Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell Host Microbe ; 30(11): 1570-1588.e7, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36309013

ABSTRACT

Upon pathogen detection, macrophages normally stay sessile in tissues while dendritic cells (DCs) migrate to secondary lymphoid tissues. The obligate intracellular protozoan Toxoplasma gondii exploits the trafficking of mononuclear phagocytes for dissemination via unclear mechanisms. We report that, upon T. gondii infection, macrophages initiate the expression of transcription factors normally attributed to DCs, upregulate CCR7 expression with a chemotactic response, and perform systemic migration when adoptively transferred into mice. We show that parasite effector GRA28, released by the MYR1 secretory pathway, cooperates with host chromatin remodelers in the host cell nucleus to drive the chemotactic migration of parasitized macrophages. During in vivo challenge studies, bone marrow-derived macrophages infected with wild-type T. gondii outcompeted those challenged with MYR1- or GRA28-deficient strains in migrating and reaching secondary organs. This work reveals how an intracellular parasite hijacks chemotaxis in phagocytes and highlights a remarkable migratory plasticity in differentiated cells of the mononuclear phagocyte system.


Subject(s)
Parasites , Toxoplasma , Mice , Animals , Toxoplasma/physiology , Dendritic Cells/physiology , Cell Movement , Macrophages
2.
Trends Parasitol ; 38(6): 450-461, 2022 06.
Article in English | MEDLINE | ID: mdl-35227615

ABSTRACT

The blood-brain barrier (BBB) efficiently protects the central nervous system (CNS) from infectious insults. Yet, the apicomplexan parasite Toxoplasma gondii has a remarkable capability to establish latent cerebral infection in humans and other vertebrates. In addition to the proposed mechanisms for access to the brain parenchyma, recent findings highlight a paramount role played by the BBB in restricting parasite passage and minimizing parasite loads in the brain. Consistent with clinically asymptomatic primary infections in humans, mounting evidence indicates that the original colonization of the brain by T. gondii encompasses previously unappreciated, nondisruptive translocation processes that precede the onset of parasite-limiting immune responses.


Subject(s)
Parasites , Toxoplasma , Animals , Blood-Brain Barrier/parasitology , Brain/parasitology , Humans , Immunity
3.
Elife ; 102021 12 08.
Article in English | MEDLINE | ID: mdl-34877929

ABSTRACT

The cellular barriers of the central nervous system proficiently protect the brain parenchyma from infectious insults. Yet, the single-celled parasite Toxoplasma gondii commonly causes latent cerebral infection in humans and other vertebrates. Here, we addressed the role of the cerebral vasculature in the passage of T. gondii to the brain parenchyma. Shortly after inoculation in mice, parasites mainly localized to cortical capillaries, in preference over post-capillary venules, cortical arterioles or meningeal and choroidal vessels. Early invasion to the parenchyma (days 1-5) occurred in absence of a measurable increase in blood-brain barrier (BBB) permeability, perivascular leukocyte cuffs or hemorrhage. However, sparse focalized permeability elevations were detected adjacently to replicative parasite foci. Further, T. gondii triggered inflammatory responses in cortical microvessels and endothelium. Pro- and anti-inflammatory treatments of mice with LPS and hydrocortisone, respectively, impacted BBB permeability and parasite loads in the brain parenchyma. Finally, pharmacological inhibition or Cre/loxP conditional knockout of endothelial focal adhesion kinase (FAK), a BBB intercellular junction regulator, facilitated parasite translocation to the brain parenchyma. The data reveal that the initial passage of T. gondii to the central nervous system occurs principally across cortical capillaries. The integrity of the microvascular BBB restricts parasite transit, which conversely is exacerbated by the inflammatory response.


Subject(s)
Blood-Brain Barrier/parasitology , Capillaries/physiology , Toxoplasma/physiology , Toxoplasmosis, Cerebral/parasitology , Animals , Brain/parasitology , Female , Male , Mice , Parasite Load , Permeability
4.
PLoS Negl Trop Dis ; 15(9): e0009764, 2021 09.
Article in English | MEDLINE | ID: mdl-34587172

ABSTRACT

The infection by Trypanosoma brucei brucei (T.b.b.), a protozoan parasite, is characterized by an early-systemic stage followed by a late stage in which parasites invade the brain parenchyma in a T cell-dependent manner. Here we found that early after infection effector-memory T cells were predominant among brain T cells, whereas, during the encephalitic stage T cells acquired a tissue resident memory phenotype (TRM) and expressed PD1. Both CD4 and CD8 T cells were independently redundant for the penetration of T.b.b. and other leukocytes into the brain parenchyma. The role of lymphoid cells during the T.b.b. infection was studied by comparing T- and B-cell deficient rag1-/- and WT mice. Early after infection, parasites located in circumventricular organs, brain structures with increased vascular permeability, particularly in the median eminence (ME), paced closed to the sleep-wake regulatory arcuate nucleus of the hypothalamus (Arc). Whereas parasite levels in the ME were higher in rag1-/- than in WT mice, leukocytes were instead reduced. Rag1-/- infected mice showed increased levels of meca32 mRNA coding for a blood /hypothalamus endothelial molecule absent in the blood-brain-barrier (BBB). Both immune and metabolic transcripts were elevated in the ME/Arc of WT and rag1-/- mice early after infection, except for ifng mRNA, which levels were only increased in WT mice. Finally, using a non-invasive sleep-wake cycle assessment method we proposed a putative role of lymphocytes in mediating sleep alterations during the infection with T.b.b. Thus, the majority of T cells in the brain during the early stage of T.b.b. infection expressed an effector-memory phenotype while TRM cells developed in the late stage of infection. T cells and parasites invade the ME/Arc altering the metabolic and inflammatory responses during the early stage of infection and modulating sleep disturbances.


Subject(s)
Central Nervous System Diseases/immunology , Central Nervous System Diseases/parasitology , T-Lymphocyte Subsets/physiology , Trypanosoma brucei brucei , Trypanosomiasis, African/immunology , Trypanosomiasis, African/pathology , Animals , Brain/parasitology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunologic Memory , Leukocytes , Mice , Mice, Knockout , Sleep
5.
Elife ; 92020 11 12.
Article in English | MEDLINE | ID: mdl-33179597

ABSTRACT

Gamma-aminobutyric acid (GABA) serves diverse biological functions in prokaryotes and eukaryotes, including neurotransmission in vertebrates. Yet, the role of GABA in the immune system has remained elusive. Here, a comprehensive characterization of human and murine myeloid mononuclear phagocytes revealed the presence of a conserved and tightly regulated GABAergic machinery with expression of GABA metabolic enzymes and transporters, GABA-A receptors and regulators, and voltage-dependent calcium channels. Infection challenge with the common coccidian parasites Toxoplasma gondii and Neospora caninum activated GABAergic signaling in phagocytes. Using gene silencing and pharmacological modulators in vitro and in vivo in mice, we identify the functional determinants of GABAergic signaling in parasitized phagocytes and demonstrate a link to calcium responses and migratory activation. The findings reveal a regulatory role for a GABAergic signaling machinery in the host-pathogen interplay between phagocytes and invasive coccidian parasites. The co-option of GABA underlies colonization of the host by a Trojan horse mechanism.


Subject(s)
Phagocytes/metabolism , Toxoplasma/physiology , Toxoplasmosis, Animal/parasitology , gamma-Aminobutyric Acid/metabolism , Adoptive Transfer , Animals , Cell Movement , Cells, Cultured , Dendritic Cells/physiology , Female , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR
6.
Am J Trop Med Hyg ; 103(6): 2244-2252, 2020 12.
Article in English | MEDLINE | ID: mdl-33078699

ABSTRACT

Human African trypanosomiasis (HAT) remains a serious public health problem with diagnostic and treatment challenges in many African countries. The absence of a gold-standard biomarker has been a major difficulty for accurate disease staging and treatment follow-up. We therefore attempted to develop a simple, affordable, and noninvasive biomarker for HAT diagnosis and staging. Simultaneous actigraphy and polysomnography as well as cerebrospinal fluid (CSF) white blood cell (WBC) count, trypanosome presence, and C-X-C motif ligand (CXCL)-10 cytokine levels were performed in 20 HAT patients and nine healthy individuals (controls) using standard procedures. The International HIV Dementia Scale (IHDS) was scored in some patients as a surrogate for clinical assessment. From actigraphic parameters, we developed a novel sleep score and used it to determine correlations with other HAT markers, and compared their performance in differentiating between patients and controls and between HAT stages. The novel actigraphy sleep score (ASS) had the following ranges: 0-25 (healthy controls), 67-103 (HAT stage I), 111-126 (HAT intermediate), and 133-250 (HAT stage II). Compared with controls, stage I patients displayed a 7-fold increase in the ASS (P < 0.01), intermediate stage patients a 10-fold increase (P < 0.001), and HAT stage II patients an almost 20-fold increase (P < 0.001). CXCL-10 showed high interindividual differences. White blood cell counts were only marked in HAT stage II patients with a high interindividual variability. The International HIV Dementia Scale score negatively correlated with the ASS. We report the development and better performance of a new biomarker, ASS, for HAT diagnosis, disease staging, and monitoring that needs to be confirmed in large cohort studies.


Subject(s)
Actigraphy/methods , Biomarkers/analysis , Trypanosoma brucei gambiense/isolation & purification , Trypanosomiasis, African/diagnosis , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Leukocyte Count , Male , Middle Aged , Sleep , Trypanosomiasis, African/cerebrospinal fluid , Trypanosomiasis, African/parasitology , Young Adult
7.
Cell Microbiol ; 21(9): e13048, 2019 09.
Article in English | MEDLINE | ID: mdl-31099453

ABSTRACT

The apicomplexan parasite Toxoplasma gondii invades tissues and traverses non-permissive biological barriers in infected humans and other vertebrates. Following ingestion, the parasite penetrates the intestinal wall and disseminates to immune-privileged sites such as the brain parenchyma, after crossing the blood-brain barrier. In the present study, we have established a protocol for high-purification of primary mouse brain endothelial cells to generate stably polarised monolayers that allowed assessment of cellular barrier traversal by T. gondii. We report that T. gondii tachyzoites translocate across polarised monolayers of mouse brain endothelial cells and human intestinal Caco2 cells without significantly perturbing barrier impermeability and with minimal change in transcellular electrical resistance. In contrast, challenge with parasite lysate or LPS increased barrier permeability by destabilising intercellular tight junctions (TJs) and accentuated transmigration of T. gondii. Conversely, reduced phosphorylation of the TJ-regulator focal adhesion kinase (FAK) was observed dose-dependently upon challenge of monolayers with live T. gondii but not with parasite lysate or LPS. Pharmacological inhibition of FAK phosphorylation reversibly altered barrier integrity and facilitated T. gondii translocation. Finally, gene silencing of FAK by shRNA facilitated transmigration of T. gondii across epithelial and endothelial monolayers. Jointly, the data demonstrate that T. gondii infection transiently alters the TJ stability through FAK dysregulation to facilitate transmigration. This work identifies the implication of the TJ regulator FAK in the transmigration of T. gondii across polarised cellular monolayers and provides novel insights in how microbes overcome the restrictiveness of biological barriers.


Subject(s)
Blood-Brain Barrier/parasitology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Toxoplasma/pathogenicity , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Brain/parasitology , Caco-2 Cells , Cell Polarity/physiology , Endothelial Cells/parasitology , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Silencing , Host-Pathogen Interactions , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Phosphorylation , RNA, Small Interfering , Tight Junctions/metabolism , Tight Junctions/parasitology , Virulence/drug effects , Virulence/immunology
8.
PLoS Pathog ; 12(2): e1005442, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26915097

ABSTRACT

Nitric oxide (NO) generated by inducible NO synthase (iNOS) is critical for defense against intracellular pathogens but may mediate inflammatory tissue damage. To elucidate the role of iNOS in neuroinflammation, infections with encephalitogenic Trypanosoma brucei parasites were compared in inos(-/-) and wild-type mice. Inos(-/-) mice showed enhanced brain invasion by parasites and T cells, and elevated protein permeability of cerebral vessels, but similar parasitemia levels. Trypanosome infection stimulated T cell- and TNF-mediated iNOS expression in perivascular macrophages. NO nitrosylated and inactivated pro-inflammatory molecules such as NF-κΒp65, and reduced TNF expression and signalling. iNOS-derived NO hampered both TNF- and T cell-mediated parasite brain invasion. In inos(-/-) mice, TNF stimulated MMP, including MMP9 activity that increased cerebral vessel permeability. Thus, iNOS-generated NO by perivascular macrophages, strategically located at sites of leukocyte brain penetration, can serve as a negative feed-back regulator that prevents unlimited influx of inflammatory cells by restoring the integrity of the blood-brain barrier.


Subject(s)
Blood-Brain Barrier/metabolism , Encephalitis/metabolism , Macrophages, Peritoneal/metabolism , Macrophages/metabolism , Nitric Oxide/metabolism , Animals , Cytokines/metabolism , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Trypanosoma brucei brucei/metabolism
9.
PLoS One ; 9(8): e104951, 2014.
Article in English | MEDLINE | ID: mdl-25111833

ABSTRACT

Numerous abnormalities of the peripheral blood T cell compartment have been reported in human chronic Trypanosoma cruzi infection and related to prolonged antigenic stimulation by persisting parasites. Herein, we measured circulating lymphocytes of various phenotypes based on the differential expression of CD19, CD4, CD27, CD10, IgD, IgM, IgG and CD138 in a total of 48 T. cruzi-infected individuals and 24 healthy controls. Infected individuals had decreased frequencies of CD19+CD27+ cells, which positively correlated with the frequencies of CD4+CD27+ cells. The contraction of CD19+CD27+ cells was comprised of IgG+IgD-, IgM+IgD- and isotype switched IgM-IgD- memory B cells, CD19+CD10+CD27+ B cell precursors and terminally differentiated CD19+CD27+CD138+ plasma cells. Conversely, infected individuals had increased proportions of CD19+IgG+CD27-IgD- memory and CD19+IgM+CD27-IgD+ transitional/naïve B cells. These observations prompted us to assess soluble CD27, a molecule generated by the cleavage of membrane-bound CD27 and used to monitor systemic immune activation. Elevated levels of serum soluble CD27 were observed in infected individuals with Chagas cardiomyopathy, indicating its potentiality as an immunological marker for disease progression in endemic areas. In conclusion, our results demonstrate that chronic T. cruzi infection alters the distribution of various peripheral blood B cell subsets, probably related to the CD4+ T cell deregulation process provoked by the parasite in humans.


Subject(s)
Antibodies, Protozoan/blood , Antigens, CD/blood , B-Lymphocyte Subsets/immunology , Chagas Disease/immunology , Immunologic Memory/immunology , Antigens, CD/biosynthesis , B-Lymphocyte Subsets/cytology , CD4-Positive T-Lymphocytes/immunology , Chagas Disease/parasitology , Chronic Disease , Humans , Immunoglobulin D/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Immunophenotyping , Plasma Cells/immunology , Precursor Cells, B-Lymphoid/immunology , Trypanosoma cruzi/immunology
10.
J Med Chem ; 56(24): 9861-73, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24283924

ABSTRACT

Novel methods for treatment of African trypanosomiasis, caused by infection with Trypanosoma brucei are needed. Cordycepin (3'-deoxyadenosine, 1a) is a powerful trypanocidal compound in vitro but is ineffective in vivo because of rapid metabolic degradation by adenosine deaminase (ADA). We elucidated the structural moieties of cordycepin required for trypanocidal activity and designed analogues that retained trypanotoxicity while gaining resistance to ADA-mediated metabolism. 2-Fluorocordycepin (2-fluoro-3'-deoxyadenosine, 1b) was identified as a selective, potent, and ADA-resistant trypanocidal compound that cured T. brucei infection in mice. Compound 1b is transported through the high affinity TbAT1/P2 adenosine transporter and is a substrate of T. b. brucei adenosine kinase. 1b has good preclinical properties suitable for an oral drug, albeit a relatively short plasma half-life. We present a rapid and efficient synthesis of 2-halogenated cordycepins, also useful synthons for the development of additional novel C2-substituted 3'-deoxyadenosine analogues to be evaluated in development of experimental therapeutics.


Subject(s)
Deoxyadenosines/pharmacology , Trypanocidal Agents/pharmacology , Trypanosomiasis, African/drug therapy , Animals , Cell Survival/drug effects , Cells, Cultured , Deoxyadenosines/chemical synthesis , Deoxyadenosines/chemistry , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry , Trypanosoma brucei brucei/drug effects , Trypanosoma brucei rhodesiense/drug effects
11.
Microbes Infect ; 12(5): 359-63, 2010 May.
Article in English | MEDLINE | ID: mdl-20123034

ABSTRACT

Trypanosoma cruzi-specific immune responses were evaluated in a total of 88 subjects living in areas endemic of Chagas disease of Argentina by IFN-gamma ELISPOT assays and immunoblotting. Positive T. cruzi antigen-induced IFN-gamma responses were detected in 42% of subjects evaluated (15/26 positive by conventional serology and 22/62 seronegative subjects). Using immunoblotting, T. cruzi-specific IgG reactivity was detected in all seropositive subjects and in 11% (7/61) of subjects negative by conventional serology. Measurements of T cell responses and antibodies by immunoblotting, in conjunction with conventional serology, might enhance the capability of detection of exposure to T. cruzi in endemic areas.


Subject(s)
Chagas Disease/immunology , Trypanosoma cruzi/immunology , Adult , Aged , Antibodies, Protozoan/blood , Argentina , Endemic Diseases , Female , Humans , Immunoblotting , Immunoglobulin G/blood , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...