Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Molecules ; 27(3)2022 Jan 20.
Article in English | MEDLINE | ID: mdl-35163920

ABSTRACT

During liver fibrogenesis, there is an imbalance between regeneration and wound healing. The current treatment is the withdrawal of the causing agent; thus, investigation of new and effective treatments is important. Studies have highlighted the action of chondroitin sulfate (CS) in different cells; thus, our aim was to analyze its effect on an experimental model of bile duct ligation (BDL). Adult Wistar rats were subjected to BDL and treated with CS for 7, 14, 21, or 28 days intraperitoneally. We performed histomorphometric analyses on Picrosirius-stained liver sections. Cell death was analyzed according to caspase-3 and cathepsin B activity and using a TUNEL assay. Regeneration was evaluated using PCNA immunohistochemistry. BDL led to increased collagen content with corresponding decreased liver parenchyma. CS treatment reduced total collagen and increased parenchyma content after 21 and 28 days. The treatment also promoted changes in the hepatic collagen type III/I ratio. Furthermore, it was observed that CS treatment reduced caspase-3 activity and the percentage of TUNEL-positive cells after 14 days and cathepsin B activity only after 28 days. The regeneration increased after 14, 21, and 28 days of CS treatment. In conclusion, our study showed a promising hepatoprotective action of CS in fibrogenesis induced by BDL.


Subject(s)
Cholestasis/complications , Chondroitin Sulfates/pharmacology , Common Bile Duct/surgery , Liver Diseases/drug therapy , Animals , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/pathology , Male , Protective Agents/pharmacology , Rats , Rats, Wistar
2.
Malar J ; 18(1): 213, 2019 Jun 24.
Article in English | MEDLINE | ID: mdl-31234939

ABSTRACT

BACKGROUND: Malaria represents a worldwide medical emergency affecting mainly poor areas. Plasmodium parasites during blood stages can release kinins to the extracellular space after internalization of host kininogen inside erythrocytes and these released peptides could represent an important mechanism in liver pathophysiology by activation of calcium signaling pathway in endothelial cells of vertebrate host. Receptors (B1 and B2) activated by kinins peptides are important elements for the control of haemodynamics in liver and its physiology. The aim of this study was to identify changes in the liver host responses (i.e. kinin receptors expression and localization) and the effect of ACE inhibition during malaria infection using a murine model. METHODS: Balb/C mice infected by Plasmodium chabaudi were treated with captopril, an angiotensin I-converting enzyme (ACE) inhibitor, used alone or in association with the anti-malarial chloroquine in order to study the effect of ACE inhibition on mice survival and the activation of liver responses involving B1R and B2R signaling pathways. The kinin receptors (B1R and B2R) expression and localization was analysed in liver by western blotting and immunolocalization in different conditions. RESULTS: It was verified that captopril treatment caused host death during the peak of malaria infection (parasitaemia about 45%). B1R expression was stimulated in endothelial cells of sinusoids and other blood vessels of mice liver infected by P. chabaudi. At the same time, it was also demonstrated that B1R knockout mice infected presented a significant reduction of survival. However, the infection did not alter the B2R levels and localization in liver blood vessels. CONCLUSIONS: Thus, it was observed through in vivo studies that the vasodilation induced by plasma ACE inhibition increases mice mortality during P. chabaudi infection. Besides, it was also seen that the anti-malarial chloroquine causes changes in B1R expression in liver, even after days of parasite clearance. The differential expression of B1R and B2R in liver during malaria infection may have an important role in the disease pathophysiology and represents an issue for clinical treatments.


Subject(s)
Gene Expression Regulation , Liver/physiopathology , Malaria/physiopathology , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B2/genetics , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Captopril/pharmacology , Chloroquine/pharmacology , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred BALB C , Plasmodium chabaudi , Receptor, Bradykinin B1/metabolism , Receptor, Bradykinin B2/metabolism
3.
Bioorg Med Chem ; 25(17): 4628-4636, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28728898

ABSTRACT

Malaria is a global human parasitic disease mainly caused by the protozoon Plasmodium falciparum. Increased parasite resistance to current drugs determines the relevance of finding new treatments against new targets. A novel target is the M1 alanyl-aminopeptidase from P. falciparum (PfA-M1), which is essential for parasite development in human erythrocytes and is inhibited by the pseudo-peptide bestatin. In this work, we used a combinatorial multicomponent approach to produce a library of peptidomimetics and screened it for the inhibition of recombinant PfA-M1 (rPfA-M1) and the in vitro growth of P. falciparum erythrocytic stages (3D7 and FcB1 strains). Dose-response studies with selected compounds allowed identifying the bestatin-based peptidomimetic KBE009 as a submicromolar rPfA-M1 inhibitor (Ki=0.4µM) and an in vitro antimalarial compound as potent as bestatin (IC50=18µM; without promoting erythrocyte lysis). At therapeutic-relevant concentrations, KBE009 is selective for rPfA-M1 over porcine APN (a model of these enzymes from mammals), and is not cytotoxic against HUVEC cells. Docking simulations indicate that this compound binds PfA-M1 without Zn2+ coordination, establishing mainly hydrophobic interactions and showing a remarkable shape complementarity with the active site of the enzyme. Moreover, KBE009 inhibits the M1-type aminopeptidase activity (Ala-7-amido-4-methylcoumarin substrate) in isolated live parasites with a potency similar to that of the antimalarial activity (IC50=82µM), strongly suggesting that the antimalarial effect is directly related to the inhibition of the endogenous PfA-M1. These results support the value of this multicomponent strategy to identify PfA-M1 inhibitors, and make KBE009 a promising hit for drug development against malaria.


Subject(s)
Antimalarials/chemistry , CD13 Antigens/antagonists & inhibitors , Dipeptides/chemistry , Plasmodium falciparum/enzymology , Protozoan Proteins/antagonists & inhibitors , Antimalarials/chemical synthesis , Antimalarials/pharmacology , Binding Sites , CD13 Antigens/genetics , CD13 Antigens/metabolism , Catalytic Domain , Cell Survival/drug effects , Dipeptides/chemical synthesis , Dipeptides/pharmacology , Erythrocytes/cytology , Erythrocytes/drug effects , Erythrocytes/parasitology , Human Umbilical Vein Endothelial Cells , Humans , Leucine/analogs & derivatives , Leucine/chemistry , Leucine/pharmacology , Molecular Docking Simulation , Peptidomimetics , Plasmodium falciparum/drug effects , Protozoan Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Structure-Activity Relationship
4.
Anal Biochem ; 468: 22-7, 2015 01 01.
Article in English | MEDLINE | ID: mdl-25281458

ABSTRACT

In the intraerythrocytic trophozoite stages of Plasmodium falciparum, the calcium-dependent cysteine protease calpain (Pf-calpain) has an important role in the parasite calcium modulation and cell development. We established specific conditions to follow by confocal microscopy and spectrofluorimetry measurements the intracellular activity of Pf-calpain in live cells. The catalytic activity was measured using the fluorogenic Z-Phe-Arg-MCA (where Z is carbobenzoxy and MCA is 4-methylcoumaryl-7-amide). The calmodulin inhibitor calmidazolium and the sarcoplasmic reticulum calcium ATPase inhibitor thapsigargin were used for modifications in the cytosolic calcium concentrations that persisted in the absence of extracellular calcium. The observed calcium-dependent peptidase activity was greatly inhibited by specific cysteine protease inhibitor E-64 and by the selective calpain inhibitor ALLN (N-acetyl-l-leucyl-l-leucyl-l-norleucinal). Taken together, we observed that intracellular Pf-calpain can be selectively detected and is the main calcium-dependent protease in the intraerythrocytic stages of the parasite. The method described here can be helpful in cell metabolism studies and antimalarial drug screening.


Subject(s)
Calpain/metabolism , Plasmodium chabaudi/enzymology , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Animals , Calcium/metabolism , Calpain/analysis , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Male , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Protozoan Proteins/analysis , Protozoan Proteins/antagonists & inhibitors , Spectrometry, Fluorescence
5.
Malar J ; 14: 183, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25927919

ABSTRACT

BACKGROUND: Parasitic diseases like malaria are a major public health problem in many countries and disrupted sleep patterns are an increasingly common part of modern life. The aim of this study was to assess the effects of paradoxical sleep deprivation (PSD) and sleep rebound (RB) on malarial parasite infection in mice. METHODS: After PSD, one group was immediately infected with parasites (PSD). The two other PSD rebound groups were allowed to sleep normally for either 24 h (24 h RB) or 48 h (48 h RB). After the recovery periods, mice were inoculated with parasites. RESULTS: The PSD group was the most affected by parasites presenting the higher death rate (0.02), higher number of infected cells (p < 0.01), and decrease in body weight (p < 0.04) compared to control and 48 h RB groups. The 24 h RB group was also different from control group in survival (p < 0.03), number of infected cells (p < 0.05) and body weight (p < 0.04). After 48 hours of sleep rebound animals were allowed to restore their response to parasitic infection similar to normal sleep animals. CONCLUSIONS: These results suggest that PSD is damaging to the immune system and leads to an increased infection severity of malaria parasites; only 48 hours of recovery sleep was sufficient to return the mice infection response to baseline values.


Subject(s)
Immunity, Innate , Malaria/complications , Malaria/immunology , Plasmodium chabaudi/physiology , Sleep Deprivation/complications , Sleep, REM , Animals , Longevity , Malaria/mortality , Malaria/parasitology , Male , Mice
6.
Biochim Biophys Acta ; 1820(12): 1997-2006, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23000491

ABSTRACT

BACKGROUND: Sleep is a physiological event that directly influences health by affecting the immune system, in which calcium (Ca(2+)) plays a critical signaling role. We performed live cell measurements of cytosolic Ca(2+) mobilization to understand the changes in Ca(2+) signaling that occur in splenic immune cells after various periods of sleep deprivation (SD). METHODS: Adult male mice were subjected to sleep deprivation by platform technique for different periods (from 12 to 72h) and Ca(2+) intracellular fluctuations were evaluated in splenocytes by confocal microscopy. We also performed spleen cell evaluation by flow cytometry and analyzed intracellular Ca(2+) mobilization in endoplasmic reticulum and mitochondria. Additionally, Ca(2+) channel gene expression was evaluated RESULTS: Splenocytes showed a progressive loss of intracellular Ca(2+) maintenance from endoplasmic reticulum (ER) stores. Transient Ca(2+) buffering by the mitochondria was further compromised. These findings were confirmed by changes in mitochondrial integrity and in the performance of the store operated calcium entry (SOCE) and stromal interaction molecule 1 (STIM1) Ca(2+) channels. CONCLUSIONS AND GENERAL SIGNIFICANCE: These novel data suggest that SD impairs Ca(2+) signaling, most likely as a result of ER stress, leading to an insufficient Ca(2+) supply for signaling events. Our results support the previously described immunosuppressive effects of sleep loss and provide additional information on the cellular and molecular mechanisms involved in sleep function.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Mitochondria/immunology , Mitochondria/metabolism , Sleep Deprivation/immunology , Spleen/cytology , Animals , Endoplasmic Reticulum/metabolism , Lysosomes/metabolism , Male , Membrane Potential, Mitochondrial , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Sleep Deprivation/metabolism , Sleep Deprivation/pathology , Spleen/immunology , Spleen/metabolism , Stromal Interaction Molecule 1
7.
Exp Parasitol ; 135(1): 166-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23830988

ABSTRACT

Malaria cysteine proteases have been shown to be immunogenic and are being exploited as serodiagnostic markers, drug and vaccine targets. Several Plasmodium spp. cysteine proteases have been described and the best characterized of these are the falcipains, a family of papain-family enzymes. Falcipain-2 and falcipain-3 act in concert with other proteases to hydrolyze host erythrocyte hemoglobin in the parasite food vacuole. Falcipain-1 has less similarity to the other falcipains and its physiological role in parasite asexual blood stage still remains uncertain. Immunolocalization studies using an antibody developed against the Plasmodium chabaudi recombinant chabaupain-1, the falcipain-1 ortholog, were performed confirming its cellular localization in both erythrocyte and mosquito ookinete stage. Immunostaining of chabaupain-1 preferentially in apical portion of parasite ookinete suggests that this protease may be related with parasite egression from mosquito midgut. Immune responses to chabaupain-1 were evaluated using two different adjuvants, chitosan nanoparticles and hydroxide aluminum. Mice immunized with the recombinant protein alone or in association with nanoparticles were challenged with P. chabaudi showing that immunization with the recombinant protein confers partial protection to blood stage infection in BALB/c animal model.


Subject(s)
Antibodies, Protozoan/biosynthesis , Cysteine Proteases/immunology , Malaria Vaccines , Malaria/prevention & control , Plasmodium chabaudi/enzymology , Plasmodium chabaudi/immunology , Animals , Anopheles/parasitology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Cysteine Proteases/analysis , Cysteine Proteases/genetics , Cytokines/metabolism , Disease Models, Animal , Erythrocytes/parasitology , Female , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Plasmodium berghei/physiology , Plasmodium chabaudi/growth & development , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Vaccines, Synthetic
8.
Malar J ; 11: 156, 2012 May 07.
Article in English | MEDLINE | ID: mdl-22564457

ABSTRACT

BACKGROUND: The malaria burden remains a major public health concern, especially in sub-Saharan Africa. The complex biology of Plasmodium, the apicomplexan parasite responsible for this disease, challenges efforts to develop new strategies to control the disease. Proteolysis is a fundamental process in the metabolism of malaria parasites, but roles for proteases in generating vasoactive peptides have not previously been explored. RESULTS: In the present work, it was demonstrated by mass spectrometry analysis that Plasmodium parasites (Plasmodium chabaudi and Plasmodium falciparum) internalize and process plasma kininogen, thereby releasing vasoactive kinins (Lys-BK, BK and des-Arg9-BK) that may mediate haemodynamic alterations during acute malaria. In addition, it was demonstrated that the P. falciparum cysteine proteases falcipain-2 and falcipain-3 generated kinins after incubation with human kininogen, suggesting that these enzymes have an important role in this process. The biologic activity of peptides released by Plasmodium parasites was observed by measuring ileum contraction and activation of kinin receptors (B1 and B2) in HUVEC cells; the peptides elicited an increase in intracellular calcium, measured by Fluo-3 AM fluorescence. This effect was suppressed by the specific receptor antagonists Des-Arg9[Leu8]-BK and HOE-140. CONCLUSIONS: In previously undescribed means of modulating host physiology, it was demonstrated that malaria parasites can generate active kinins by proteolysis of plasma kininogen.


Subject(s)
Cysteine Endopeptidases/metabolism , Kininogens/metabolism , Kinins/metabolism , Plasmodium chabaudi/enzymology , Plasmodium falciparum/enzymology , Animals , Calcium/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Guinea Pigs , Humans , Ileum/drug effects , Mass Spectrometry , Muscle Contraction/drug effects , Plasmodium chabaudi/metabolism , Plasmodium falciparum/metabolism , Proteolysis
9.
Ann Anat ; 241: 151891, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35114378

ABSTRACT

Cell-to-cell interactions mediated by intercellular junctions (IJs) are crucial for beta-cell functioning and proper insulin secretion, however, their role in type-2 diabetes is still unclear. This work aimed to evaluate the cellular distribution and expression of proteins associated with adherens (AJs) and gap junctions (GJs) in pancreatic islets of C57BL6 mice fed a high-fat (HF) diet. The administration of HF diet for 30 days induced an increase in body weight, post-prandial glycemia, insulinemia, glucose intolerance, and moderate insulin resistance associated with mild perturbations in insulin secretion. The intercellular content of the AJ-associated proteins (namely, E-, N-cadherins, and α-, ß-catenins) was significantly higher in islet cells of HF-fed mice. Inversely, the gap junctional content of Cx36 was significantly decreased, as revealed by immunofluorescence, which was paralleled by a reduction in the frequency of calcium oscillations in islets of prediabetic mice. In conclusion, the endocrine pancreas displays significant changes in the content of several junctional proteins at the cell-cell contact region following short-term HF diet administration, indicating that IJs may be involved in the adaptive response of beta cells seen during this state.


Subject(s)
Insulin-Secreting Cells , Islets of Langerhans , Animals , Cell Adhesion Molecules/metabolism , Diet, High-Fat/adverse effects , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL
10.
Pharmaceuticals (Basel) ; 15(7)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35890113

ABSTRACT

Malaria is a parasitic disease caused by protozoan parasites from the genus Plasmodium. Plasmodium falciparum is the most prevalent species worldwide and the causative agent of severe malaria. The spread of resistance to the currently available antimalarial therapy is a major concern. Therefore, it is imperative to discover and develop new antimalarial drugs, which not only treat the disease but also control the emerging resistance. Brussonol is an icetexane derivative and a member of a family of diterpenoids that have been isolated from several terrestrial plants. Here, the synthesis and antiplasmodial profiling of a series of brussonol derivatives are reported. The compounds showed inhibitory activities in the low micromolar range against a panel of sensitive and resistant P. falciparum strains (IC50s = 5-16 µM). Moreover, brussonol showed fast-acting in vitro inhibition and an additive inhibitory behavior when combined with the antimalarial artesunate (FICindex~1). The mode of action investigation indicated that brussonol increased the cytosolic calcium levels within the parasite. Hence, the discovery of brussonol as a new scaffold endowed with antiplasmodial activity will enable us to design derivatives with improved properties to deliver new lead candidates for malaria.

11.
Int J Parasitol Drugs Drug Resist ; 20: 121-128, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36375339

ABSTRACT

Malaria is among the tropical diseases that cause the most deaths in Africa. Around 500,000 malaria deaths are reported yearly among African children under the age of five. Chloroquine (CQ) is a low-cost antimalarial used worldwide for the treatment of Plasmodium vivax malaria. Due to resistance mechanisms, CQ is no longer effective against most malaria cases caused by P. falciparum. The World Health Organization recommends artemisinin combination therapies for P. falciparum malaria, but resistance is emerging in Southeast Asia and some parts of Africa. Therefore, new medicines for treating malaria are urgently needed. Previously, our group identified the 4-aminoquinoline DAQ, a CQ analog containing an acetylenic bond in its side chain, which overcomes CQ resistance in K1 P. falciparum strains. In this work, the antiplasmodial profile, drug-like properties, and pharmacokinetics of DAQ were further investigated. DAQ showed no cross-resistance against standard CQ-resistant strains (e.g., Dd2, IPC 4912, RF12) nor against P. falciparum and P. vivax isolates from patients in the Brazilian Amazon. Using drug pressure assays, DAQ showed a low propensity to generate resistance. DAQ showed considerable solubility but low metabolic stability. The main metabolite was identified as a mono N-deethylated derivative (DAQM), which also showed significant inhibitory activity against CQ-resistant P. falciparum strains. Our findings indicated that the presence of a triple bond in CQ-analogues may represent a low-cost opportunity to overcome known mechanisms of resistance in the malaria parasite.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria, Vivax , Malaria , Plasmodium , Child , Humans , Chloroquine/pharmacology , Chloroquine/therapeutic use , Plasmodium falciparum , Acetylene/pharmacology , Acetylene/therapeutic use , Alkynes/pharmacology , Alkynes/therapeutic use , Drug Resistance , Antimalarials/therapeutic use , Malaria, Falciparum/drug therapy , Malaria, Falciparum/parasitology , Malaria, Vivax/drug therapy , Malaria/drug therapy
12.
J Pineal Res ; 50(1): 64-70, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20964707

ABSTRACT

Calcium (Ca(2+) ) is a critical regulator of many aspects of the Plasmodium reproductive cycle. In particular, intra-erythrocyte Plasmodium parasites respond to circulating levels of the melatonin in a process mediated partly by intracellular Ca(2+) . Melatonin promotes the development and synchronicity of parasites, thereby enhancing their spread and worsening the clinical implications. The signalling mechanisms underlying the effects of melatonin are not fully established, although both Ca(2+) and cyclic AMP (cAMP) have been implicated. Furthermore, it is not clear whether different strains of Plasmodium use the same, or divergent, signals to control their development. The aim of this study was to explore the signalling mechanisms engaged by melatonin in P. chabaudi, a virulent rodent parasite. Using parasites at the throphozoite stage acutely isolated from mice erythrocytes, we demonstrate that melatonin triggers cAMP production and protein kinase A (PKA) activation. Interestingly, the stimulation of cAMP/PKA signalling by melatonin was dependent on elevation of Ca(2+) within the parasite, because buffering Ca(2+) changes using the chelator BAPTA prevented cAMP production in response to melatonin. Incubation with melatonin evoked robust Ca(2+) signals within the parasite, as did the application of a membrane-permeant analogue of cAMP. Our data suggest that P. chabaudi engages both Ca(2+) and cAMP signalling systems when stimulated by melatonin. Furthermore, there is positive feedback between these messengers, because Ca(2+) evokes cAMP elevation and vice versa. Melatonin more than doubled the observed extent of parasitemia, and the increase in cAMP concentration and PKA activation was essential for this effect. These data support the possibility to use melatonin antagonists or derivates in therapeutic approach.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Melatonin/pharmacology , Plasmodium chabaudi/enzymology , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Enzyme Activation/drug effects , Malaria/parasitology , Mice , Microscopy, Confocal
13.
Pathogens ; 10(11)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34832608

ABSTRACT

Plasmodium falciparum, the most virulent of the human malaria parasite, is responsible for high mortality rates worldwide. We studied the M1 alanyl-aminopeptidase of this protozoan (PfA-M1), which is involved in the final stages of hemoglobin cleavage, an essential process for parasite survival. Aiming to help in the rational development of drugs against this target, we developed a new strain of P. falciparum overexpressing PfA-M1 without the signal peptide (overPfA-M1). The overPfA-M1 parasites showed a 2.5-fold increase in proteolytic activity toward the fluorogenic substrate alanyl-7-amido-4-methylcoumarin, in relation to the wild-type group. Inhibition studies showed that overPfA-M1 presented a lower sensitivity against the metalloaminopeptidase inhibitor bestatin and to other recombinant PfA-M1 inhibitors, in comparison with the wild-type strain, indicating that PfA-M1 is a target for the in vitro antimalarial activity of these compounds. Moreover, overPfA-M1 parasites present a decreased in vitro growth, showing a reduced number of merozoites per schizont, and also a decrease in the iRBC area occupied by the parasite in trophozoite and schizont forms when compared to the controls. Interestingly, the transgenic parasite displays an increase in the aminopeptidase activity toward Met-, Ala-, Leu- and Arg-7-amido-4-methylcoumarin. We also investigated the potential role of calmodulin and cysteine proteases in PfA-M1 activity. Taken together, our data show that the overexpression of PfA-M1 in the parasite cytosol can be a suitable tool for the screening of antimalarials in specific high-throughput assays and may be used for the identification of intracellular molecular partners that modulate their activity in P. falciparum.

14.
J Cell Physiol ; 223(2): 335-42, 2010 May.
Article in English | MEDLINE | ID: mdl-20082302

ABSTRACT

Mucopolysaccharidosis type I (MPS I) is caused by a deficiency of alpha-iduronidase (IDUA), which leads to intralysosomal accumulation of glysosaminoglycans. Patients with MPS I present a wide range of clinical manifestations, but the mechanisms by which these alterations occur are still not fully understood. Genotype-phenotype correlations have not been well established for MPS I; hence, it is likely that secondary and tertiary alterations in cellular metabolism and signaling may contribute to the physiopathology of the disease. The aim of this study was to analyze Ca(2+) and H(+) homeostasis, lysosomal leakage of cysteine proteases, and apoptosis in a murine model of MPS I. After exposition to specific drugs, cells from Idua-/- mice were shown to release more Ca(2+) from the lysosomes and endoplasmic reticulum than Idua+/+ control mice, suggesting a higher intraorganelle store of this ion. A lower content of H(+) in the lysosomes and in the cytosol was found in cells from Idua-/- mice, suggesting an alteration of pH homeostasis. In addition, Idua-/- cells presented a higher activity of cysteine proteases in the cytosol and an increased rate of apoptotic cells when compared to the control group, indicating that lysosomal membrane permeabilization might occur in this model. Altogether, our results suggest that secondary alterations-as changes in Ca(2+) and H(+) homeostasis and lysosomal membrane permeabilization-may contribute for cellular damage and death in the physiopathology of MPS I.


Subject(s)
Calcium Signaling/physiology , Homeostasis/physiology , Intracellular Membranes/metabolism , Lysosomes/metabolism , Mucopolysaccharidosis I/metabolism , Protons , Animals , Apoptosis/genetics , Calcium/metabolism , Calcium Signaling/drug effects , Cell Death/drug effects , Cell Death/physiology , Cell Membrane Permeability/physiology , Cells, Cultured , Cysteine Proteases/genetics , Cysteine Proteases/metabolism , Cytosol/drug effects , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Intracellular Membranes/drug effects , Lysosomes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Mucopolysaccharidosis I/physiopathology
15.
J Psychopharmacol ; 34(4): 467-477, 2020 04.
Article in English | MEDLINE | ID: mdl-31916893

ABSTRACT

BACKGROUND: Interaction of nuclear-distribution element-like 1 with disrupted-in-schizophrenia 1 protein is crucial for neurite outgrowth/neuronal migration, and this interaction competitively inhibits nuclear-distribution element-like 1 peptidase activity. Nuclear-distribution element-like 1 activity is reduced in antipsychotic-naïve first-episode psychosis and in medicated chronic schizophrenia, with even lower activity in treatment-resistant schizophrenia. AIMS: The purpose of this study was to investigate in a rat model overexpressing human non-mutant disrupted-in-schizophrenia 1, with consequent dysfunctional disrupted-in-schizophrenia 1 signaling, the relation of nuclear-distribution element-like 1 activity with neurodevelopment and dopamine-related phenotypes. METHODS: We measured cell distribution in striatum and cortex by histology and microtomography, and quantified the basal and amphetamine-stimulated locomotion and nuclear-distribution element-like 1 activity (in blood and brain) of transgenic disrupted-in-schizophrenia 1 rat vs wild-type littermate controls. RESULTS: 3D assessment of neuronal cell body number and spatial organization of mercury-impregnated neurons showed defective neuronal positioning, characteristic of impaired cell migration, in striatum/nucleus accumbens, and prefrontal cortex of transgenic disrupted-in-schizophrenia 1 compared to wild-type brains. Basal nuclear-distribution element-like 1 activity was lower in the blood and also in several brain regions of transgenic disrupted-in-schizophrenia 1 compared to wild-type. Locomotion and nuclear-distribution element-like 1 activity were both significantly increased by amphetamine in transgenic disrupted-in-schizophrenia 1, but not in wild-type. CONCLUSIONS: Our findings in the transgenic disrupted-in-schizophrenia 1 rat allow us to state that decreased nuclear-distribution element-like 1 activity reflects both a trait (neurodevelopmental phenotype) and a state (amphetamine-induced dopamine release). We thus define here a role for decreased nuclear-distribution element-like 1 peptidase activity both for the developing brain (the neurodevelopmental phenotype) and for the adult (interaction with dopaminergic responses), and present nuclear-distribution element-like 1 activity in a novel way, as unifying neurodevelopmental with dysfunctional dopamine response phenotypes.


Subject(s)
Amphetamine/pharmacology , Cell Nucleus/enzymology , Central Nervous System Stimulants/pharmacology , Cysteine Endopeptidases/metabolism , Nerve Tissue Proteins/genetics , Neurodevelopmental Disorders/genetics , Schizophrenia/genetics , Animals , Animals, Genetically Modified , Brain/diagnostic imaging , Cell Count , Disease Models, Animal , Motor Activity , Rats , Rats, Sprague-Dawley , Rats, Transgenic , Schizophrenia/diagnostic imaging
16.
J Cell Biol ; 161(1): 103-10, 2003 Apr 14.
Article in English | MEDLINE | ID: mdl-12682086

ABSTRACT

Malaria parasites, Plasmodia, spend most of their asexual life cycle within red blood cells, where they proliferate and mature. The erythrocyte cytoplasm has very low [Ca2+] (<100 nM), which is very different from the extracellular environment encountered by most eukaryotic cells. The absence of extracellular Ca2+ is usually incompatible with normal cell functions and survival. In the present work, we have tested the possibility that Plasmodia overcome the limitation posed by the erythrocyte intracellular environment through the maintenance of a high [Ca2+] within the parasitophorous vacuole (PV), the compartment formed during invasion and within which the parasites grow and divide. Thus, Plasmodia were allowed to invade erythrocytes in the presence of Ca2+ indicator dyes. This allowed selective loading of the Ca2+ probes within the PV. The [Ca2+] within this compartment was found to be approximately 40 microM, i.e., high enough to be compatible with a normal loading of the Plasmodia intracellular Ca2+ stores, a prerequisite for the use of a Ca2+-based signaling mechanism. We also show that reduction of extracellular [Ca2+] results in a slow depletion of the [Ca2+] within the PV. A transient drop of [Ca2+] in the PV for a period as short as 2 h affects the maturation process of the parasites within the erythrocytes, with a major reduction 48 h later in the percentage of schizonts, the form that re-invades the red blood cells.


Subject(s)
Calcium Signaling/physiology , Calcium/deficiency , Cell Compartmentation/physiology , Erythrocytes/parasitology , Host-Parasite Interactions/physiology , Plasmodium/metabolism , Vacuoles/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Cytoplasm/metabolism , Cytoplasm/parasitology , Erythrocytes/metabolism , Erythrocytes/ultrastructure , Female , Homeostasis/physiology , Intracellular Fluid/metabolism , Intracellular Fluid/parasitology , Mice , Mice, Inbred BALB C , Plasmodium/pathogenicity , Plasmodium chabaudi/metabolism , Plasmodium falciparum/metabolism , Vacuoles/ultrastructure
17.
Parasitol Int ; 67(2): 233-236, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29288140

ABSTRACT

Malaria is a disease caused by Plasmodium parasites that affects hundreds of millions of people. Plasmodium proteases are involved in invasion, erythrocyte egress and degradation of host proteins. Falcipains are well-studied cysteine peptidases located in P. falciparum food vacuoles that participate in hemoglobin degradation. Cystatins are natural cysteine protease inhibitors that are implicated in a wide range of regulatory processes. Here, we report that a cystatin from sugarcane, CaneCPI-4, is selectively internalized into P. falciparum infected erythrocytes and is not processed by the parasite proteolytic machinery. Furthermore, we demonstrated the inhibition of P. falciparum cysteine proteases by CaneCPI-4, suggesting that it can exert inhibitory functions inside the parasites. The inhibition of the proteolytic activity of parasite cells is specific to this cystatin, as the addition of an anti-CaneCPI-4 antibody completely abolished the inhibition. We extended the studies to recombinant falcipain-2 and falcipain-3 and demonstrated that CaneCPI-4 strongly inhibits these enzymes, with IC50 values of 12nM and 42nM, respectively. We also demonstrated that CaneCPI-4 decreased the hemozoin formation in the parasites, affecting the parasitemia. Taken together, this study identified a natural molecule as a potential antimalarial that specifically targets falcipains and also contributes to a better understanding of macromolecule acquisition by Plasmodium falciparum infected RBCs.


Subject(s)
Antimalarials/pharmacology , Cystatins/pharmacology , Cysteine Proteases/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Plant Proteins/pharmacology , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Antimalarials/isolation & purification , Cystatins/chemistry , Cysteine Endopeptidases/drug effects , Cysteine Endopeptidases/genetics , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/isolation & purification , Erythrocytes/drug effects , Erythrocytes/parasitology , Erythrocytes/physiology , Hemeproteins/drug effects , Humans , Inhibitory Concentration 50 , Plant Proteins/chemistry , Plasmodium falciparum/enzymology
18.
Cell Signal ; 28(3): 125-135, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26689736

ABSTRACT

Calcium and calmodulin (CaM) are important players in eukaryote cell signaling. In the present study, by using a knockin approach, we demonstrated the expression and localization of CaM in all erythrocytic stages of Plasmodium falciparum. Under extracellular Ca(2+)-free conditions, calmidazolium (CZ), a potent CaM inhibitor, promoted a transient cytosolic calcium ([Ca(2+)]cyt) increase in isolated trophozoites, indicating that CZ mobilizes intracellular sources of calcium. In the same extracellular Ca(2+)-free conditions, the [Ca(2+)]cyt rise elicited by CZ treatment was ~3.5 fold higher when the endoplasmic reticulum (ER) calcium store was previously depleted ruling out the mobilization of calcium from the ER by CZ. The effects of the Ca(2+)/H(+) ionophore ionomycin (ION) and the Na(+)/H(+) ionophore monensin (MON) suggest that the [Ca(2+)]cyt-increasing effect of CZ is driven by the removal of Ca(2+) from at least one Ca(2+)-CaM-related (CaMR) protein as well as by the mobilization of Ca(2+) from intracellular acidic calcium stores. Moreover, we showed that the mitochondrion participates in the sequestration of the cytosolic Ca(2+) elicited by CZ. Finally, the modulation of membrane Ca(2+) channels by CZ and thapsigargin (THG) was demonstrated. The opened channels were blocked by the unspecific calcium channel blocker Co(2+) but not by 2-APB (capacitative calcium entry inhibitor) or nifedipine (L-type Ca(2+) channel inhibitor). Taken together, the results suggested that one CaMR protein is an important modulator of calcium signaling and homeostasis during the Plasmodium intraerythrocytic cell cycle, working as a relevant intracellular Ca(2+) reservoir in the parasite.


Subject(s)
Calcium/metabolism , Imidazoles/pharmacology , Plasmodium falciparum/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/chemistry , Calcium Channels/metabolism , Calmodulin/genetics , Calmodulin/metabolism , Calmodulin/pharmacology , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Gene Knock-In Techniques , Humans , Microscopy, Confocal , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development , Thapsigargin/pharmacology , Trophozoites/drug effects , Trophozoites/metabolism
19.
Parasitol Int ; 65(1): 20-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26384965

ABSTRACT

Hypervalent organotellurium compounds (organotelluranes) have shown several promising applications, including their use as potent and selective cysteine protease inhibitors and antiprotozoal agents. Here, we report the antimalarial activities of three organotellurane derivatives (RF05, RF07 and RF19) in two Plasmodium falciparum strains (CQS 3D7 and CQR W2), which demonstrated significant decreases in parasitemia in vitro. The inhibition of intracellular P. falciparum proteases by RF05, RF07 and RF19 was determined and the IC50 values were 3.7±1.0µM, 1.1±0.2µM and 0.2±0.01µM, respectively. Using an assay performed in the presence of the ER Ca(2+)-ATPase inhibitor we showed that the main enzymatic targets were cysteine proteases stimulated by calcium (calpains). None of the compounds tested caused haemolysis or a significant decrease in endothelial cell viability in the concentration range used for the inhibition assay. Taken together, the results suggest promising compounds for the development of antimalarial drugs.


Subject(s)
Antimalarials/pharmacology , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Organometallic Compounds/pharmacology , Plasmodium falciparum/drug effects , Plasmodium falciparum/enzymology , Tellurium/pharmacology , Antimalarials/toxicity , Calcium/metabolism , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/toxicity , Drug Discovery , Erythrocytes/drug effects , Erythrocytes/parasitology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/parasitology , Humans , Inhibitory Concentration 50 , Malaria, Falciparum/drug therapy , Organometallic Compounds/toxicity , Tellurium/toxicity
20.
Mol Biochem Parasitol ; 141(1): 71-9, 2005 May.
Article in English | MEDLINE | ID: mdl-15811528

ABSTRACT

Bloodstage malaria parasites require proteolytic activity for key processes as invasion, hemoglobin degradation and merozoite escape from red blood cells (RBCs). We investigated by confocal microscopy the presence of cysteine-protease activity elicited by calcium stimulus in Plasmodium chabaudi and Plasmodium falciparum in free trophozoites or for the later parasite within RBC using fluorescence resonance energy transfer (FRET) peptides. Peptide probes access, to either free or intraerythrocytic parasites, was also tested by selecting a range of fluorescent peptides (653-3146 Da molecular mass) labeled with Abz or FITC. In the present work we show that Ca2+ stimulus elicited by treatment with either melatonin, thapsigargin, ionomicin or nigericin, promotes an increase of substrate hydrolysis, which was blocked by the specific cysteine-protease inhibitor E-64 and the intracellular Ca2+ chelator, BAPTA. When parasites were treated with cytoplasmic Ca2+ releasing compounds, a cysteine-protease was labeled in the parasite cytoplasm by the fluorescent specific irreversible inhibitor, Ethyl-Eps-Leu-Tyr-Cap-Lys(Abz)-NH2, where Ethyl-Eps is Ethyl-(2S,3S)-oxirane-2,3-dicarboxylate. In summary, we demonstrate that P. chabaudi and P. falciparum have a cytoplasmic dependent cysteine-protease activity elicited by Ca2+.


Subject(s)
Calcium/metabolism , Cysteine Endopeptidases/metabolism , Erythrocytes/parasitology , Plasmodium chabaudi/metabolism , Plasmodium falciparum/metabolism , Amino Acid Sequence , Animals , Calcium Signaling , Cytoplasm/enzymology , Enzyme Inhibitors/pharmacology , Erythrocytes/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemical synthesis , Ionomycin/pharmacology , Ionophores/pharmacology , Melatonin/pharmacology , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Molecular Sequence Data , Nigericin/pharmacology , Plasmodium chabaudi/drug effects , Plasmodium falciparum/drug effects , Thapsigargin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL