of distinguishing infection from inflammation in rats, we have decided to collect information on the potential toxicity of the tracer in view of its possible use for imaging in humans. MATERIALS AND METHODS: Recombinant HBD-3 underwent labeling with (99m)Tc. The CD1 mice were selected as standard rodent species. Ten mice, 5 male and 5 female, were subjected to physical examination and housed in a dedicated room in 5 per cage. After 9 days pre-test period, all mice were weighted for dose adjustment and received intravenously 6mcg/mouse of (99m)Tc-HBD-3. Mortality was recorded daily, while body weight was registered once a week. Clinical observation of animals was performed daily for sickness symptoms due to the drug treatment. At day 19 a second dose of 6mcg/mouse (99m)Tc-HBD-3, was administered. Twenty-four hours after the second dose (day 20) the animals were euthanized. A piece of liver, kidneys, heart and lungs was collected for histopathological analysis. RESULTS: Our results showed that the labelled-HBD-3 dose did not induce significant toxicity in mice. Of course these parameters were not sufficient to authorize use in humans. This non-toxic dose of HBD-3 when translated from animals to humans resulted in an equivalent dose of approximately 25 times higher than that needed for imaging. CONCLUSION: Our non toxicity data of using (99m)Tc-beta-defensin-3 in mice offer a further indication in favour of the clinical use of this radiopharmaceutical in all cases where discrimination between infection and inflammation is needed.


Subject(s)
Radiopharmaceuticals/toxicity , Technetium/administration & dosage , Technetium/toxicity , beta-Defensins/administration & dosage , beta-Defensins/toxicity , Animals , Dose-Response Relationship, Drug , Female , Isotope Labeling , Lethal Dose 50 , Mice , Radiopharmaceuticals/administration & dosage , Survival Rate , Technetium/chemistry , beta-Defensins/chemistry
8.
J Immunol ; 188(2): 735-43, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22174455

ABSTRACT

ß-Defensins are antimicrobial peptides of the innate immune system produced in the skin by various stimuli, including proinflammatory cytokines, bacterial infection, and exposure to UV radiation (UVR). In this study we demonstrate that the UVR-inducible antimicrobial peptide murine ß-defensin-14 (mBD-14) switches CD4(+)CD25(-) T cells into a regulatory phenotype by inducing the expression of specific markers like Foxp3 and CTLA-4. This is functionally relevant because mBD-14-treated T cells inhibit sensitization upon adoptive transfer into naive C57BL/6 mice. Accordingly, injection of mBD-14, comparable to UVR, suppresses the induction of contact hypersensitivity and induces Ag-specific regulatory T cells (Tregs). Further evidence for the ability of mBD-14 to induce Foxp3(+) T cells is provided using DEREG (depletion of Tregs) mice in which Foxp3-expressing cells can be depleted by injecting diphtheria toxin. mBD-14 does not suppress sensitization in IL-10 knockout mice, suggesting involvement of IL-10 in mBD-14-mediated immunosuppression. However, unlike UVR, mBD-14 does not appear to mediate its immunosuppressive effects by affecting dendritic cells. Accordingly, UVR-induced immunosuppression is not abrogated in mBD-14 knockout mice. Together, these data suggest that mBD-14, like UVR, has the capacity to induce Tregs but does not appear to play a major role in UVR-induced immunosuppression. Through this capacity, mBD-14 may protect the host from microbial attacks on the one hand, but tame T cell-driven reactions on the other hand, thereby enabling an antimicrobial defense without collateral damage by the adaptive immune system.


Subject(s)
Cell Differentiation/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , beta-Defensins/administration & dosage , beta-Defensins/physiology , Adoptive Transfer , Animals , CTLA-4 Antigen/biosynthesis , Cell Differentiation/genetics , Cell Differentiation/radiation effects , Dinitrofluorobenzene/administration & dosage , Female , Forkhead Transcription Factors/biosynthesis , Immunophenotyping , Injections, Intravenous , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropilins/biosynthesis , T-Lymphocytes, Regulatory/radiation effects , Ultraviolet Rays , beta-Defensins/deficiency
9.
Biochem Pharmacol ; 227: 116451, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39059771

ABSTRACT

The increasing threat of antibiotic resistance among pathogenic microorganisms and the urgent demand for new antibiotics require immediate attention. Antimicrobial peptides exhibit effectiveness against microorganisms, fungi, viruses, and protozoa. The discovery of human ß-defensins represents a major milestone in biomedical research, opening new avenues for scientific investigation into the innate immune system and its resistance mechanisms against pathogenic microorganisms. Multiple defensins present a promising alternative in the context of antibiotic abuse. However, obstacles to the practical application of defensins as anti-infective therapies persist due to the unique properties of human ß-defensins themselves and serious pharmacological and technical challenges. To overcome these challenges, diverse delivery vehicles have been developed and progressively improved for the conjugation or encapsulation of human ß-defensins. This review briefly introduces the biology of human ß-defensins, focusing on their multistage structure and diverse functions. It also discusses several heterologous systems for producing human ß-defensins, various delivery systems created for these peptides, and patent applications related to their utilization, concluding with a summary of current challenges and potential solutions.


Subject(s)
beta-Defensins , Humans , beta-Defensins/chemistry , beta-Defensins/pharmacology , beta-Defensins/administration & dosage , Animals , Drug Delivery Systems/methods , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/administration & dosage
10.
Protein Expr Purif ; 89(1): 33-43, 2013 May.
Article in English | MEDLINE | ID: mdl-23459290

ABSTRACT

Five variants of human ß-defensins (HBDs) were expressed in Escherichia coli using two vector systems (pET28a(+) and pQE30) with inducible expression by IPTG. The last vector has not been previously reported as an expression system for HBDs. The recombinant peptides were different in their lengths and overall charge. The HBDs were expressed as soluble or insoluble proteins depending on the expression system used, and the final protein yields ranged from 0.5 to 1.6 mg of peptide/g of wet weight cells, with purities higher than 90%. The recombinant HBDs demonstrated a direct correlation between antimicrobial activity and the number of basic charged residues; that is, their antimicrobial activity was as follows: HBD3-M-HBD2 > HBD3 = HBD3-M = HB2-KLK > HBD2 when assayed against E. coli, Staphylococcus aureus and Pseudomonas aeruginosa. Interestingly, HBD2 had the best antimicrobial activity against the Mycobacterium tuberculosis strain H37Rv (1.5 µM) and the heterologous tandem peptide, HBD3-M-HBD2, had the best minimal inhibitory concentration (MIC) value (2.7 µM) against a multidrug resistance strain (MDR) of M. tuberculosis, demonstrating the feasibility of the use of HBDs against pathogenic M. tuberculosis reported to be resistant to commercial antibiotics.


Subject(s)
Peptides/genetics , Recombinant Proteins/genetics , beta-Defensins/genetics , Amino Acid Sequence , Escherichia coli/drug effects , Escherichia coli/pathogenicity , Gene Expression Regulation, Bacterial , Humans , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Peptides/administration & dosage , Peptides/chemistry , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , beta-Defensins/administration & dosage , beta-Defensins/chemistry
11.
Microbiol Immunol ; 55(4): 217-23, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21272062

ABSTRACT

The purpose of this study was to assess human ß-defensin-2 (hBD-2) gene transfection in human bladder epithelial cells and its therapeutic efficacy in a rat urinary tract infection (UTI) model via liposome mediated gene transfer. A large amount of hBD2 production (36.5 ± 3.2 ng/10(6) cells) was demonstrated in transfected cells' supernatants. In addition, a detectable amount of hBD-2 was identified in rats' urine (4.77 ± 1.4 ng/mL) by ELISA. Expression of the transgene hBD-2 in transfected cells and rats' bladders was also confirmed by RT-PCR and Western blotting. Immunohistochemistry revealed that the transgene hBD-2 expressed in the entire epithelial layer of the transduced bladders. Numbers of bacterial colony-forming units in urine and bladders from hBD2 gene treated UTI rats were significantly lower than those from the UTI rats administered PBS at 24, 36, and 72 hr after infection (P < 0.05). In addition, in vivo expression of hBD-2 reduced mucosal damage, interstitial edema and inflammatory cell infiltration in UTI animals. The results indicate that successful inhibition of UTI progression can be produced by hBD2 gene therapy. The liposome-mediated hBD2 plasmid DNA transfection system appears to be a promising method for antimicrobial gene therapy of UTI.


Subject(s)
Genetic Therapy/methods , Transfection/methods , Urinary Tract Infections/genetics , Urinary Tract Infections/therapy , beta-Defensins/genetics , Administration, Intravesical , Animals , Cell Line , Disease Models, Animal , Female , Genetic Therapy/instrumentation , Humans , Liposomes , Mice , Rats , Rats, Wistar , Transfection/instrumentation , Urinary Tract Infections/microbiology , beta-Defensins/administration & dosage , beta-Defensins/therapeutic use
12.
J Leukoc Biol ; 110(5): 951-963, 2021 11.
Article in English | MEDLINE | ID: mdl-33682193

ABSTRACT

As components of the innate immune response, antimicrobial peptides (AMPs) efficiently contribute to infection control and maintenance of a latent state in pulmonary tuberculosis (TB). As a therapeutic strategy, the administration of recombinant AMPs could be limited by enzymatic degradation and high production costs. Likewise, strategies based on the induction of AMPs have generated controversial results. In this study, 2 recombinant type-5 adenoviruses (Ad) expressing the human ß-defensin 3 (HßD3) or cathelicidin (LL37) were assessed in a murine pulmonary TB model. Mice infected with either a high dose of a drug-sensitive (H37Rv) or a multidrug-resistant (MDR) strain of Mycobacterium tuberculosis (Mtb) were treated with a single administration of AdHßD3, AdLL37, AdGFP (control vector expressing a green fluorescent protein), or saline solution (SS). Lungs were obtained to determine the bacterial burden, histologic damage, and cytokine expression at different time points. Mice treated with AdHßD3 or AdLL37 showed significantly lower bacterial load and pneumonia, and higher proinflammatory cytokine expression than the control groups AdGFP and SS. A synergistic therapeutic effect could be observed when first- or second-line antibiotics (ABs) were administered with adenoviral therapy in animals infected with H37Rv or MDR strains, respectively. Adenovirus-delivered AMP's administration constitutes a promising adjuvant therapy for current anti-TB drugs by enhancing a protective immune response and potentially reducing current AB regimes' duration.


Subject(s)
Antimicrobial Cationic Peptides/administration & dosage , Antitubercular Agents/administration & dosage , Tuberculosis, Pulmonary/pathology , beta-Defensins/administration & dosage , Adenoviridae , Animals , Drug Therapy, Combination/methods , Genetic Vectors , Humans , Mice , Tuberculosis, Multidrug-Resistant/pathology , Cathelicidins
13.
Fundam Clin Pharmacol ; 34(1): 102-108, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31313350

ABSTRACT

The aims of this study were: (i) To investigate the activity of recombinant AMPs HNP-1 and hBD-1 in combination with cefotaxime against Staphylococcus aureus strains (MSSA and MRSA) in vitro using checkerboard method; (ii) To investigate the activity of HNP-1 and hBD-1 encapsulated in silicon nanoparticles (niosomes) in the treatment of MRSA-infected wound in rats. For this S. aureus strains (MSSA and MRSA) were isolated from patients with diabetic foot infection. Cefotaxime, recombinant HNP-1 and hBD-1 (in all possible combinations with each other) were used for testing by the checkerboard method. Two niosomal topical gels with HNP-1/hBD-1 were prepared to treat MRSA-infected wounds in rats. Gels were administered once a day, the control group-without treatment. Wound healing rate was calculated on the 4th, 9th and 16th days of the experiment and compared using one-way ANOVA with Bonferroni correction. MIC of HNP-1 for MSSA and MRSA was the same-1 mg/L. MIC of hBD-1 for MSSA and MRSA was also the same-0.5 mg/L. Topical gels with niosomal HNP-1 (or hBD-1) showed a significantly faster wound healing in comparison with the control. The data obtained open up prospects for use of AMPs encapsulated in silica nanoparticles for the development of new antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Staphylococcal Infections/drug therapy , alpha-Defensins/pharmacology , beta-Defensins/pharmacology , Administration, Topical , Animals , Anti-Bacterial Agents/administration & dosage , Cefotaxime/administration & dosage , Cefotaxime/pharmacology , Diabetic Foot/drug therapy , Diabetic Foot/microbiology , Drug Therapy, Combination , Gels , Humans , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Nanoparticles , Rats , Rats, Wistar , Silicon Dioxide/chemistry , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/isolation & purification , Wound Healing/drug effects , alpha-Defensins/administration & dosage , beta-Defensins/administration & dosage
14.
Dev Comp Immunol ; 103: 103511, 2020 02.
Article in English | MEDLINE | ID: mdl-31580833

ABSTRACT

ß-defensin is a cationic host defense peptide actively participating in host innate immune response against pathogens. In teleost fish, ß-defensin exhibits a diversity in genotypes and functions. Herein, a ß-defensin homolog (PaBD) was identified from ayu, Plecoglossus altivelis, showing multiple tissues' upregulation against Vibrio anguillarum challenge. In vivo experiments revealed that intraperitoneal injection of chemically synthesized mature PaBD (mPaBD) increased the survival rate of V. anguillarum-infected ayu, accompanied by reduced bacterial load and decreased tissue mRNA levels of tumor necrosis factor α (PaTNF-α) and interleukin 1ß (PaIL-1ß). However, in vitro, mPaBD showed weak bactericidal activity against V. anguillarum. Interestingly, mPaBD enhanced phagocytosis, intracellular bacterial killing, and respiratory burst of ayu monocytes/macrophages (MO/MΦ). Moreover, it inhibited mRNA levels of PaIL-1ß and PaTNF-α in MO/MФ upon V. anguillarum infection. In conclusion, PaBD protects ayu against V. anguillarum challenge not only through its direct antibacterial ability, but also through its immunomodulation in MO/MΦ.


Subject(s)
Fish Diseases/immunology , Fish Proteins/metabolism , Osmeriformes/immunology , Vibrio Infections/veterinary , Vibrio/physiology , beta-Defensins/metabolism , Amino Acid Sequence , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Bacterial Load/drug effects , Cytokines/genetics , Fish Diseases/microbiology , Fish Diseases/prevention & control , Fish Proteins/administration & dosage , Fish Proteins/genetics , Immunomodulation , Macrophages/immunology , Macrophages/microbiology , Monocytes/immunology , Monocytes/microbiology , Osmeriformes/classification , Osmeriformes/genetics , Phagocytosis , Phylogeny , Respiratory Burst , Sequence Alignment , Survival Rate , Tissue Distribution , Vibrio/drug effects , Vibrio Infections/immunology , Vibrio Infections/microbiology , Vibrio Infections/prevention & control , beta-Defensins/administration & dosage , beta-Defensins/genetics
15.
J Microbiol Biotechnol ; 19(11): 1447-55, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19996700

ABSTRACT

Two novel avian beta-defensins (AvBDs) isolated from duck liver were characterized and their homologies with other AvBDs were analyzed. They were shown to be duck AvBD9 and AvBD10. The mRNA expression of the two genes was analyzed in 17 different tissues from 1-28-dayold ducks. AvBD9 was differentially expressed in the tissues, with especially high levels of expression in liver, kidney, crop, and trachea, whereas AvBD10 was only expressed in the liver and kidney of ducks at all the ages investigated. We produced and purified GST-tagged recombinant AvBD9 and AvBD10 by expressing the two genes in Escherichia coli. Both recombinant proteins exhibited antimicrobial activity against several bacterial strains. The results revealed that both recombinant proteins retained their antimicrobial activities against Staphylococcus aureus under a range of different temperatures (-70 degrees C -100 degrees C) and pH values (pH 3-12).


Subject(s)
Anti-Infective Agents/administration & dosage , beta-Defensins/administration & dosage , beta-Defensins/physiology , Amino Acid Sequence , Animals , Anti-Infective Agents/isolation & purification , Cloning, Molecular , Crop, Avian/metabolism , Ducks , Escherichia coli/metabolism , Gene Expression Profiling , Gene Expression Regulation , Kidney/metabolism , Liver/metabolism , Molecular Sequence Data , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Sequence Homology , Staphylococcus aureus/drug effects , Trachea/metabolism , beta-Defensins/isolation & purification
16.
Histol Histopathol ; 32(10): 1017-1027, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28032328

ABSTRACT

A number of pathogens for which there are no effective treatments infect the cells via endocytosis. Once in the endosomes, the pathogens complete their life cycle by overriding normal lysosomal functions. Recently, our laboratory identified the lysosomal targeting signal of prosaposin, which is recognized by the sorting receptor "sortilin". Based on this evidence, we tested whether the antimicrobial peptide ß-Defensin linked to the targeting sequence of prosaposin (ßD-PSAP) could be redirected from its secretory pathway to the endolysosomal compartment. To this effect, ßD-PSAP was transfected into COS-7 cells. The sub-cellular distribution of ßD-PSAP was analyzed by confocal microscopy and differential centrifugation. Confocal microscopy demonstrated that ßD-PSAP overlaid with the lysosomal marker LAMP1, indicating that the construct reached endosomes and lysosomes. Differential centrifugation also showed that ßD-PSAP was in the lysosomal fractions. In addition, our binding inhibition assay demonstrated that ßD-PSAP bound specifically to sortilin. Similarly, the delivery of ßD-PSAP was abolished after overexpressing a truncated sortilin. These results indicate that the prosaposin C-terminus and D/C-domain (prosaposin targeting sequence) was an effective "guidance system" to redirect ßD-PSAP to the endolysosomal compartment. In the future, this and other fusion proteins with antimicrobial properties will be assembled to our "biotic vehicle" to target pathogens growing within these compartments.


Subject(s)
Anti-Infective Agents/pharmacology , Drug Delivery Systems/methods , Lysosomes/drug effects , Pharmaceutical Vehicles , beta-Defensins/pharmacology , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Anti-Infective Agents/administration & dosage , COS Cells , Chlorocebus aethiops , Endosomes/metabolism , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Protein Binding/drug effects , Saposins/metabolism , Subcellular Fractions/metabolism , beta-Defensins/administration & dosage
17.
Vaccine ; 35(6): 981-986, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28087147

ABSTRACT

Inclusion body hepatitis (IBH) is one of the major viral infections causing substantial economic loss to the global poultry industry. The disease is characterized by a sudden onset of mortality (2-30%) and high morbidity (60-70%). IBH is caused by a number of serotypes of fowl adenovirus with substantially low levels of serotype cross protection. Thus far, there is no effective and safe vaccine commercially available in the North America for the control of IBH in chickens. Poly[di(sodium carboxylatoethylphenoxy)]phosphazene (PCEP) is a high molecular weight, biodegradable water soluble polymer that has been well characterized as a safe and effective adjuvant for a number of experimental veterinary vaccines. Similarly, host defence peptides, including ß-defensins, have also been shown to exhibit strong adjuvant potential. In this study, we evaluated the adjuvant activity of PCEP and avian beta defensin (ABD) in a vaccine formulation containing inactivated fowl adenovirus (FAdV) serotype 8b administered in ovo. Our data showed that a combination of PCEP and inactivated virus is capable of inducing a robust and long lasting antibody response. Moreover, significant enhancement of IFN-γ, IFN-α, IL-12(p40) and IL-6 gene expression under the influence of PCEP suggests that as an in ovo adjuvant PCEP has the ability to activate a substantial balanced immune response in chickens. To our knowledge, these are the first studies in which PCEP and ABD have been characterized as adjuvants for the development of an in ovo poultry vaccine. It is expected that these preliminary studies will be helpful in the development of safer and more effective in ovo vaccine against IBH and other infectious diseases affecting chickens.


Subject(s)
Adenoviridae Infections/prevention & control , Adenovirus Vaccines/administration & dosage , Chickens/immunology , Fowl adenovirus A/immunology , Phenylpropionates/administration & dosage , Polymers/administration & dosage , Poultry Diseases/prevention & control , beta-Defensins/administration & dosage , Adenoviridae Infections/immunology , Adenoviridae Infections/veterinary , Adenoviridae Infections/virology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Viral/biosynthesis , Chick Embryo , Chickens/virology , Fowl adenovirus A/growth & development , Fowl adenovirus A/pathogenicity , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Interferon-alpha/biosynthesis , Interferon-alpha/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-12 Subunit p40/biosynthesis , Interleukin-12 Subunit p40/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Poultry Diseases/immunology , Poultry Diseases/virology , Serogroup , Vaccines, Attenuated
18.
Anim Sci J ; 87(10): 1258-1266, 2016 Oct.
Article in English | MEDLINE | ID: mdl-26707031

ABSTRACT

Synthetic porcine beta-defensin-2 (pBD-2) was tested as an alternative to antimicrobial growth-promoters in pig production. Thirty 21-day weaned piglets were challenged with enterotoxigenic Escherichia coli, and orally dosed with either sterile water (CON), pBD-2 (BD) or neomycin sulphate (NS) twice daily for 21 days. pBD-2 and NS led to higher growth performance, jejunum villus height and increased expression of insulin-like growth factor-I compared with the CON group (P < 0.05). Hemolytic E. coli scores from rectal swabs, and copy numbers of E. coli, Bacteroides fragilis and Streptococcus in the cecal digesta of the BD- or NS-treated piglets were lower than those in the CON group (P < 0.05). Messenger RNA levels of toll-like receptor 4, tumor necrosis factor-α, interleukin (IL)-1ß, and IL-8 in the jejunum mucosa of the BD and NS groups were lower than those in the CON group (P < 0.05). Copy numbers of Lactobacilli and Bifidobacteria in the cecal digesta of the BD group were higher than those of the CON and NS groups (P < 0.05). Therefore, pBD-2 has antimicrobial activity in piglets, and it can improve growth performance, reduce inflammatory cytokine expression and affect intestinal morphological indices in the same way as probiotics. © 2015 Japanese Society of Animal Science.


Subject(s)
Cecum/microbiology , Cytokines/genetics , Cytokines/metabolism , Down-Regulation/drug effects , Enterotoxigenic Escherichia coli/immunology , Gastrointestinal Microbiome , Gene Expression/drug effects , Inflammation Mediators/metabolism , Swine/growth & development , Swine/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , beta-Defensins/administration & dosage , beta-Defensins/pharmacology , Administration, Oral , Animals , Male , Probiotics , Swine/microbiology , Weaning
19.
Peptides ; 82: 92-100, 2016 08.
Article in English | MEDLINE | ID: mdl-27298203

ABSTRACT

Human beta-defensin 3 (hBD3) is an antimicrobial peptide showing immunomodulatory effect on both innate and acquired immune response. Atherosclerosis is an inflammatory disease characterized by accumulation of lipids in the vascular wall. In this study, we evaluated whether hBD3 could attenuate the atherosclerosis development accelerated by Porphyromonas gingivalis lipopolysaccharide (Pg-LPS) with apolipoprotein E-deficient (ApoE(-/-)) mice. We observed that, in vivo, hBD3 inhibited serum MCP-1, sICAM-1 levels of ApoE-deficient mice exposed to Pg-LPS in a chronic inflammation model. Serum levels of total cholesterol (TC) and low-density lipoprotein (LDL) were also markedly reduced with hBD3 intervention. In addition, thinned vascular walls, less macrophage infiltration and the formation of atherosclerotic lesions were observed in the hBD3-treated group. Furthermore, in vitro, hBD3 profoundly suppressed the production of TNF-α and IL-6 in RAW 264.7 cells induced by Pg-LPS in a dose-dependent manner. Moreover, hBD3 attenuated the phosphorylation of p38 and ERK1/2 in the mitogen-activated protein kinase (MAPK) pathway. Taken together, our work has revealed that hBD3 exhibits potent anti-inflammatory properties both in vitro and in vivo, and this effect might be correlated with inhibition of MAPK pathway.


Subject(s)
Atherosclerosis/drug therapy , Inflammation/drug therapy , beta-Defensins/administration & dosage , Animals , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/chemically induced , Atherosclerosis/microbiology , Chemokine CCL2/blood , Humans , Inflammation/blood , Inflammation/chemically induced , Intercellular Adhesion Molecule-1/blood , Interleukin-6/genetics , Lipopolysaccharides/chemistry , Lipopolysaccharides/toxicity , Mice , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/genetics , Porphyromonas gingivalis/chemistry , Porphyromonas gingivalis/pathogenicity , Tumor Necrosis Factor-alpha/genetics , beta-Defensins/metabolism
20.
Sci Rep ; 6: 26790, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27225034

ABSTRACT

Post-weaning diarrhoea (PWD) in piglets is associated with colonization of the intestine with bacterial pathogens. In this study, we evaluated the use of recombinant porcine ß-defensin 2 (rpBD2) as a medicated feed additive for weaned piglets. The crude extract from the culture supernatant of rpBD2-expressing Pichia pastoris was used as a medicated feed additive for weaned piglets. Dietary treatments included a positive control (basal diet + antibiotics, designated PC) and three different rpBD2 treatments without antibiotics (basal diet supplemented with 1, 5, or 15 g of crude rpBD2/kg basal diet, designated 1PD, 5PD, and 15PD, respectively). Of all the treatments, 5PD had the greatest impact on the weaned piglets. It increased their body weight, average daily weight gain, average daily feed intake, and intestinal villus height in the duodenum and jejunum, and reduced the incidence of PWD. The diversity of the cecal digesta and mucosa microflora was compared between the weaned piglets in the PC and 5PD groups. Piglets treated with 5PD had lower diversity indices and fewer bacterial pathogens in their cecal digesta and mucosa than the PC group. Our results demonstrate that crude rpBD2 could provide an alternative to the traditional antibiotic feed additives given to weaned piglets.


Subject(s)
Animal Feed , Anti-Infective Agents/therapeutic use , Diarrhea/veterinary , Food Additives/therapeutic use , Swine Diseases/prevention & control , beta-Defensins/therapeutic use , Animals , Anti-Infective Agents/administration & dosage , Bacteria/classification , Bacteria/isolation & purification , Body Weight/drug effects , Cecum/microbiology , Diarrhea/microbiology , Diarrhea/prevention & control , Drug Evaluation, Preclinical , Eating , Food Additives/administration & dosage , Gastrointestinal Microbiome/drug effects , Intestine, Small/drug effects , Intestine, Small/ultrastructure , Microvilli/ultrastructure , Pichia , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Ribotyping , Sequence Alignment , Staphylococcus aureus/drug effects , Swine , Swine Diseases/microbiology , Weaning , beta-Defensins/administration & dosage , beta-Defensins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL