Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Trop Biomed ; 38(3): 338-342, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34508341

ABSTRACT

Leishmaniasis is a neglected and endemic disease that affects poorest population mainly in developing countries. Thymus provides an essential complex environment for T cell maturation and differentiation during leishmania infection. The aim of this study was to investigate the pathological alterations of the Thymus during early Leishmania amazonensis murine infection. BALB/c mice were infected with 105 amastigotes for 24 h, 3 days, 7 days, 15 days or 30 days. At different times of infection, the relative weight of the Thymus was obtained, and the Thymus cellularity was determined by counting total cells of one thymic lobe. The thymic lobe was, alternatively, processed for standard Haematoxylin and Eosin protocol. Our results suggest thymic alteration during the early days of BALB/c mice infection with L. amazonensis. The thymic hypertrophy was accompanied by histological alterations in Thymus architecture with thickening cortex at 3 days p.i. and loss of an evident delimitation between the cortex and medulla at 7 days p.i. when compared to the control mice. That is the first time that Thymus hypertrophy was observed during the early leishmaniasis. However, how it may contribute to infection susceptibility requires further investigation.


Subject(s)
Leishmaniasis , Thymus Gland , Animals , Hypertrophy , Leishmania mexicana , Leishmaniasis/pathology , Mice , Mice, Inbred BALB C , Thymus Gland/parasitology , Thymus Gland/pathology
2.
Front Immunol ; 11: 953, 2020.
Article in English | MEDLINE | ID: mdl-32508833

ABSTRACT

Background: Visceral leishmaniasis/HIV-co-infected patients (VL/HIV) accounts for around 8% of VL reported cases in Brazil. Relapses of Leishmania infection after anti-leishmanial treatment constitute a great challenge in the clinical practice because of the disease severity and drug resistance. We have shown that non-relapsing-VL/HIV (NR-) evolved with increase of CD4+ T-cell counts and reduction of activated CD4+ and CD8+ T cells after anti-leishmanial treatment. This immune profile was not observed in relapsing-VL/HIV patients (R-), indicating a more severe immunological compromising degree. Elevated activation status may be related to a deficient immune reconstitution and could help to explain the frequent relapses in VL/HIV co-infection. Our aim was to evaluate if this gain of T cells was related to changes in the peripheral TCRVß repertoire and inflammatory status, as well as the possible thymus involvement in the replenishment of these newly formed T lymphocytes. Methods: VL/HIV patients, grouped into non-relapsing (NR- = 6) and relapsing (R- = 12) were evaluated from the active phase up to 12 months post-treatment (mpt). HIV-infected patients (non-VL) and healthy subjects (HS) were included. The TCRVß repertoire was evaluated ex vivo by flow cytometry, whereas the plasmatic cytokine levels were assessed by Luminex assay. To evaluate the thymic output, DNA was extracted from PBMCs for TCR rearrangement excision circles (TREC) quantification by qPCR. Results: VL/HIV cases presented an altered mobilization profile (expansions or retractions) of the TCRVß families when compared to HS independent of the follow-up phase (p < 0.05). TCRVß repertoire on CD4+ T-cells was more homogeneous in the NR-VL/HIV cases, but heterogeneous on CD8+ T-cells, since different Vß-families were mobilized. NR-VL/HIV had the inflammatory pattern reduced after 6 mpt. Importantly, VL/HIV patients showed number of TREC copies lower than controls during all follow-up. An increase of recent thymic emigrants was observed in NR-VL/HIV individuals at 10 mpt compared to R- patients (p < 0.01), who maintained lower TREC contents than the HIV controls. Conclusions: VL/HIV patients that maintain the thymic function, thus generating new T-cells, seem able to replenish the T lymphocyte compartment with effector cells, then enabling parasite control.


Subject(s)
Coinfection , HIV Infections/immunology , Leishmaniasis, Visceral/immunology , Lymphocyte Activation , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , CD4-CD8 Ratio , Case-Control Studies , Cell Proliferation , Cytokines/blood , HIV Infections/drug therapy , HIV Infections/metabolism , HIV Infections/virology , Humans , Leishmaniasis, Visceral/drug therapy , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Phenotype , Prospective Studies , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recurrence , T-Lymphocyte Subsets/parasitology , T-Lymphocyte Subsets/virology , Thymus Gland/parasitology , Thymus Gland/virology , Time Factors , Treatment Outcome
3.
Immun Inflamm Dis ; 8(2): 127-139, 2020 06.
Article in English | MEDLINE | ID: mdl-32207879

ABSTRACT

INTRODUCTION: In canine visceral leishmaniasis (CVL), lymphopenia, and the disorganization of lymphoid organs such as spleen and lymph nodes have been demonstrated. However, the involvement of thymus in CVL has not been evaluated so far. Herein, we investigated whether the thymus can be colonized by Leishmania infantum in naturally infected dogs. METHODS: Thymus were obtained from 16 of 58 dogs and samples of this organ were submitted to immunohistochemistry for laminin and fibronectin detection, histopathology, in situ hybridization and polymerase chain reaction (PCR) targeting the gene ITS-1 for Leishmania and sequenced. Samples of spleen, skin and popliteal lymph nodes were collected and submitted to immunohistochemistry and parasitological culture followed by multilocus enzyme electrophoresis. RESULTS: L. infantum was identified in all dogs. DNA and amastigote forms of Leishmania were detected in the thymus from 16 dogs by PCR and in eight by immunohistochemistry. Besides thymus, parasites were detected in spleen, lymph nodes, and skin. A granulomatous or pyogranulomatous thymitis was observed in eight dogs associated to intact amastigotes forms of this parasite. Fibronectin deposition in thymus was higher in dogs with more clinical signs. CONCLUSIONS: These results demonstrate that the thymus of dogs can be parasitized by L. infantum, which may generate inflammatory reactions leading to alterations in thymic microarchitecture.


Subject(s)
DNA, Protozoan/isolation & purification , Leishmania infantum/isolation & purification , Leishmaniasis, Visceral/veterinary , Thymus Gland/parasitology , Animals , DNA, Protozoan/genetics , Dog Diseases/parasitology , Dogs , Female , Leishmania infantum/genetics , Leishmaniasis, Visceral/parasitology , Leishmaniasis, Visceral/pathology , Lymph Nodes/parasitology , Male , Parasite Load , Skin/parasitology , Spleen/parasitology
4.
Article in English | MEDLINE | ID: mdl-31355153

ABSTRACT

Detrimental effects of malnutrition on immune responses to pathogens have long been recognized and it is considered a main risk factor for various infectious diseases, including visceral leishmaniasis (VL). Thymus is a target of both malnutrition and infection, but its role in the immune response to Leishmania infantum in malnourished individuals is barely studied. Because we previously observed thymic atrophy and significant reduction in cellularity and chemokine levels in malnourished mice infected with L. infantum, we postulated that the thymic microenvironment is severely compromised in those animals. To test this, we analyzed the microarchitecture of the organ and measured the protein abundance in its interstitial space in malnourished BALB/c mice infected or not with L. infantum. Malnourished-infected animals exhibited a significant reduction of the thymic cortex:medulla ratio and altered abundance of proteins secreted in the thymic interstitial fluid. Eighty-one percent of identified proteins are secreted by exosomes and malnourished-infected mice showed significant decrease in exosomal proteins, suggesting that exosomal carrier system, and therefore intrathymic communication, is dysregulated in those animals. Malnourished-infected mice also exhibited a significant increase in the abundance of proteins involved in lipid metabolism and tricarboxylic acid cycle, suggestive of a non-proliferative microenvironment. Accordingly, flow cytometry analysis revealed decreased proliferation of single positive and double positive T cells in those animals. Together, the reduced cortical area, decreased proliferation, and altered protein abundance suggest a dysfunctional thymic microenvironment where T cell migration, proliferation, and maturation are compromised, contributing for the thymic atrophy observed in malnourished animals. All these alterations could affect the control of the local and systemic infection, resulting in an impaired response to L. infantum infection.


Subject(s)
Host-Pathogen Interactions/immunology , Leishmania infantum/immunology , Leishmaniasis, Visceral/immunology , Malnutrition/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Animals , Biological Transport , Cell Movement , Cell Proliferation , Citric Acid Cycle/genetics , Citric Acid Cycle/immunology , Exosomes/immunology , Exosomes/metabolism , Exosomes/parasitology , Extracellular Fluid/immunology , Extracellular Fluid/metabolism , Extracellular Fluid/parasitology , Galectin 1/genetics , Galectin 1/immunology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Immunity, Innate , Leishmania infantum/growth & development , Leishmaniasis, Visceral/genetics , Leishmaniasis, Visceral/metabolism , Leishmaniasis, Visceral/parasitology , Lipid Metabolism , Male , Malnutrition/genetics , Malnutrition/metabolism , Malnutrition/parasitology , Mice , Mice, Inbred BALB C , Plasminogen/genetics , Plasminogen/immunology , Proteome/genetics , Proteome/immunology , T-Lymphocytes/parasitology , Thymus Gland/metabolism , Thymus Gland/parasitology
5.
J Pineal Res ; 65(3): e12510, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29781553

ABSTRACT

Although the exact etiology of Chagas disease is not completely elucidated, thymic atrophy and oxidative stress are believed to be important contributors to the pathogenesis during acute Trypanosoma cruzi (T. cruzi) infection. We hypothesized that exogenous melatonin, administered by gavage (5 mg/kg, p.o., gavage) to young (5 weeks old) and middle-aged (18 months old) male Wistar rats, would modulate thymic oxidative damage and reverse the age-related thymus regression during T. cruzi acute infection. Increased levels of superoxide anion (O2- ) were detected in the thymus of infected animals, and treatment with melatonin reverted this response. We found reduced TBARS levels as well as a significant increase in superoxide dismutase (SOD) activity in the thymus of all middle-aged melatonin-treated animals, infected or not with T. cruzi. Furthermore, melatonin increased the thymic expression of SOD1 and SOD2 in middle-aged control animals. Nox2 expression was not affected by melatonin treatment in young or middle-aged animals. Melatonin reverted the age-related thymic regression as revealed by the increase in thymus weight, total number of thymocytes, and reduction in age-related accumulation of double-negative thymocytes. This is the first report to directly examine the effects of melatonin treatment on the thymic antioxidant/oxidant status and thymic changes during T. cruzi infection. Our results revealed new antioxidant features that turn melatonin a potentially useful compound for the treatment of Chagas disease, a condition in which an excessive oxidative damage occurs.


Subject(s)
Antioxidants/pharmacology , Chagas Disease/metabolism , Melatonin/pharmacology , Oxidative Stress/drug effects , Thymus Gland/metabolism , Trypanosoma cruzi , Animals , Chagas Disease/drug therapy , Chagas Disease/pathology , Male , Rats , Rats, Wistar , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism , Superoxides/metabolism , Thymocytes/metabolism , Thymocytes/parasitology , Thymocytes/pathology , Thymus Gland/parasitology , Thymus Gland/pathology
6.
PLoS One ; 10(10): e0122566, 2015.
Article in English | MEDLINE | ID: mdl-26469517

ABSTRACT

Chagas disease is a worldwide public health problem. Although the vectorial transmission of Chagas disease has been controlled in Brazil there are other ways of transmission, such as the ingestion of T. cruzi contaminated food, which ensures the continuation of this zoonosis. Here, we demonstrate the influence of the inoculation route on the establishment and development of the SC2005 T. cruzi strain infection in mice. Groups of Swiss mice were infected intragastrically (IG) or intraperitoneally (IP) with the T. cruzi SC2005 strain derived from an outbreak of oral Chagas disease. The results revealed that 100% of IP infected mice showed parasitemia, while just 36% of IG infected showed the presence of the parasite in blood. The parasitemia peaks were later and less intense in the IG infected mice. Mortality of the IP infected animals was more intense and earlier when compared to the IG infected mice. In the IP infected mice leucopenia occurred in the early infection followed by leucocytosis, correlating positively with the increase of the parasites. However, in the IG infected mice only an increase in monocytes was observed, which was positively correlated with the increase of the parasites. Histopathological analyses revealed a myotropic pattern of the SC2005 strain with the presence of inflammatory infiltrates and parasites in different organs of the animals infected by both routes as well as fibrosis foci and collagen redistribution. The flow cytometric analysis demonstrated a fluctuation of the T lymphocyte population in the blood, spleen and mesenteric lymph nodes of the infected animals. T. cruzi DNA associated with the presence of inflammatory infiltrates was detected by PCR in the esophagus, stomach and intestine of all infected mice. These findings are important for the understanding of the pathogenesis of T. cruzi infection by both inoculation routes.


Subject(s)
Chagas Disease/epidemiology , Chagas Disease/parasitology , Disease Outbreaks , Animals , Brazil/epidemiology , Chagas Disease/blood , Chagas Disease/transmission , Female , Humans , Leukocyte Count , Mice , Parasitemia/blood , Parasitemia/epidemiology , Parasitemia/parasitology , Parasitemia/transmission , Spleen/parasitology , Thymus Gland/parasitology
7.
Parasite Immunol ; 37(7): 349-61, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25886201

ABSTRACT

Although thymic atrophy and apoptosis of the double-positive (DP) T cells have been reported in murine malaria, comparative studies investigating the effect of lethal and nonlethal Plasmodium infections on the thymus are lacking. We assessed the effects of P. yoelii lethal (17XL) and nonlethal (17XNL) infections on thymic T cells. Both strains affected the thymus. 17XL infection induced DP T-cell apoptosis and a selective decrease in surface CD8 expression on developing thymocytes. By contrast, more severe but reversible effects were observed during 17XNL infection. DP T cells underwent apoptosis, and proliferation of both DN and DP cells was affected around peak parasitemia. A transient increase in surface CD8 expression on thymic T cells was also observed. Adult thymic organ culture revealed that soluble serum factors, but not IFN-γ or TNF-α, contributed to the observed effects. Thus, lethal and nonlethal malarial infections led to multiple disparate effects on thymus. These parasite-induced thymic changes are expected to impact the naïve T-cell repertoire and the subsequent control of the immune response against the parasite. Further investigations are required to elucidate the mechanism responsible for these disparate effects, especially the reversible involution of the thymus in case of nonlethal infection.


Subject(s)
Apoptosis/immunology , CD8 Antigens/metabolism , Malaria/immunology , Plasmodium yoelii/immunology , T-Lymphocytes/immunology , Animals , Cell Proliferation , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Malaria/parasitology , Male , Mice , Mice, Inbred BALB C , Parasitemia/immunology , Plasmodium yoelii/pathogenicity , Thymus Gland/parasitology , Thymus Gland/pathology , Tumor Necrosis Factor-alpha/metabolism
9.
Article in Chinese | MEDLINE | ID: mdl-24812878

ABSTRACT

OBJECTIVE: To study the pathological damage of thymus and thymus cell apoptosis of male rats infected with Toxoplasma gondii. METHODS: Fifty Wistar male rats (7-8-week-old) were randomly divided into infection group (40) and control group (10). Rats in infection group were infected with 5 x 10(4) tachyzoites by intraperitoneal injection, while those in control group received same volume of PBS. On the 3rd, 6th, 9th and 12th day post infection, ten rats from infection group and two from control group were sacrificed, the thymus glands were removed. The thymus tissue sections were stained with hematoxylin and eosin (HE) for observation on histopathological changes. Single thymus cell suspensions were prepared. Cell cycle analysis was performed by flow cytometry, and proliferation index was calculated. Thymus frozen sections were stained with Hoechst 33258, and morphologic changes in apoptotic nuclei were observed under fluorescence microscope. Expression of Bcl-2 and Bax proteins were determined by using immunohistochemistry. RESULTS: Microscopic examination showed that pathological changes occurred in thymus grand on the 3rd day after infection. The space between connective tissue capsules was widened, cells in cortex and medulla cells were sparse, and more phagocytes and extravasated blood were found in thymus. On the 6th day post infection the thymus damage was aggravated, and no significant improvement was seen on day 12. On the 3rd, 6th, 9th and 12th day after infection, thymocyte proliferation index was (11.15 +/- 0.99)%, (6.17 +/- 1.02)%, (5.45 +/- 0.96)% and (6.63 +/- 1.52)%, respectively, and each of them was significantly lower than that of the control [(13.81 +/- 1.18)%] (P < 0.01). On the 3rd day after infection, the number of apoptotic cells increased, significantly increased on day 6, and there was no much difference in the number of apoptotic cells between day 6 and day 12. The immunohistochemistry results showed that on the 3rd, 6th, 9th and 12th day post-infection, the gray scale value of Bax positive cells was 88.21 +/- 4.74, 64.69 +/- 6.82, 83.62 +/- 5.79, and 101.09 +/- 6.72, respectively, and each of them was significantly lower than that of the control (128.69 +/- 8.95) (P < 0.01), while there was no significant change in the Bcl-2 protein level (P > 0.05). CONCLUSION: T. gondii causes severe pathological damage in host thymus tissue with a decrease in the proliferation index, an increase in the number of apoptotic cells, and high expression of Bax protein.


Subject(s)
Apoptosis , Thymus Gland/pathology , Thymus Gland/parasitology , Toxoplasmosis/pathology , Animals , Male , Rats , Rats, Wistar , Thymus Gland/cytology , Toxoplasma , bcl-2-Associated X Protein/metabolism
10.
Cell Immunol ; 279(1): 53-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23089194

ABSTRACT

The thymus plays a crucial role in the generation of T-cells, and so our laboratory has been interested in the study of the intrathymic events that occur during infection diseases and may cause disruption in its functions. Previously, we showed that thymus from experimentally Plasmodium berghei-infected mice present histological alterations with high levels of apoptosis, changes in cell migration-related molecules, and premature egress of immature thymocytes to periphery. In addition, parasites were found inside the thymus. In this work we investigated alterations in the expression pattern and activity of matrix metalloproteinases MMP-2 and -9, and their tissue inhibitors, TIMP-1 and TIMP-2. Our results show enhanced expression and widespread distribution of these molecules in thymus from infected animals. Also, the presence of active MMP-2 was detected. These data are suggestive of MMPs and TIMPs importance in the earlier observed changes in the extracellular matrix during thymic alterations after plasmodium infection.


Subject(s)
Malaria/parasitology , Parasitemia/parasitology , Plasmodium berghei/physiology , Thymus Gland/parasitology , Animals , Gene Expression Profiling , Host-Parasite Interactions , Immunohistochemistry , Malaria/genetics , Malaria/metabolism , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Parasitemia/genetics , Parasitemia/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thymus Gland/metabolism , Thymus Gland/pathology , Time Factors , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Tissue Inhibitor of Metalloproteinase-2/genetics , Tissue Inhibitor of Metalloproteinase-2/metabolism
11.
J Immunol ; 189(9): 4371-8, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23024277

ABSTRACT

CD4(+) Th2 cytokine responses promote the development of allergic inflammation and are critical for immunity to parasitic helminth infection. Recent studies highlighted that basophils can promote Th2 cytokine-mediated inflammation and that phenotypic and functional heterogeneity exists between classical IL-3-elicited basophils and thymic stromal lymphopoietin (TSLP)-elicited basophils. However, whether distinct basophil populations develop after helminth infection and their relative contributions to anti-helminth immune responses remain to be defined. After Trichinella spiralis infection of mice, we show that basophil responses are rapidly induced in multiple tissue compartments, including intestinal-draining lymph nodes. Trichinella-induced basophil responses were IL-3-IL-3R independent but critically dependent on TSLP-TSLPR interactions. Selective depletion of basophils after Trichinella infection impaired infection-induced CD4(+) Th2 cytokine responses, suggesting that TSLP-dependent basophils augment Th2 cytokine responses after helminth infection. The identification and functional classification of TSLP-dependent basophils in a helminth infection model, coupled with their recently described role in promoting atopic dermatitis, suggests that these cells may be a critical population in promoting Th2 cytokine-associated inflammation in a variety of inflammatory or infectious settings. Collectively, these data suggest that the TSLP-basophil pathway may represent a new target in the design of therapeutic intervention strategies to promote or limit Th2 cytokine-dependent immunity and inflammation.


Subject(s)
Basophils/immunology , Cytokines/biosynthesis , Intestinal Mucosa/immunology , Th2 Cells/immunology , Thymus Gland/immunology , Trichinella spiralis/immunology , Trichinellosis/immunology , Animals , Basophils/parasitology , Basophils/pathology , Cytokines/physiology , Inflammation/immunology , Inflammation/metabolism , Inflammation/parasitology , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Stromal Cells/immunology , Stromal Cells/parasitology , Stromal Cells/pathology , Th2 Cells/parasitology , Th2 Cells/pathology , Thymus Gland/parasitology , Thymus Gland/pathology , Trichinellosis/metabolism , Trichinellosis/pathology , Thymic Stromal Lymphopoietin
12.
Immunobiology ; 216(10): 1148-54, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21601941

ABSTRACT

We have previously showed alterations in the thymus during experimental infection with Plasmodium berghei, the causative agent of Malaria. Such alterations comprised histological changes with loss of delimitation between cortical and medullar regions, a profound atrophy with depletion of CD4(+)CD8(+) double-positive (DP) thymocytes, and severe changes in the expression of cell migration-related molecules, belonging to the extracellular matrix and chemokine protein families. Taken together, these considerations prompted us to evaluate if the acute thymic atrophy observed during Plasmodium infection was correlated with increased apoptotic levels of thymocytes or with their premature emigration to the periphery. Our results confirmed that the marked reduction of the thymus weight in infected animals was accompanied by histological alterations, which included a very large number of cells showing nuclear condensation and karyorrhectic changes surrounded by histiocytes suggesting increased levels of apoptosis. This was confirmed by immunohistochemistry and flow cytometry techniques. In order to verify if an accelerated emigration of thymic cells to the peripheral lymphoid organs was also occurring we analyzed the spleen and mesenteric lymph nodes from control and infected mice. No significant differences were found in the spleen, but were seen after 14 days of infection between control and infected mice in the mesenteric lymph nodes. The main alteration was the presence of double negative (CD4(-)CD8(-)) and double positive (CD4(+)CD8(+)) cells. We concluded that both apoptosis of thymocytes and premature egress of immature cells take place during infection. Additional studies will be necessary to verify how such alterations might influence the systemic immune response to the parasite.


Subject(s)
Apoptosis/immunology , Malaria/immunology , Plasmodium berghei/immunology , Thymocytes/immunology , Thymus Gland/immunology , Animals , CD4 Antigens/analysis , CD8 Antigens/analysis , Immunophenotyping , Lymph Nodes/immunology , Lymph Nodes/metabolism , Malaria/parasitology , Malaria/pathology , Male , Mice , Mice, Inbred BALB C , Thymocytes/metabolism , Thymus Gland/parasitology , Thymus Gland/pathology
13.
PLoS Pathog ; 6(5): e1000877, 2010 May 06.
Article in English | MEDLINE | ID: mdl-20463809

ABSTRACT

Immunization with irradiated sporozoites is currently the most effective vaccination strategy against liver stages of malaria parasites, yet the mechanisms underpinning the success of this approach are unknown. Here we show that the complete development of protective CD8+ T cell responses requires prolonged antigen presentation. Using TCR transgenic cells specific for the malaria circumsporozoite protein, a leading vaccine candidate, we found that sporozoite antigen persists for over 8 weeks after immunization--a remarkable finding since irradiated sporozoites are incapable of replication and do not differentiate beyond early liver stages. Persisting antigen was detected in lymphoid organs and depends on the presence of CD11c+ cells. Prolonged antigen presentation enhanced the magnitude of the CD8+ T cell response in a number of ways. Firstly, reducing the time primed CD8+ T cells were exposed to antigen in vivo severely reduced the final size of the developing memory population. Secondly, fully developed memory cells expanded in previously immunized mice but not when transferred to naïve animals. Finally, persisting antigen was able to prime naïve cells, including recent thymic emigrants, to become functional effector cells capable of eliminating parasites in the liver. Together these data show that the optimal development of protective CD8+ T cell immunity against malaria liver stages is dependent upon the prolonged presentation of sporozoite-derived antigen.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , Liver/immunology , Malaria/immunology , Plasmodium yoelii/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/parasitology , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/parasitology , Female , Immunization , Immunologic Memory/immunology , Liver/parasitology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Plasmodium yoelii/growth & development , Spleen/immunology , Spleen/parasitology , Sporozoites/growth & development , Sporozoites/immunology , Thymus Gland/immunology , Thymus Gland/parasitology
14.
Morfologiia ; 137(5): 36-9, 2010.
Article in Russian | MEDLINE | ID: mdl-21500430

ABSTRACT

Using the methods of light microscopy, eosinophil topography, quantitative and qualitative changes (degranulation level of and a cationic protein content) were studied in the thymus and bursa of 36 herring gull nestlings Larus argentatus mongolicus (Suskin, 1925) 2 weeks after experimental infestation with gull-tapeworm Diphyllobothrium dendriticum (Nitsch, 1824). Eosinophils in the thymus were located in trabecules, mainly close to the blood vessels, thymic (Hassall's) corpuscles and also directly inside them, while in the bursa they were found within the internodular space. As compared with the control bird counts, relative eosinophil count in the birds with an average invasion intensity was increased 3.8 times in the thymus and 2.5 times in the bursa. In birds with high invasion intensity, these counts were increased 4 times in the thymus and 1.2 times in the bursa.


Subject(s)
Bursa of Fabricius , Charadriiformes , Diphyllobothriasis , Diphyllobothrium/immunology , Eosinophils/immunology , Thymus Gland , Animals , Bursa of Fabricius/immunology , Bursa of Fabricius/parasitology , Bursa of Fabricius/pathology , Charadriiformes/immunology , Charadriiformes/parasitology , Diphyllobothriasis/immunology , Diphyllobothriasis/parasitology , Diphyllobothriasis/pathology , Thymus Gland/immunology , Thymus Gland/parasitology , Thymus Gland/pathology
15.
Immunology ; 129(2): 248-56, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19824923

ABSTRACT

We previously showed alterations in the thymus during experimental infection with Plasmodium berghei. Such alterations comprised histological changes, with loss of cortical-medullary limits, and the intrathymic presence of parasites. As the combination of chemokines, adhesion molecules and extracellular matrix (ECM) is critical to appropriate thymocyte development, we analysed the thymic expression of ECM ligands and receptors, as well as chemokines and their respective receptors during the experimental P. berghei infection. Increased expression of ECM components was observed in thymi from infected mice. In contrast, down-regulated surface expression of fibronectin and laminin receptors was observed in thymocytes from these animals. Moreover, in thymi from infected mice there was increased CXCL12 and CXCR4, and a decreased expression of CCL25 and CCR9. An altered thymocyte migration towards ECM elements and chemokines was seen when the thymi from infected mice were analysed. Evaluation of ex vivo migration patterns of CD4/CD8-defined thymocyte subpopulations revealed that double-negative (DN), and CD4(+) and CD8(+) single-positive (SP) cells from P. berghei-infected mice have higher migratory responses compared with controls. Interestingly, increased numbers of DN and SP subpopulations were found in the spleens of infected mice. Overall, we show that the thymic atrophy observed in P. berghei-infected mice is accompanied by thymic microenvironmental changes that comprise altered expression of thymocyte migration-related molecules of the ECM and chemokine protein families, which in turn can alter the thymocyte migration pattern. These thymic disturbances may have consequences for the control of the immune response against this protozoan.


Subject(s)
Cell Movement/immunology , Malaria/immunology , Plasmodium berghei/immunology , Precursor Cells, T-Lymphoid/metabolism , Thymus Gland/metabolism , Animals , CD4 Antigens/biosynthesis , CD8 Antigens/biosynthesis , Cells, Cultured , Chemokines/biosynthesis , Chemokines/genetics , Chemokines/immunology , Gene Expression Regulation , Malaria/parasitology , Malaria/pathology , Male , Mice , Mice, Inbred BALB C , Models, Animal , Plasmodium berghei/pathogenicity , Precursor Cells, T-Lymphoid/immunology , Precursor Cells, T-Lymphoid/parasitology , Precursor Cells, T-Lymphoid/pathology , Receptors, Cytoadhesin/biosynthesis , Receptors, Cytoadhesin/genetics , Receptors, Cytoadhesin/immunology , Receptors, Fibronectin/biosynthesis , Receptors, Fibronectin/genetics , Receptors, Fibronectin/immunology , Receptors, Laminin/biosynthesis , Receptors, Laminin/genetics , Receptors, Laminin/immunology , Thymus Gland/immunology , Thymus Gland/parasitology , Thymus Gland/pathology
16.
Proc Natl Acad Sci U S A ; 106(40): 17123-8, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19805095

ABSTRACT

Interleukin (IL)-10 is an immunoregulatory cytokine that is produced by diverse cell populations. Studies in mice suggest that the cellular source of IL-10 is a key determinant in various disease pathologies, yet little is known regarding the control of tissue-specific human IL-10 expression. To assess cell type-specific human IL-10 regulation, we created a human IL-10 transgenic mouse with a bacterial artificial chromosome (hIL10BAC) in which the IL10 gene is positioned centrally. Since human IL-10 is biologically active in the mouse, we could examine the in vivo capacity of tissue-specific human IL-10 expression to recapitulate IL-10-dependent phenotypes by reconstituting Il10(-/-) mice (Il10(-/-)/hIL10BAC). In response to LPS, Il10(-/-)/hIL10BAC mice proficiently regulate IL-10-target genes and normalize sensitivity to LPS toxicity via faithful human IL-10 expression from macrophages and dendritic cells. However, in the Leishmania donovani model of pathogen persistence, Il10(-/-)/hIL10BAC mice did not develop the characteristic IL-10(+)IFN-gamma(+)CD4 T cell subset thought to mediate persistence and, like Il10(-/-) mice, cleared the parasites. Furthermore, the IL-10-promoting cytokine IL-27 failed to regulate transgenic human IL-10 production in CD4(+) T cells in vitro which together suggests that the hIL10BAC encodes for weak T cell-specific IL-10 expression. Thus, the hIL10BAC mouse is a model of human gene structure and function revealing tissue-specific regulatory requirements for IL-10 expression which impacts disease outcomes.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Gene Expression Profiling , Interleukin-10/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Female , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Interleukin-10/metabolism , Leishmania donovani/growth & development , Leishmaniasis/genetics , Leishmaniasis/metabolism , Leishmaniasis/parasitology , Lipopolysaccharides/pharmacology , Liver/metabolism , Liver/parasitology , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism , Spleen/parasitology , Spleen/pathology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Thymus Gland/metabolism , Thymus Gland/parasitology , Thymus Gland/pathology
17.
Parasitol Res ; 103(4): 829-38, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18604654

ABSTRACT

P2X7 is a member of the purinergic receptors family, with extracellular adenosine triphosphate (ATP) as the main agonist, promoting cations influx and membrane permeabilization that can lead to cell death. We previously proposed that extracellular ATP is involved in thymus atrophy induced by Trypanosoma cruzi infection through the induction of CD4+/CD8+ double-positive cell death and that P2X7 could be involved in this process. To further elucidate this possibility raised by in vitro assays, in this study, we used P2X7-/- mice and observed no difference in thymus atrophy or parasitemia when compared to C57Bl/6. We then decided to investigate other aspects of purinergic receptor interplay that could be better evidenced by the infection and observed that (1) thymocytes from infected and noninfected C57Bl/6 mice express P2X4 and P2X7 receptors (Western blotting), but ATP-induced membrane permeabilization only occurs in thymocytes from infected mice; (2) peritoneal macrophages from noninfected C57Bl/6 mice (P2X4+ and P2X7+) are permeabilized by ATP. Although macrophages from infected C57Bl/6 mice are P2X7- but P2X4+, they are resistant to ATP, either through permeabilization or Ca++ influx (fluorimetry); (3) using noninfected P2X7-/- mice, C57Bl/6 infected mice, and different agonistic stimuli, we observed interesting cross-talks among P2X and P2Y receptors (flow cytometry).


Subject(s)
Receptors, Purinergic P2/metabolism , Thymus Gland/pathology , Thymus Gland/parasitology , Trypanosoma cruzi/physiology , Adenosine Triphosphate , Animals , Atrophy , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Membrane Permeability , Cells, Cultured , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parasitemia , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2X7
18.
Cell Immunol ; 253(1-2): 1-4, 2008.
Article in English | MEDLINE | ID: mdl-18635160

ABSTRACT

The primary function of the thymus is to develop immature T-cells into cells that further in the periphery will be able to carry out immune functions. The Literature has shown that thymus can be a target for many pathogens and severe structural alterations take place in this organ during infectious diseases. Here, we investigated if thymus is also a target organ during experimental malaria infection by analyzing the presence of parasites inside the organ and histological alterations in thymuses from Plasmodium berghei NK65-infected BALB/c. After 14 days of infection, parasites were found inside the thymus that presented a profound atrophy with total loss of its architecture. We propose that the presence of parasites in the thymus induces histological modifications that alter the microenvironment, impairing by consequence the successful T cell development. Additional studies are currently being developed in our laboratory to verify if such thymic alterations can influence the systemic immune response to the parasite.


Subject(s)
Malaria , Plasmodium berghei/immunology , Thymus Gland , Animals , Malaria/immunology , Malaria/parasitology , Malaria/pathology , Male , Mice , Mice, Inbred BALB C , Plasmodium berghei/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , T-Lymphocytes/physiology , Thymus Gland/immunology , Thymus Gland/parasitology , Thymus Gland/pathology
19.
Brain Behav Immun ; 21(7): 890-900, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17412557

ABSTRACT

C57BL/6 mice infected with Trypanosoma cruzi, the causal agent of Chagas' disease, develop severe thymocyte depletion paralleled by an inflammatory syndrome mediated by tumor necrosis factor-alpha (TNF-alpha). The exacerbated inflammatory reaction induces the activation of hypothalamus-pituitary-adrenal (HPA) axis with the consequent release of corticosterone (CT) into the circulation as a protective response. Thymocyte apoptosis has been related to a rise in TNF-alpha and CT levels, and both mediators are increased in T. cruzi-infected C57BL/6 mice. The depletion of immature CD4(+)CD8(+) thymocytes by apoptosis following infection with the parasite was still present in mice defective in both types of TNF-receptors (double knockout). However, thymic atrophy was prevented by adrenalectomy combined with RU486 administration, demonstrating that this is a CT-driven phenomenon. Our results put emphasis on the importance of an appropriated immuno-endocrine balance during T. cruzi infection and show that functional deviations in the immuno-endocrine equilibrium have profound effects on the thymus and disease outcome.


Subject(s)
Chagas Disease/immunology , Neuroimmunomodulation/immunology , Thymus Gland/immunology , Thymus Gland/parasitology , Trypanosoma cruzi/immunology , Acute Disease , Animals , Apoptosis/immunology , Atrophy , CD4-CD8 Ratio , Chagas Disease/mortality , Chagas Disease/pathology , Corticosterone/blood , Corticosterone/immunology , Hormone Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mifepristone/pharmacology , Neurosecretory Systems/immunology , Neurosecretory Systems/parasitology , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type I/genetics , Thymus Gland/pathology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
20.
Am J Pathol ; 170(2): 546-56, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17255323

ABSTRACT

During acute infection with Trypanosoma cruzi, the causative agent of Chagas' disease, the thymus undergoes intense atrophy followed by a premature escape of CD4+CD8+ immature cortical thymocytes. Here we report a pivotal role for the endogenous lectin galectin-3 in accelerating death of thymocytes and migration of these cells away from the thymus after T. cruzi infection. We observed a pronounced increase in galectin-3 expression that paralleled the extensive depletion of CD4+CD8+ immature thymocytes after infection. In vitro, recombinant galectin-3 induced increased levels of death in cortical immature thymocytes. Consistent with the role of galectin-3 in promoting cell death, thymuses from gal-3-/- mice did not show cortical thymocyte depletion after parasite infection in vivo. In addition, galectin-3 accelerated laminin-driven CD4+CD8+ thymocyte migration in vitro and in vivo induced exportation of CD4+CD8+ cells from the thymus to the peripheral compartment. Our findings provide evidence of a novel role for galectin-3 in the regulation of thymus physiology and identify a potential mechanism based on protein-glycan interactions in thymic atrophy associated with acute T. cruzi infection.


Subject(s)
CD4 Antigens/metabolism , CD8 Antigens/metabolism , Chagas Disease/metabolism , Galectin 3/metabolism , Thymus Gland/metabolism , Trypanosoma cruzi , Animals , Atrophy/genetics , Atrophy/metabolism , Atrophy/pathology , Cell Death/genetics , Cell Differentiation/genetics , Chagas Disease/genetics , Galectin 3/deficiency , Gene Expression Regulation/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Thymus Gland/parasitology , Thymus Gland/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...