Article preview
- Abstract
- Section snippets
- References (42)
- Cited by (41)
- Recommended articles (6)
International Immunopharmacology
Volume 8, Issue 10,
October 2008
, Pages 1338-1343
Author links open overlay panel, , , ,
Abstract
Cellular immune responses directed against protozoan parasites are key for controlling pathogen replication and disease resolution. However, an uncontrolled, or improperly controlled, response can be deleterious to the host in terms of both allowing for the establishment of pathology, as well as less effective establishment of memory responses. Human cutaneous leishmaniasis is a disease caused by the infection with Leishmania spp. following a bite from the sandfly, the natural vector of this disease. Tens of millions worldwide are currently infected with Leishmania and no effective vaccines have been developed to date. In the face of the complexity presented by the interaction between a host (humans) with the parasite, Leishmania, and the fact that this parasite is inoculated by another complex, biologically active, vector, the sandfly, it is clearly important to study the immunoregulatory mechanisms that are induced in humans naturally infected by this parasite if we hope to develop effective vaccines and immunotherapeutic treatments in the future. Our laboratory has focused over the years on the study of the local and systemic T cell response during the first episode of cutaneous leishmaniasis suffered by individuals before they undergo antimony treatment. The goal of this review is to briefly outline our findings with hopes of putting our most recent studies concerning the dichotomy between alpha/beta TCR and gamma/delta TCR expressing, CD4−CD8− (double negative—DN) T cells in the context of a balanced immune response against Leishmania and to discuss the implications of these findings toward our understanding of human leishmaniasis.
Section snippets
Human leishmaniasis — a complex disease resulting from the host–parasite–vector interaction
Human leishmaniasis is caused by the infection with the obligate intracellular protozoan parasite, Leishmania, and is expressed as one or more of several possible clinical forms including principally the visceral and tegumentary diseases (localized cutaneous, mucosal or disseminated) [1]. Several factors influence the progression of an infected individual into these clinical forms of disease including, but not limited to, host genetics (protective and susceptibility factors), host environment
Protection vs. pathology vs. memory…a three-way balancing act
The study of the cellular immune response and determination of cytokine profiles in both murine models and in humans has been determined over the years through the application of a number of quantitative and semi-quantitative methodologies. These methods have ranged from single-cell cytoplasmic staining of cytokines within specific cell populations to ELISA from culture supernatants or body fluids, and quantitative PCR. In tissues investigators have used in situ detection of cytokines by
CD4−CD8− (double negative—DN) T cells in human cutaneous leishmaniasis: alpha/beta DN T cells likely have a role in protection and pathology, while gamma/delta DN T cells could play a more important role in negative regulation
In addition to classic CD4+ and CD8+ T cells, there is a minority subpopulation of T cells that express neither CD4 nor CD8, thus termed double negative (DN) T cells. Within this DN T cell population several subpopulations can be found. In the broad sense, they contain T cells expressing either the gamma/delta or alpha/beta TCR, and within each of these populations, several subpopulations can be defined. Amongst the alpha/beta TCR DN T cells in humans, studies have identified cells of both
Acknowledgements
We would like to thank the collaboration over the past ten years of Dr. Edgar Carvalho and his group at Hospital Edgard Santos, UFBA, in Salvador, Bahia, Brazil, without which the studies described in this overview of our work would not have been possible. We would also like to thank the following funding agencies for support of our work over the years: WHO/TDR, CNPq, FINEP, FAPEMIG, and NIH-NIAID.
References (42)
- L. Van Kaer
NKT cells: T lymphocytes with innate effector functions
Curr Opin Immunol
(Jun 2007)
- D.I. Godfrey et al.
NKT cells: facts, functions and fallacies
Immunol Today
(Nov 2000)
- K. Fischer et al.
Isolation and characterization of human antigen-specific TCR alpha beta+ CD4(−)CD8− double-negative regulatory T cells
Blood
(Apr 1 2005)
- L.R. Antonelli et al.
Activated inflammatory T cells correlate with lesion size in human cutaneous leishmaniasis
Immunol Lett
(Nov 15 2005)
- R.L. Coffman et al.
Leishmania major infection in mice: a model system for the study of CD4+ T-cell subset differentiation
Res Immunol
(Jan 1991)
- S.G. Coutinho et al.
Parasitological and immunological follow-up of American tegumentary leishmaniasis patients
Trans R Soc Trop Med Hyg
(Apr 2002)
- B.L. Herwaldt
Leishmaniasis
Lancet
(Oct 2 1999)
- M.P. de Oliveira-Neto et al.
American tegumentary leishmaniasis (ATL) in Rio de Janeiro State, Brazil: main clinical and epidemiologic characteristics
Int J Dermatol
(Jul 2000)
- A. Barral et al.
Leishmaniasis in Bahia, Brazil: evidence that Leishmania amazonensis produces a wide spectrum of clinical disease
See AlsoCancer-Associated Fibroblasts Are Positively Correlated With Metastatic Potential - [PDF Document]Alpha II Antiplasmin Deficiency Complicating Pregnancy: A Case ReportAm J Trop Med Hyg
(May 1991)
- P. Scott
T-cell subsets and T-cell antigens in protective immunity against experimental leishmaniasis
Curr Top Microbiol Immunol
(1990)
The development of effector T cell subsets in murine Leishmania major infection
Ciba Found Symp
(1995)
Experimental Leishmania major infection in mice: role of IL-10
Parasite Immunol
(Apr 1999)
The role of interleukin (IL)-10 in the persistence of Leishmania major in the skin after healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile cure
J Exp Med
(Nov 19 2001)
Immune privilege in sites of chronic infection: Leishmania and regulatory T cells
Immunol Rev
(Oct 2006)
Re-examination of the immunosuppressive mechanisms mediating non-cure of Leishmania infection in mice
Immunol Rev
(Oct 2004)
Development and regulation of cell-mediated immunity in experimental leishmaniasis
Immunol Res
(2003)
The immunopathology of experimental visceral leishmaniasis
Immunol Rev
(Oct 2004)
The role of CD4(+)CD25(+) regulatory T cells in Leishmania infection
Expert Opin Biol Ther
(Sep 2003)
Immunologic memory in cutaneous leishmaniasis
Cell Microbiol
(Dec 2005)
Central memory T cells mediate long-term immunity to Leishmania major in the absence of persistent parasites
Nat Med
(Oct 2004)
Mucosal leishmaniasis patients display an activated inflammatory T-cell phenotype associated with a nonbalanced monocyte population
Scand J Immunol
(Jan 2006)
Cited by (41)
Host's immune response in unresponsive and responsive patients with anthroponotic cutaneous leishmaniasis treated by meglumine antimoniate: A case-control study of Th1 and Th2 pathways
2019, International Immunopharmacology
Citation Excerpt :
Generally, the disease susceptibility in uncontrolled or non-healed infections has been related to Th2 proliferation and production of IL-4, IL-5 and IL-10. In contrast, healing responses and resistance to infection have been associated with expansion of IFN-γ-producing Th1 cells [18–21]. T-cell responses in individuals with mild infection involve a mixture of Th1 and Th2, whereas patients with exacerbating disease show an absence of Th1 response to specific Leishmania antigens.
The objective of the present study was to compare the host's immune responses between unresponsive and responsive patients with anthroponotic cutaneous leishmaniasis (ACL) treated by meglumine antimoniate.
A case-control study was carried out in an endemic focus in Iran. Blood samples were taken from patients and peripheral blood mononuclear cells (PBMCs) were isolated. Two wells were considered for each isolate of unresponsive and responsive patients; one was exposed to L. tropica (Lt-stimulated cells) and the other remained non-exposed (non-stimulated cells). After 24 h of incubation, whole RNA was extracted from each sample. Real-time quantitative PCR was carried out to confirm the differences in expression levels of IL-12 P40, IFN-γ, IL-1β, IL-4 and IL-10 among isolates. Data were analyzed and P < 0.05 was considered to be statistically significant.
In our study, Lt-stimulated cells and non-stimulated cells in unresponsive groups demonstrated significantly lower expression levels of IL-1β, IL-12 P40 and IFN-γ genes and higher expression levels of IL-4 and IL-10 genes, compared to Lt-stimulated cells and non-stimulated cells in responsive groups. There was a negative correlation between IL-12 P40 with IL-10 and IL-1β with IL-10 in ACL Lt-stimulated cells in unresponsive group, while a positive correlation between IL-12 P40 with IL-1β and IL-12 P40 with IFN-γ in ACL Lt-stimulated cells in responsive group.
Probably, different immune responses caused by various factors play a major role in the pathogenesis and development of unresponsiveness in ACL patients. The profile and timing of cytokine production correlated well with the treatment outcome of Leishmania infection.
Gene expression profile of cytokines produced in biopsies from patients with American cutaneous leishmaniasis
2019, Acta Tropica
American cutaneous leishmaniasis (ACL) causes a local inflammatory process, inducing expression of several cytokine genes. Particularly, IFN-γ can predict to disease susceptibility. Based in these data, this study was aimed to investigate the gene expression profile of IFN-γ, IL-10, IL-27, TNF-γ, TGF-β and IL-6 produced in biopsies from ACL patients; and whether the gene expression profile of IFN-γ could determine the disease evolution. Gene expression of 6 cytokines was investigated in 40 formalin-fixed paraffin embedded (FFPE) biopsies from patients with cutaneous leishmaniosis (CL); and 10 FFPE biopsies from patients with mucosal leishmaniasis (ML) (control). All 50 patients were infected with Leishmania (Viannia) braziliensis. Gene expression was determined by qPCR; and a normal control group was used for calculations (5 normal biopsies). Values were expressed as Relative Quantification (RQ). The 40 CL patients were classified into 2 groups. CLlowIFN-γ, 35 patients with RQ for IFN-γ below 100; and CLhighIFN-γ, 5 (12.5%) patients with RQ above 100. Significant increase of mRNA levels of IFN-γ, IL-10 and IL-27 was shown in CLhighIFN-γ group when compared with CLlowIFN-γ and ML groups. TNF-α levels in CLlowIFN-γ group were higher than CLhighIFN-γ and ML groups. TGF-β and IL-6 were similar in 3 groups. Comparison of cytokine expression/group showed that CLlowIFN-γ group had an equilibrium between the cytokines analyzed. In ML group, IFN-γ was over-expressed; but in CLhighIFN-γ group, besides IFN-γ, IL-27 was also over-expressed. The immune response to Leishmania induces to identification of some markers, which can be determined by analysis by gene expression of cytokines produced in biopsies.
Immunization with Leishmania donovani protein disulfide isomerase DNA construct induces Th1 and Th17 dependent immune response and protection against experimental visceral leishmaniasis in Balb/c mice
2017, Molecular Immunology
Citation Excerpt :
Subsequently, patients develop various clinical conditions such as splenomegaly, pancytopenia, anemia, and disseminated hemorrhages (Murray et al., 2005). Two distinct lineages of T-cells, namely CD4 + Th1 andCD4 + Th17, are responsible for the controlled and programmed immune response mediated against Leishmania and supported by various cytokines (such as interleukin-2 [IL-2], interferon-γ[IFN-γ], IL-6, and the tumor necrosis factor [TNF]), and chemokine(such as CXCL8 [also known as IL-8], CXCL10 [also known as IP-10], CXCL1, and CXCL6; Bhattacharya and Ali, 2013; Sanjabi et al., 2009; Gollob et al., 2008). The Th1 group of CD4+ T cells counters Leishmania infection within macrophages in an infected spleen and liver through support from IL-12, IL-2, IFN-γ, etc.
In the present study, the efficacy of Leishmania donovani protein disulfide isomerase (LdPDI) as a DNA vaccine was evaluated in BALB/C mice. Mice immunized with the LdPDI-DNA construct were found to be the most immuno-reactive, as the construct induced higher T-cell proliferation. The increased T-cell proliferation was associated with a substantial rise in Th1 and Th17+ CD4 cell response and triggered a higher proportion of CD8+ T cells for the release of interferon-gamma along with a reduced splenic parasite load on Days20 and 60 post challenge (PC). Furthermore, the vaccine construct triggered increased interferon (IFN)-γ, interleukin(IL)-17A, and IL-22 release accompanied by decreased extracellular signal-regulated kinases (ERK) 1/2 signaling and increased mitogen-activated protein kinase (MAPK) signaling coinciding with an increase in the amount of nitrite and reactive oxygen species (ROS)in vaccinating the splenocyts. We summarize from our data that the PDI-DNA construct of Leishmania donovani has the potential to elicit protective immunity through the pro-inflammatory cytokines of CD8+ and CD4+(Th1 and Th17) following an intervention in the downstream signaling event of ERK1/2 (probably through p38MAPK signaling). Therefore, the study suggests a new control against visceral leishmaniasis in the future.
American tegumentary leishmaniasis: mRNA expression for Th1 and Treg mediators are predominant in patients with recent active disease
2016, Immunobiology
Citation Excerpt :
In humans, the spectrum of the disease may vary from subclinical infection to manifestations such as localized, disseminated or diffuse cutaneous lesions, or even the involvement of the nasal mucosa, pharynx and larynx. In all these possibilities, the expansion of T helper cells exerts a fundamental influence (Gollob et al., 2008). Classically, a Th1 profile is responsible for producing interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), which activates the inducible nitric-oxide synthase (iNOS) to catalyze NO secretion by macrophages, eliminating the parasite.
Besides the Th1×Th2 paradigm, Treg and Th17 cytokines may play a role in the response to American tegumentary leishmaniasis. Considering the sensitivity and accuracy of qPCR and the lack of studies using this approach, we evaluated mRNA expression for IFN-γ, TNF-α, IL-4, IL-10, IL-6, IL-17A, IL-22, TGF-β, Foxp3 and RORC in peripheral blood mononuclear cells (PBMC) from patients with active disease, after stimulation with L. (V.) braziliensis soluble or insoluble fractions. Our results show that the antigens promoted specific mRNA expression related to the immune response in patients with ATL, and the insoluble fraction seems to stimulate the immune response in a higher intensity. The pro-inflammatory response was also fueled by IFN-γ and TNF-α, probably due to the active disease. IL-4, in certain way, seems to regulate this response along with IL-10 that may be produced by Treg cells, which are supposedly present in the patients’ samples due the evidenced expression of Foxp3, in the presence of AgIns. In contrast, down-regulated RORC suggests that the significant levels of IL-6 expressed in response to AgSol were not able to induce an expressive Th17 profile along with TGF-β, which might have predominantly contributed to the development of a regulatory profile in the active disease.
Leishmania infection and neuroinflammation: Specific chemokine profile and absence of parasites in the brain of naturally-infected dogs
2015, Journal of Neuroimmunology
Citation Excerpt :
Recently, different subsets of T lymphocytes gained focus on parasitic diseases, especially the double negative (DN) T cells, which express neither CD4 nor CD8, including αβ DN T cells, γδ DN T cells, NK T cells (D'Acquisto and Crompton, 2011). In cases of human cutaneous leishmaniasis, elevated numbers of circulating αβ DN T cells are described, associated with elevated production of IFN-γ and TNF-α and high levels of the activation markers CD69 and CD56 (Antonelli et al., 2006; Gollob et al., 2008). Further, the association between NK T cells and immune response during VL has already been studied in the liver of mice infected with L. donovani (Amprey et al., 2004).
Visceral leishmaniasis is a chronic disease caused by Leishmania infantum. We aimed to detect the parasite in the brain of fifteen naturally-infected dogs using in situ hybridization and immunohistochemistry, and the gene expression of selected chemokines by RT-qPCR. We detected no parasite in the brain, but perivascular deposition of parasite DNA and IgG in the choroid plexus. We noticed up-regulation of CCL-3, CCL-4 and CCL-5, coherent with T lymphocyte accumulation, stating the brain as a pro-inflammatory environment. Indeed, not necessarily the parasite itself, but rather its DNA seems to act as a trigger to promote brain inflammation during visceral leishmaniasis.
Cytokines and NO in American tegumentary leishmaniasis patients: Profiles in active disease, after therapy and in self-healed individuals
2013, Microbial Pathogenesis
Citation Excerpt :
According to the authors, susceptibility to the mucocutaneous form of disease may be directly associated with regulatory polymorphisms affecting TNF-α production. Besides the benefits of Th1 response, Gollob et al. [32] suggest that IL-10 production may be critical for the disease control. Thus, it is postulated that IL-10 levels presented by the patients in this study regulate the production of proinflammatory cytokines, consequently leading to the clinical resolution of the disease.
Studies suggest the influence of immune response on the successful treatment of American tegumentary leishmaniasis (ATL), and indicate the existence of protective immunity in self-healed patients. Thus, the aim of this work was to quantify interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), interleukin (IL-) 10, IL-17, IL-22 and nitric oxide (NO) in culture supernatants of PBMC from patients with active disease (AD), after treatment (AT), and from self-healed (SH) and healthy subjects (CT), in response to Leishmania (Viannia) braziliensis insoluble antigen (AgIns). All groups of patients produced IFN-γ, indicating a predominant proinflammatory profile. AD and AT patients presented TNF-α levels, with a slight increase after therapy, whereas it was weakly quantified in SH. Interestingly, NO secretion was significant in these individuals, whereas IL-17 appeared in low levels and seems to be regulated by NO. Although IL-22 was detected in AD, its role is still questionable. The presence of IL-10 in all groups of patients suggests that the cytokine plays distinct roles in the disease. These results indicate that specific cellular immunity takes part against Leishmania, but with some similarities between the different clinical states herein described; these mediators seem to be necessary for the cure to occur.
Recommended articles (6)
Research article
Lack of both α2-antiplasmin and plasminogen activator inhibitor type-1 induces high IgE production
Life Sciences, Volume 93, Issues 2–3, 2013, pp. 89-95
We investigated the pathophysiological changes in mice lacking α2-antiplasmin (α2-AP) and plasminogen activator inhibitor type-1 (PAI-1) genes, and elucidated the involvement of these inhibitors for fibrinolysis in immune response.
The pathophysiological changes induced by a lack of both α2-AP and PAI-1 were investigated using double knockout (KO) mice. The lung, liver, kidney and spleen tissues from α2-AP/PAI-1-double KO mice were compared with those from wild-type (WT) mice. Furthermore, the bone marrow cells from α2-AP/PAI-1-double KO mice were transplanted into 10-Gy X ray irradiated WT mice, and then the effects of the transplantation were studied.
Plasma IgE levels in the α2-AP/PAI-1-double KO mice increased with age and exceeded 1000ng/mL after 6months of age. The plasma cells that produced IgE were detected in perivascular assembled lymphocytes. In the α2-AP/PAI-1-double KO mice, perivascular lymphocyte infiltration was observed in the lung, liver, and kidneys and peribronchial lymphocyte infiltration was present in the lung. When the bone marrow cells from α2-AP/PAI-1-double KO mice were transplanted into 10-Gy X ray irradiated WT mice, the phenotypes of the recipients were similar to those of α2-AP/PAI-1-double KO mice.
The simultaneous expression of both the α2-AP and PAI-1 genes contributes to the maintenance of immunological functions that are related to IgE. Moreover, it is suggested that both α2-AP and PAI-1 are involved in the recruitment of lymphocytes in the peripheral tissues.
Research article
Rapamycin can restore the negative regulatory function of transforming growth factor beta 1 in high grade lymphomas
Cytokine, Volume 73, Issue 2, 2015, pp. 219-224
TGF-β1 (transforming growth factor beta 1) is a negative regulator of lymphocytes, inhibiting proliferation and switching on the apoptotic program in normal lymphoid cells. Lymphoma cells often lose their sensitivity to proapoptotic/anti-proliferative regulators such as TGF-β1. Rapamycin can influence both mTOR (mammalian target of rapamycin) and TGF-β signaling, and through these pathways it is able to enhance TGF-β induced anti-proliferative and apoptotic responses. In the present work we investigated the effect of rapamycin and TGF-β1 combination on cell growth and on TGF-β and mTOR signalling events in lymphoma cells.
Rapamycin, an inhibitor of mTORC1 (mTOR complex 1) did not elicit apoptosis in lymphoma cells; however, the combination of rapamycin with exogenous TGF-β1 induced apoptosis and restored TGF-β1 dependent apoptotic machinery in several lymphoma cell lines with reduced TGF-β sensitivity in vitro. In parallel, the phosphorylation of p70 ribosomal S6 kinase (p70S6K) and ribosomal S6 protein, targets of mTORC1, was completely eliminated. Knockdown of Smad signalling by Smad4 siRNA had no influence on apoptosis induced by the rapamycin+TGF-β1, suggesting that this effect is independent of Smad signalling. However, apoptosis induction was dependent on early protein phosphatase 2A (PP2A) activity, and in part on caspases. Rapamycin+TGF-β1 induced apoptosis was not completely eliminated by a caspase inhibitor.
These results suggest that high mTOR activity contributes to TGF-β resistance and lowering mTORC1 kinase activity may provide a tool in high grade B-cell lymphoma therapy by restoring the sensitivity to normally available regulators such as TGF-β1.
Research article
Avidity of anticardiolipin antibodies—A factor that could be important for their detection by ELISA methods
Journal of Applied Biomedicine, Volume 12, Issue 4, 2014, pp. 277-284
Avidity is an important feature of antibodies associated with their pathogenic effects. Anticardiolipin antibodies (aCLs) are the most commonly examined antiphospholipid antibodies, however, their avidity has been investigated only marginally. The aim of the study was to compare the avidity of the antibodies specifically bound to the cardiolipin-coated microtitrate wells with those bound to the wells bearing no cardiolipin in various conditions. We analysed 22 serum samples with high, medium and low levels of aCL IgG. The avidity of aCL IgG was determined in serially diluted sera by the ELISA method in the presence of increasing urea concentrations (from 2 to 8mol/L) as a chaotropic agent. The serum dilution 1:50 and 1:100 and the concentrations of urea 6mol/L and 8mol/L seemed to be suitable for the determination of aCL avidity. The avidity of antibodies specifically bound to the cardiolipin and those bound to the cardiolipin-free wells significantly differed in their avidity. The simultaneous determination of higher-avidity aCL and lower-avidity polyspecific antibodies, whose presence complicates the ELISA methods by an increase of the background signal, might limit the shortcomings of some ELISA methods.
Research article
Serological signatures of clinical cure following successful treatment with sodium stibogluconate in Ethiopian visceral leishmaniasis
Cytokine, Volume 91, 2017, pp. 6-9
In Ethiopia, visceral leishmaniasis (VL) is a growing public health threat. Among the key challenges in VL control in Ethiopia is lack of an effective test of cure. The recommended test of cure is parasite detection. As sterile cure is not expected with the current widely used drugs, the value of parasite detection as test of cure is questionable. Moreover, the sampling is invasive, requires a well-equipped facility and highly skilled personnel, which are all hardly found in endemic set-ups.
Our aim was to assess the value of sCD40L, MMP9 and IL-10 serum levels as signature biomarkers of clinical cure in VL cases from Ethiopia.
A total of 45 VL cases before and after treatment and 30 endemic healthy controls were included in the study. Sandwich ELISA was used to measure serum levels of sCD40L, MMP9 and IL-10.
The mean sCD40L, MMP9 and IL-10 serum levels changed significantly at clinical cure. At individual case level sCD40L and MMP9 showed an increasing trend. Yet, the degree of increase in serum level of MMP9 seems to be affected by nutritional status of the individual VL case. The mean IL-10 serum level was significantly reduced at clinical cure. As seen on case by case basis, all demonstrated a declining trend except that two VL cases had a high IL10 level at clinical cure.
Our result is suggestive of the possibility of developing a signature biomarker to monitor VL treatment in Ethiopia using one or a combination of parameters.
Research article
Prediction Forecast for Culex tritaeniorhynchus Populations in Korea
Osong Public Health and Research Perspectives, Volume 5, Issue 3, 2014, pp. 131-137
Japanese encephalitis is considered as a secondary legal infectious disease in Korea and is transmitted by mosquitoes in the summer season. The purpose of this study was to predict the ratio of Culex tritaeniorhynchus to all the species of mosquitoes present in the study regions.
From 1999 to 2012, black light traps were installed in 10 regions in Korea (Busan, Gyeonggi, Gangwon, Chungbuk, Chungnam, Jeonbuk, Jeonnam, Gyeongbuk, Gyeongnam, and Jeju) to capture mosquitoes for identification and classification under a dissecting microscope. The number of mosquitoes captured/week was used to calculate its daily occurrence (mosquitoes/trap/night). To predict the characteristics of the mosquito population, an autoregressive model of order p (AR(p)) was used to execute the out-of-sample prediction and the in-sample estimation after presumption.
Compared with the out-of-sample method, the sample-weighted regression method's case was relatively superior for prediction, and this method predicted a decrease in the frequency of Cx. tritaeniorhynchus for 2013. However, the actual frequency of this species showed an increase in frequency. By contrast, the frequency rate of all the mosquitoes including Cx. tritaeniorhynchus gradually decreased.
The number of patients with Japanese encephalitis has been strongly associated with the occurrence and density of vector mosquitoes, and the importance of this infectious disease has been highlighted since 2010. The 2013 prediction indicated an increase after an initial decrease, although the ratio of the two mosquito species decreased. The increase in vector density may be due to changes in temperature and the environment. Thus, continuous prevalence prediction is warranted.
Research article
Belatacept-Resistant Rejection Is Associated With CD28+ Memory CD8 T Cells
American Journal of Transplantation, Volume 17, Issue 9, 2017, pp. 2285-2299
Recently, newer therapies have been designed to more specifically target rejection in an effort to improve efficacy and limit unwanted toxicity. Belatacept, a CD28-CD80/86 specific reagent, is associated with superior patient survival and graft function compared with traditional therapy, but its adoption as a mainstay immunosuppressive therapy has been tempered by increased rejection rates. It is essential that the underlying mechanisms associated with this rejection be elucidated before belatacept is more widely used. To that end, we designed a study in a nonhuman primate kidney transplant model where animals were treated with either a belatacept- or a tacrolimus-based immunosuppressive regimen. Interestingly, we found that elevated pretransplant frequencies of CD28+CD8+TEMRA cells are associated with rejection on belatacept but not tacrolimus treatment. Further analysis showed that the CD28+CD8+TEMRA cells rapidly lose CD28 expression after transplant in those animals that go on to reject with the allograft infiltrate being predominantly CD28−. These data suggest that CD28+ memory T cells may be resistant to belatacept, capable of further differentiation including loss of CD28 expression while maintaining effector function. The unique signaling requirements of CD28+ memory T cells provide opportunities for the development of targeted therapies, which may synergize with belatacept to prevent costimulation-independent rejection.
Copyright © 2008 Elsevier B.V. All rights reserved.
FAQs
What is the mechanism of immunity in leishmaniasis? ›
Lymphocytes are involved in adaptive immune responses to Leishmania infection, primarily through the elaboration of cytokines that activate or dampen the antiparasitic activity of macrophages. It is well known that T cells play a major role in immunity to the various forms of Leishmania.
What is the T cell response in leishmaniasis? ›Studies in the literature have shown the important role played by CD4 T cells in protection against human leishmaniasis by producing cytokines able to activate the macrophages that kill the parasites.
Which immune response is host protective during Leishmania infection Th1 or Th2? ›The control of leishmanial infection is mediated by a Th1-type immune response, and experimental studies in murine models of CL have established a clear-cut dichotomy between Th1-mediated protection and Th2-mediated disease susceptibility (reviewed by Sacks & Noben-Trauth, 2002).
Is it painful to have Leishmaniasis? ›The sores may start out as papules (bumps) or nodules (lumps) and may end up as ulcers (like a volcano, with a raised edge and central crater); skin ulcers may be covered by scab or crust. The sores usually are painless but can be painful.
How can you protect yourself from getting infected with Leishmania? ›...
When indoors:
- Stay in well-screened or air-conditioned areas.
- Keep in mind that sand flies are much smaller than mosquitoes and therefore can get through smaller holes.
- Spray living/sleeping areas with an insecticide to kill insects.
Leishmaniasis is transmitted by the bite of infected female phlebotomine sand flies. The sand flies inject the infective stage (i.e., promastigotes) from their proboscis during blood meals .
Which cells of your body are infected by Leishmania? ›Although Leishmania parasites interact and infect a variety of host cell types, macrophages and dendritic cells (DCs) are arguably the most important cells that regulate the outcome of infection.
What is CD4 T cell response? ›CD4+T cells are crucial in achieving a regulated effective immune response to pathogens. Naive CD4+T cells are activated after interaction with antigen-MHC complex and differentiate into specific subtypes depending mainly on the cytokine milieu of the microenvironment.
What are the responses of T helper 2 cell? ›Abstract. T helper 2 (TH2) cells orchestrate protective type 2 immune responses, such as those that target helminths and facilitate tissue repair, but also contribute to chronic inflammatory diseases, such as asthma and allergy.
What is Th1 Th2 response in autoimmune disease? ›Th1 cells promote cellular immunity and are involved in the development of autoimmune diseases; Th2 cells mediate humoral immunity and are involved in allergic immune responses.
What is the difference between Th1 and Th2 response leishmaniasis? ›
Th1-type cellular immune responses play an important role in defense against infection with Leishmania parasites, whereas activation of Th2-type cells results in progressive disease (4, 5).
What is the difference between Th1 and Th2 in Leishmania? ›Th1 and Th2 cells can be distinguished by the cytokines they secrete: Th1 cells secrete activators of cell-mediated immunity such as interferon (IFN)-γ, while Th2 cells secrete cytokines such as interleukin (IL)-4 which promote antibody responses.
What is the most severe form of Leishmania? ›There are three main forms of the disease: cutaneous leishmaniasis (CL), visceral leishmaniasis (VL), also known as kala-azar, and mucocutaneous leishmaniasis (MCL). CL is the most common form, VL is the most severe form and MCL is the most disabling form of the disease.
What is the most serious form of leishmaniasis? ›Visceral leishmaniasis (VL), also known as kala-azar, is fatal if left untreated in over 95% of cases.
Can leishmaniasis go away on its own? ›The skin sores of cutaneous leishmaniasis usually heal on their own, even without treatment. But this can take months or even years, and the sores can leave ugly scars.
What are the symptoms of Leishmania in humans? ›- Fever. This can start suddenly and come and go for several weeks.
- Chills.
- Cold sweats.
- Swollen lymph nodes.
- Swollen abdomen (belly) from an enlarged spleen.
- Weight loss.
- Fatigue.
- Weakness.
Liposomal amphotericin B is an effective treatment for cutaneous leishmaniasis and mucosal leishmaniasis, but requires administration by trained health-care professionals and cold chain transport. Miltefosine is a phospholipid analogue and is currently the only oral drug available.
What is the best treatment for Leishmania? ›Liposomal amphotericin B is FDA-approved for treatment of visceral leishmaniasis and generally is the treatment of choice for U.S. patients.
Can mosquitoes spread Leishmania? ›It is prevalent in tropical and subtropical countries and become a challenge to a highly endemic area of East Africa including Ethiopia [2, 3]. It is transmitted via the bite of infected female anopheles mosquito.
Which of the following is a vector for the parasite that transmits leishmaniasis? ›Lutzomyia longipalpis sand flies are the major natural vector of Leishmania infantum parasites, responsible for transmission of visceral leishmaniasis in the New World.
What is the main host of leishmaniasis? ›
Their primary hosts are vertebrates; Leishmania commonly infects hyraxes, canids, rodents, and humans.
What are the stages of the leishmaniasis disease? ›The Two Life Stages of Leishmania: Amastigote (A) and Promastigotes (B). Leishmaniasis is a parasitic disease caused by hemoflagellate, Leishmania spp. The parasite is transmitted through the bites of an infected female phlebotomine sandfly. Leishmaniasis is prevalent throughout the world and in at least 88 countries.
What is the reservoir host for Leishmania parasites affecting humans? ›There are two main sources of human leishmaniases, zoonotic leishmaniases, in which the reservoir hosts are wild animals, commensals or domestic animals, and anthroponotic leishmaniases, in which the reservoir host is human.
What is the prognosis for leishmaniasis? ›Outlook (Prognosis)
Cutaneous leishmaniasis may lead to disfigurement. Death is usually caused by complications (such as other infections), rather than from the disease itself. Death often occurs within 2 years.
Primary T cell activation involves the integration of three distinct signals delivered in sequence: (1) anti- gen recognition, (2) costimulation, and (3) cytokine- mediated differentiation and expansion.
What disease CD4 T cells are affected? ›CD4+ T cells play an essential role in the pathogenesis of autoimmune disorders. The aberrant CD4+ T cell responses have been demonstrated in patients with autoimmune disorders, including inflammatory bowel diseases (IBD), 1 systemic lupus erythematosus (SLE),2 and rheumatoid arthritis (RA).
What causes CD4 T cell dysfunction? ›Virus-specific CD4 T cell dysfunction results from a combination of an exhaustion program and skewing in Thelper lineage differentiation which impact function.. The CD4 and CD8 T cell exhaustion programs present similarities and distinct features.
What is type 2 inflammatory disease? ›As mentioned, it is now well documented that type 2 inflammatory responses are at the basis of atopic dermatitis (AD), asthma, and CRSwNP, as well as for eosinophilic esophagitis, some forms of COPD, and other conditions [4-6].
Is Th2 anti-inflammatory? ›Th2-related cytokinees (IL-4 and IL-10) are associated with humoral immunity and anti-inflammatory properties.
Why is Th2 anti-inflammatory? ›The Th2-type cytokines include interleukins 4, 5, and 13, which are associated with the promotion of IgE and eosinophilic responses in atopy, and also interleukin-10, which has more of an anti-inflammatory response. In excess, Th2 responses will counteract the Th1 mediated microbicidal action.
Is Hashimoto's Th1 or Th2 dominant? ›
Because Graves' hyperthyroidism is directly caused by thyroid-stimulating antibodies (TSAbs), it is logical to assume that the disease belongs to the Th2 category. Conversely, the dominance of cellular immunity and thyroid tissue damage in Hashimoto's thyroiditis implies a Th1 origin.
What is the difference between Th1 and Th2 inflammation? ›Th1 diseases are characterized by a strong Th1 response, while Th2 diseases are characterized by a weak or absent Th1 response. Th1 cytokines like IFNγ and TNFα promote inflammation, while Th2 cytokines like IL-21 and IL-22 inhibit inflammation.
What is the difference between Th1 and Th2 lupus? ›Th1 cells protect against intracellular pathogens, activate phagocytes, induce IgG2a antibodies, and promote delayed-type hypersensitivity responses, whereas Th2 cells protect against extracellular pathogens, activate eosinophils, induce IgE-mediated allergic reactions, and promote other humoral responses in which IgG1 ...
What is humoral immunity in Leishmania? ›Humoral immunity is characterized by the production of antibodies capable of promoting neutralization, opsonization, and activation of the complement system. In this scenario, B lymphocytes produce antibodies that play an important role in Leishmania infection although neglected for a long time.
What are the mechanisms of human immunity to fungal infections? ›Host defence mechanisms against fungi involve innate immunity and adaptive immunity. The two systems are intimately linked and controlled by sets of molecules and receptors that act to generate a highly coordinate and unitary process for protection against fungal pathogens.
What adaptations does leishmaniasis have? ›Once inside the macrophage, Leishmania is protected from phagolysosome degradation by a variety of adaptations to inhibit cellular defense mechanisms. These include the inhibition of phagosome-endosome fusion, hydrolytic enzymes, cell signaling pathways, nitric oxide production, and cytokine production.
What is the role of cytokines in leishmaniasis? ›IL-8 is a strong proinflammatory cytokine that plays an essential role in the recruitment and activation of neutrophils in the course of inflammation. This cytokine is secreted by tissue-resident macrophages in response to Leishmania infections and plays a key role in the initial stages of infection or tissue damage.