Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Cell Rep ; 43(6): 114317, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38848213

ABSTRACT

Naive CD4+ T cells must differentiate in order to orchestrate immunity to Plasmodium, yet understanding of their emerging phenotypes, clonality, spatial distributions, and cellular interactions remains incomplete. Here, we observe that splenic polyclonal CD4+ T cells differentiate toward T helper 1 (Th1) and T follicular helper (Tfh)-like states and exhibit rarer phenotypes not elicited among T cell receptor (TCR) transgenic counterparts. TCR clones present at higher frequencies exhibit Th1 skewing, suggesting that variation in major histocompatibility complex class II (MHC-II) interaction influences proliferation and Th1 differentiation. To characterize CD4+ T cell interactions, we map splenic microarchitecture, cellular locations, and molecular interactions using spatial transcriptomics at near single-cell resolution. Tfh-like cells co-locate with stromal cells in B cell follicles, while Th1 cells in red pulp co-locate with activated monocytes expressing multiple chemokines and MHC-II. Spatial mapping of individual transcriptomes suggests that proximity to chemokine-expressing monocytes correlates with stronger effector phenotypes in Th1 cells. Finally, CRISPR-Cas9 gene disruption reveals a role for CCR5 in promoting clonal expansion and Th1 differentiation. A database of cellular locations and interactions is presented: https://haquelab.mdhs.unimelb.edu.au/spatial_gui/.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Differentiation , Malaria , Phenotype , Animals , Malaria/immunology , Malaria/parasitology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Th1 Cells/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , Spleen/immunology
2.
Nat Commun ; 15(1): 5497, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38944658

ABSTRACT

Children in malaria-endemic regions can experience repeated Plasmodium infections over short periods of time. Effects of re-infection on multiple co-existing CD4+ T cell subsets remain unresolved. Here, we examine antigen-experienced CD4+ T cells during re-infection in mice, using scRNA-seq/TCR-seq and spatial transcriptomics. TCR transgenic TEM cells initiate rapid Th1/Tr1 recall responses prior to proliferating, while GC Tfh counterparts are refractory, with TCM/Tfh-like cells exhibiting modest non-proliferative responses. Th1-recall is a partial facsimile of primary Th1-responses, with no upregulated effector-associated genes being unique to recall. Polyclonal, TCR-diverse, CD4+ T cells exhibit similar recall dynamics, with individual clones giving rise to multiple effectors including highly proliferative Th1/Tr1 cells, as well as GC Tfh and Tfh-like cells lacking proliferative capacity. Thus, we show substantial diversity in recall responses mounted by multiple co-existing CD4+ T cell subsets in the spleen, and present graphical user interfaces for studying gene expression dynamics and clonal relationships during re-infection.


Subject(s)
CD4-Positive T-Lymphocytes , Malaria , Reinfection , Animals , Malaria/immunology , Malaria/parasitology , CD4-Positive T-Lymphocytes/immunology , Mice , Reinfection/immunology , Th1 Cells/immunology , Mice, Inbred C57BL , Spleen/immunology , Spleen/parasitology , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/genetics , Mice, Transgenic , Female , Immunologic Memory
4.
JCI Insight ; 8(24)2023 Dec 22.
Article in English | MEDLINE | ID: mdl-37917177

ABSTRACT

Control of visceral leishmaniasis (VL) depends on proinflammatory Th1 cells that activate infected tissue macrophages to kill resident intracellular parasites. However, proinflammatory cytokines produced by Th1 cells can damage tissues and require tight regulation. Th1 cell IL-10 production is an important cell-autologous mechanism to prevent such damage. However, IL-10-producing Th1 (type 1 regulatory; Tr1) cells can also delay control of parasites and the generation of immunity following drug treatment or vaccination. To identify molecules to target in order to alter the balance between Th1 and Tr1 cells for improved antiparasitic immunity, we compared the molecular and phenotypic profiles of Th1 and Tr1 cells in experimental VL caused by Leishmania donovani infection of C57BL/6J mice. We also identified a shared Tr1 cell protozoan signature by comparing the transcriptional profiles of Tr1 cells from mice with experimental VL and malaria. We identified LAG3 as an important coinhibitory receptor in patients with VL and experimental VL, and we reveal tissue-specific heterogeneity of coinhibitory receptor expression by Tr1 cells. We also discovered a role for the transcription factor Pbx1 in suppressing CD4+ T cell cytokine production. This work provides insights into the development and function of CD4+ T cells during protozoan parasitic infections and identifies key immunoregulatory molecules.


Subject(s)
Interleukin-10 , Protozoan Infections , Th1 Cells , Th1 Cells/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/immunology , Mice, Inbred C57BL , Leishmania donovani , Leishmaniasis, Visceral/immunology , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/parasitology , Protozoan Infections/immunology , Humans , Animals , Mice , Lymphocyte Activation Gene 3 Protein/antagonists & inhibitors , Interferon-gamma/metabolism , Protein Binding , Promoter Regions, Genetic/immunology , Disease Models, Animal
5.
J Clin Invest ; 133(19)2023 10 02.
Article in English | MEDLINE | ID: mdl-37781920

ABSTRACT

The development of highly effective malaria vaccines and improvement of drug-treatment protocols to boost antiparasitic immunity are critical for malaria elimination. However, the rapid establishment of parasite-specific immune regulatory networks following exposure to malaria parasites hampers these efforts. Here, we identified stimulator of interferon genes (STING) as a critical mediator of type I interferon production by CD4+ T cells during blood-stage Plasmodium falciparum infection. The activation of STING in CD4+ T cells by cyclic guanosine monophosphate-adenosine monophosphate (cGAMP) stimulated IFNB gene transcription, which promoted development of IL-10- and IFN-γ-coproducing CD4+ T (type I regulatory [Tr1]) cells. The critical role for type I IFN signaling for Tr1 cell development was confirmed in vivo using a preclinical malaria model. CD4+ T cell sensitivity to STING phosphorylation was increased in healthy volunteers following P. falciparum infection, particularly in Tr1 cells. These findings identified STING expressed by CD4+ T cells as an important mediator of type I IFN production and Tr1 cell development and activation during malaria.


Subject(s)
Interferon Type I , Malaria, Falciparum , T-Lymphocytes, Regulatory , Humans , CD4-Positive T-Lymphocytes , Interferon Type I/immunology , Malaria, Falciparum/immunology , T-Lymphocytes, Regulatory/immunology
6.
mBio ; 14(4): e0112923, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37449844

ABSTRACT

Maturation rates of malaria parasites within red blood cells (RBCs) can be influenced by host nutrient status and circadian rhythm; whether host inflammatory responses can also influence maturation remains less clear. Here, we observed that systemic host inflammation induced in mice by an innate immune stimulus, lipopolysaccharide (LPS), or by ongoing acute Plasmodium infection, slowed the progression of a single cohort of parasites from one generation of RBC to the next. Importantly, plasma from LPS-conditioned or acutely infected mice directly inhibited parasite maturation during in vitro culture, which was not rescued by supplementation, suggesting the emergence of inhibitory factors in plasma. Metabolomic assessments confirmed substantial alterations to the plasma of LPS-conditioned and acutely infected mice, and identified a small number of candidate inhibitory metabolites. Finally, we confirmed rapid parasite responses to systemic host inflammation in vivo using parasite scRNA-seq, noting broad impairment in transcriptional activity and translational capacity specifically in trophozoites but not rings or schizonts. Thus, we provide evidence that systemic host inflammation rapidly triggered transcriptional alterations in circulating blood-stage Plasmodium trophozoites and predict candidate inhibitory metabolites in the plasma that may impair parasite maturation in vivo. IMPORTANCE Malaria parasites cyclically invade, multiply, and burst out of red blood cells. We found that a strong inflammatory response can cause changes to the composition of host plasma, which directly slows down parasite maturation. Thus, our work highlights a new mechanism that limits malaria parasite growth in the bloodstream.


Subject(s)
Malaria , Parasites , Mice , Animals , Transcriptome , Lipopolysaccharides , Malaria/parasitology , Inflammation , Erythrocytes/parasitology
8.
J Clin Invest ; 133(1)2023 01 03.
Article in English | MEDLINE | ID: mdl-36594463

ABSTRACT

Control of intracellular parasites responsible for malaria requires host IFN-γ+T-bet+CD4+ T cells (Th1 cells) with IL-10 produced by Th1 cells to mitigate the pathology induced by this inflammatory response. However, these IL-10-producing Th1 (induced type I regulatory [Tr1]) cells can also promote parasite persistence or impair immunity to reinfection or vaccination. Here, we identified molecular and phenotypic signatures that distinguished IL-10-Th1 cells from IL-10+Tr1 cells in Plasmodium falciparum-infected people who participated in controlled human malaria infection studies, as well as C57BL/6 mice with experimental malaria caused by P. berghei ANKA. We also identified a conserved Tr1 cell molecular signature shared between patients with malaria, dengue, and graft-versus-host disease. Genetic manipulation of primary human CD4+ T cells showed that the transcription factor cMAF played an important role in the induction of IL-10, while BLIMP-1 promoted the development of human CD4+ T cells expressing multiple coinhibitory receptors. We also describe heterogeneity of Tr1 cell coinhibitory receptor expression that has implications for targeting these molecules for clinical advantage during infection. Overall, this work provides insights into CD4+ T cell development during malaria that offer opportunities for creation of strategies to modulate CD4+ T cell functions and improve antiparasitic immunity.


Subject(s)
Malaria , T-Lymphocytes, Regulatory , Mice , Animals , Humans , Th1 Cells , Interleukin-10 , Mice, Inbred C57BL , Malaria/genetics , CD4-Positive T-Lymphocytes
9.
Clin Transl Immunology ; 11(6): e1396, 2022.
Article in English | MEDLINE | ID: mdl-35663920

ABSTRACT

Objectives: There is an urgent need to be able to identify individuals with asymptomatic Leishmania donovani infection, so their risk of progressing to VL and transmitting parasites can be managed. This study examined transcriptional markers expressed by CD4+ T cells that could distinguish asymptomatic individuals from endemic controls and visceral leishmaniasis (VL) patients. Methods: CD4+ T cells were isolated from individuals with asymptomatic L. donovani infection, endemic controls and VL patients. RNA was extracted and RNAseq employed to identify differentially expressed genes. The expression of one gene and its protein product during asymptomatic infection were evaluated. Results: Amphiregulin (AREG) was identified as a distinguishing gene product in CD4+ T cells from individuals with asymptomatic L. donovani infection, compared to VL patients and healthy endemic control individuals. AREG levels in plasma and antigen-stimulated whole-blood assay cell culture supernatants were significantly elevated in asymptomatic individuals, compared to endemic controls and VL patients. Regulatory T (Treg) cells were identified as an important source of AREG amongst CD4+ T-cell subsets in asymptomatic individuals. Conclusion: Increased Treg cell AREG expression was identified in individuals with asymptomatic L. donovani infection, suggesting the presence of an ongoing inflammatory response in these individuals required for controlling infection and that AREG may play an important role in preventing inflammation-induced tissue damage and subsequent disease in asymptomatic individuals.

10.
Ecancermedicalscience ; 15: 1278, 2021.
Article in English | MEDLINE | ID: mdl-34567263

ABSTRACT

BACKGROUND: The existence of oestrogen receptor-negative (ER-)/progesterone receptor-positive (PR+) breast cancer continues to be an area of controversy amongst oncologists and pathologists. METHODS: To re-evaluate breast cancers originally classified as ER-/PR+ via Oncotype DX® assay and compare molecular phenotype with Recurrence Score® (RS) result, clinicopathologic features and clinical outcomes were retrospectively obtained from electronic health records between January 1998 and June 2005. Archived formalin-fixed, paraffin-embedded (FFPE) tumour specimens were tested for the expression of ER, PR and human-epidermal-growth-factor-2. The number of positive ER-/PR+ samples confirmed by transcriptional analysis was the primary outcome of interest with event-free and overall survival as secondary outcomes. Biopsies from 26 patients underwent Oncotype DX testing and analysis. RESULTS: Approximately 60% were middle-aged (40-50 years old) women, and 84.6% had invasive ductal carcinoma. Based on the Oncotype DX assay, approximately 65% (N = 17) had ER+/PR+ status; 23% (N = 6) had ER-/PR- status; and 12% had a single hormone positive receptor (1 ER-/PR+, 2 ER+/PR-) status. Almost one-quarter of patients were stratified into the low-RS (<18) or intermediate-RS (18-30) results, and half of the patients had a high-RS (>30) result. CONCLUSION: Our findings suggest the ER-/PR+ subtype is not a reproducible entity and emphasises the value of retesting this subtype via molecular methods for appropriate treatment selection and patient outcomes. Multigene assay analysis may serve as a second-line or confirming tool for clinical determination of ER/PR phenotype in breast cancer patients for targeted therapies.

11.
Clin Med Res ; 19(2): 64-71, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33789952

ABSTRACT

Objective: Perioperative chemotherapy can potentially downstage esophageal cancer, reducing the risk of early systemic dissemination. One recommended neoadjuvant regimen for managing gastroesophageal junction and esophageal cancer is docetaxel, cisplatin, and 5-fluorouracil (DCF). To address the high toxicity profile of DCF, modifications in dosages and treatment intervals have been studied. We integrated a modified DCF regimen (mDCF) into a multimodal treatment approach for non-metastatic esophageal cancer (nMEC). Retrospectively, we sought to describe our community experience of administrating neoadjuvant mDCF to patients with nMEC.Design: Patients diagnosed with nMEC between August 2008 and November 2017 and prescribed mDCF were identified for retrospective review. Outcomes of interest included disease-free survival (DFS), overall survival (OS), and hematologic toxicities. Analyses were performed using SAS 9.4.Results: Thirty patients met inclusion criteria with a median age of 64.9 years; 90% were male. The 2-year and 5-year DFS was 60.8% and 41.7%, respectively, for adenocarcinoma and 71.4% and 71.4% for squamous cell carcinoma (SCC). The 2-year and 5-year OS was 64.9% and 44.5%, respectively, for adenocarcinoma and 71.4% and 71.4% for SCC. Both DFS and OS decreased with increasing disease stage, histology (adenocarcinoma versus squamous), esophageal compared to esophagogastric-junction involvement, and without surgical intervention. Frequent toxicity grades for leukopenia and thrombocytopenia were Grades I and II.Conclusion: Using an mDCF regimen in combination with chemoradiation +/- surgical resection in a community setting appears to have an acceptable toxicity profile as well as DFS and OS outcomes compared to chemotherapeutic regimens reported in other similar studies.


Subject(s)
Esophageal Neoplasms , Stomach Neoplasms , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cisplatin/therapeutic use , Docetaxel/therapeutic use , Esophageal Neoplasms/drug therapy , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Neoadjuvant Therapy , Retrospective Studies , Stomach Neoplasms/drug therapy , Taxoids/therapeutic use
12.
Nat Immunol ; 21(12): 1597-1610, 2020 12.
Article in English | MEDLINE | ID: mdl-33046889

ABSTRACT

The dynamics of CD4+ T cell memory development remain to be examined at genome scale. In malaria-endemic regions, antimalarial chemoprevention protects long after its cessation and associates with effects on CD4+ T cells. We applied single-cell RNA sequencing and computational modelling to track memory development during Plasmodium infection and treatment. In the absence of central memory precursors, two trajectories developed as T helper 1 (TH1) and follicular helper T (TFH) transcriptomes contracted and partially coalesced over three weeks. Progeny of single clones populated TH1 and TFH trajectories, and fate-mapping suggested that there was minimal lineage plasticity. Relationships between TFH and central memory were revealed, with antimalarials modulating these responses and boosting TH1 recall. Finally, single-cell epigenomics confirmed that heterogeneity among effectors was partially reset in memory. Thus, the effector-to-memory transition in CD4+ T cells is gradual during malaria and is modulated by antiparasitic drugs. Graphical user interfaces are presented for examining gene-expression dynamics and gene-gene correlations ( http://haquelab.mdhs.unimelb.edu.au/cd4_memory/ ).


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immunologic Memory , Malaria/immunology , Plasmodium/immunology , Transcriptome , Adoptive Transfer , Animals , Antimalarials/pharmacology , Biomarkers , Chromatin/genetics , Disease Models, Animal , Gene Expression Profiling , Humans , Malaria/parasitology , Malaria/therapy , Mice , Plasmodium/drug effects
13.
Immunol Cell Biol ; 98(8): 620-622, 2020 09.
Article in English | MEDLINE | ID: mdl-32895977

ABSTRACT

Holz et al. report a glycolipid-conjugate vaccine that provides sterile immunity in mice against Plasmodium berghei ANKA sporozoite challenge by inducing long-lasting tissue-resident memory (TRM) CD8+ T cells in the liver.


Subject(s)
Cancer Vaccines , Malaria Vaccines , Malaria , Animals , CD8-Positive T-Lymphocytes/immunology , Glycolipids , Immunologic Memory , Liver/immunology , Malaria/prevention & control , Mice , Peptides , Rodentia , Vaccination , Vaccines, Subunit
14.
Nat Immunol ; 21(10): 1205-1218, 2020 10.
Article in English | MEDLINE | ID: mdl-32839608

ABSTRACT

Immune-modulating therapies have revolutionized the treatment of chronic diseases, particularly cancer. However, their success is restricted and there is a need to identify new therapeutic targets. Here, we show that natural killer cell granule protein 7 (NKG7) is a regulator of lymphocyte granule exocytosis and downstream inflammation in a broad range of diseases. NKG7 expressed by CD4+ and CD8+ T cells played key roles in promoting inflammation during visceral leishmaniasis and malaria-two important parasitic diseases. Additionally, NKG7 expressed by natural killer cells was critical for controlling cancer initiation, growth and metastasis. NKG7 function in natural killer and CD8+ T cells was linked with their ability to regulate the translocation of CD107a to the cell surface and kill cellular targets, while NKG7 also had a major impact on CD4+ T cell activation following infection. Thus, we report a novel therapeutic target expressed on a range of immune cells with functions in different immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Leishmania donovani/physiology , Leishmaniasis, Visceral/immunology , Malaria/immunology , Membrane Proteins/metabolism , Plasmodium/physiology , Animals , Cells, Cultured , Cytotoxicity, Immunologic , Disease Models, Animal , Exocytosis , Humans , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Secretory Vesicles/metabolism
15.
JCI Insight ; 5(13)2020 07 09.
Article in English | MEDLINE | ID: mdl-32484791

ABSTRACT

Acute gastrointestinal (GI) graft-versus-host disease (GVHD) is a primary determinant of mortality after allogeneic hematopoietic stem cell transplantation (alloSCT). The condition is mediated by alloreactive donor CD4+ T cells that differentiate into pathogenic subsets expressing IFN-γ, IL-17A, or GM-CSF and is regulated by subsets expressing IL-10 and/or Foxp3. Developmental relationships between Th cell states during priming in mesenteric lymph nodes (mLNs) and effector function in the GI tract remain undefined at genome scale. We applied scRNA-Seq and computational modeling to a mouse model of donor DC-mediated GVHD exacerbation, creating an atlas of putative CD4+ T cell differentiation pathways in vivo. Computational trajectory inference suggested emergence of pathogenic and regulatory states along a single developmental trajectory in mLNs. Importantly, we inferred an unexpected second trajectory, categorized by little proliferation or cytokine expression, reduced glycolysis, and high tcf7 expression. TCF1hi cells upregulated α4ß7 before gut migration and failed to express cytokines. These cells exhibited recall potential and plasticity following secondary transplantation, including cytokine or Foxp3 expression, but reduced T cell factor 1 (TCF1). Thus, scRNA-Seq suggested divergence of alloreactive CD4+ T cells into quiescent and effector states during gut GVHD exacerbation by donor DC, reflecting putative heterogeneous priming in vivo. These findings, which are potentially the first at a single-cell level during GVHD over time, may assist in examination of T cell differentiation in patients undergoing alloSCT.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Lymphocyte Activation/immunology , Transcriptome/genetics , Animals , Gastrointestinal Microbiome/genetics , Graft vs Host Disease/genetics , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous/methods
16.
Case Rep Oncol ; 13(1): 321-329, 2020.
Article in English | MEDLINE | ID: mdl-32308599

ABSTRACT

Hypercalcemia is an ominous development in the course of malignancy associated with a mean survival of only several months. A majority of cases of hypercalcemia are related to humoral hypercalcemia of malignancy (HHM), where hypercalcemia is caused by increased levels of circulating parathyroid hormone-related protein (PTHrP). Mainstay treatments in the management of HHM are intravenous fluids, intravenous bisphosphonates, and subcutaneous denosumab, although hypercalcemia oftentimes recurs despite these efforts. We present a case of advanced non-small cell lung cancer with PTHrP-mediated hypercalcemia that proved resistant to standard therapy. A trial of oral cinacalcet was initiated and improved calcium levels for 2 months despite a progressive rise in PTHrP and prior to subsequent disease progression. Based on the current body of literature, we propose that this calcium-lowering effect of cinacalcet occurs due to a potential effect on renal calcium excretion.

17.
Immunol Cell Biol ; 97(7): 617-624, 2019 08.
Article in English | MEDLINE | ID: mdl-31120158

ABSTRACT

The ability of circulating CD4+ T cells to retain memories of previous antigenic encounters is a cardinal feature of the adaptive immune system. Over the past two decades, since the first description of central and effector memory T cells, many studies have examined molecular mechanisms controlling CD8+ T-cell memory, with comparatively less research into CD4+ T-cell memory. Here, we review a number of seminal studies showing that circulating memory CD4+ T cells develop directly from effector cells; and in so doing, preserve features of their effector precursors. We examine mechanisms controlling the development and phenotypes of memory CD4+ T cells, and provide an updated model that accommodates both the central and effector memory paradigm and the diverse T helper cell classification system.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Immunologic Memory , Animals , Biomarkers , Cell Differentiation/genetics , Cell Differentiation/immunology , Energy Metabolism , Gene Expression Regulation , Humans , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Models, Biological , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcription, Genetic
18.
PLoS Pathog ; 15(2): e1007599, 2019 02.
Article in English | MEDLINE | ID: mdl-30811498

ABSTRACT

Plasmodium parasites invade and multiply inside red blood cells (RBC). Through a cycle of maturation, asexual replication, rupture and release of multiple infective merozoites, parasitised RBC (pRBC) can reach very high numbers in vivo, a process that correlates with disease severity in humans and experimental animals. Thus, controlling pRBC numbers can prevent or ameliorate malaria. In endemic regions, circulating parasite-specific antibodies associate with immunity to high parasitemia. Although in vitro assays reveal that protective antibodies could control pRBC via multiple mechanisms, in vivo assessment of antibody function remains challenging. Here, we employed two mouse models of antibody-mediated immunity to malaria, P. yoelii 17XNL and P. chabaudi chabaudi AS infection, to study infection-induced, parasite-specific antibody function in vivo. By tracking a single generation of pRBC, we tested the hypothesis that parasite-specific antibodies accelerate pRBC clearance. Though strongly protective against homologous re-challenge, parasite-specific IgG did not alter the rate of pRBC clearance, even in the presence of ongoing, systemic inflammation. Instead, antibodies prevented parasites progressing from one generation of RBC to the next. In vivo depletion studies using clodronate liposomes or cobra venom factor, suggested that optimal antibody function required splenic macrophages and dendritic cells, but not complement C3/C5-mediated killing. Finally, parasite-specific IgG bound poorly to the surface of pRBC, yet strongly to structures likely exposed by the rupture of mature schizonts. Thus, in our models of humoral immunity to malaria, infection-induced antibodies did not accelerate pRBC clearance, and instead co-operated with splenic phagocytes to block subsequent generations of pRBC.


Subject(s)
Malaria/immunology , Malaria/metabolism , Plasmodium/growth & development , Animals , Antibodies, Protozoan/metabolism , Disease Models, Animal , Erythrocytes/microbiology , Erythrocytes/physiology , Humans , Mice , Parasites , Phagocytes , Plasmodium/metabolism , Plasmodium/pathogenicity , Plasmodium chabaudi/immunology , Plasmodium chabaudi/pathogenicity , Plasmodium yoelii/immunology , Plasmodium yoelii/pathogenicity
19.
Exp Parasitol ; 198: 7-16, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30682336

ABSTRACT

Plasmodium falciparum histone deacetylases (PfHDACs) are an important class of epigenetic regulators that alter protein lysine acetylation, contributing to regulation of gene expression and normal parasite growth and development. PfHDACs are therefore under investigation as drug targets for malaria. Despite this, our understanding of the biological roles of these enzymes is only just beginning to emerge. In higher eukaryotes, HDACs function as part of multi-protein complexes and act on both histone and non-histone substrates. Here, we present a proteomics analysis of PfHDAC1 immunoprecipitates, identifying 26 putative P. falciparum complex proteins in trophozoite-stage asexual intraerythrocytic parasites. The co-migration of two of these (P. falciparum heat shock proteins 70-1 and 90) with PfHDAC1 was validated using Blue Native PAGE combined with Western blot. These data provide a snapshot of possible PfHDAC1 interactions and a starting point for future studies focused on elucidating the broader function of PfHDACs in Plasmodium parasites.


Subject(s)
Histone Deacetylase 1/analysis , Plasmodium falciparum/enzymology , Proteomics , Protozoan Proteins/chemistry , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Histone Deacetylase 1/chemistry , Immunoprecipitation , Mass Spectrometry/methods
20.
Clin Med Res ; 16(3-4): 69-72, 2018 12.
Article in English | MEDLINE | ID: mdl-30587560

ABSTRACT

A man, aged 61 years, with a history of hypogonadism and family history of cancer experienced persistent urinary difficulties with no visible prostate abnormalities. Laboratory testing and diagnostic imaging revealed a primary lesion in the prostate with lymph node involvement and multiple bone metastases. Treatment with androgen-deprivation therapy, 17,20-lyase inhibition, and bisphosphonates for 7 months was unsuccessful in preventing disease progression, but second-line chemotherapy and continued androgen-deprivation therapy improved prostate specific antigen levels. During the patient's second treatment regimen, his daughter received a diagnosis of breast cancer. The patient's daughter underwent genetic testing for oncogenic mutations, and it was discovered that she carried a mutation in RAD51C, a gene encoding a protein involved in DNA repair and genomic maintenance. Subsequent genetic testing of the patient revealed mutation in RAD51C as well. For patients with metastatic prostate cancer who are unresponsive to standard treatment and who have a positive family history of cancer, genetic testing may be warranted to develop alternative treatment regimens for the patient and guide family discussions regarding cancer risk. Targeted agents like poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors may be a consideration in prostate cancer patients with DNA repair mutations and with refractory disease.


Subject(s)
DNA-Binding Proteins/genetics , Mutation , Prostatic Neoplasms/genetics , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , DNA Repair , DNA-Binding Proteins/metabolism , Humans , Male , Middle Aged , Neoplasm Metastasis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...