Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Future Microbiol ; 16(3): 143-157, 2021 02.
Article in English | MEDLINE | ID: mdl-33528272

ABSTRACT

Aim: Iron uptake and metabolism pathways are promising targets in vaccine development as an alternative strategy for antibiotics. Methods & methods: HemTR, a putative heme receptor of Acinetobacter baumannii, was expressed and its protectivity against A. baumannii was determined singly or in combination with the siderophore receptor, BauA, in mice. Results: High level of IgG was elicited. There was a delay in mice mortality with reduced bacterial loads in internal organs in the sublethal challenge. Protection was better in the HemTR-BauA group in both lethal and sublethal challenges. Passive transfer of anti-HemTR and anti-BauA partially protected mice against A. baumannii infection. Conclusion: HemTR in combination with other iron receptors could contribute to the development of protective vaccines against A. baumannii.


Subject(s)
Acinetobacter Infections/prevention & control , Acinetobacter baumannii/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Receptors, Cell Surface/immunology , Sepsis/prevention & control , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Animals , Bacterial Load , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Disease Models, Animal , Female , Heme/immunology , Humans , Mice , Mice, Inbred BALB C , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/genetics , Sepsis/immunology , Sepsis/microbiology
3.
Microb Pathog ; 144: 104161, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32194179

ABSTRACT

Klebsiella pneumoniae, a multi drug resistant nosocomial pathogen is associated with pneumonia and immunization gives a hope to fight its infections. A possible vaccine candidate is the conserved protein, yersiniabactin receptor FyuA. Its expression along with the siderophore yersiniabactin increases in bacteria under iron starving conditions prevailing in lungs. In this study, the potential of recombinant FyuA of K. pneumoniae has been evaluated against lung infection in BALB/c mice. Immunization generated both humoral and cell mediated response which conferred protection against the lethal dose of bacteria. Bacterial burden in lungs reduced by 6 log10 CFU/ml after 2nd day post infection as compared to control. Similarly, the levels of pro-inflammatory cytokines IL-17, TNF-α and IL-1ß were also reduced significantly; reduced tissue damage was evident from histopathology of lungs in immunized mice. These results indicate the protective role of FyuA which can be a potential vaccine candidate.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Klebsiella Infections/prevention & control , Klebsiella pneumoniae/immunology , Pneumonia, Bacterial/prevention & control , Receptors, Cell Surface/immunology , Animals , Bacterial Load , Bacterial Proteins/administration & dosage , Female , Immunization , Interleukin-17/blood , Interleukin-1beta/blood , Lung/pathology , Mice , Mice, Inbred BALB C , Phenols/metabolism , Pneumonia, Bacterial/microbiology , Receptors, Cell Surface/administration & dosage , Thiazoles/metabolism , Tumor Necrosis Factor-alpha/blood
4.
Hypertension ; 74(4): 1014-1020, 2019 10.
Article in English | MEDLINE | ID: mdl-31378099

ABSTRACT

Obesity-related hypertension is a major public health concern. We recently demonstrated that plasma levels of the soluble form of the prorenin receptor (sPRR) were elevated in obesity-associated hypertension. Therefore, in the present study, we investigated the contribution of sPRR to blood pressure (BP) elevation in the context of obesity. High fat-fed C57BL/6 male mice were infused with vehicle or sPRR (30 µg/kg per day) via subcutaneously implanted osmotic minipump for 4 weeks. BP parameters were recorded using radiotelemetry devices. Male mice infused with sPRR exhibited higher systolic BP and mean arterial pressure and lower spontaneous baroreflex sensitivity than mice infused with vehicle. To define mechanisms involved in systolic BP elevation, mice were injected with an AT1R (Ang II [angiotensin II] type 1 receptor) antagonist (losartan), a muscarinic receptor antagonist (atropine), a ß-adrenergic antagonist (propranolol), and a ganglionic blocker (chlorisondamine). Losartan did not blunt sPRR-induced elevation in systolic BP. Chlorisondamine treatment exacerbated the decrease in mean arterial pressure in male mice infused with sPRR. These results demonstrated that sPRR induced autonomic nervous dysfunction. Interestingly, plasma leptin levels were increased in high fat-fed C57BL/6 male mice infused with sPRR. Overall, our results indicated that sPRR increased systolic BP through an impairment of the baroreflex sensitivity and an increase in the sympathetic tone potentially mediated by leptin in high fat-fed C57BL/6 male mice.


Subject(s)
Blood Pressure/drug effects , Diet, High-Fat , Receptors, Cell Surface/administration & dosage , Adrenergic beta-Antagonists/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Atropine/pharmacology , Baroreflex/drug effects , Chlorisondamine/pharmacology , Ganglionic Blockers/pharmacology , Infusions, Subcutaneous , Leptin/blood , Losartan/pharmacology , Male , Mice , Mice, Inbred C57BL , Muscarinic Antagonists/pharmacology , Propranolol/pharmacology , Prorenin Receptor
5.
Int Immunopharmacol ; 63: 211-219, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30103196

ABSTRACT

Ruminants are the major reservoirs of Escherichia coli O157:H7 and its fecal shedding mainly act as a source of entry of this pathogen into the human food chain. In humans, E. coli O157:H7 infection causes diarrhea, hemorrhagic colitis and hemolytic uremic syndrome. Intimate adherence of E. coli O157:H7 is mediated by Translocated intimin receptor (Tir) to which intimin binds in the host cell. Since E. coli O157:H7 colonizes intestinal epithelium, the mucosal vaccine has a potential to prevent its colonization. Zonula occludens toxin (Zot) of Vibrio cholerae transiently, reversibly alters epithelial tight junction structure to increase mucosal permeability of macromolecules via paracellular route. The C-terminal region of Zot (ΔG) responsible for this function could be used for mucosal antigen delivery. Therefore, we employed individual (Tir), cocktail (ΔG + Tir), fusion protein (ΔG-Tir) and assessed the efficacy of its intranasal immunization on immunogenicity and fecal shedding of E. coli O157:H7 in streptomycin treated mouse model. Compared to control, ΔG + Tir, ΔG-Tir immunized mice elicited significant antigen specific antibody titers in serum (IgG, IgA) and feces (IgA), whereas Tir immunized mice induced only serum IgG titer. Cytokine analysis revealed mixed Th1/Th2 type immune response in case of ΔG + Tir, ΔG-Tir group while that of Tir group was solely Th2 type. Tir, ΔG + Tir and ΔG-Tir immunized mice showed reduction in shedding of E. coli O157:H7 compared to control group. However, ΔG-Tir immunized group performed better than ΔG + Tir, Tir group in reducing fecal shedding. Overall, our results demonstrate that intranasal immunization of ΔG-Tir induces effective systemic, mucosal, cellular immune responses and represents a promising mucosal subunit vaccine to prevent E. coli O157:H7 colonization.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cholera Toxin/administration & dosage , Escherichia coli Proteins/administration & dosage , Receptors, Cell Surface/administration & dosage , Recombinant Fusion Proteins/administration & dosage , Vaccines, Subunit/administration & dosage , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Cytokines/immunology , Endotoxins , Escherichia coli Infections/immunology , Escherichia coli O157 , Feces/microbiology , Female , Immunization , Immunoglobulin A/blood , Immunoglobulin G/blood , Mice, Inbred BALB C
6.
Vet Microbiol ; 219: 1-7, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29778179

ABSTRACT

Porcine reproductive and respiratory syndrome (PRRS) is one of the most important swine diseases affecting pig industry worldwide. Sialoadehesin (Sn) and CD163 are the two specific receptors for PRRSV infection of porcine alveolar macrophages. Our previous study showed that the soluble Sn receptor Sn4D-Fc and soluble CD163 receptor SRCR59-Fc expressed by the two recombinant adenoviral (rAd) vectors have an additive anti-PRRSV effect in vitro. In the present study, rAd-Sn4D-Fc and rAd-SRCR59-Fc were inoculated into pigs, and the efficient expression of Sn4D-Fc and SRCR59-Fc proteins was detected by ELISA. Then, PRRSV-naïve pigs were inoculated with rAd-Sn4D-Fc and/or rAd-SRCR59-Fc before contagious infection with different PRRSV strains. Among the three rAd inoculation groups, simultaneous inoculation with the two rAd vectors provided the best protection against highly pathogenic JXA1 strain PRRSV, followed by rAd-SRCR59-Fc inoculation and rAd-Sn4D-Fc inoculation. Clinical observation and quantitative RT-PCR analyses showed that all of the double rAd-inoculated pigs (n = 9) survived from the contagious infection with highly pathogenic JXA1, JS07 or SH1705 strain PRRSV with significantly alleviated clinical scores, viremia, fecal viral emission and tissue virus loads. These data suggest that rAd-Sn4D-Fc and rAd-SRCR59-Fc can be developed further as the universal therapeutic vaccine to facilitate PRRSV eradication.


Subject(s)
Adenoviridae/genetics , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/immunology , Receptors, Cell Surface/immunology , Sialic Acid Binding Ig-like Lectin 1/immunology , Animals , Antibodies, Viral , Antigens, CD/administration & dosage , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/administration & dosage , Antigens, Differentiation, Myelomonocytic/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Genetic Vectors , Lung/immunology , Lung/pathology , Lung/virology , Macrophages, Alveolar/virology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/pathogenicity , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/genetics , Sialic Acid Binding Ig-like Lectin 1/administration & dosage , Sialic Acid Binding Ig-like Lectin 1/genetics , Swine , Viral Load , Viremia
7.
Ann Rheum Dis ; 75(9): 1706-13, 2016 09.
Article in English | MEDLINE | ID: mdl-26452536

ABSTRACT

OBJECTIVE: Netrin-1 is a chemorepulsant and matrix protein expressed during and required for osteoclast differentiation, which also plays a role in inflammation by preventing macrophage egress. Because wear particle-induced osteolysis requires osteoclast-mediated destruction of bone, we hypothesised that blockade of Netrin-1 or Unc5b, a receptor for Netrin-1, may diminish this pathological condition. METHODS: C57BL/6 mice, 6-8 weeks old, had 3 mg of ultrahigh-molecular-weight polyethylene particles implanted over the calvaria and then received 10 µg of monoclonal antibodies for Netrin-1 or its receptors, Unc5b and deleted in colon cancer (DCC), injected intraperitoneally on a weekly basis. After 2 weeks, micro-computed tomography and histology analysis were performed. Netrin-1 expression was analysed in human tissue obtained following primary prosthesis implantation or after prosthesis revision for peri-implant osteolysis and aseptic implant loosening. RESULTS: Weekly injection of anti-Netrin-1 or anti-Unc5b-antibodies significantly reduced particle-induced bone pitting in calvaria exposed to wear particles (46±4% and 49±3% of control bone pitting, respectively, p<0.001) but anti-DCC antibody did not affect inflammatory osteolysis (80±7% of control bone pitting, p=ns). Anti-Netrin-1 or anti-Unc5b, but not anti-DCC, antibody treatment markedly reduced the inflammatory infiltrate and the number of tartrate resistance acid phosphatase (TRAP)-positive osteoclasts (7±1, 4±1 and 14±1 cells/high power field (hpf), respectively, vs 12±1 cells/hpf for control, p<0.001), with no significant changes in alkaline phosphatase-positive osteoblasts on bone-forming surfaces in any antibody-treated group. Netrin-1 immunostaining colocalised with CD68 staining for macrophages. The peri-implant tissues of patients undergoing prosthesis revision surgery showed an increase in Netrin-1 expression, whereas there was little Netrin-1 expression in soft tissues removed at the time of primary joint replacement. CONCLUSIONS: These results demonstrate a unique role for Netrin-1 in osteoclast biology and inflammation and may be a novel target for prevention/treatment of inflammatory osteolysis.


Subject(s)
Nerve Growth Factors/physiology , Osteoclasts/physiology , Osteolysis/metabolism , Tumor Suppressor Proteins/physiology , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Inflammation , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors/antagonists & inhibitors , Netrin Receptors , Netrin-1 , Osteolysis/chemically induced , Osteolysis/pathology , Osteolysis/prevention & control , Polyethylenes , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/antagonists & inhibitors , Skull/metabolism , Skull/pathology , Tumor Suppressor Proteins/antagonists & inhibitors
8.
Wei Sheng Wu Xue Bao ; 56(8): 1326-34, 2016 Aug 04.
Article in Chinese | MEDLINE | ID: mdl-29738202

ABSTRACT

Objective: The purpose of this study is to reduce the colonization level of Campylobacter jejuni in chicken intestine by oral immunization of recombinant Lactococcus lactis expressing the ferric enterobactin receptor CfrA of C. jejuni. Methods: The whole cfrA gene and its N-terminal fragments were amplified by PCR, inserted into the expression vector pNZ8149 and transformed into L. lactis NZ3900. Based on the expression of CfrA in recombinant L. lactis by Western blot, the expression level was optimized by screening nisin concentration, induction temperature and time. Then the recombinant L. lactis strains were used to orally immunize specific-pathogen-free chickens. After oral immunization, the duration of recombinant L. lactis in chickens was determined by PCR, and the antibody levels of anti-CfrA serum IgG and intestinal mucosal sIgA were measured by ELISA. Finally, the immunized chickens were orally inoculated with C. jejuni to evaluate the inhibitory effect of recombinant L. lactis on colonization of C. jejuni. Results: Western blot results determined that the whole cfrA gene and its N-terminal fragments were both expressed in recombinant L. lactis in soluble forms whereas no secreted CfrA protein was detected outside bacterial cells. The optimal conditions for inducing the expression were grown at 37℃ for 1 h with nisin concentration of 25 ng/mL. Detection of chicken cloacal swabs showed that the duration of oral L. lactis was less than 10 days in chicken. The immunized groups produced higher antibody titers of anti-CfrA specific serum IgG and mucosal sIgA than the control groups. Moreover, the colonization rate of C. jejuni in the immunized groups was significantly lower than that in the control groups. Conclusion: Oral immunization of chickens with recombinant L. lactis expressing CfrA inhibited the colonization of C. jejuni. Our findings can be useful to develop oral vaccines with recombinant Lactobacillus for control of C. jejuni infection in chickens.


Subject(s)
Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Campylobacter Infections/veterinary , Campylobacter jejuni/immunology , Carrier Proteins/immunology , Lactococcus lactis/genetics , Poultry Diseases/immunology , Receptors, Cell Surface/immunology , Animals , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Campylobacter Infections/immunology , Campylobacter Infections/microbiology , Campylobacter Infections/prevention & control , Campylobacter jejuni/genetics , Carrier Proteins/administration & dosage , Carrier Proteins/genetics , Chickens , Gene Expression , Immunization , Lactococcus lactis/metabolism , Poultry Diseases/microbiology , Poultry Diseases/prevention & control , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/genetics
9.
J Infect ; 71(3): 326-37, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25982025

ABSTRACT

OBJECTIVES: Outer membrane vesicle (OMV) vaccines are used against outbreaks of capsular group B Neisseria meningitidis (MenB) caused by strains expressing particular PorA outer membrane proteins (OMPs). Ferric enterobactin receptor (FetA) is another variable OMP that induces type-specific bactericidal antibodies, and the combination of judiciously chosen PorA and FetA variants in vaccine formulations is a potential approach to broaden protection of such vaccines. METHODS: The OMV vaccine MenPF-1 was generated by genetically modifying N. meningitidis strain 44/76 to constitutively express FetA. Three doses of 25 µg or 50 µg of MenPF-1 were delivered intra-muscularly to 52 healthy adults. RESULTS: MenPF-1 was safe and well tolerated. Immunogenicity was measured by serum bactericidal assay (SBA) against wild-type and isogenic mutant strains. After 3 doses, the proportion of volunteers with SBA titres ≥1:4 (the putative protective titre) was 98% for the wild-type strain, and 77% for the strain 44/76 FetA(on)PorA(off) compared to 51% in the strain 44/76 FetA(off)PorA(off), demonstrating that vaccination with MenPF-1 simultaneously induced FetA and PorA bactericidal antibodies. CONCLUSION: This study provides a proof-of-concept for generating bactericidal antibodies against FetA after OMV vaccination in humans. Prevalence-based choice of PorA and FetA types can be used to formulate a vaccine for broad protection against MenB disease.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Carrier Proteins/genetics , Carrier Proteins/immunology , Meningococcal Vaccines/administration & dosage , Neisseria meningitidis, Serogroup B/genetics , Neisseria meningitidis, Serogroup B/immunology , Porins/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Adolescent , Adult , Antibodies, Bacterial/blood , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/administration & dosage , Female , Humans , Male , Meningococcal Vaccines/adverse effects , Meningococcal Vaccines/immunology , Middle Aged , Molecular Epidemiology , Porins/genetics , Receptors, Cell Surface/administration & dosage , Serum Bactericidal Antibody Assay , Young Adult
10.
Invest Ophthalmol Vis Sci ; 56(2): 1357-66, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25655801

ABSTRACT

PURPOSE: Glaucoma is a major cause of vision loss due to retinal ganglion cell (RGC) degeneration. Therapeutic intervention controls increased IOP, but neuroprotection is unavailable. NogoReceptor1 (NgR1) limits adult central nervous system (CNS) axonal sprouting and regeneration. We examined NgR1 blocking decoy as a potential therapy by defining the pharmacokinetics of intravitreal NgR(310)-Fc, its promotion of RGC axonal regeneration following nerve crush, and its neuroprotective effect in a microbead glaucoma model. METHODS: Human NgR1(310)-Fc was administered intravitreally, and levels were monitored in rat vitreal humor and retina. Axonal regeneration after optic nerve crush was assessed by cholera toxin ß anterograde labeling. In a microbead model of glaucoma with increased IOP, the number of surviving and actively transporting RGCs was determined after 4 weeks by retrograde tracing with Fluro-Gold (FG) from the superior colliculus. RESULTS: After intravitreal bolus administration, the terminal half-life of NgR1(310)-Fc between 1 and 7 days was approximately 24 hours. Injection of 5 µg protein once per week after optic nerve crush injury significantly increased RGCs with regenerating axons. Microbeads delivered to the anterior chamber increased pressure, and caused 15% reduction in FG-labeled RGCs of control rats, with a 40% reduction in large diameter RGCs. Intravitreal treatment with NgR1(310)-Fc did not reduce IOP, but maintained large diameter RGC density at control levels. CONCLUSIONS: Human NgR1(310)-Fc has favorable pharmacokinetics in the vitreal space and rescues large diameter RGC counts from increased IOP. Thus, the NgR1 blocking decoy protein may have efficacy as a disease-modifying therapy for glaucoma.


Subject(s)
Axons/pathology , Glaucoma/prevention & control , Nerve Regeneration/drug effects , Optic Nerve Injuries/drug therapy , Optic Nerve/pathology , Receptors, Cell Surface/administration & dosage , Retinal Ganglion Cells/drug effects , Animals , Axons/drug effects , Cell Line , Disease Models, Animal , Female , GPI-Linked Proteins/administration & dosage , Glaucoma/etiology , Glaucoma/pathology , Humans , Intravitreal Injections , Male , Nerve Crush , Nogo Receptors , Optic Nerve/drug effects , Optic Nerve/metabolism , Optic Nerve Injuries/complications , Optic Nerve Injuries/pathology , Rats , Rats, Sprague-Dawley , Retinal Ganglion Cells/pathology
11.
Vaccine ; 33(1): 133-40, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25448113

ABSTRACT

Zona pellucida (ZP) glycoproteins based contraceptive vaccines have been proposed for the management of wildlife population. In the present study, a fusion protein encompassing promiscuous T cell epitope of tetanus toxoid [TT; amino acid (aa) residues 830-844] followed by a dilysine linker and an ectodomain of dog ZP3 (ZP3; aa residues 23-348) without any affinity tag (TT-KK-ZP3) has been expressed in Escherichia coli. The recombinant protein was successfully produced in fed-batch fermentor and purified. The average yield of purified refolded protein was 12.20 ± 0.61 mg/2g wet cell pellet. Female FvB/J mice immunized with the varying doses of recombinant TT-KK-ZP3 supplemented with alum/PetGel A as adjuvants following a three injection schedule, showed dose dependent increase in serum IgG titer. Antibodies against TT-KK-ZP3 recognized native mouse/dog ZP and significantly inhibited mouse in-vitro fertilization (p=0.012). Immunized mice showed significant reduction in fertility (p<0.05). Higher antibody titers were associated with a decrease in the number of pups born to the immunized female mice. To reduce the number of injections, two injection schedule using various dose combinations of TT-KK-ZP3 supplemented with alum revealed lower immunogenicity and contraceptive efficacy as compared to the three injection schedule. To overcome this, CpG motif was included in addition to alum and both intraperitoneal and intranasal route of immunization following the two injection schedule was investigated. Inclusion of CpG significantly enhanced the antibody titer and improved contraceptive efficacy. In the mice immunized following intraperitoneal route, serum/vaginal IgG and in the mice immunized through intranasal route, vaginal IgA seemed to be important for curtailment in fertility. To conclude, the recombinant protein described herein may be a good candidate for developing contraceptive vaccine for the wildlife population management, in particular street dogs.


Subject(s)
Egg Proteins/immunology , Egg Proteins/isolation & purification , Fertility , Immunization/methods , Membrane Glycoproteins/immunology , Membrane Glycoproteins/isolation & purification , Receptors, Cell Surface/immunology , Receptors, Cell Surface/isolation & purification , Vaccines, Contraceptive/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies/blood , Cloning, Molecular , Dogs , Egg Proteins/administration & dosage , Escherichia coli/genetics , Escherichia coli/growth & development , Female , Gene Expression , Immunoglobulin G/blood , Litter Size , Membrane Glycoproteins/administration & dosage , Mice , Receptors, Cell Surface/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Treatment Outcome , Vaccines, Contraceptive/administration & dosage , Vaccines, Contraceptive/isolation & purification , Zona Pellucida Glycoproteins
12.
J Neurotrauma ; 31(24): 1955-66, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-24964223

ABSTRACT

Axonal growth and neurological recovery after traumatic spinal cord injury (SCI) is limited by the presence of inhibitory proteins in myelin, several of which act via the NgR1 protein in neurons. A truncated soluble ligand-binding fragment of NgR1 serves as a decoy and promotes recovery in acute and chronic rodent SCI models. To develop the translational potential of these observations, we created a human sequence-derived NgR1(310)-Fc protein. This protein is active in vitro. When the human NgR1 decoy is administered by continuous intracerebroventricular infusion to rats with a spinal contusion injury at doses of 0.09-0.53 mg/kg/d, neurological recovery is improved. Effective doses double the percentage of rats able to bear weight on their hindlimbs. Next, we considered the half-life and distribution of NgR1(310)-Fc after bolus delivery to the lumbar intrathecal space. The protein is found throughout the neuraxis and has a tissue half-life of approximately 2 days in the rat, and 5 days in the nonhuman primate. At an intermittent, once every 4 day, lumbar bolus dosing schedule of 0.14 mg/kg/d, NgR1(310)-Fc promoted locomotor rat recovery from spinal cord contusion at least as effectively as continuous infusion in open field and grid walking tasks. Moreover, the intermittent lumbar NgR1(310)-Fc treatment increased the growth of raphespinal axons into the lumbar spinal cord after injury. Thus, human NgR1(310)-Fc provides effective treatment for recovery from traumatic SCI in this preclinical model with a simplified administration regimen that facilitates clinical testing.


Subject(s)
Myelin Proteins/administration & dosage , Neuroprotective Agents/administration & dosage , Receptors, Cell Surface/administration & dosage , Receptors, Fc/administration & dosage , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Disease Models, Animal , Female , GPI-Linked Proteins/administration & dosage , Humans , Injections, Spinal , Nogo Receptor 1 , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage
13.
Lab Invest ; 94(8): 881-92, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24955893

ABSTRACT

Vascular endothelial cells (ECs) are ideal gene therapy targets as they provide widespread tissue access and are the first contact surfaces following intravenous vector administration. Human recombinant adenovirus serotype 5 (Ad5) is the most frequently used gene transfer system because of its appreciable transgene payload capacity and lack of somatic mutation risk. However, standard Ad5 vectors predominantly transduce liver but not the vasculature following intravenous administration. We recently developed an Ad5 vector with a myeloid cell-binding peptide (MBP) incorporated into the knob-deleted, T4 fibritin chimeric fiber (Ad.MBP). This vector was shown to transduce pulmonary ECs presumably via a vector handoff mechanism. Here we tested the body-wide tropism of the Ad.MBP vector, its myeloid cell necessity, and vector-EC expression dose response. Using comprehensive multi-organ co-immunofluorescence analysis, we discovered that Ad.MBP produced widespread EC transduction in the lung, heart, kidney, skeletal muscle, pancreas, small bowel, and brain. Surprisingly, Ad.MBP retained hepatocyte tropism albeit at a reduced frequency compared with the standard Ad5. While binding specifically to myeloid cells ex vivo, multi-organ Ad.MBP expression was not dependent on circulating monocytes or macrophages. Ad.MBP dose de-escalation maintained full lung-targeting capacity but drastically reduced transgene expression in other organs. Swapping the EC-specific ROBO4 for the CMV promoter/enhancer abrogated hepatocyte expression but also reduced gene expression in other organs. Collectively, our multilevel targeting strategy could enable therapeutic biological production in previously inaccessible organs that pertain to the most debilitating or lethal human diseases.


Subject(s)
Adenoviridae/genetics , Endothelium, Vascular/metabolism , Gene Transfer Techniques , Genetic Vectors/physiology , Receptors, Cell Surface/administration & dosage , Viral Tropism , Adenoviridae/physiology , Animals , Cytomegalovirus/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/virology , Genetic Vectors/administration & dosage , Green Fluorescent Proteins/administration & dosage , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Injections, Intravenous , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/metabolism , Myeloid Cells/virology , Peptides/administration & dosage , Peptides/metabolism , Promoter Regions, Genetic , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/metabolism , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Proteins/metabolism , Virion/physiology
14.
Cancer Imaging ; 13(4): 466-81, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24334439

ABSTRACT

Brain tumors are one of the most challenging disorders encountered, and early and accurate diagnosis is essential for the management and treatment of these tumors. In this article, diagnostic modalities including single-photon emission computed tomography, positron emission tomography, magnetic resonance imaging, and optical imaging are reviewed. We mainly focus on the newly emerging, specific imaging probes, and their potential use in animal models and clinical settings.


Subject(s)
Brain Neoplasms/diagnosis , Animals , Antigens, CD/administration & dosage , Endoglin , Gadolinium DTPA , Humans , Liposomes , Magnetic Resonance Imaging , Magnetite Nanoparticles/administration & dosage , Optical Imaging , Positron-Emission Tomography , Ranitidine , Receptors, Cell Surface/administration & dosage , Tomography, Emission-Computed, Single-Photon
15.
Insect Biochem Mol Biol ; 43(12): 1133-41, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24128609

ABSTRACT

We have identified the full-length cDNA encoding a vitellogenin receptor (VgR) from the African bont tick Amblyomma hebraeum Koch (1844). VgRs are members of the low-density lipoprotein receptor superfamily that promote the uptake of the yolk protein vitellogenin (Vg), from the haemolymph. The AhVgR (GenBank accession No. JX846592) is 5703 bp, and encodes an 1801 aa protein with a 196.5 kDa molecular mass following cleavage of a 22 aa signal peptide. Phylogenetic analysis indicates that AhVgR is highly similar to other tick VgRs. AhVgR is expressed in only the ovary of mated, engorged females, and is absent in all other female tissues and in both fed and unfed males. Unfed, adult females injected with a VgR-dsRNA probe to knock-down VgR expression experienced a significant delay in ovary development and started oviposition significantly later than controls. These results indicate that the expression of AhVgR is important for the uptake of Vg and subsequent maturation of the oocytes.


Subject(s)
Egg Proteins/genetics , Ovary/growth & development , Receptors, Cell Surface/genetics , Ticks/genetics , Animals , DNA, Complementary , Egg Proteins/administration & dosage , Egg Proteins/chemistry , Egg Proteins/isolation & purification , Female , Gene Expression Regulation, Developmental , Male , Ovary/drug effects , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/isolation & purification
16.
Crit Care Med ; 41(11): e309-18, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23928834

ABSTRACT

OBJECTIVES: Macrophages are important cells in immunity and the main producers of pro-inflammatory cytokines. The main objective was to evaluate if specific delivery of glucocorticoid to the macrophage receptor CD163 is superior to systemic glucocorticoid therapy in dampening the cytokine response to lipopolysaccharide infusion in pigs. DESIGN: Two randomized, placebo-controlled trials. SETTING: University hospital laboratory. SUBJECTS: Female farm-bred pigs (26-31 kg). DESIGN: A humanized antibody that binds to pig and human CD163 was produced, characterized, and conjugated with dexamethasone. In the first study (total n = 12), pigs were randomly assigned to four groups: 1) saline; 2) dexamethasone (1.0 mg/kg); 3) dexamethasone (0.02 mg/kg); and 4) anti-CD163-conjugated dexamethasone (0.02 mg/kg). In the second study (total n = 36), two additional groups were included in addition to the four original groups: 5) anti-CD163-conjugated dexamethasone (0.005 mg/kg); 6) unconjugated anti-CD163. Treatments were given 20 hours prior to infusion of lipopolysaccharide (1 µg × kg × h) for 5 hours. Blood samples were analyzed for cytokines, cortisol, and adrenocorticotropic hormone. RESULTS: In the saline group, lipopolysaccharide increased cytokine and plasma cortisol levels. In both studies, dexamethasone (1 mg/kg) and anti-CD163 dexamethasone (0.02 mg/kg) uniformly attenuated tumor necrosis factor-α peak levels (both p < 0.05) compared with low-dose dexamethasone (0.02 mg/kg). However, dexamethasone 1 mg/kg significantly suppressed plasma cortisol and adrenocorticotropic hormone levels compared with anti-CD163 dexamethasone (0.02 mg/kg; p < 0.05). No significant hemodynamic difference existed between groups. The anti-CD163 dexamethasone drug conjugate exhibited a fast plasma clearance, with a half-life of approximately 5-8 minutes. CONCLUSION: Targeted delivery of dexamethasone to macrophages using a humanized CD163 antibody as carrier exhibits anti-inflammatory effects comparable with 50 times higher concentrations of free dexamethasone and does not inhibit endogenous cortisol production. This antibody-drug complex showing similar affinity and specificity for human CD163 is, therefore, a promising drug candidate in this novel type of anti-inflammatory therapy.


Subject(s)
Antigens, CD/administration & dosage , Antigens, Differentiation, Myelomonocytic/administration & dosage , Dexamethasone/administration & dosage , Drug Carriers/pharmacology , Endotoxemia/drug therapy , Glucocorticoids/administration & dosage , Macrophages/metabolism , Receptors, Cell Surface/administration & dosage , Animals , Antigens, CD/pharmacology , Antigens, Differentiation, Myelomonocytic/pharmacology , Dexamethasone/pharmacology , Dose-Response Relationship, Drug , Female , Glucocorticoids/pharmacology , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Random Allocation , Swine
17.
Vaccine ; 31(40): 4382-8, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-23916294

ABSTRACT

The Duffy binding protein (DBP) of Plasmodium vivax is vital for host erythrocyte invasion. DBP region II (DBPII) contains critical residues for receptor recognition and anti-DBPII antibodies have been shown to inhibit erythrocyte binding and invasion, thereby making the molecule an attractive vaccine candidate against P. vivax blood stages. Similar to other blood-stage antigens, allelic variation within the DBPII and associated strain-specific immunity is a major challenge for development of a broadly effective vaccine against P. vivax malaria. We hypothesized that immunization with a vaccine composed of multiple DBP alleles or a modified epitope DBP (DEKnull) will be more effective in producing a broadly reactive and inhibitory antibody response to diverse DBPII alleles than a single allele vaccine. In this study, we compared single, naturally occurring DBPII allele immunizations (Sal1, 7.18, P) and DEKnull with a combination of (Sal1, 7.18, P) alleles. Quantitative analysis by ELISA demonstrated that the multiple allele vaccine tend to be more immunogenic than any of the single allele vaccines when tested for reactivity against a panel of DBPII allelic variants whereas DEKnull was less immunogenic than the mixed-allele vaccine but similar in reactivity to the single allele vaccines. Further analysis for functional efficacy by in vitro erythrocyte-binding inhibition assays demonstrated that the multiple allele immunization produced a stronger strain-neutralizing response than the other vaccination strategies even though inhibition remained biased toward some alleles. Overall, there was no correlation between antibody titer and functional inhibition. These data suggest that a multiple allele vaccine may enhance immunogenicity of a DBPII vaccine but further investigation is required to optimize this vaccine strategy to achieve broader coverage against global P. vivax strains.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Erythrocytes/parasitology , Malaria Vaccines/immunology , Plasmodium vivax/immunology , Protozoan Proteins/immunology , Receptors, Cell Surface/immunology , Animals , Antibodies, Protozoan/immunology , Antibody Formation/immunology , Antigens, Protozoan/administration & dosage , COS Cells , Cell Line , Chlorocebus aethiops , Genetic Variation/immunology , Humans , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Mice , Mice, Inbred BALB C , Protozoan Proteins/administration & dosage , Receptors, Cell Surface/administration & dosage
18.
Andrology ; 1(4): 624-31, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23657873

ABSTRACT

Sperm agglutinating factor (SAF) isolated from Staphylococcus aureus immobilizes spermatozoa by agglutination and causes sperm death. This interaction of SAF with spermatozoa is receptor mediated and this receptor has been isolated and purified from human spermatozoa. In this study we attempt to study whether the receptor could ameliorate the detrimental effects of SAF on sperm parameters. Receptor was evaluated against SAF mediated compromised sperm parameters such as Mg(2+) dependent ATPase activity, acrosome status and apoptosis, in vitro using fluorescent microscopy and flow cytometry as well as in vivo by studying the impact on fertility in mice. Incubation of SAF (80 µg) with spermatozoa resulted in reduced Mg(2+) dependent ATPase activity and premature acrosomal loss whereas a higher concentration (100 µg), induced apoptosis. However, in the presence of receptor a dose dependent blockage of SAF induced inhibition of Mg(2+) dependent ATPase activity was observed. At higher concentrations 100 and 125 µg, receptor could inhibit both the premature acrosomal loss and apoptosis. In vivo studies showed that receptor (50 µg) could alleviate SAF induced infertility in female Balb/c mice following a single intravaginal application before mating. The work highlights the efficacy of the receptor as a corrective measure against negative influence of SAF on functional parameters of spermatozoa as well as fertility and presents receptor as a potential therapeutic intervention against SAF induced infertility.


Subject(s)
Receptors, Cell Surface/metabolism , Sperm Agglutination , Spermatozoa/metabolism , Staphylococcus aureus/immunology , Acrosome Reaction , Adenosine Triphosphatases/metabolism , Animals , Apoptosis , Dose-Response Relationship, Drug , Female , Fertility , Flow Cytometry , Humans , Infertility, Female/immunology , Infertility, Female/metabolism , Infertility, Female/physiopathology , Infertility, Female/prevention & control , Male , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Receptors, Cell Surface/administration & dosage , Signal Transduction , Sperm Agglutination/drug effects , Spermatozoa/drug effects , Spermatozoa/immunology , Spermatozoa/pathology
19.
Nat Chem Biol ; 9(4): 250-6, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23416331

ABSTRACT

Targeted delivery of antigens to dendritic cells (DCs) is a promising vaccination strategy. However, to ensure immunity, the approach depends on coadministration of an adjuvant. Here we ask whether targeting of both adjuvant and antigen to DCs is sufficient to induce immunity. Using a protein ligation method, we develop a general approach for linking the immune stimulant, poly dA:dT (pdA:dT), to a monoclonal antibody (mAb) specific for DEC205 (DEC). We show that DEC-specific mAbs deliver pdA:dT to DCs for the efficient production of type I interferon in human monocyte-derived DCs and in mice. Notably, adaptive T-cell immunity is elicited when mAbs specific for DEC-pdA:dT are used as the activation stimuli and are administered together with a DC-targeted antigen. Collectively, our studies indicate that DCs can integrate innate and adaptive immunity in vivo and suggest that dual delivery of antigen and adjuvant to DCs might be an efficient approach to vaccine development.


Subject(s)
Adaptive Immunity/drug effects , Antibodies, Monoclonal/immunology , Antigens, CD/immunology , Antigens/immunology , Dendritic Cells/drug effects , Immunity, Innate/drug effects , Immunoconjugates/immunology , Lectins, C-Type/immunology , Poly dA-dT/immunology , Receptors, Cell Surface/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/chemistry , Antigens/administration & dosage , Antigens/chemistry , Antigens, CD/administration & dosage , Antigens, CD/chemistry , Dendritic Cells/immunology , Drug Delivery Systems , Genetic Vectors , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/chemistry , Interferon Type I/biosynthesis , Interferon Type I/immunology , Lectins, C-Type/administration & dosage , Lectins, C-Type/chemistry , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Plasmids , Poly dA-dT/administration & dosage , Poly dA-dT/chemistry , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/chemistry
20.
Front Biosci (Elite Ed) ; 5(2): 697-705, 2013 01 01.
Article in English | MEDLINE | ID: mdl-23277024

ABSTRACT

Prorenin-induced intracellular signaling pathway is not fully elucidated. We investigated whether the (pro)renin receptor mediates epidermal growth factor (EGF) receptor transactivation through angiotensin (Ang) II-dependent and -independent pathways in human embryo kidney 293 cells. Prorenin (2 nmol/L) caused biphasic phosphorylation of EGF receptor (Tyr992) and extracellular signal-regulated kinase (ERK) 1/2, peaking at 5 minutes followed by a decrease and a second peak at 60-120 minutes, whereas EGF receptor (Tyr1068) and Src were phosphorylated at only 120 minutes. These prorenin-induced phosphorylation processes were inhibited by (pro)renin receptor siRNA. Similarly, Ang II type 1 (AT1) receptor blocker (ARB) or AT1 receptor siRNA completely inhibited prorenin-induced phosphorylation of EGF receptor (Tyr1068) and Src, as well as the late peaks of EGF receptor (Tyr992) and ERK 1/2. However, early peaks of EGF receptor (Tyr992) and ERK 1/2 at 5 minutes were not effectively blocked by ARB or AT1 receptor siRNA. Incubation with prorenin significantly increased Ang II levels of cell lysate. These data indicate that the (pro)renin receptor mediates EGF receptor transactivation in both Ang II-dependent and -independent pathways.


Subject(s)
Angiotensin II/metabolism , ErbB Receptors/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Transcriptional Activation/physiology , Analysis of Variance , Blotting, Western , DNA Primers/genetics , ErbB Receptors/physiology , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/genetics , Time Factors , Prorenin Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...