Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Eukaryot Microbiol ; 69(3): e12903, 2022 05.
Article in English | MEDLINE | ID: mdl-35279903

ABSTRACT

Cellular invasion by Trypanosoma cruzi metacyclic trypomastigotes (MTs) or tissue culture trypomastigotes (TCTs) is a complex process involving host-parasite cellular and molecular interactions. Particularly, the involvement of host cell actin cytoskeleton during trypomastigote invasion is poorly investigated, and still, the results are controversial. In the present work, we compare side by side both trypomastigote forms and employ state-of-the-art live-cell imaging showing for the first time the dynamic mobilization of host cell actin cytoskeleton to MT and TCT invasion sites. Moreover, cytochalasin D, latrunculin B, and jasplakinolide-pretreated cells inhibited MT and TCT invasion. Furthermore, our results demonstrated that TCT invasion decreased in RhoA, Rac1, and Cdc-42 GTPase-depleted cells, whereas MT invasion decreased only in Cdc42-and RhoA-depleted cells. Interestingly, depletion of the three studied GTPases induced a scattered lysosomal distribution throughout the cytosol. These observations indicate that GTPase depletion is sufficient to impair parasite invasion despite the importance of lysosome spread in trypomastigote invasion. Together, our results demonstrate that the host cell actin cytoskeleton plays a direct role during TCT and MT invasion.


Subject(s)
Trypanosoma cruzi , Actin Cytoskeleton/metabolism , Lysosomes/metabolism , Lysosomes/parasitology , Trypanosoma cruzi/metabolism
2.
PLoS Pathog ; 15(6): e1007834, 2019 06.
Article in English | MEDLINE | ID: mdl-31199856

ABSTRACT

V-ATPases are part of the membrane components of pathogen-containing vacuoles, although their function in intracellular infection remains elusive. In addition to organelle acidification, V-ATPases are alternatively implicated in membrane fusion and anti-inflammatory functions controlled by ATP6V0d2, the d subunit variant of the V-ATPase complex. Therefore, we evaluated the role of ATP6V0d2 in the biogenesis of pathogen-containing vacuoles using ATP6V0d2 knock-down macrophages infected with the protozoan parasite Leishmania amazonensis. These parasites survive within IFNγ/LPS-activated inflammatory macrophages, multiplying in large/fusogenic parasitophorous vacuoles (PVs) and inducing ATP6V0d2 upregulation. ATP6V0d2 knock-down decreased macrophage cholesterol levels and inhibited PV enlargement without interfering with parasite multiplication. However, parasites required ATP6V0d2 to resist the influx of oxidized low-density lipoprotein (ox-LDL)-derived cholesterol, which restored PV enlargement in ATP6V0d2 knock-down macrophages by replenishing macrophage cholesterol pools. Thus, we reveal parasite-mediated subversion of host V-ATPase function toward cholesterol retention, which is required for establishing an inflammation-resistant intracellular parasite niche.


Subject(s)
Cholesterol/metabolism , Gene Expression Regulation, Enzymologic , Homeostasis , Leishmania/metabolism , Macrophages/metabolism , Up-Regulation , Vacuolar Proton-Translocating ATPases/biosynthesis , Vacuoles/metabolism , Animals , Lipoproteins, LDL/metabolism , Macrophages/parasitology , Macrophages/pathology , Mice , Mice, Inbred BALB C , RAW 264.7 Cells , Vacuoles/parasitology , Vacuoles/pathology
3.
Am J Physiol Cell Physiol ; 315(3): C367-C379, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29874111

ABSTRACT

Hypertension is a global health problem, and angiotensin I (ANG I)-converting enzyme (ACE) inhibitors are largely used to control this pathology. Recently, it has been shown that ACE can also act as a transducer signal molecule when its inhibitors or substrates bind to it. This new role of ACE could contribute to understanding some of the effects not explained by its catalytic activity only. In this study, we investigated signaling pathway activation in Chinese hamster ovary (CHO) cells stably expressing ACE (CHO-ACE) under different conditions. We also investigated gene modulation after 4 h and 24 h of captopril treatment. Our results demonstrated that CHO-ACE cells when stimulated with ANG I, ramipril, or captopril led to JNK and ERK1/2 phosphorylation. To verify any physiological role at the endogenous level, we made use of primary cultures of mesangial cells from spontaneously hypertensive rats (SHR) and Wistar rats. Our results showed that ERK1/2 activation occurred mainly in primary cultures of mesangial cells from SHR rats upon captopril stimulation, suggesting that this signaling pathway could be differentially regulated during hypertension. Our results also showed that captopril treatment leads to a decrease of cyclooxygenase 2, interleukin-1ß, and ß-arrestin2 and a significant increase of AP2 gene expression levels. Our findings strengthen the fact that, in addition to the blockage of enzymatic activity, ACE inhibitors also trigger signaling pathway activation, and this may contribute to their beneficial effects in the treatment of hypertension and other pathologies.


Subject(s)
Angiotensin I/metabolism , Captopril/pharmacology , Peptidyl-Dipeptidase A/metabolism , Signal Transduction/drug effects , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , CHO Cells , Cell Line , Cricetulus , Hypertension/drug therapy , Hypertension/metabolism , MAP Kinase Signaling System/drug effects , Male , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Phosphorylation/drug effects , Rats , Rats, Inbred SHR , Rats, Wistar
4.
BMC Microbiol ; 17(1): 195, 2017 Sep 13.
Article in English | MEDLINE | ID: mdl-28903728

ABSTRACT

BACKGROUND: Outbreaks of infections caused by rapidly growing mycobacteria have been reported worldwide generally associated with medical procedures. Mycobacterium abscessus subsp. massiliense CRM0019 was obtained during an epidemic of postsurgical infections and was characterized by increased persistence in vivo. To better understand the successful survival strategies of this microorganism, we evaluated its infectivity and proliferation in macrophages (RAW and BMDM) and alveolar epithelial cells (A549). For that, we assessed the following parameters, for both M. abscessus CRM0019 as well as the reference strain M. abscessus ATCC 19977: internalization, intracellular survival for up 3 days, competence to subvert lysosome fusion and the intracellular survival after cell reinfection. RESULTS: CRM0019 and ATCC 19977 strains showed the same internalization rate (approximately 30% after 6 h infection), in both A549 and RAW cells. However, colony forming units data showed that CRM0019 survived better in A549 cells than the ATCC 19977 strain. Phagosomal characteristics of CRM0019 showed the bacteria inside tight phagosomes in A549 cells, contrasting to the loosely phagosomal membrane in macrophages. This observation holds for the ATCC 19977 strain in both cell types. The competence to subvert lysosome fusion was assessed by acidification and acquisition of lysosomal protein. For M. abscessus strains the phagosomes were acidified in all cell lines; nevertheless, the acquisition of lysosomal protein was reduced by CRM0019 compared to the ATCC 19977 strain, in A549 cells. Conversely, in macrophages, both M. abscessus strains were located in mature phagosomes, however without bacterial death. Once recovered from macrophages M. abscessus could establish a new intracellular infection. Nevertheless, only CRM0019 showed a higher growth rate in A549, increasing nearly 10-fold after 48 and 72 h. CONCLUSION: M. abscessus CRM0019 creates a protective and replicative niche in alveolar epithelial cells mainly by avoiding phagosome maturation. Once recovered from infected macrophages, CRM0019 remains infective and displays greater intracellular growth in A549 cells compared to the ATCC 19977 strain. This evasion strategy in alveolar epithelial cells may contribute to the long survival of the CRM0019 strain in the host and thus to the inefficacy of in vivo treatment.


Subject(s)
Alveolar Epithelial Cells/microbiology , Cell Proliferation , Host-Pathogen Interactions/physiology , Microbial Viability , Mycobacterium abscessus/physiology , Mycobacterium abscessus/pathogenicity , A549 Cells , Animals , Colony Count, Microbial , Humans , Immune Evasion , Lysosomes/metabolism , Macrophages/microbiology , Mice , Phagosomes/microbiology , RAW 264.7 Cells
5.
Cell Microbiol ; 18(6): 779-83, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26639617

ABSTRACT

Although imaging the live Trypanosoma cruzi parasite is a routine technique in most laboratories, identification of the parasite in infected tissues and organs has been hindered by their intrinsic opaque nature. We describe a simple method for in vivo observation of live single-cell Trypanosoma cruzi parasites inside mammalian host tissues. BALB/c or C57BL/6 mice infected with DsRed-CL or GFP-G trypomastigotes had their organs removed and sectioned with surgical blades. Ex vivo organ sections were observed under confocal microscopy. For the first time, this procedure enabled imaging of individual amastigotes, intermediate forms and motile trypomastigotes within infected tissues of mammalian hosts.


Subject(s)
Single-Cell Analysis/methods , Trypanosoma cruzi/cytology , Trypanosoma cruzi/pathogenicity , Animals , Chagas Disease/parasitology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Confocal
6.
Infect Immun ; 84(5): 1603-1614, 2016 05.
Article in English | MEDLINE | ID: mdl-26975994

ABSTRACT

The trypanosomatids Leishmania amazonensis and Trypanosoma cruzi are excellent models for the study of the cell biology of intracellular protozoan infections. After their uptake by mammalian cells, the parasitic protozoan flagellates L. amazonensis and T. cruzi lodge within acidified parasitophorous vacuoles (PVs). However, whereas L. amazonensis develops in spacious, phagolysosome-like PVs that may enclose numerous parasites, T. cruzi is transiently hosted within smaller vacuoles from which it soon escapes to the host cell cytosol. To investigate if parasite-specific vacuoles are required for the survival and differentiation of T. cruzi, we constructed chimeric vacuoles by infection of L. amazonensis amastigote-infected macrophages with T. cruzi epimastigotes (EPIs) or metacyclic trypomastigotes (MTs). These chimeric vacuoles, easily observed by microscopy, allowed the entry and fate of T. cruzi in L. amazonensis PVs to be dynamically recorded by multidimensional imaging of coinfected cells. We found that although T. cruzi EPIs remained motile and conserved their morphology in chimeric vacuoles, T. cruzi MTs differentiated into amastigote-like forms capable of multiplying. These results demonstrate that the large adaptive vacuoles of L. amazonensis are permissive to T. cruzi survival and differentiation and that noninfective EPIs are spared from destruction within the chimeric PVs. We conclude that T. cruzi differentiation can take place in Leishmania-containing vacuoles, suggesting this occurs prior to their escape into the host cell cytosol.


Subject(s)
Cell Differentiation , Leishmania/physiology , Macrophages/parasitology , Trypanosoma cruzi/physiology , Vacuoles/parasitology , Animals , Coinfection/parasitology , Leishmania/growth & development , Mice , Microscopy, Confocal , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Fluorescence , RAW 264.7 Cells , Trypanosoma cruzi/growth & development
7.
Cell Immunol ; 300: 18-25, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26632272

ABSTRACT

Myeloperoxidase (MPO) is an important enzyme in the front-line protection against microorganisms. In peripheral blood, it is accepted that MPO is only produced by myeloid-lineage cells. Thus, MPO presence is unexpected in lymphocytes. We showed recently that B1-lymphocytes from mice have MPO. Here, we showed that subsets of human peripheral B, CD4(+) and CD8(+) T lymphocytes express MPO. The content of MPO in lymphocytes was very low compared to neutrophils/monocytes with a preferential distribution in the nucleus and perinuclear region. Also, we performed a MPO mRNA expression analysis from human blood cells derived from microarray raw data publicly available, showing that MPO is modulated in infectious disease. MPO was increased in CD4(+) T lymphocytes from HIV chronic infection and in CD8(+) T lymphocytes from HCV-positive patients. Our study points out MPO as a multifunctional protein due to its subcellular localization and expression modulation in lymphocytes indicating alternative unknown functions for MPO in lymphocytes.


Subject(s)
B-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/enzymology , Peroxidase/biosynthesis , B-Lymphocytes/immunology , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Separation , Flow Cytometry , HIV Infections/enzymology , HIV Infections/immunology , Hepatitis C/enzymology , Hepatitis C/immunology , Humans , Immunophenotyping , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Oligonucleotide Array Sequence Analysis , Peroxidase/immunology , Real-Time Polymerase Chain Reaction
8.
Cell Microbiol ; 17(12): 1797-810, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26096820

ABSTRACT

Trypanosoma cruzi extracellular amastigotes (EAs) display unique mechanisms for cell invasion that are highly dependent on host actin filaments. Protein kinase D1 (PKD1) phosphorylates and modulates the activity of cortactin, a key regulator of actin dynamics. We evaluated the role of host cortactin and PKD1 in actin filament dynamics during HeLa cell invasion by EAs. Host cortactin, PKD1 and actin are recruited by EAs based on experiments in fixed and live cells by time lapse confocal microscopy. EAs trigger PKD1 and extracellular signal-regulated kinase 1/2 activation, but not Src family kinases, and selectively phosphorylate cortactin. Heat-killed EAs and non-infective epimastigotes both triggered distinct host responses and did not recruit the molecules studied herein. EA invasion was influenced by depletion or overexpression of host cortactin and PKD1, respectively, suggesting the involvement of both proteins in this event. Collectively, these results show new host cell mechanisms subverted during EA internalization into non-phagocytic cells.


Subject(s)
Actins/metabolism , Cortactin/metabolism , Endocytosis , Host-Pathogen Interactions , Protein Kinase C/metabolism , Signal Transduction , Trypanosoma cruzi/physiology , Epithelial Cells/parasitology , Epithelial Cells/physiology , HeLa Cells , Humans , Microscopy, Confocal , Sequence Analysis, DNA , Time-Lapse Imaging
9.
Cell Microbiol ; 16(10): 1549-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24824158

ABSTRACT

The last step of Leishmania intracellular life cycle is the egress of amastigotes from the host cell and their uptake by adjacent cells. Using multidimensional live imaging of long-term-infected macrophage cultures we observed that Leishmania amazonensis amastigotes were transferred from cell to cell when the donor host macrophage delivers warning signs of imminent apoptosis. They were extruded from the macrophage within zeiotic structures (membrane blebs, an apoptotic feature) rich in phagolysosomal membrane components. The extrusions containing amastigotes were selectively internalized by vicinal macrophages and the rescued amastigotes remain viable in recipient macrophages. Host cell apoptosis induced by micro-irradiation of infected macrophage nuclei promoted amastigotes extrusion, which were rescued by non-irradiated vicinal macrophages. Using amastigotes isolated from LAMP1/LAMP2 knockout fibroblasts, we observed that the presence of these lysosomal components on amastigotes increases interleukin 10 production. Enclosed within host cell membranes, amastigotes can be transferred from cell to cell without full exposure to the extracellular milieu, what represents an important strategy developed by the parasite to evade host immune system.


Subject(s)
Leishmania/pathogenicity , Leishmaniasis/transmission , Lysosomal Membrane Proteins/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Macrophages/parasitology , Animals , Apoptosis , Cell Line , Cell Membrane/parasitology , Fibroblasts , Interleukin-10/biosynthesis , Leishmaniasis/pathology , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
10.
Glycobiology ; 24(2): 179-84, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24225883

ABSTRACT

The invasion of host cells by the intracellular protozoan Trypanosoma cruzi requires interactions with host cell molecules, and the replication of the parasite requires escape from a parasitophorous vacuole into the host cell cytosol. Galectin-3, a member of ß-galactosidase-binding lectin family, has numerous extracellular and intracellular functions. In this study, we investigated the role of galectin-3 during the invasion and intracellular trafficking of T. cruzi extracellular amastigotes (EAs). Endogenous galectin-3 from mouse peritoneal macrophages accumulated around the pathogen during cell invasion by EAs. In addition, galectin-3 accumulated around parasites after their escape from the parasitophorous vacuole. Thus, galectin-3 behaved as a novel marker of phagolysosome lysis during the infection of host cells by T. cruzi.


Subject(s)
Galectin 3/metabolism , Trypanosoma cruzi/physiology , Trypanosoma cruzi/pathogenicity , Animals , Biological Transport , Cells, Cultured , Cytoplasm/parasitology , Embryonic Stem Cells/parasitology , Endocytosis , Humans , Macrophages, Peritoneal/parasitology , Mice , Mice, Inbred C57BL , Protein Binding
11.
Anticancer Res ; 44(1): 71-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38160013

ABSTRACT

BACKGROUND/AIM: The Brain-Specific Homeobox/POU Domain Protein 2 (BRN2) transcription factor supports melanoma progression by regulating the expression of several genes involved in cell migration and invasion. We hypothesized that a peptide designed based on the POU domain of BRN2 could block the BRN2 transcription activity and, consequently, reduce metastasis. MATERIALS AND METHODS: Cell viability was accessed by Trypan Blue exclusion dye assay and xCelligence platform. Wound-healing scratch assay and transwell invasion with matrigel membrane assay were performed to analyze cell migration and invasion. The internalization mechanism of the L13S peptide was investigated using confocal microscopy and wound-healing scratch assay. The impact of L13S on cell protein expression was analyzed through western blotting. In vivo assays were conducted to evaluate the protective effect and toxicity of L13S in a metastatic model using murine melanoma cells. RESULTS: Here, we show that the peptide named L13S can inhibit the migration and invasion of murine melanoma cells (B16F10-Nex2) as well as the migration of human melanoma cells (SK-MEL-25 and A375) by regulating the expression of proteins involved in motility. Mechanistically, we found that L13S is internalized by murine melanoma cells via macropinocytosis and binds actin filaments and nuclei. More importantly, in vivo studies indicated that the peptide was able to significantly inhibit lung metastasis in syngeneic models without off-target effects and with virtually no cytotoxicity toward normal organs. CONCLUSION: L13S peptide is a strong candidate for further development as an anticancer agent for the treatment of melanoma metastasis.


Subject(s)
Antineoplastic Agents , Melanoma , Humans , Mice , Animals , Melanoma/pathology , Antineoplastic Agents/pharmacology , Peptides/pharmacology , Peptides/therapeutic use , Cell Movement , Cell Line, Tumor , Cell Proliferation , Neoplasm Invasiveness
12.
Front Cell Infect Microbiol ; 14: 1412345, 2024.
Article in English | MEDLINE | ID: mdl-38988814

ABSTRACT

P21 is a protein secreted by all forms of Trypanosoma cruzi (T. cruzi) with recognized biological activities determined in studies using the recombinant form of the protein. In our recent study, we found that the ablation of P21 gene decreased Y strain axenic epimastigotes multiplication and increased intracellular replication of amastigotes in HeLa cells infected with metacyclic trypomastigotes. In the present study, we investigated the effect of P21 in vitro using C2C12 cell lines infected with tissue culture-derived trypomastigotes (TCT) of wild-type and P21 knockout (TcP21-/-) Y strain, and in vivo using an experimental model of T. cruzi infection in BALB/c mice. Our in-vitro results showed a significant decrease in the host cell invasion rate by TcP21-/- parasites as measured by Giemsa staining and cell count in bright light microscope. Quantitative polymerase chain reaction (qPCR) analysis showed that TcP21-/- parasites multiplied intracellularly to a higher extent than the scrambled parasites at 72h post-infection. In addition, we observed a higher egress of TcP21-/- trypomastigotes from C2C12 cells at 144h and 168h post-infection. Mice infected with Y strain TcP21-/- trypomastigotes displayed higher systemic parasitemia, heart tissue parasite burden, and several histopathological alterations in heart tissues compared to control animals infected with scrambled parasites. Therewith, we propose that P21 is important in the host-pathogen interaction during invasion, cell multiplication, and egress, and may be part of the mechanism that controls parasitism and promotes chronic infection without patent systemic parasitemia.


Subject(s)
Chagas Disease , Protozoan Proteins , Trypanosoma cruzi , Animals , Humans , Mice , Cell Line , Chagas Disease/parasitology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Models, Animal , Gene Knockout Techniques , Host-Parasite Interactions , Mice, Inbred BALB C , Parasitemia , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Trypanosoma cruzi/genetics , Trypanosoma cruzi/pathogenicity , Trypanosoma cruzi/physiology , Trypanosoma cruzi/metabolism , Virulence
13.
J Neuroinflammation ; 10: 61, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23663962

ABSTRACT

Aging is often accompanied by cognitive decline, memory impairment and an increased susceptibility to neurodegenerative disorders. Most of these age-related alterations have been associated with deleterious processes such as changes in the expression of inflammatory cytokines. Indeed, higher levels of pro-inflammatory cytokines and lower levels of anti-inflammatory cytokines are found in the aged brain. This perturbation in pro- and anti-inflammatory balance can represent one of the mechanisms that contribute to age-associated neuronal dysfunction and brain vulnerability. We conducted an experimental study to investigate whether an aerobic exercise program could promote changes in inflammatory response in the brains of aged rats. To do so, we evaluated the levels of tumor necrosis factor alpha (TNFα), interleukin 1 beta (IL1ß), interleukin 6 (IL6) and interleukin 10 (IL10) in the hippocampal formation of 18 month old rats that underwent treadmill training over 10 consecutive days. Quantitative immunoassay analyses showed that the physical exercise increased anti-inflammatory cytokine levels IL10 in the hippocampal formation of aged rats, when compared to the control group. The hippocampal levels of pro-inflammatory cytokines IL1ß, IL6 and TNFα were not statistically different between the groups. However, a significant reduction in IL1ß/IL10, IL6/IL10 and TNFα/IL10 ratio was observed in the exercised group in relation to the control group. These findings indicate a favorable effect of physical exercise in the balance between hippocampal pro- and anti-inflammatory during aging, as well as reinforce the potential therapeutic of exercise in reducing the risk of neuroinflammation-linked disorders.


Subject(s)
Aging/physiology , Hippocampus/physiology , Inflammation/physiopathology , Physical Conditioning, Animal/physiology , Animals , Fluorescent Antibody Technique , Immunoassay , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Neuronal Plasticity/physiology , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/metabolism
14.
Cell Immunol ; 281(1): 27-30, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23434459

ABSTRACT

Generation of hypochlorous acid (HOCl), an important microbicidal agent, is considered to be the main function of myeloperoxidase (MPO), an enzyme present in phagocytes. High amounts of MPO are present in neutrophil azurophilic granules, which are mobilized into the phagolysosome vacuole during phagocytosis. MPO is also present in monocytes and macrophages, although to a lesser degree than in neutrophils. In the present study, we investigated the distribution of MPO in murine peritoneal cells using flow cytometry, confocal microscopy (CM) and transmission electron microscopy (TEM). MPO was observed in macrophages, and surprisingly, we detected MPO in B lymphocytes, specifically in B1-a. MPO was present in cytoplasmic granules, vesicles, mitochondria and the nucleus of murine peritoneal cells. Together, these findings suggest that, in addition to its known microbicidal activity, MPO has a myriad of other unanticipated cellular functions.


Subject(s)
Ascitic Fluid/cytology , B-Lymphocytes , Macrophages , Peritoneal Cavity/cytology , Peroxidase/metabolism , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Nucleus/metabolism , Cytoplasmic Granules/metabolism , Cytoplasmic Vesicles/metabolism , Flow Cytometry , Hypochlorous Acid/metabolism , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Mice , Microscopy, Confocal , Mitochondria/metabolism , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/metabolism , Peroxidase/immunology
15.
Hippocampus ; 22(2): 347-58, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21136521

ABSTRACT

There is a great deal of evidence showing the capacity of physical exercise to enhance cognitive function, reduce anxiety and depression, and protect the brain against neurodegenerative disorders. Although the effects of exercise are well documented in the mature brain, the influence of exercise in the developing brain has been poorly explored. Therefore, we investigated the morphological and functional hippocampal changes in adult rats submitted to daily treadmill exercise during the adolescent period. Male Wistar rats aged 21 postnatal days old (P21) were divided into two groups: exercise and control. Animals in the exercise group were submitted to daily exercise on the treadmill between P21 and P60. Running time and speed gradually increased over this period, reaching a maximum of 18 m/min for 60 min. After the aerobic exercise program (P60), histological and behavioral (water maze) analyses were performed. The results show that early-life exercise increased mossy fibers density and hippocampal expression of brain-derived neurotrophic factor and its receptor tropomyosin-related kinase B, improved spatial learning and memory, and enhanced capacity to evoke spatial memories in later stages (when measured at P96). It is important to point out that while physical exercise induces hippocampal plasticity, degenerative effects could appear in undue conditions of physical or psychological stress. In this regard, we also showed that the exercise protocol used here did not induce inflammatory response and degenerating neurons in the hippocampal formation of developing rats. Our findings demonstrate that physical exercise during postnatal development results in positive changes for the hippocampal formation, both in structure and function.


Subject(s)
Hippocampus/cytology , Hippocampus/physiology , Memory/physiology , Neuronal Plasticity/physiology , Physical Conditioning, Animal/physiology , Animals , Blotting, Western , Cell Count , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Immunohistochemistry , Male , Maze Learning/physiology , Mossy Fibers, Hippocampal/physiology , Rats , Rats, Wistar , Spatial Behavior/physiology
16.
Front Cell Infect Microbiol ; 12: 807172, 2022.
Article in English | MEDLINE | ID: mdl-35573777

ABSTRACT

Trypanosomatids are flagellate protozoans that can infect several invertebrate and vertebrate hosts, including insects and humans. The three most studied species are the human pathogens Trypanosoma brucei, Trypanosoma cruzi and Leishmania spp. which are the causative agents of Human African Trypanosomiasis (HAT), Chagas disease and different clinical forms of leishmaniasis, respectively. These parasites possess complex dixenous life cycles, with zoonotic and anthroponotic stages, and are transmitted by hematophagous insects. To colonize this myriad of hosts, they developed mechanisms, mediated by virulence factors, to infect, propagate and survive in different environments. In insects, surface proteins play roles in parasite attachment and survival in the insect gut, whilst in the mammalian host, the parasites have a whole group of proteins and mechanisms that aid them invading the host cells and evading its immune system components. Many studies have been done on the impact of these molecules in the vertebrate host, however it is also essential to notice the importance of these virulence factors in the insect vector during the parasite life cycle. When inside the insect, the parasites, like in humans, also need to survive defense mechanisms components that can inhibit parasite colonization or survival, e.g., midgut peritrophic membrane barrier, digestive enzymes, evasion of excretion alongside the digested blood meal, anatomic structures and physiological mechanisms of the anterior gut. This protection inside the insect is often implemented by the same group of virulence factors that perform roles of immune evasion in the mammalian host with just a few exceptions, in which a specific protein is expressed specifically for the insect vector form of the parasite. This review aims to discuss the roles of the virulence molecules in the insect vectors, showing the differences and similarities of modes of action of the same group of molecules in insect and humans, exclusive insect molecules and discuss possible genetic events that may have generated this protein diversity.


Subject(s)
Chagas Disease , Parasites , Trypanosoma cruzi , Animals , Chagas Disease/parasitology , Humans , Insect Vectors/parasitology , Insecta , Mammals , Membrane Proteins , Trypanosoma cruzi/physiology , Virulence Factors/genetics
17.
Front Immunol ; 13: 1038332, 2022.
Article in English | MEDLINE | ID: mdl-36389843

ABSTRACT

Trypanosoma cruzi is the causative protozoan of Chagas' Disease, a neglected tropical disease that affects 6-7 million people worldwide. Interaction of the parasite with the host immune system is a key factor in disease progression and chronic symptoms. Although the human immune system is capable of controlling the disease, the parasite has numerous evasion mechanisms that aim to maintain intracellular persistence and survival. Due to the pronounced genetic variability of T. cruzi, co-infections or mixed infections with more than one parasite strain have been reported in the literature. The intermodulation in such cases is unclear. This study aimed to evaluate the co-infection of T. cruzi strains G and CL compared to their individual infections in human macrophages derived from THP-1 cells activated by classical or alternative pathways. Flow cytometry analysis demonstrated that trypomastigotes were more infective than extracellular amastigotes (EAs) and that strain G could infect more macrophages than strain CL. Classically activated macrophages showed lower number of infected cells and IL-4-stimulated cells displayed increased CL-infected macrophages. However, co-infection was a rare event. CL EAs decreased the production of reactive oxygen species (ROS), whereas G trypomastigotes displayed increased ROS detection in classically activated cells. Co-infection did not affect ROS production. Monoinfection by strain G or CL mainly induced an anti-inflammatory cytokine profile by decreasing inflammatory cytokines (IFN-γ, TNF-α, IL-1ß) and/or increasing IL-4, IL-10, and TGF-ß. Co-infection led to a predominant inflammatory milieu, with reduced IL-10 and TGF-ß, and/or promotion of IFN-γ and IL-1ß release. Infection by strain G reduced activation of intracellular signal transducer and activator of transcription (STAT) factors. In EAs, monoinfections impaired STAT-1 activity and promoted phosphorylation of STAT-3, both changes may prolong cell survival. Coinfected macrophages displayed pronounced activation of all STATs examined. These activations likely promoted parasite persistence and survival of infected cells. The collective results demonstrate that although macrophages respond to both strains, T. cruzi can modulate the intracellular environment, inducing different responses depending on the strain, parasite infective form, and co-infection or monoinfection. The modulation influences parasite persistence and survival of infected cells.


Subject(s)
Chagas Disease , Coinfection , Trypanosoma cruzi , Humans , Coinfection/metabolism , Interleukin-10/metabolism , Interleukin-4/metabolism , Macrophages , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism
18.
Nanomaterials (Basel) ; 12(18)2022 Sep 10.
Article in English | MEDLINE | ID: mdl-36144933

ABSTRACT

Inflammasomes are cytosolic complexes composed of a Nod-like receptor, NLR, the adaptor protein, ASC, and a proteolytic enzyme, caspase-1. Inflammasome activation leads to caspase-1 activation and promotes functional maturation of IL-1ß and IL-18, two prototypical inflammatory cytokines. Besides, inflammasome activation leads to pyroptosis, an inflammatory type of cell death. Inflammasomes are vital for the host to cope with foreign pathogens or tissue damage. Herein, we show that quantum-dot-based iron oxide nanoparticles, MNP@QD, trigger NLRP3 inflammasome activation and subsequent release of proinflammatory interleukin IL-1ß by murine bone marrow-derived dendritic cells (BMDCs). This activation is more pronounced if these cells endocytose the nanoparticles before receiving inflammatory stimulation. MNP@QD was characterized by using imaging techniques like transmission electron microscopy, fluorescence microscopy, and atomic force microscopy, as well as physical and spectroscopical techniques such as fluorescence spectroscopy and powder diffraction. These findings may open the possibility of using the composite MNP@QD as both an imaging and a therapeutic tool.

19.
Front Cell Infect Microbiol ; 12: 799668, 2022.
Article in English | MEDLINE | ID: mdl-35252026

ABSTRACT

P21 is an immunomodulatory protein expressed throughout the life cycle of Trypanosoma cruzi, the etiologic agent of Chagas disease. In vitro and in vivo studies have shown that P21 plays an important role in the invasion of mammalian host cells and establishment of infection in a murine model. P21 functions as a signal transducer, triggering intracellular cascades in host cells and resulting in the remodeling of the actin cytoskeleton and parasite internalization. Furthermore, in vivo studies have shown that P21 inhibits angiogenesis, induces inflammation and fibrosis, and regulates intracellular amastigote replication. In this study, we used the CRISPR/Cas9 system for P21 gene knockout and investigated whether the ablation of P21 results in changes in the phenotypes associated with this protein. Ablation of P21 gene resulted in a lower growth rate of epimastigotes and delayed cell cycle progression, accompanied by accumulation of parasites in G1 phase. However, P21 knockout epimastigotes were viable and able to differentiate into metacyclic trypomastigotes, which are infective to mammalian cells. In comparison with wild-type parasites, P21 knockout cells showed a reduced cell invasion rate, demonstrating the role of this protein in host cell invasion. However, there was a higher number of intracellular amastigotes per cell, suggesting that P21 is a negative regulator of amastigote proliferation in mammalian cells. Here, for the first time, we demonstrated the direct correlation between P21 and the replication of intracellular amastigotes, which underlies the chronicity of T. cruzi infection.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Actin Cytoskeleton/physiology , Animals , Chagas Disease/parasitology , Gene Knockout Techniques , Life Cycle Stages/physiology , Mammals/genetics , Mice , Trypanosoma cruzi/physiology
20.
Mem Inst Oswaldo Cruz ; 106(8): 1014-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22241126

ABSTRACT

It has been recently shown that Trypanosoma cruzi trypomastigotes subvert a constitutive membrane repair mechanism to invade HeLa cells. Using a membrane extraction protocol and high-resolution microscopy, the HeLa cytoskeleton and T. cruzi parasites were imaged during the invasion process after 15 min and 45 min. Parasites were initially found under cells and were later observed in the cytoplasm. At later stages, parasite-driven protrusions with parallel filaments were observed, with trypomastigotes at their tips. We conclude that T. cruzi trypomastigotes induce deformations of the cortical actin cytoskeleton shortly after invasion, leading to the formation of pseudopod-like structures.


Subject(s)
Cell Membrane/parasitology , Cytoskeleton/parasitology , Trypanosoma cruzi/physiology , Cell Membrane/ultrastructure , Cytoskeleton/ultrastructure , HeLa Cells/parasitology , HeLa Cells/ultrastructure , Humans , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL