Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell Mol Life Sci ; 79(5): 245, 2022 Apr 18.
Article in English | MEDLINE | ID: mdl-35435504

ABSTRACT

BACKGROUND: Blastocystis is a common gut protistan parasite in humans and animals worldwide, but its interrelationship with the host gut microbiota and mucosal immune responses remains poorly understood. Different murine models of Blastocystis colonization were used to examine the effect of a common Blastocystis subtype (ST4) on host gut microbial community and adaptive immune system. RESULTS: Blastocystis ST4-colonized normal healthy mice and Rag1-/- mice asymptomatically and was able to alter the microbial community composition, mainly leading to increases in the proportion of Clostridia vadinBB60 group and Lachnospiraceae NK4A136 group, respectively. Blastocystis ST4 colonization promoted T helper 2 (Th2) response defined by interleukin (IL)-5 and IL-13 cytokine production, and T regulatory (Treg) induction from colonic lamina propria in normal healthy mice. Additionally, we observed that Blastocystis ST4 colonization can maintain the stability of bacterial community composition and induce Th2 and Treg immune responses to promote faster recovery from experimentally induced colitis. Furthermore, fecal microbiota transplantation of Blastocystis ST4-altered gut microbiome to colitis mice reduced the severity of colitis, which was associated with increased production of short-chain fat acids (SCFAs) and anti-inflammatory cytokine IL-10. CONCLUSIONS: The data confirm our hypothesis that Blastocystis ST4 is a beneficial commensal, and the beneficial effects of Blastocystis ST4 colonization is mediated through modulating of the host gut bacterial composition, SCFAs production, and Th2 and Treg responses in different murine colonization models.


Subject(s)
Blastocystis , Colitis , Gastrointestinal Microbiome , Animals , Bacteria , Colitis/chemically induced , Cytokines , Disease Models, Animal , Immunity , Mice , Mice, Inbred C57BL
2.
Article in English | MEDLINE | ID: mdl-29311064

ABSTRACT

Plasmodium falciparum infections leading to malaria have severe clinical manifestations and high mortality rates. Chloroquine (CQ), a former mainstay of malaria chemotherapy, has been rendered ineffective due to the emergence of widespread resistance. Recent studies, however, have unveiled a novel mode of action in which low-micromolar levels of CQ permeabilized the parasite's digestive vacuole (DV) membrane, leading to calcium efflux, mitochondrial depolarization, and DNA degradation. These phenotypes implicate the DV as an alternative target of CQ and suggest that DV disruption is an attractive target for exploitation by DV-disruptive antimalarials. In the current study, high-content screening of the Medicines for Malaria Venture (MMV) Pathogen Box (2015) was performed to select compounds which disrupt the DV membrane, as measured by the leakage of intravacuolar Ca2+ using the calcium probe Fluo-4 AM. The hits were further characterized by hemozoin biocrystallization inhibition assays and dose-response half-maximal (50%) inhibitory concentration (IC50) assays across resistant and sensitive strains. Three hits, MMV676380, MMV085071, and MMV687812, were shown to demonstrate a lack of CQ cross-resistance in parasite strains and field isolates. Through systematic analyses, MMV085071 emerged as the top hit due to its rapid parasiticidal effect, low-nanomolar IC50, and good efficacy in triggering DV disruption, mitochondrial degradation, and DNA fragmentation in P. falciparum These programmed cell death (PCD)-like phenotypes following permeabilization of the DV suggests that these compounds kill the parasite by a PCD-like mechanism. From the drug development perspective, MMV085071, which was identified to be a potent DV disruptor, offers a promising starting point for subsequent hit-to-lead generation and optimization through structure-activity relationships.


Subject(s)
Antimalarials/pharmacology , Calcium/metabolism , High-Throughput Screening Assays , Plasmodium falciparum/drug effects , Small Molecule Libraries/pharmacology , Vacuoles/drug effects , Aniline Compounds/chemistry , Antimalarials/chemistry , Chloroquine/chemistry , Chloroquine/pharmacology , Crystallization , Databases, Pharmaceutical , Drug Resistance , Erythrocytes/drug effects , Erythrocytes/metabolism , Erythrocytes/parasitology , Fluorescent Dyes/chemistry , Hemeproteins/chemistry , Hemeproteins/drug effects , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Intracellular Membranes/parasitology , Permeability , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Vacuoles/metabolism , Vacuoles/parasitology , Xanthenes/chemistry
3.
Article in English | MEDLINE | ID: mdl-29866860

ABSTRACT

Blastocystis is an enteric parasite with extensive global prevalence. Studies have linked infection with this protist with a variety of gastrointestinal disorders, including irritable bowel syndrome. Due to the polymorphic nature of Blastocystis, studies on the parasite could be complicated, as results can be easily misinterpreted. Metronidazole is the commonly prescribed drug for Blastocystis infection, although there have been increasing reports of drug resistance. Hence, there is a need to identify alternative drugs to eliminate Blastocystis infection. In this study, LOPAC1280 was screened and drugs that can decrease the viability of three Blastocystis isolates in cultures were identified. Using apoptosis assay and imaging flow cytometry, phenotypic changes in Blastocystis cells after treatment were also analyzed to obtain insights into the possible mechanism of action of these drugs. Three drugs-diphenyleneiodonium chloride, auranofin, and BIX 01294 trihydrochloride hydrate-were effective against all three isolates tested. Repurposing of these drugs for Blastocystis treatment could be a way of combating metronidazole resistance relatively quickly and at a lower cost.


Subject(s)
Antiprotozoal Agents/pharmacology , Auranofin/pharmacology , Azepines/pharmacology , Blastocystis/drug effects , Onium Compounds/pharmacology , Quinazolines/pharmacology , Small Molecule Libraries/pharmacology , Antiprotozoal Agents/chemistry , Antirheumatic Agents/chemistry , Antirheumatic Agents/pharmacology , Apoptosis/drug effects , Auranofin/chemistry , Azepines/chemistry , Blastocystis/classification , Blastocystis/growth & development , Blastocystis/isolation & purification , Blastocystis Infections/parasitology , Drug Repositioning , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Inhibitory Concentration 50 , Onium Compounds/chemistry , Phosphorylation/drug effects , Quinazolines/chemistry , Small Molecule Libraries/chemistry
4.
Methods ; 112: 211-220, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27389304

ABSTRACT

Malaria, despite being one of the world's oldest infectious diseases, remains difficult to eradicate because the parasite is rapidly developing resistance to frontline chemotherapies. Previous studies have shown that the parasite exhibits features resembling programmed cell death upon treatment with drugs that disrupt its digestive vacuole (DV), providing a phenotypic readout that can be detected using the imaging flow cytometer. Large compound collections can thus be screened to identify drugs that are able to disrupt the DV of the malaria parasite using this high-content high-throughput screening platform. As a proof-of-concept, 4440 compounds were screened using this platform in 4months and 254 hits (5.7% hit rate) were obtained. Additionally, 25 compounds (0.6% top hit rate) were observed to retain potent DV disruption activity that was comparable to the canonical DV disruptive drug chloroquine when tested at a ten-fold lower concentration from the original screen. This pilot study demonstrates the robustness and high-throughput capability of the imaging flow cytometer and we report herein the methodology of this screening assay.


Subject(s)
Erythrocytes/parasitology , Flow Cytometry/methods , Image Cytometry/methods , Life Cycle Stages/drug effects , Plasmodium falciparum/drug effects , Vacuoles/drug effects , Aniline Compounds/chemistry , Antimalarials/chemistry , Antimalarials/pharmacology , Benzimidazoles/chemistry , Carbocyanines/chemistry , Cells, Cultured , Erythrocytes/drug effects , Erythrocytes/ultrastructure , Fluorescent Dyes/chemistry , High-Throughput Screening Assays , Humans , Life Cycle Stages/physiology , Plasmodium falciparum/growth & development , Plasmodium falciparum/ultrastructure , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Staining and Labeling/methods , Vacuoles/ultrastructure , Xanthenes/chemistry
5.
Molecules ; 23(10)2018 Oct 14.
Article in English | MEDLINE | ID: mdl-30322183

ABSTRACT

Chloroquine was among the first of several effective drug treatments against malaria until the onset of chloroquine resistance. In light of diminished clinical efficacy of chloroquine as an antimalarial therapeutic, there is potential in efforts to adapt chloroquine for other clinical applications, such as in combination therapies and in diagnostics. In this context, we designed and synthesized a novel asymmetrical squaraine dye coupled with chloroquine (SQR1-CQ). In this study, SQR1-CQ was used to label live Plasmodium falciparum (P. falciparum) parasite cultures of varying sensitivities towards chloroquine. SQR1-CQ positively stained ring, mature trophozoite and schizont stages of both chloroquine⁻sensitive and chloroquine⁻resistant P. falciparum strains. In addition, SQR1-CQ exhibited significantly higher fluorescence, when compared to the commercial chloroquine-BODIPY (borondipyrromethene) conjugate CQ-BODIPY. We also achieved successful SQR1-CQ labelling of P. falciparum directly on thin blood smear preparations. Drug efficacy experiments measuring half-maximal inhibitory concentration (IC50) showed lower concentration of effective inhibition against resistant strain K1 by SQR1-CQ compared to conventional chloroquine. Taken together, the versatile and highly fluorescent labelling capability of SQR1-CQ and promising preliminary IC50 findings makes it a great candidate for further development as diagnostic tool with drug efficacy against chloroquine-resistant P. falciparum.


Subject(s)
Antimalarials/pharmacology , Chloroquine/pharmacology , Cyclobutanes/chemistry , Fluorescent Dyes/chemistry , Phenols/chemistry , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Blood/parasitology , Chloroquine/chemistry , Drug Resistance , Humans , Inhibitory Concentration 50 , Microscopy, Confocal , Molecular Imaging , Molecular Structure
6.
J Infect Dis ; 213(1): 100-4, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26136472

ABSTRACT

Recent clinical trials revealed a surprisingly rapid clearance of red blood cells (RBCs) infected with malaria parasites by the spiroindolone KAE609. Here, we show that ring-stage parasite-infected RBCs exposed to KAE609 become spherical and rigid, probably through osmotic dysregulation consequent to the disruption of the parasite's sodium efflux pump (adenosine triphosphate 4). We also show that this peculiar drug effect is likely to cause accelerated splenic clearance of the rheologically impaired Plasmodium vivax- and Plasmodium falciparum-infected RBCs.


Subject(s)
Antimalarials/pharmacology , Indoles/pharmacology , Malaria, Falciparum/parasitology , Plasmodium falciparum/drug effects , Plasmodium vivax/drug effects , Spiro Compounds/pharmacology , Erythrocytes/parasitology , Humans , Plasmodium falciparum/genetics , Plasmodium vivax/genetics
7.
Infect Immun ; 84(8): 2220-2232, 2016 08.
Article in English | MEDLINE | ID: mdl-27217421

ABSTRACT

Blastocystis is one of the most common eukaryotic organisms found in humans and many types of animals. Several reports have identified its role in gastrointestinal disorders, although its pathogenicity is yet to be clarified. Blastocystis is transmitted via the fecal-to-oral route and colonizes the large intestines. Epithelial cells lining the intestine secrete antimicrobial peptides (AMPs), including beta-defensins and cathelicidin, as a response to infection. This study explores the effects of host colonic antimicrobial peptides, particularly LL-37, a fragment of cathelicidin, on different Blastocystis subtypes. Blastocystis is composed of several subtypes that have genetic, metabolic, and biological differences. These subtypes also have various outcomes in terms of drug treatment and immune response. In this study, Blastocystis isolates from three different subtypes were found to induce intestinal epithelial cells to secrete LL-37. We also show that among the antimicrobial peptides tested, only LL-37 has broad activity on all the subtypes. LL-37 causes membrane disruption and causes Blastocystis to change shape. Blastocystis subtype 7 (ST7), however, showed relative resistance to LL-37. An isolate, ST7 isolate B (ST7-B), from this subtype releases proteases that can degrade the peptide. It also makes the environment acidic, which causes attenuation of LL-37 activity. The Blastocystis ST7-B isolate was also observed to have a thicker surface coat, which may protect the parasite from direct killing by LL-37. This study determined the effects of LL-37 on different Blastocystis isolates and indicates that AMPs have significant roles in Blastocystis infections.


Subject(s)
Blastocystis Infections/parasitology , Blastocystis/drug effects , Cathelicidins/pharmacology , Drug Resistance , Animals , Antimicrobial Cationic Peptides , Blastocystis/ultrastructure , Blastocystis Infections/metabolism , Cathelicidins/biosynthesis , Cell Line , Cell Membrane/drug effects , Disease Models, Animal , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/parasitology , Mice , Parasitic Sensitivity Tests
8.
J Infect Dis ; 212(4): 635-44, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25612733

ABSTRACT

Vitamin D level is linked to susceptibility to infections, but its relevance in candidemia is unknown. We aimed to investigate the in vivo sequelae of vitamin D3 supplementation in systemic Candida infection. Implicating the role of vitamin D in Candida infections, we showed that candidemic patients had significantly lower 25-OHD concentrations. Candida-infected mice treated with low-dose 1,25(OH)2D3 had reduced fungal burden and better survival relative to untreated mice. Conversely, higher 1,25(OH)2D3 doses led to poor outcomes. Mechanistically, low-dose 1,25(OH)2D3 induced proinflammatory immune responses. This was mediated through suppression of SOCS3 and induction of vitamin D receptor binding with the vitamin D-response elements in the promoter of the gene encoding interferon γ. These beneficial effects were negated with higher vitamin D3 doses. While the antiinflammatory effects of vitamin D3 are well described, we found that, conversely, lower doses conferred proinflammatory benefits in Candida infection. Our study highlights caution against extreme deviations of vitamin D levels during infections.


Subject(s)
Candidiasis/drug therapy , Cholecalciferol/pharmacology , Vitamin D/blood , Animals , Candidiasis/immunology , Cholecalciferol/administration & dosage , Cohort Studies , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Inflammation/immunology , Inflammation/metabolism , Interferon-gamma/metabolism , Leukocytes, Mononuclear , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
9.
J Biol Chem ; 289(48): 33425-41, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25305013

ABSTRACT

Artesunate (ART) is an anti-malaria drug that has been shown to exhibit anti-tumor activity, and functional lysosomes are reported to be required for ART-induced cancer cell death, whereas the underlying molecular mechanisms remain largely elusive. In this study, we aimed to elucidate the molecular mechanisms underlying ART-induced cell death. We first confirmed that ART induces apoptotic cell death in cancer cells. Interestingly, we found that ART preferably accumulates in the lysosomes and is able to activate lysosomal function via promotion of lysosomal V-ATPase assembly. Furthermore, we found that lysosomes function upstream of mitochondria in reactive oxygen species production. Importantly, we provided evidence showing that lysosomal iron is required for the lysosomal activation and mitochondrial reactive oxygen species production induced by ART. Finally, we showed that ART-induced cell death is mediated by the release of iron in the lysosomes, which results from the lysosomal degradation of ferritin, an iron storage protein. Meanwhile, overexpression of ferritin heavy chain significantly protected cells from ART-induced cell death. In addition, knockdown of nuclear receptor coactivator 4, the adaptor protein for ferritin degradation, was able to block ART-mediated ferritin degradation and rescue the ART-induced cell death. In summary, our study demonstrates that ART treatment activates lysosomal function and then promotes ferritin degradation, subsequently leading to the increase of lysosomal iron that is utilized by ART for its cytotoxic effect on cancer cells. Thus, our data reveal a new mechanistic action underlying ART-induced cell death in cancer cells.


Subject(s)
Antimalarials/pharmacology , Artemisinins/pharmacology , Ferritins/metabolism , Lysosomes/metabolism , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Proteolysis/drug effects , Artesunate , Cell Death/drug effects , HeLa Cells , Hep G2 Cells , Humans , Iron/metabolism , Neoplasms/metabolism , Neoplasms/physiopathology , Nuclear Receptor Coactivators/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
10.
Infect Immun ; 82(11): 4789-801, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25156742

ABSTRACT

Blastocystis is a common enteric protistan parasite that can cause acute, as well as chronic, infection and is associated with irritable bowel syndrome (IBS). However, the pathogenic status of Blastocystis infection remains unclear. In this study, we found that Blastocystis antigens induced abundant expression of proinflammatory cytokines, including interleukin 1ß (IL-1ß), IL-6, and tumor necrosis factor alpha (TNF-α), in mouse intestinal explants, in mouse colitis colon, and in macrophages. Further investigation utilizing RAW264.7 murine macrophages showed that Blastocystis treatment in RAW264.7 macrophages induced the activation of ERK, JNK, and p38, the three major groups of mammalian mitogen-activated protein (MAP) kinases that play essential roles in the expression of proinflammatory cytokines. ERK inhibition in macrophages significantly suppressed both mRNA and protein expression of IL-6 and TNF-α and mRNA expression of IL-1ß. On the other hand, JNK inhibition resulted in reductions in both c-Jun and ERK activation and significant suppression of all three proinflammatory cytokines at both the mRNA and protein levels. Inhibition of p38 suppressed only IL-6 protein expression with no effect on the expression of IL-1ß and TNF-α. Furthermore, we found that serine proteases produced by Blastocystis play an important role in the induction of ERK activation and proinflammatory cytokine expression by macrophages. Our study thus demonstrated for the first time that Blastocystis could induce the expression of various proinflammatory cytokines via the activation of MAP kinases and that infection with Blastocystis may contribute to the pathogenesis of inflammatory intestinal diseases through the activation of inflammatory pathways in host immune cells, such as macrophages.


Subject(s)
Blastocystis Infections/metabolism , Blastocystis/immunology , Cytokines/metabolism , Gene Expression Regulation/immunology , Macrophages/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Animals , Blastocystis Infections/immunology , Cell Line , Cytokines/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/genetics , Tissue Culture Techniques
11.
Mediators Inflamm ; 2014: 243713, 2014.
Article in English | MEDLINE | ID: mdl-25157202

ABSTRACT

Malaria is one of the most serious infectious diseases in humans and responsible for approximately 500 million clinical cases and 500 thousand deaths annually. Acquired adaptive immune responses control parasite replication and infection-induced pathologies. Most infections are clinically silent which reflects on the ability of adaptive immune mechanisms to prevent the disease. However, a minority of these can become severe and life-threatening, manifesting a range of overlapping syndromes of complex origins which could be induced by uncontrolled immune responses. Major players of the innate and adaptive responses are interferons. Here, we review their roles and the signaling pathways involved in their production and protection against infection and induced immunopathologies.


Subject(s)
Interferon Regulatory Factors/metabolism , Interferons/metabolism , Malaria/metabolism , Animals , Humans
12.
Trends Parasitol ; 40(1): 1-4, 2024 01.
Article in English | MEDLINE | ID: mdl-37980258

ABSTRACT

Blastocystis is a common intestinal parasite that has been linked to gut pathology in humans. In this article, we highlight recent publications that offer insight into how these organisms can influence human cognition and the gut microbiome. We also suggest a potential mechanism of action by which this might occur.


Subject(s)
Blastocystis Infections , Blastocystis , Gastrointestinal Microbiome , Humans , Blastocystis Infections/parasitology , Brain , Feces/parasitology
13.
Cell Death Differ ; 31(3): 280-291, 2024 03.
Article in English | MEDLINE | ID: mdl-38383887

ABSTRACT

Detection of cytosolic nucleic acids by pattern recognition receptors, including STING and RIG-I, leads to the activation of multiple signalling pathways that culminate in the production of type I interferons (IFNs) which are vital for host survival during virus infection. In addition to protective immune modulatory functions, type I IFNs are also associated with autoimmune diseases. Hence, it is important to elucidate the mechanisms that govern their expression. In this study, we identified a critical regulatory function of the DUSP4 phosphatase in innate immune signalling. We found that DUSP4 regulates the activation of TBK1 and ERK1/2 in a signalling complex containing DUSP4, TBK1, ERK1/2 and IRF3 to regulate the production of type I IFNs. Mice deficient in DUSP4 were more resistant to infections by both RNA and DNA viruses but more susceptible to malaria parasites. Therefore, our study establishes DUSP4 as a regulator of nucleic acid sensor signalling and sheds light on an important facet of the type I IFN regulatory system.


Subject(s)
Interferon Type I , Membrane Proteins , Protein Tyrosine Phosphatases , Receptors, Cell Surface , Roundabout Proteins , Virus Diseases , Animals , Mice , Immunity, Innate , Interferon Type I/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Virus Diseases/immunology , Virus Diseases/metabolism , Membrane Proteins/metabolism , Roundabout Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptors, Cell Surface/metabolism
14.
Theranostics ; 13(3): 1165-1179, 2023.
Article in English | MEDLINE | ID: mdl-36793854

ABSTRACT

Rationale: The gut microbiota plays a significant role in the pathogenesis of inflammatory bowel disease (IBD). However, the role of Blastocystis infection and Blastocystis-altered gut microbiota in the development of inflammatory diseases and their underlying mechanisms are not well understood. Methods: We investigated the effect of Blastocystis ST4 and ST7 infection on the intestinal microbiota, metabolism, and host immune responses, and then explored the role of Blastocystis-altered gut microbiome in the development of dextran sulfate sodium (DSS)-induced colitis in mice. Results: This study showed that prior colonization with ST4 conferred protection from DSS-induced colitis through elevating the abundance of beneficial bacteria, short-chain fatty acid (SCFA) production and the proportion of Foxp3+ and IL-10-producing CD4+ T cells. Conversely, prior ST7 infection exacerbated the severity of colitis by increasing the proportion of pathogenic bacteria and inducing pro-inflammatory IL-17A and TNF-α-producing CD4+ T cells. Furthermore, transplantation of ST4- and ST7-altered microbiota resulted in similar phenotypes. Conclusions: Our data showed that ST4 and ST7 infection exert strikingly differential effects on the gut microbiota, and these could influence the susceptibility to colitis. ST4 colonization prevented DSS-induced colitis in mice and may be considered as a novel therapeutic strategy against immunological diseases in the future, while ST7 infection is a potential risk factor for the development of experimentally induced colitis that warrants attention.


Subject(s)
Blastocystis , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Microbiota , Animals , Mice , Colitis/pathology , Dextran Sulfate/adverse effects , Mice, Inbred C57BL , Disease Models, Animal , Colon/pathology
15.
NPJ Biofilms Microbiomes ; 9(1): 22, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185924

ABSTRACT

Blastocystis is a species complex that exhibits extensive genetic diversity, evidenced by its classification into several genetically distinct subtypes (ST). Although several studies have shown the relationships between a specific subtype and gut microbiota, there is no study to show the effect of the ubiquitous Blastocystis ST1 on the gut microbiota and host health. Here, we show that Blastocystis ST1 colonization increased the proportion of beneficial bacteria Alloprevotella and Akkermansia, and induced Th2 and Treg cell responses in normal healthy mice. ST1-colonized mice showed decreases in the severity of DSS-induced colitis when compared to non-colonized mice. Furthermore, mice transplanted with ST1-altered gut microbiota were refractory to dextran sulfate sodium (DSS)-induced colitis via induction of Treg cells and elevated short-chain fat acid (SCFA) production. Our results suggest that colonization with Blastocystis ST1, one of the most common subtypes in humans, exerts beneficial effects on host health through modulating the gut microbiota and adaptive immune responses.


Subject(s)
Blastocystis , Colitis , Gastrointestinal Microbiome , Microbiota , Humans , Mice , Animals , Blastocystis/genetics , Colitis/chemically induced , Colitis/microbiology , Bacteria
16.
Front Cell Infect Microbiol ; 12: 1019789, 2022.
Article in English | MEDLINE | ID: mdl-36389146

ABSTRACT

Blastocystis is a genus of single-celled protist belonging to the stramenopile group. Prior studies have shown that isolates of Blastocystis subtype 7 (ST7) induced higher levels of intestinal epithelial cell damage and gut microbiota dysbiosis in comparison to other subtypes in in vivo and in vitro settings. Prior research has shown a link between gut dysbiosis and exposure to extracellular vesicles (EVs) produced by pathogenic microorganisms. This study demonstrates a protocol for the isolation of EVs from Blastocystis ST7 via ultracentrifugation. Nanoparticle tracking analysis and transmission electron microscopy were used to assess EV size and morphology. The protein content of isolated EVs was assessed by mass spectrophotometry and the presence of EV markers were evaluated by Western blotting. Finally, the EVs were cocultured with prominent human gut microbiome species to observe their effect on prokaryote growth. Our data shows that Blastocystis ST7 secretes EVs that are similar in morphology to previously characterized EVs from other organisms and that these EVs contain a limited yet unique protein cargo with functions in host-parasite intercellular communication and cell viability. This cargo may be involved in mediating the effects of Blastocystis on its surrounding environment.


Subject(s)
Blastocystis , Extracellular Vesicles , Parasites , Animals , Humans , Dysbiosis , Extracellular Vesicles/metabolism , Ultracentrifugation , Proteins/metabolism
17.
J Biol Chem ; 285(3): 1790-8, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-19915007

ABSTRACT

Programmed cell death (PCD) is crucial for cellular growth and development in multicellular organisms. Although distinct PCD features have been described for unicellular eukaryotes, homology searches have failed to reveal clear PCD-related orthologues among these organisms. Our previous studies revealed that a surface-reactive monoclonal antibody (mAb) 1D5 could induce multiple PCD pathways in the protozoan Blastocystis. In this study, we identified, by two-dimensional gel electrophoresis and mass spectrometry, the target of mAb 1D5 as a surface-localized legumain, an asparagine endopeptidase that is usually found in lysosomal/acidic compartments of other organisms. Recombinant Blastocystis legumain displayed biphasic pH optima in substrate assays, with peaks at pH 4 and 7.5. Activity of Blastocystis legumain was greatly inhibited by the legumain-specific inhibitor carbobenzyloxy-Ala-Ala-AAsn-epoxycarboxylate ethyl ester (APE-RR) (where AAsn is aza-asparagine) and moderately inhibited by mAb 1D5, cystatin, and caspase-1 inhibitor. Interestingly, inhibition of legumain activity induced PCD in Blastocystis, observed by increased externalization of phosphatidylserine residues and in situ DNA fragmentation. In contrast to plants, in which legumains have been shown to play a pro-death role, legumain appears to display a pro-survival role in Blastocystis.


Subject(s)
Blastocystis/cytology , Blastocystis/enzymology , Cysteine Endopeptidases/metabolism , Protease Inhibitors/pharmacology , Amino Acid Sequence , Animals , Annexin A5/metabolism , Antibodies, Monoclonal/immunology , Blastocystis/genetics , Blastocystis/metabolism , Cattle , Cell Death , Cell Survival , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/immunology , DNA Fragmentation , Escherichia coli/genetics , Humans , Hydrogen-Ion Concentration , In Situ Nick-End Labeling , Mice , Molecular Sequence Data , Phosphatidylserines/metabolism , Protein Transport , Rats , Substrate Specificity
18.
PLoS Negl Trop Dis ; 15(9): e0009730, 2021 09.
Article in English | MEDLINE | ID: mdl-34492012

ABSTRACT

In recent years, the human gut microbiome has been recognised to play a pivotal role in the health of the host. Intestinal homeostasis relies on this intricate and complex relationship between the gut microbiota and the human host. While much effort and attention has been placed on the characterization of the organisms that inhabit the gut microbiome, the complex molecular cross-talk between the microbiota could also exert an effect on gastrointestinal conditions. Blastocystis is a single-cell eukaryotic parasite of emerging interest, as its beneficial or pathogenic role in the microbiota has been a subject of contention even to-date. In this study, we assessed the function of the Blastocystis tryptophanase gene (BhTnaA), which was acquired by horizontal gene transfer and likely to be of bacterial origin within Blastocystis. Bioinformatic analysis and phylogenetic reconstruction revealed distinct divergence of BhTnaA versus known bacterial homologs. Despite sharing high homology with the E. coli tryptophanase gene, we show that Blastocystis does not readily convert tryptophan into indole. Instead, BhTnaA preferentially catalyzes the conversion of indole to tryptophan. We also show a direct link between E. coli and Blastocystis tryptophan metabolism: In the presence of E. coli, Blastocystis ST7 is less able to metabolise indole to tryptophan. This study examines the potential for functional variation in horizontally-acquired genes relative to their canonical counterparts, and identifies Blastocystis as a possible producer of tryptophan within the gut.


Subject(s)
Blastocystis/enzymology , Protozoan Proteins/metabolism , Tryptophanase/metabolism , Amino Acid Sequence , Bacteria/classification , Bacteria/enzymology , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Blastocystis/genetics , Blastocystis/metabolism , Gene Transfer, Horizontal , Humans , Indoles/metabolism , Kinetics , Phylogeny , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Sequence Alignment , Tryptophan/metabolism , Tryptophanase/chemistry , Tryptophanase/genetics
19.
Lab Chip ; 10(19): 2605-13, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20689864

ABSTRACT

In blood vessels with luminal diameter less than 300 µm, red blood cells (RBCs) which are smaller in size and more deformable than leukocytes, migrate to the axial centre of the vessel due to flow velocity gradient within the vessels. This phenomenon displaces the leukocytes to the vessel wall and is aptly termed as margination. Here, we demonstrate using microfluidics that stiffer malaria-infected RBCs (iRBCs) behave similar to leukocytes and undergo margination towards the sidewalls. This provides better understanding of the hemodynamic effects of iRBCs in microcirculation and its contribution to pathophysiological outcome relating to cytoadherence to endothelium. In this work, cell margination is mimicked for the separation of iRBCs from whole blood based on their reduced deformability. The malaria infected sample was tested in a simple long straight channel microfluidic device fabricated in polydimethylsiloxane. In this microchannel, cell margination was directed along the channel width with the iRBCs aligning near each sidewall and then subsequently removed using a 3-outlet system, thus achieving separation. Tests were conducted using ring stage and late trophozoite/schizont stage iRBCs. Device performance was quantified by analyzing the distribution of these iRBCs across the microchannel width at the outlet and also conducting flow cytometry analysis. Results indicate recovery of approximately 75% for early stage iRBCs and >90% for late stage iRBCs at the side outlets. The simple and passive system operation makes this technique ideal for on-site iRBCs enrichment in resource-limited settings, and can be applied to other blood cell diseases, e.g. sickle cell anemia and leukemia, characterized by changes in cell stiffness.


Subject(s)
Cell Separation/instrumentation , Erythrocyte Aggregation/physiology , Erythrocytes/pathology , Malaria/blood , Microfluidic Analytical Techniques/instrumentation , Equipment Design , Equipment Failure Analysis , Humans
20.
Microbiome ; 7(1): 30, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30853028

ABSTRACT

BACKGROUND: Blastocystis is a common gut eukaryote detected in humans and animals. It has been associated with gastrointestinal disease in the past although recent metagenomic studies also suggest that it is a member of normal microbiota. This study investigates interactions between pathogenic human isolates belonging to Blastocystis subtype 7 (ST7) and bacterial representatives of the gut microbiota. RESULTS: Generally, Blastocystis ST7 exerts a positive effect on the viability of representative gut bacteria except on Bifidobacterium longum. Gene expression analysis and flow cytometry indicate that the bacterium may be undergoing oxidative stress in the presence of Blastocystis. In vitro assays demonstrate that Blastocystis-induced host responses are able to decrease Bifidobacterium counts. Mice infected with Blastocystis also reveal a decrease in beneficial bacteria Bifidobacterium and Lactobacillus. CONCLUSIONS: This study shows that particular isolates of Blastocystis ST7 cause changes in microbiota populations and potentially lead to an imbalance of the gut microbiota. This study suggests that certain isolates of Blastocystis exert their pathogenic effects through disruption of the gut microbiota and provides a counterpoint to the increasing reports indicating the commensal nature of this ubiquitous parasite.


Subject(s)
Blastocystis Infections/microbiology , Blastocystis/growth & development , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome , Gene Expression Profiling/methods , Animals , Bacterial Proteins/genetics , Bifidobacterium longum/genetics , Bifidobacterium longum/growth & development , Blastocystis/classification , Blastocystis/isolation & purification , Blastocystis/pathogenicity , Coculture Techniques , Disease Models, Animal , Feces/microbiology , Gene Expression Regulation, Bacterial , HT29 Cells , Humans , Lactobacillus/genetics , Lactobacillus/growth & development , Metagenomics , Mice
SELECTION OF CITATIONS
SEARCH DETAIL