Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
iScience ; 27(4): 109463, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38562521

ABSTRACT

Antimalarial drug resistance and unavailability of effective vaccine warrant for newer drugs and drug targets. Hence, anti-inflammatory activity of phyto-compound (oleuropein; OLP) was determined in antigen (LPS)-stimulated human THP-1 macrophages (macrophage model of inflammation; MMI). Reduction in the inflammation was controlled by the PI3K-Akt1 signaling to establish the "immune-homeostasis." Also, OLP treatment influenced the cell death/autophagy axis leading to the modulated inflammation for extended cell survival. The findings with MII prompted us to detect the antimalarial activity of OLP in the wild type (3D7), D10-expressing GFP-Atg18 parasite, and chloroquine-resistant (Dd2) parasite. OLP did not show the parasite inhibition in the routine in vitro culture of P. falciparum whereas OLP increased the antimalarial activity of artesunate. The molecular docking of autophagy-related proteins, investigations with MMI, and parasite inhibition assays indicated that the host activated the autophagy to survive OLP pressure. The challenge model of P. berghei infection showed to induce autophagy for circumventing anti-plasmodial defenses.

2.
Biomedicines ; 11(11)2023 Nov 14.
Article in English | MEDLINE | ID: mdl-38002042

ABSTRACT

In recent years, there has been a captivating focus of interest in elucidating the intricate crosstalk between adiponectin (APN), a versatile fat-associated adipokine and ocular pathologies. Unveiling the intricate relationship between adipocytokine APN and its receptors (AdipoRs) with aging eye disorders has emerged as a fascinating frontier in medical research. This review article delves into this connection, illuminating the hidden influence of APN on retinal health. This comprehensive review critically examines the latest findings and breakthroughs that underscore the pivotal roles of APN/AdipoRs signaling in maintaining ocular homeostasis and protecting against eye ailments. Here, we meticulously explore the intriguing mechanisms by which APN protein influences retinal function and overall visual acuity. Drawing from an extensive array of cutting-edge studies, the article highlights APN's multifaceted functions, ranging from anti-inflammatory properties and oxidative stress reduction to angiogenic regulation within retinal and macula tissues. The involvement of APN/AdipoRs in mediating these effects opens up novel avenues for potential therapeutic interventions targeting prevalent aging eye conditions. Moreover, this review unravels the interplay between APN signaling pathways and age-related macular degeneration (AMD). The single-cell RNA-seq results validate the expression of both the receptor isoforms (AdipoR1/R2) in retinal cells. The transcriptomic analysis showed lower expression of AdipoR1/2 in dry AMD pathogenesis compared to healthy subjects. The inhibitory adiponectin peptide (APN1) demonstrated over 75% suppression of CNV, whereas the control peptide did not exert any inhibitory effect on choroidal neovascularization (CNV). The elucidation of these relationships fosters a deeper understanding of adipose tissue's profound influence on ocular health, presenting new prospects for personalized treatments and preventative measures. Because APN1 inhibits CNV and leakage, it can be used to treat human AMD, although the possibility to treat human AMD is in the early stage and more clinical research is needed. In conclusion, this review provides a captivating journey into the enthralling world of APN, intertwining the realms of adipose biology and ophthalmology in aging.

3.
Biomolecules ; 13(7)2023 07 14.
Article in English | MEDLINE | ID: mdl-37509157

ABSTRACT

Age-related macular degeneration (AMD) has two phenotypes: dry AMD and wet AMD [...].


Subject(s)
Geographic Atrophy , Wet Macular Degeneration , Humans , Retina
4.
Biochem Biophys Res Commun ; 530(2): 367-373, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32800337

ABSTRACT

Choroidal neovascularization (CNV) is the hallmark of wet age-related macular degeneration (AMD), a leading cause of irreversible blindness in the modern world. The objective for this study was to investigate the therapeutic potential of known antiangiogenic agents: thalidomide, senicapoc, and sodium butyrate. Dose-dependent effect of the agents on growth of ARPE-19 cells and human umbilical vein endothelial cells (HUVECs) was investigated with cell counting assays. Half-maximal inhibitory concentrations of thalidomide (765 µM and 1520 µM), senicapoc (50 µM and 79 µM), and sodium butyrate (933 µM and 557 µM) were determined for HUVECs and ARPE-19 cells, respectively. Immunofluorescence analysis showed decrease of VEGFA expression in both ARPE-19 cells and HUVECs after treatment only with thalidomide but not with senicapoc or sodium butyrate. Efficacy of the agents was studied in vivo with laser-induced CNV in C57BL/6 mice. Thalidomide (24 µg), senicapoc (4 µg), or sodium butyrate (100 µg) was intravitreally injected the day after CNV induction. Thalidomide, senicapoc, and sodium butyrate inhibited CNV size by 56%, 24%, and 21% respectively on day 7 post-laser. Thalidomide also reduced cobalt chloride induced increase of VEGFA mRNA in ARPE-19 (-33%) and protein in culture medium (-20%). Our results suggest that thalidomide may have more therapeutic potential than senicapoc or sodium butyrate for treatment of CNV or wet AMD.


Subject(s)
Acetamides/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Butyric Acid/therapeutic use , Choroidal Neovascularization/drug therapy , Thalidomide/therapeutic use , Trityl Compounds/therapeutic use , Acetamides/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Butyric Acid/pharmacology , Cell Line , Cell Proliferation/drug effects , Choroidal Neovascularization/pathology , Disease Models, Animal , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice, Inbred C57BL , Thalidomide/pharmacology , Trityl Compounds/pharmacology
5.
Biochimie ; 168: 41-52, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31676315

ABSTRACT

Type 2 diabetes (T2D) is the most common endocrine and metabolic disorder, leading to reproductive impairments and infertility in male. Our recent study showed crucial role of adiponectin in the regulation of testicular functions, and the circulating level of adiponectin declines in diabetes. The current study thus aimed to examine the efficacy of adiponectin in improving testicular dysfunction in high-fat diet/streptozotocin-induced T2D mice. T2D was induced in pre-pubertal mice fed with high-fat diet for ∼10 weeks followed by a single dose of streptozotocin. T2D mice showed presence of increased body mass, hyperglycemia, hyperinsulinemia, insulin resistance, increased oxidative stress, and declined serum testosterone compared to vehicle-treated control mice. The spermatogenic, steroidogenic, metabolic, and antioxidative parameters were evaluated in T2D mice treated with adiponectin for both two and four weeks. The exogenous administration of adiponectin to T2D mice showed enhanced serum testosterone and expression of testicular steroidogenic markers proteins, insulin receptor and GLUT8 proteins, increase in intra-testicular concentrations of glucose and lactate and activity of LDH and antioxidant enzymes compared to the levels in untreated T2D mice. This suggests that treatment of adiponectin effectively improves testicular functions by increasing expression of insulin receptor-mediated increased transport of energy substrate (glucose and lactate) and a marked reduction in oxidative stress are the possible mechanism by which adiponectin effectively improves testicular function in T2D mice.


Subject(s)
Adiponectin/pharmacology , Antioxidants/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Testis/drug effects , Animals , Diabetes Mellitus, Experimental/pathology , Glucose/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Lactic Acid/metabolism , Male , Mice , Oxidative Stress/drug effects , Receptor, Insulin/metabolism , Testosterone/blood
6.
Graefes Arch Clin Exp Ophthalmol ; 257(5): 953-960, 2019 May.
Article in English | MEDLINE | ID: mdl-30719689

ABSTRACT

PURPOSE: Experimental autoimmune anterior uveitis (EAAU) is a clinically relevant animal model for human idiopathic anterior uveitis (IAU). The role of the immunomodulator transforming growth factor ß2 (TGF-ß2) in EAAU pathology is unknown. In this study, we investigated the regulatory role of TGF-ß2 in EAAU. METHODS: EAAU was induced in male Lewis rats by footpad injection of melanin-associated antigen (MAA). TGF-ß2 was administered intravenously (iv) in MAA-sensitized rats during the induction of EAAU, or after the clinical onset of uveitis. MAA-sensitized rats injected similarly with an equal volume of PBS served as control. Animals were examined daily between days 7 and 30 post-injection for the clinical signs of uveitis using slit lamp biomicroscopy. Animals were sacrificed at various time points and eyes were harvested for histological analysis to assess the course and severity of inflammation. For histopathological analysis, paraffin sections of harvested eyes were stained with hematoxylin and eosin. Popliteal lymph nodes (LNs) were used for CD4+CD25+FoxP3+ T regulatory (Tregs) population analysis and for CD4+ T cell proliferation assay. RESULTS: Administration of recombinant TGF-ß2 during the early stages of EAAU prevented the induction of uveitis. Compared to PBS, the presence of TGF-ß2 in the cell culture significantly (p < 0.05) inhibited the proliferation of CD4+ T cells in response to MAA. In MAA-sensitized Lewis rats, iv treatment with recombinant TGF-ß2 resulted in significantly (p < 0.05) increased percentage of Tregs compared to animals treated similarly with PBS. Thus, TGF-ß2 inhibited the induction of EAAU by inhibiting CD4+ T cell proliferation and increasing the number of Tregs. Injection of TGF-ß2 in rats with active EAAU resulted in diminished disease activity. Unfortunately, this treatment did not lead to the early resolution of EAAU. CONCLUSIONS: TGF-ß2 plays a critical role in regulation of intraocular inflammation in EAAU. Findings reported in this study improve our understanding of immunopathology of IAU and suggest that recombinant TGF-ß2 may be a promising therapeutic agent for human IAU.


Subject(s)
Autoimmune Diseases/drug therapy , Transforming Growth Factor beta2/pharmacology , Uveitis, Anterior/drug therapy , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Proliferation , Disease Models, Animal , Injections, Intraocular , Male , Rats , Rats, Inbred Lew , Recombinant Proteins , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Uveitis, Anterior/immunology , Uveitis, Anterior/pathology
7.
Gen Comp Endocrinol ; 279: 1-11, 2019 08 01.
Article in English | MEDLINE | ID: mdl-29908833

ABSTRACT

Obesity is a major health problem that is linked to decreased sperm count. It is hypothesized that an obesity-associated reduction in adiponectin secretion may be responsible for impairment of spermatogenesis. Therefore, the aim of the study was to evaluate the direct role of adiponectin in spermatogenesis and steroid synthesis in adult mice. This study showed that adiponectin receptors (AdipoR1 and AdipoR2) were localized in Leydig cells and seminiferous tubules in the testis of adult mice. The result of the in vitro study showed the direct action of adiponectin on spermatogenesis by stimulating cell proliferation (PCNA) and survival (Bcl2) and by suppressing cell apoptosis. Treatment of testis with adiponectin also enhanced transport of the energetic substrates glucose and lactate to protect cells from undergoing apoptosis. Adiponectin treatment further showed a significant reduction in oxidative stress and nitric oxide. Our findings suggest that adiponectin effectively facilitates cell survival and proliferation, as well as protects from apoptosis. Thus, adiponectin treatment may be responsible for enhancing sperm counts. Interestingly, this study showed the stimulatory effect of adiponectin in spermatogenesis but showed an inhibitory effect on testosterone and estradiol synthesis in the testes. Based on the present study, it is hypothesized that systemic adiponectin treatment may be a promising therapeutic strategy for the improvement of spermatogenesis and sperm count.


Subject(s)
Adiponectin/pharmacology , Antioxidants/metabolism , Reproduction/drug effects , Testis/enzymology , Testis/metabolism , Adenylate Kinase/metabolism , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Estradiol/metabolism , Glucose/metabolism , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Male , Mice , Models, Biological , Nitric Oxide/metabolism , Receptors, Adiponectin/metabolism , Testis/drug effects , Testosterone/metabolism
8.
Biochim Biophys Acta Mol Basis Dis ; 1865(2): 413-427, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30471430

ABSTRACT

The mechanisms by which testicular functions decline with aging remain largely speculative. Our recent finding showed the importance of adiponectin in the regulation of testicular functions, whereas its concentration declines during male infertility. Thus, the aim of present study was to explore the potential role of adiponectin during aging. The changes in adiponectin, adiponectin-receptors, and insulin receptor proteins expression in the testis were evaluated and compared with the testicular parameters, mass, and testosterone level in the mice from early post-natal to late senescence period. Further, the current study has examined the effect of exogenous adiponectin treatment on testicular functions in aged mice. The results showed a significant decline in adiponectin/adiponectin-receptors expression simultaneously with a significant decline in testicular mass, insulin receptor expression and testosterone synthesis in the testis of aged mice. Exogenous treatment of adiponectin to aged mice resulted in marked improvements in testicular mass, histological features (cells proliferation), insulin receptor expression, testicular glucose uptake, anti-oxidative enzymes activity and testosterone synthesis as compared with the control. Based on these findings, it may be concluded that a marked decline in adiponectin synthesis and action results in decreased insulin sensitivity (development of insulin resistance) and increased oxidative stress which consequently suppresses testicular functions during aging. This study further showed that treatment with adiponectin ameliorates reduced testicular functions by enhanced expression of insulin receptor in the testis of senescent mice. It is thus hypothesized that systemic adiponectin treatment could be a promising therapeutic strategy for improvement of testosterone production and sperm counts during aging.


Subject(s)
Adiponectin/metabolism , Aging/metabolism , Testis/metabolism , Animals , Animals, Newborn , Antioxidants/metabolism , Apoptosis , Biomarkers/metabolism , Blood Glucose/metabolism , Body Weight , Caspase 3/metabolism , Cell Proliferation , Cell Survival , Extracellular Signal-Regulated MAP Kinases/metabolism , Lipid Peroxidation , Male , Mice , Models, Biological , Organ Size , Phosphorylation , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptor, Insulin/metabolism , Receptors, Adiponectin/metabolism , Signal Transduction , Testis/cytology , Testosterone/blood
9.
Am J Pathol ; 186(8): 2088-2104, 2016 08.
Article in English | MEDLINE | ID: mdl-27295359

ABSTRACT

In the mouse, membrane cofactor protein (CD46), a key regulator of the alternative pathway of the complement system, is only expressed in the eye and on the inner acrosomal membrane of spermatozoa. We noted that although Cd46(-/-) mice have normal systemic alternative pathway activating ability, lack of CD46 leads to dysregulated complement activation in the eye, as evidenced by increased deposition of C5b-9 in the retinal pigment epithelium (RPE) and choroid. A knockout of CD46 induced the following cardinal features of human dry age-related macular degeneration (AMD) in 12-month-old male and female mice: accumulation of autofluorescent material in and hypertrophy of the RPE, dense deposits in and thickening of Bruch's membrane, loss of photoreceptors, cells in subretinal space, and a reduction of choroidal vessels. Collectively, our results demonstrate spontaneous age-related degenerative changes in the retina, RPE, and choroid of Cd46(-/-) mice that are consistent with human dry AMD. These findings provide the exciting possibility of using Cd46(-/-) mice as a convenient and reliable animal model for dry AMD. Having such a relatively straight-forward model for dry AMD should provide valuable insights into pathogenesis and a test model system for novel drug targets. More important, tissue-specific expression of CD46 gives the Cd46(-/-) mouse model of dry AMD a unique advantage over other mouse models using knockout strains.


Subject(s)
Disease Models, Animal , Geographic Atrophy/genetics , Macular Degeneration/genetics , Membrane Cofactor Protein/deficiency , Animals , Blotting, Western , Female , Geographic Atrophy/pathology , Macular Degeneration/pathology , Male , Membrane Cofactor Protein/genetics , Mice , Mice, Knockout
10.
Clin Exp Ophthalmol ; 44(7): 610-617, 2016 Sep.
Article in English | MEDLINE | ID: mdl-26861462

ABSTRACT

BACKGROUND: To characterize the effect of aspirin (ASA) in mouse models of choroidal neovascularization (CNV) and retinal degeneration. METHODS: In vivo: Male C57BL/6 mice were given ASA in food or regular rodent diet. CNV was induced by argon laser photocoagulation. Subretinal injections of polyethylene glycol 400 (PEG-400) were administered to induce retinal degeneration. CNV size, laser spot area and mean intensity of VEGF in the laser injured zones were measured. In the PEG injected eyes the thickness of retinal pigment epithelium (RPE) and choroid was measured. In vitro: Human ARPE-19 cells were treated with 0.5 or 2.0 mM/L of ASA for 72 h. ELISA was used to measure the concentration of VEGF and CCL-2 in the supernatants. Additionally, damaged RPE monolayer was treated with ASA (0.5 or 2.0 mM/L) and vehicle separately. Size of damaged area was measured. ELISA was used to measure secretion of VEGF-A and CCL-2 by damaged cells after 24 h. RESULTS: No statistically significant effect of ASA on CNV size, laser spot size or VEGF expression was noted in CNV model. In the PEG model, ASA did not have any effect on RPE and choroid thickness; however, a significant increase in RPE atrophy was observed (P = 0.02 + 38%). In addition, ASA had a significant effect on the ability of the RPE cells to regenerate and become confluent after mechanical damage. CONCLUSIONS: ASA at doses consumed clinically for various medical causes may not worsen CNV in human subjects. However, ASA may increase RPE atrophy when consumed over long periods of time.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , Choroidal Neovascularization/drug therapy , Disease Models, Animal , Retinal Degeneration/drug therapy , Retinal Pigment Epithelium/drug effects , Animals , Cell Line , Chemokine CCL2/metabolism , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Diet , Enzyme-Linked Immunosorbent Assay , Humans , Male , Mice , Mice, Inbred C57BL , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects
11.
Mol Immunol ; 63(2): 176-83, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25074023

ABSTRACT

Studies performed over the past decade in humans and experimental animals have been a major source of information and improved our understanding of how dysregulation of the complement system contributes to age-related macular degeneration (AMD) pathology. Drusen, the hall-mark of dry-type AMD are reported to be the by-product of complement mediated inflammatory processes. In wet AMD, unregulated complement activation results in increased production of angiogenic growth factors leading to choroidal neovascularization both in humans and in animal models. In this review article we have linked the complement system with modifiable and non-modifiable AMD risk factors as well as with prediction models of AMD. Understanding the association between the complement system, risk factors and prediction models will help improve our understanding of AMD pathology and management of this disease.


Subject(s)
Complement System Proteins/immunology , Macular Degeneration/immunology , Models, Biological , Animals , Disease Models, Animal , Humans , Risk Factors
12.
Exp Eye Res ; 127: 143-52, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25088354

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of irreversible blindness. This study was done to characterize dry AMD-like changes in mouse retinal pigment epithelium (RPE) and retina after polyethylene glycol (PEG) treatment. We injected male C57BL/6 mice subretinally with PBS, 0.025, 0.25, 0.5 and 1.0 mg of PEG-400 and the animals were sacrificed on day 5. Eyes were harvested and processed for histological analysis. In all other experiments 0.5 mg PEG was injected and animals were sacrificed on days 1, 3, 5 or 14. Paraffin, 5 µm and plastic, 1 µm and 80 nm sections were used for further analysis. Subretinal injection of 0.5 mg PEG induced a 32% reduction of outer nuclear layer (ONL) thickness, 61% decrease of photoreceptor outer and inner segment length, 49% decrease of nuclear density in the ONL and 31% increase of RPE cell density by day 5 after injection. The maximum level of TUNEL positive nuclei in the ONL (6.8 + 1.99%) was detected at day 5 after PEG injection and co-localized with Casp3act. Histological signs of apoptosis were observed in the ONL by light or electron microscopy. Degeneration of RPE cells was found in PEG injected eyes. Gene expression data identified several genes reported to be involved in human AMD. C3, Cfi, Serping1, Mmp9, Htra1 and Lpl were up-regulated in PEG injected eyes compared to PBS controls. PEG leads to morphological and gene expression changes in RPE and retina consistent with dry AMD. This model will be useful to investigate dry AMD pathogenesis and treatment.


Subject(s)
Disease Models, Animal , Geographic Atrophy/pathology , Photoreceptor Cells, Vertebrate/pathology , Polyethylene Glycols/toxicity , Retinal Pigment Epithelium/pathology , Aminopeptidases/genetics , Animals , Apoptosis , Autophagy , Complement C1 Inhibitor Protein/genetics , Complement C3/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/physiology , Geographic Atrophy/chemically induced , Geographic Atrophy/genetics , High-Temperature Requirement A Serine Peptidase 1 , In Situ Nick-End Labeling , Injections, Intraocular , Male , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Serine Endopeptidases/genetics , Time Factors
13.
Am J Pathol ; 184(9): 2537-48, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25019227

ABSTRACT

Dysregulation of the complement system is increasingly recognized as a contributing factor in age-related macular degeneration. Although the complement regulator CD46 is expressed ubiquitously in humans, in mouse it was previously thought to be expressed only on spermatozoa. We detected CD46 mRNA and protein in the posterior ocular segment (neuronal retina, retinal pigment epithelium, and choroid) of wild-type (WT) C57BL/6J mice. Cd46(-/-) knockout mice exhibited increased levels of the membrane attack complex and of vascular endothelial growth factor (VEGF) in the retina and choroid. The Cd46(-/-) mice were also more susceptible to laser-induced choroidal neovascularization (CNV). In Cd46(-/-) mice, 19% of laser spots were positive for CNV at day 2 after treatment, but no positive spots were detected in WT mice. At day 3, 42% of laser spots were positive in Cd46(-/-) mice, but only 11% in WT mice. A fully developed CNV complex was noted in both Cd46(-/-) and WT mice at day 7; however, lesion size was significantly (P < 0.05) increased in Cd46(-/-) mice. Our findings provide evidence for expression of CD46 in the mouse eye and a role for CD46 in protection against laser-induced CNV. We propose that the Cd46(-/-) mouse has a greater susceptibility to experimental CNV because of insufficient complement inhibition, which leads to increased membrane attack complex deposition and VEGF expression.


Subject(s)
Choroidal Neovascularization/metabolism , Membrane Cofactor Protein/metabolism , Animals , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
14.
PLoS One ; 7(10): e47227, 2012.
Article in English | MEDLINE | ID: mdl-23077573

ABSTRACT

The Plasmodium falciparum cysteine proteases falcipain-2 and falcipain-3 are major hemoglobinases and potential antimalarial drug targets. Our previous studies demonstrated that these enzymes are equipped with specific domains for specific functions. Structural and functional analysis of falcipains showed that they have unique domains including a refolding domain and a hemoglobin binding domain. As with many proteases, falcipain-2 and falcipain-3 are synthesized as inactive zymogens. However, it is not known how these enzymes get activated for hemoglobin hydrolysis. In this study, we are presenting the first evidence that salt bridges and hydrophobic interactions are required for the auto activation of cysteine proteases of P.falciparum. To investigate the mechanism of activation of these enzymes, we expressed the wild type protein as well as different mutants in E.coli. Refolding was assessed by circular dichroism. Both CD and trans activation data showed that the wild type enzymes and mutants are rich in secondary structures with similar folds. Our study revealed that prodomain-mature domain of falcipain-2 and falcipain-3 interacts via salt bridges and hydrophobic interactions. We mutated specific residues of falcipain-2 and falcipain-3, and evaluated their ability to undergo auto processing. Mutagenesis result showed that two salt bridges (Arg¹85- Glu²²¹, Glu²¹°- Lys4°³) in falcipain-2, and one salt bridge (Arg²°²-Glu²³8) in falcipain-3, play crucial roles in the activation of these enzymes. Further study revealed that hydrophobic interactions present both in falcipain-2 (Phe²¹4 Trp449 Trp45³) and falcipain-3 (Phe²³¹ Trp457 Trp46¹) also play important roles in the activation of these enzymes. Our results revealed the interactions involved in auto processing of two major hemoglobinases of malaria parasite.


Subject(s)
Cysteine Endopeptidases/genetics , Enzyme Activation/genetics , Malaria, Falciparum/enzymology , Plasmodium falciparum/enzymology , Amino Acid Sequence , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Hemoglobins/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Malaria, Falciparum/parasitology , Protein Binding , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Structure-Activity Relationship
15.
Biochim Biophys Acta ; 1823(8): 1264-72, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22633972

ABSTRACT

Age-related macular degeneration (AMD) is a leading cause of central blindness in the elderly population. The wet type of AMD is characterized by extensive growth of new vessels. One of the effective strategies to treat wet AMD is to limit the choroidal neovascularization (CNV). We studied the effects of adiponectin peptide I (APNpI) on new vessel growth in laser-induced rat model of wet AMD and on rat choroidal endothelial cell (CEC) culture. CNV size and vessel density were investigated by microscopy. Immunohistochemical staining (IHC) for von Willebrand Factor (vWF), APN, APN receptors 1 (AdipoR1), 2 (AdipoR2), VEGF, VEGF receptor 2 (VEGF-R2), proliferating cell nuclear antigen (PCNA) was performed in CNV area. The mRNA expression of VEGF and VEGF-R2 in RPE-choroid was investigated by RT-PCR and real-time PCR. APNpI inhibited area of CNV by 4 fold, number of vWF positive vessels by 99% and area of subretinal tissue by 40%. The expression of VEGF and VEGF-R2 at mRNA and protein levels decreased after APNpI treatment in vivo. Proliferative index (PCNA) was 5 folds less in laser spots of APNpI treated rats compared to controls. In conclusion, APNpI inhibited formation of new vessels in rat model of CNV by decreasing VEGF, VEGF-R2 expression and cell proliferation. Thus, APNpI may have potential therapeutic use for AMD treatment since it significantly inhibited CNV.


Subject(s)
Adiponectin/pharmacology , Choroid/blood supply , Choroidal Neovascularization/prevention & control , Peptide Fragments/pharmacology , Wet Macular Degeneration/drug therapy , Adiponectin/therapeutic use , Animals , Cell Proliferation/drug effects , Cells, Cultured , Choroid/drug effects , Choroid/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/physiology , Gene Expression/drug effects , Male , Peptide Fragments/therapeutic use , Proliferating Cell Nuclear Antigen/metabolism , Rats, Inbred BN , Receptors, Adiponectin/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Retina ; 32(6): 1171-80, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22088983

ABSTRACT

PURPOSE: The purpose of this article is to evaluate the effect of nicotine on anti-vascular endothelial growth factor therapy in the treatment of neovascular age-related macular degeneration. METHODS: One group of mice received nicotine in drinking water and the other group received water only. Choroidal neovascularization (CNV) was induced with a laser. Nicotinic acetylcholine receptor-α7 (nAChRα7) expression was evaluated by immunohistochemistry. Bevacizumab or adiponectin peptide II (APNpII) was injected intravitreally on Day 7 postlaser, and the effects were evaluated on Days 14 and 21. α-Bungarotoxin was injected intraperitoneally on Days 2 to 5, and its effect was evaluated on Day 14. RESULTS: Expression of nAChRα7 was 2 to 7 times higher between Days 3 and 7 postlaser compared with naive mice. In water-fed mice, APNpII, bevacizumab, and α-bungarotoxin significantly reduced CNV size. In nicotine-fed mice, treatment with APNpII or bevacizumab did not significantly reduce CNV size, whereas α-bungarotoxin did have an effect. Comparing water- and nicotine-fed mice, CNV size was 61% to 86% smaller in water-fed mice except for the α-bungarotoxin group, where there was no difference. Platelet-derived growth factor and vascular endothelial growth factor expression was 1.5- to 2.5-fold higher at Day 14 in nicotine-treated mice. CONCLUSION: Nicotine significantly blocks the effect of anti-vascular endothelial growth factor therapy in the treatment of laser-induced neovascular age-related macular degeneration. nAChRα7 is significantly upregulated during the formation of CNV, and treatment with an nAChRα7 antagonist decreases CNV size irrespective of nicotine administration.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Choroidal Neovascularization/drug therapy , Nicotine/pharmacology , Vascular Endothelial Growth Factors/antagonists & inhibitors , Adiponectin/therapeutic use , Analysis of Variance , Animals , Bevacizumab , Bungarotoxins/pharmacology , Choroid/metabolism , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Immunohistochemistry , Intravitreal Injections , Male , Mice , Mice, Inbred C57BL , Receptors, Nicotinic/metabolism , Retinal Pigment Epithelium/metabolism , Vascular Endothelial Growth Factors/metabolism , alpha7 Nicotinic Acetylcholine Receptor
17.
Curr Eye Res ; 36(9): 787-96, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21812661

ABSTRACT

PURPOSE: To investigate the effect of topically applied honey on intact corneas, surgically induced corneal abrasions and endotoxin induced keratitis. MATERIALS AND METHODS: The effect of honey on the cornea was investigated by application of honey on intact corneas, wounded corneas and endotoxin-induced keratitis in Lewis rats. The corneas were wounded by creating an epithelial defect using a surgical blade, and the keratitis was induced by topically applying Pseudomonas aeruginosa endotoxin to scarified corneas. After treatment rats were sacrificed and cornea harvested in each case. Corneas were processed for paraffin embedding for histological and immuno-fluorescence staining. Corneas were also harvested and processed for total ribonucleic acid (RNA) isolation for reverse transcriptase-polymerase chain reaction (RT-PCR) analysis for various growth factors and inflammatory chemokines/cytokines). RESULTS: Histological analysis revealed that no inflammation or morphological changes occurred after honey treatment in naive intact corneas. Vascular endothelial growth factor (VEGF) levels were also not altered after honey treatment. Topical application of honey to injured corneas resulted in faster epithelial healing and decreased expression of VEGF, transforming growth factor beta (TGF-ß), interferon gamma (IFN-γ), interleukin 12 (IL-12) and tumor necrosis factor alpha (TNF-α) in injured corneas. Our results also established that honey treatment reduced the inflammation in endotoxin-induced keratitis by reducing the levels of angiogenic factors (VEGF and TGF-ß), inflammatory cytokines (IL-12) and chemokines (CC chemokine receptor 5(CCR-5)). CONCLUSION: Short term use of honey on intact corneas can be safe. Honey has anti-angiogenic and anti-inflammatory properties that can be explored in several corneal inflammatory and infectious conditions.


Subject(s)
Corneal Injuries , Eye Infections, Bacterial/therapy , Eye Injuries/therapy , Honey , Keratitis/therapy , Pseudomonas Infections/therapy , Wound Healing , Administration, Topical , Animals , Cornea/metabolism , Cornea/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Endotoxins/toxicity , Eye Infections, Bacterial/chemically induced , Eye Infections, Bacterial/pathology , Eye Injuries/pathology , Follow-Up Studies , Keratitis/chemically induced , Keratitis/pathology , Male , Pseudomonas Infections/pathology , Pseudomonas aeruginosa , RNA/analysis , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , Treatment Outcome
18.
J Biol Chem ; 286(18): 16229-37, 2011 May 06.
Article in English | MEDLINE | ID: mdl-21454496

ABSTRACT

In this study, we describe a new method for inducing choroidal neovascularization (CNV) in C57BL/6 mice, an animal model of wet age-related macular degeneration (AMD). AMD is a disease that causes central blindness in humans. We injected PEG-8 subretinally in different doses (0.125-2 mg) to induce CNV. After PEG-8 injection, we examined CNV at several time points (days 3-42). We also used Western blotting, immunohistochemistry, and ELISA to examine the complement component C3 split products, C9, VEGF, TGF-ß2, and basic FGF. As early as day 1 after treatment, we found that a single subretinal injection of 1 mg of PEG-8 increased the C3 split products and the C9, TGF-ß2, and basic FGF levels in the retinal pigment epithelium-choroid tissue. By day 3 after PEG-8 injection, the intraocular activation of the complement system caused induction and progression of CNV, including new vessels penetrating the Bruch's membrane. At day 5 after PEG-8 injection, we observed a fully developed CNV and retinal degeneration. Thus, in this study, we present a new, inexpensive, and accelerated mouse model of CNV that may be useful to study AMD.


Subject(s)
Choroidal Neovascularization/chemically induced , Disease Models, Animal , Drug Carriers/adverse effects , Macular Degeneration , Polyethylene Glycols/adverse effects , Animals , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Complement C3/metabolism , Complement C9/metabolism , Drug Carriers/pharmacology , Mice , Polyethylene Glycols/pharmacology , Time Factors , Transforming Growth Factor beta2/metabolism , Vascular Endothelial Growth Factor A/metabolism
19.
J Biol Chem ; 286(23): 20991-1001, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21515678

ABSTRACT

The present study investigated the interactions among the complement membrane attack complex (MAC), CCL2, and VEGF that occur in vivo during the development of choroidal neovascularization (CNV). We first investigated the sequential expression of MAC, CCL2, and VEGF during laser-induced CNV in C57BL/6 mice. Increased MAC deposition was detected at 1 h, CCL2 increased at 3 h, and VEGF was up-regulated at day 3 post-laser treatment. These results suggested that during laser-induced CNV, MAC, CCL2 and VEGF are formed and/or expressed in the following order: MAC → CCL2 → VEGF. To determine the cross-talk between MAC, CCL2, and VEGF during laser-induced CNV, neutralizing antibodies were injected both systemically and locally to block the bioactivity of each molecule. Blocking MAC formation inhibited CCL2 and VEGF expression and also limited CNV formation, whereas neutralization of CCL2 bioactivity did not affect MAC deposition; however, it reduced VEGF expression and CNV formation. When bioactivity of VEGF was blocked, CNV formation was significantly inhibited, but MAC deposition was not affected. Together, our results demonstrate that MAC is an upstream mediator and effect of MAC on the development of laser-induced CNV can be attributed to its direct effect on VEGF as well as its effect on VEGF that is mediated by CCL2. Understanding the interplay between immune mediators is critical to gain insight into the pathogenesis of CNV.


Subject(s)
Chemokine CCL2/biosynthesis , Choroidal Neovascularization/metabolism , Complement Membrane Attack Complex/metabolism , Gene Expression Regulation , Lasers/adverse effects , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Antibodies, Neutralizing/pharmacology , Choroidal Neovascularization/etiology , Choroidal Neovascularization/pathology , Disease Models, Animal , Mice , Time Factors
20.
J Biol Chem ; 286(10): 8472-8480, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21216963

ABSTRACT

The objective of the current study was to delineate the pathway of complement activation that is crucial for the induction of experimental autoimmune anterior uveitis (EAAU). We studied the development of EAAU in melanin-associated antigen (MAA)-sensitized Lewis rats treated with antibody against C4 or factor B. Control animals received isotype IgG control. Antibody against C4 had no effect on EAAU, and all of the animals developed EAAU similar to those injected with control IgG. In contrast, EAAU was completely inhibited in all MAA-sensitized Lewis rats injected with factor B antibody. Treatment with anti-factor B antibody resulted in suppression of ocular complement activation. Adoptive transfer of T lymphocytes harvested from draining lymph nodes of donor animals treated with anti-factor B did not transfer EAAU to naïve syngenic rats. Anti-factor B antibody inhibited the ability of MAA-specific CD4(+) T cells to proliferate (in vitro) in response to MAA in a dose-dependent manner. Level of TNF-α and IFN-γ decreased in the presence of anti-factor B. Collectively, our results provide the novel finding that complement activation via the alternative pathway contributes to intraocular inflammation in EAAU, and anti-factor B-mediated inhibition of EAAU is due to diminished antigen-specific CD4(+) T cell responses to MAA. Our findings explain the interactions between the complement system and T cells that are critical for the induction of EAAU and may lead to the development of therapy for idiopathic anterior uveitis based on selective blockade of the alternative pathway.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Complement C4/immunology , Complement Factor B/immunology , Complement Pathway, Alternative/immunology , Uveitis, Anterior/immunology , Adoptive Transfer , Animals , Autoantibodies/immunology , Autoantibodies/pharmacology , Autoimmune Diseases/chemically induced , Complement Pathway, Alternative/drug effects , Disease Models, Animal , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Male , Melanins/adverse effects , Melanins/immunology , Melanins/pharmacology , Rats , Rats, Inbred Lew , Uveitis, Anterior/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...