Next Article in Journal
Advances in Babesia Vaccine Development: An Overview
Next Article in Special Issue
Regional Differences in Intestinal Parasitic Infections among Army Recruits in a Southern Mozambique Training Center: A Cross-Sectional Study
Previous Article in Journal
HPV and Cervical Cancer: A Review of Epidemiology and Screening Uptake in the UK
Previous Article in Special Issue
Interleukin 10 (IL-10) Production and Seroprevalence of Entamoeba histolytica Infection among HIV-Infected Patients in South Africa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

A Systematic Review of Apicomplexa Looking into Epigenetic Pathways and the Opportunity for Novel Therapies

by
Yara de Oliveira Brandão
* and
Marcelo Beltrão Molento
Graduate Program in Microbiology, Parasitology, Pathology, Laboratory of Veterinary Clinical Parasitology, Federal University of Paraná, Curitiba 80035-050, PR, Brazil
*
Author to whom correspondence should be addressed.
Pathogens 2023, 12(2), 299; https://doi.org/10.3390/pathogens12020299
Submission received: 26 December 2022 / Revised: 3 February 2023 / Accepted: 9 February 2023 / Published: 11 February 2023
(This article belongs to the Special Issue Intestinal Parasites Infection)

Abstract

:
Interest in host epigenetic changes during apicomplexan infections increased in the last decade, mainly due to the emergence of new therapies directed to these alterations. This review aims to carry out a bibliometric analysis of the publications related to host epigenetic changes during apicomplexan infections and to summarize the main studied pathways in this context, pointing out those that represent putative drug targets. We used four databases for the article search. After screening, 116 studies were included. The bibliometric analysis revealed that the USA and China had the highest number of relevant publications. The evaluation of the selected studies revealed that Toxoplasma gondii was considered in most of the studies, non-coding RNA was the most frequently reported epigenetic event, and host defense was the most explored pathway. These findings were reinforced by an analysis of the co-occurrence of keywords. Even though we present putative targets for repurposing epidrugs and ncRNA-based drugs in apicomplexan infections, we understand that more detailed knowledge of the hosts’ epigenetic pathways is still needed before establishing a definitive drug target.

1. Introduction

Parasites and hosts have experienced an intense coevolution process [1]. Both genetic and epigenetic changes have occurred, leading to the rise of more resilient hosts and more virulent or adapted parasites [1]. The epigenetic concept came to light in 1942 with Waddington’s epigenetic landscape [2]. This concept proposed that the environment played an essential role in gene regulation, connecting the genotype to the phenotype, which could enlighten embryo development and cell differentiation. Later, the epigenetic concept underwent many interpretations and modifications, resulting in the current definition. Therefore, molecular epigenetics is an alteration in gene expression due to molecular mechanisms other than the change in the nucleotide sequence, such as histone chemical modifications, changes in chromatin structure, and non-coding RNA (ncRNA) [3].
The coordinated action of post-translational modifications of histones (PTHM), histone variants, DNA methylation, chromatin remodeling complexes, and ncRNAs (i.e., micro-RNA—miRNA, long non-coding RNA—lncRNA, and piwi RNA—piRNA) promotes changes in the chromatin structure that can allow or prevent gene transcription. In addition, ncRNAs can regulate gene expression at a post-transcriptional level by mRNA degradation or translation blockage [4]. These epigenetic mechanisms implicate the development and progression of many pathologies, such as cancer and neurological, metabolic, immunological, and infectious diseases in human and domestic animals [5,6]. Thus, enzymes that mediate the epigenetic gene regulation pathways have been used as drug targets, and epidrugs are currently used in clinical practice to treat hematological disorders [7].
Azacitidine and decitabine are examples of epidrugs that target epigenetic activity. These drugs inhibit DNA methyltransferases (DNMTs), which add methyl groups in CpG locations of DNA, leading to DNA methylation. Other enzymes responsible for epigenetic modifications are the writers and erasers that mediate histone marks by adding (e.g., histone acetyltransferase (HAT) or histone methyltransferase (HMT)) or removing (e.g., histone deacetylase (HADC) or histone demethylase (HDM)) chemical groups from histone tails. Histone deacetylases inhibitor (HDACi) drugs (e.g., vorinostat, romidepsin) and HMTi tazemetostat are approved by the Food and Drug Administration (FDA) to treat some types of lymphoma, while other HMT inhibitor (HMTi) drugs and HDM inhibitor (HDMi) drugs are in preclinical or clinical trial phases [8]. In addition to their therapeutical potential, epigenetic marks have been investigated for use in diagnosis. For example, DNA methylation of specific genes is already used for prostate and colon carcinoma diagnosis [7]. However, despite the advances in epigenetic-targeted therapy and diagnosis, no epigenetic-based drug or biomarker is available for parasitic diseases.
Parasitic diseases of humans and animals include the Apicomplexa phylum, such as Toxoplasma gondii, Cryptosporidium spp., Plasmodium spp., and Theileria spp. These unicellular, eukaryotic, obligate intracellular parasites reside in the parasitophorous vacuole (PV) derived from the host cell membrane (except for Theileria spp.). Their virulence factors are responsible for host cell adhesion, invasion, and the modulation of the host’s immune response and metabolism [9]. The success of parasites’ invasion, survival, and reproduction relies on their close interaction with the host’s genome, which has been demonstrated to implicate epigenetic mechanisms [10]. For example, parasites can induce chromatin remodeling in genes related to the host immune response, changing their expression and leading to a more beneficial environment for their development. In addition, parasites may modulate the expression of miRNAs involved in cell cycle regulation, aiming to keep the infected cell alive and allowing the parasite to survive and proliferate [10].
Given the importance and the therapeutic potential of the epigenetic pathways employed by Apicomplexa in infected host cells, this review aims to carry out a bibliometric analysis of the publications related to host epigenetic changes during apicomplexan infections and to summarize the main studied pathways, pointing out those that represent putative drug targets. Understanding which host pathways apicomplexan parasites modulate and how they do it can unravel new drug targets.

2. Material and Methods

2.1. Information Sources and Search Strategy

The protocol of this study was registered in the International Prospective Register of Systematic Reviews (PROSPERO: http://www.crd.york.ac.uk/prospero, accessed on 25 January 2023), CRD42023391155.
We screened relevant reports in Scopus, Science Direct, Web of Science, and PUBMED databases. In Scopus, PUBMED, and Science Direct, we searched under “title, keywords, and abstract”; in Web of Science, we searched under “topic”. All searches were limited to “article” (Web of Science and Scopus), with the following search term combinations: epigen* AND (“parasitic diseases” OR parasite OR protozoan); (“DNA methylation” OR “histone modification” OR “non-coding RNA”) AND (“parasitic diseases” OR parasite OR protozoan); epigen* AND parasite AND host; epigen* AND (“host gene” OR “host response” OR “host cell”) AND (“parasitic diseases” OR parasite OR protozoan); “chromatin remodeling” AND (“parasitic diseases” OR parasite OR protozoan); (“micro RNA” OR “miRNA” OR “lnc RNA” OR “small non-coding RNA” OR “long non-coding RNA” OR “piwi RNA” OR “pi RNA”) AND (“parasitic disease” OR parasite OR protozoan); (“modification* of histone*” OR “histone* modification*”) AND (“parasitic diseases” OR parasite OR protozoan); “DNA methylation” AND (“parasitic diseases” OR parasite OR protozoan). As the Science Direct database does not accept wildcards, the words “epigenetic”, “modification”, and “histone” were applied instead of “epigen*”, “modification*”, and “histone*”. The searches were performed in August 2020 and the last update was made in October 2022. Papers listed as a reference in selected articles were also evaluated. We carried out two new searches in January 2023, using the same keywords and replacing the term “protozoan” with “protist*.” However, no paper found exclusively under the “protist” term fulfilled the inclusion criteria, and we retained the results of the search that was carried out in October 2022.

2.2. Search Analysis—Eligibility Criteria

  • Research article: only original research articles were included.
  • Epigenetic approaches: papers showed one or more tests to demonstrate the epigenetic event.
  • Host–parasite interaction: studies investigating modifications due to the interaction between the parasite and the host.
  • Host species: only studies in mammalian hosts were included.
  • Parasite species: only Apicomplexa parasites were included.
  • Time period: articles were included independently of the year of publication.
  • Idiom: articles published in any language were considered. For those published in idioms other than Portuguese or English, Google Translate (https://translate.google.com/, accessed on 25 January 2023) was used for translation into English.

2.3. Study Records, Data Items, Outcome, and Bias Risk Evaluation

Title, abstract, and keywords information were obtained in RIS format and imported to the reference manager Mendeley Desktop 1.19.8 software (Mendeley Ltd, London, UK). After excluding duplicated documents, the articles were screened by analyzing the title and abstract, addressing the inclusion criteria. The selected papers were fully accessed, and those not fulfilling the inclusion criteria were excluded. A Microsoft Excel table was filled with information regarding parasite species, the gene expression regulatory mechanism, the targeted process, the research models, the country, and the complete reference. In the topic “regulatory mechanism,” the type of event was recorded: i.e., DNA methylation, post-translational modifications of histones, or ncRNA (miRNA, lncRNA, piRNA, circRNA). The “targeted process” refers to the main cell function pathways cited in each study. To assess the bias risk, title, abstract screening, and full paper accessing were carried out by the two authors independently.

2.4. Additional Analysis

For keyword and country co-occurrence analysis, the selected papers were imported to VOS Viewer 1.1.16 (Leiden University, Leiden, The Netherlands). A world map with the distribution of the selected reports was constructed using Microsoft Excel software.

3. Results and Discussion

3.1. Document Selection

The search returned 6237 documents, of which 3595 remained after duplicate exclusion. Upon screening the title and abstract, 3442 records did not meet the eligibility criteria. Of the 153 remaining records that were fully accessed, 43 did not meet the eligibility criteria. Six docs were added from the references of fully accessed articles, resulting in a total of 116 included records (Figure 1). Of the 43 excluded studies, 14 did not present epigenetic events; four reported epigenetic events that were not related to the host-parasite interaction; one study investigated a host that was not a mammal; two studies did not show sufficient evidence; one study was a pre-printed article; 21 studies were not research articles (i.e., opinion letters, methodology or hypothesis papers, case reports or addenda to articles, and 16 review articles).

3.2. Analysis of Affiliated Countries

Table 1 includes all collected articles showing a limited range of parasites, possible regulatory mechanisms, target processes, and research models. The United States of America (USA) had the highest number of publications on the topic of this review (33.6%), followed by China (29.3%) (Figure 2). Those two countries also showed strong cooperation in paper publications (Figure 3). In the country co-authorship analysis, the closer the node, the higher the cooperation. Countries with the same node color were included in the same cluster, which was formed by the frequency of the co-occurrences of the countries’ names. The node size depicts the number of publications (the larger the node, the higher the number of publications), and the thickness of the lines represents the number of collaborations. China and the USA had the largest nodes, belonging to the same cluster and linked by a thick line. These characteristics reflected the findings above: i.e., many publications and strong cooperation.
Interestingly, the most representative countries in this review (USA, China, and Germany) did not present the highest prevalence of Apicomplexan-related diseases. For example, a recent meta-analysis showed IgG seroprevalence against T. gondii in less than 10% of pregnant women in the USA, compared to more than 50% in low and middle-income countries (LMICs), such as Brazil [127].
The same pattern was also observed to concerning malaria and cryptosporidiosis [13,14,128,129]. The lack of studies in the most-affected countries was related to their socioeconomic status and research and development investments [15]. Diseases such as cryptosporidiosis and toxoplasmosis can be prevented by having access to clean water and education, which is not a reality for most of the populations in LMICs [14]. Therefore, these countries cannot guarantee ways to prevent these diseases or support quality research. The lower research investment in LMICs reflects the smaller number of publications. However, this could be enriched by more research collaborations between the North and South.

3.3. Keywords Co-Occurrence Analysis

The keywords co-occurrence network analysis resulted in three clusters (Figure 4). The clusters are identified by different colors showing words related to the findings presented in Table 1 and summarized in Table 2. The red cluster displays the words related to T. gondii, the most represented parasite. T. gondii is an intestinal coccidian with felids as definitive hosts, infecting a wide variety of avian and mammal (i.e., mice and humans) intermediate hosts. In its acute phase, T. gondii invades and replicates into the host cells, especially macrophages, causing mild clinical signs. In the chronic phase, tissue cysts are responsible for neurological and ocular disorders in offspring through congenital transmission [130]. The significant number of studies concerning T. gondii may be due to its pathogenicity, high prevalence in humans, and worldwide distribution. Other reasons are that tachyzoites are successfully maintained in culture, are tractable for genetic manipulation, and evoke a vigorous immune response in the host; therefore, they are used as models for apicomplexan parasites studies [127,131]. However, despite the number of articles selected, only three studies used samples from toxoplasmosis patients [91,106,107], and only one article investigated T. gondii in its definitive host [90]. Although felids usually do not present severe clinical signs associated with toxoplasmosis, they are an important contamination source; thus, understanding how the parasite manipulates the felid epigenetics would be of great value.
The second and third most cited parasites were Plasmodium spp. and Cryptosporidium parvum. They are represented in the yellow and blue clusters, respectively. Malaria, a disease caused by Plasmodium spp., was estimated to account for 229 million cases and 409.000 deaths in 2019, according to the World Malaria Report of the World Health Organization [128]. Despite its epidemiological importance, the number of studies of Plasmodium spp. was a bit more than half of the number of studies of T. gondii. This low number of reports may have three possible reasons: (1) the characteristic of the inclusion criteria (as we excluded studies in invertebrate hosts, such as Plasmodium spp. Vectors); (2) difficulties in maintaining this parasite in culture, which often restricted clinical sampling in experiments [132]; and (3) the infected cell type of the host cell. The mature erythrocyte is one of the targets of Plasmodium spp. but it lacks a nucleus, DNA, and chromatin. Thus, DNA methylation, post-translational histone modifications, chromatin remodeling, and change in miRNA expression cannot occur in these cells, regardless of the infection. The selected Plasmodium spp. studies focused on the epigenetic modifications in leukocytes, other infected cell types (i.e., hepatocytes), ncRNA in the plasma, and the ncRNA content of extracellular vesicles secreted by infected erythrocytes. Therefore, more studies are needed covering the possible effects of Plasmodium spp. infection in epigenetic events during the erythropoiesis process. These modifications can, for example, be responsible for alterations in the mature erythrocytes, such as the microRNA composition of the secreted exosomes [58].
C. parvum is an intestinal protist responsible for epithelial barrier disruption and enterocyte apoptosis. These lesions result in nutrient malabsorption and persistent diarrhea [133]. The disease is more severe in immunocompromised human patients and young animals; however, none of the selected studies investigated the C. parvum-related epigenetic changes in these individuals. Eight studies use in vivo models, and none included samples from human patients or domestic animals. One probable limitation for studies with clinical samples is the strict location of the parasite in the intestine, making sample collection difficult. However, intestinal tissue from biopsies or even the extraction of genetic material from feces could be used to investigate the epigenetic modifications during acute cryptosporidiosis. Other words highlighted in the co-occurrence analysis, such as “microrna”, “dna methylation”, “phosphorylation”, “transcriptome”, and “gene expression profiling”, were related to the epigenetic regulation of gene expression. ncRNAs were the targets of most of the studies, followed by PTHM and DNA methylation (Table 2).
In recent decades, the interest in ncRNAs, such as miRNA, lncRNA, siRNA, and piRNA, has increased considerably. The next-generation sequencing (NGS) technology, and therefore RNA sequencing (RNAseq), has enabled the investigation of the total transcriptome and the discovery of many ncRNAs [134]. Fifty-one of the 88 ncRNAs studies (58%) focused on identifying the different profiles of ncRNA expression in multiple cell lineages and host species infected by apicomplexan parasites, covering a high number of transcripts. The high specificity of miRNAs and lncRNAs for cell type and host species highlights the importance of these studies [134]. However, it is crucial to clarify the role of ncRNA in each parasite species before suggesting new targets for drugs or biomarkers for diagnosis. The lower number of studies of ncRNA function (42%) confirmed the relatively recent interest in this RNA in apicomplexan infection [46]. We expect that the ncRNAs identified by the studies on the difference of expression will be further investigated to unravel their pathways in apicomplexan infections. The co-occurrence network of keywords also showed that the different apicomplexan parasites (T. gondii, Plasmodium spp., and C. parvum) drive epigenetic changes in the same host pathways. Words such as “immunology”, “cell proliferation”, and “nf kappa-B” displayed in the blue cluster were linked to nodes in red and yellow clusters, referring to the host process that suffered interference from the parasite (Table 1). As expected, the host defense was the most epigenetically influenced route. In healthy individuals, infections elicit an immune response by activating pathways regulated by epigenetic mechanisms. In addition to activating these defense pathways, the apicomplexan parasites may drive changes in epigenetic events to evade the immune response and guarantee their survival. The parasite preserves its habitat by interfering in apoptosis regulation, favoring its survival and reproduction.
Epigenetic changes are also related to the pathogenesis of apicomplexan parasites infection. For example, the pathogenic B cell proliferation in Theileria annulata infection can be related to changes in miRNA expressions that regulate cancer pathways. Epigenetic changes in cell migration in C. parvum infection are related to the lesion and clinical signs associated with the disease [23,69]. Multiple pathways share many of the epigenetic mechanisms reported here. Changes in miRNAs expression and the activation or inhibition of enzymes related to DNA methylation or PTHM can alter multiple routes. Thus, future studies are needed to identify the exact pathways modulated by parasites to ensure the specificity of putative drug targets.

3.4. Epidrugs: Drug Targets and Host-Directed Therapy

The current treatment for apicomplexan parasite infections relies on drugs directed at parasite-specific molecules or pathways essential for their survival or replication. The rapid reproductive life cycle of the organism favors the emergence of mutations, leading to drug resistance that is not a major problem in host-directed therapy (HDT) [135]. Another benefit of HDT is the possibility of repurposing preapproved drugs. HDT may also activate or modulate host pathways, such as innate and adaptative immune responses, inflammation, cell cycle regulation, and apoptosis [136]. Although epidrugs aimed at the parasite have been suggested [137], studies on the use of these drugs to modulate host response are still lacking (Table 3; Supplementary Table S2). Understanding which host pathways are epigenetically modulated by apicomplexan parasites and how they do it can unravel new drug targets. In this section, we summarize the current knowledge regarding epigenetic modifications induced by Apicomplexan parasites and highlight putative targets for repurposing epidrugs.

3.4.1. Histone Post-Translational Modifications and Chromatin Remodeling

Drugs targeting PTHM enzymes HDAC, HMT, and HDM were previously approved for cancer treatment or were in pre-clinical or clinical trials. Here, we describe the host process that could be related to these drugs in apicomplexan infections.
T. gondii infection epigenetically modulates the IFNγ, NFκB, and TNFα pathways, which interfere with monocyte differentiation and activation [105]. For example, in murine macrophages, T. gondii secretes the effector T. gondii inhibitor of STAT1 transcriptional activity (TgIST), which reaches the nucleus and complexes with STAT-1 (signal transducer and activator of transcription 1) and Mi-2/NuRD, repressing the transcription of IFNγ response genes [96] (Figure 5A). The exact mechanism of the TgIST-induced gene-expression inhibition has not been completely elucidated; however, the gene-expression regulation seems not to be the same for all IFNγ responsive genes. Lang et al. (2012) [75] observed a decrease in the histone H3 acetylation associated with a lower expression of the secondary IFNγ response gene Ciita, which was restored after treatment with MS-275. Entinostat, or MS-275, which is an HDAC1 inhibitor with a lesser action in HDAC2 and 3 and was in a clinical trial for breast cancer treatment. The intracellular pathogen Mycobacterium tuberculosis also silenced the Ciita gene through H3 deacetylation, and treatment with sodium butyrate partially rescued the gene expression [138]. In addition to MS-275, the FDA-approved drugs vorinostat and romidepsin inhibit HDAC1 and 2 and could be tested to restore Ciita and other secondary IFNγ response gene expression, increasing host immune defense (Figure 5B). Moreover, these HDACi enzymes may rescue TNFα expression, whose gene silencing function seems to be due to a decrease in H3 acetylation in T. gondii-infected macrophages [72].
The toxoplasma E2F4-associated EZH2-inducing gene regulator (TEEGR) is another T. gondii granule that reaches the host cell nuclei, inducing epigenetic modifications. In HFF and BMDM cells, this effector was demonstrated to recruit the histone methyltransferase enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) to the promoter of NFκB-dependent genes, increasing the histone closing mark H3K27me3, thereby repressing the expression of interleukin 8, 6, and 1β. The negative modulation of these genes contributes to parasite persistence in the host [97]. The parasite Leishmania donovani uses a similar mechanism in mice macrophages to silence the inflammation-related gene inos. The in vitro and in vivo knockdown of EZH2 followed by L. donovani infection restored the iNOS expression [139]. The FDA-approved drug tazemetostat is a selective HMTi that specifically inhibits EZH2 and is also a promising epidrug option to restore the expression of NFκB pathway genes that are inhibited in T. gondii infection.
Post-translational modifications in histones have been determined in HCT8 cells and the intestinal cells of mice infected with C. parvum [28,35]. C. parvum transcripts were demonstrated to interact with the methyltransferase G9a, promoting the trimethylation of H3K9 and the consequent silencing of the immune response, cell differentiation, cell adhesion, and migration-related genes [22,23,24]. Therefore, the hypothesis that G9a methyltransferase inhibition in C. parvum infection decreases cell damage, boosts cell differentiation, and increases enteric epithelium repair should be investigated. When confirmed, the G9a may be a target for the HMTi drug for cryptosporidiosis treatment.
Mathy et al. (2022) [34] showed an increase in the expression of the lncRNA Nostrill in IEC4.1 cells and the intestinal cells of mice infected with C. parvum. This lncRNA mediates the increment of the open H3K4me3 mark and the recruitment of RNA polymerase II to the promotor of the gene Ifr7, increasing this gene expression, which is a regulator of the I-IFN pathway. Further inquiry on the role of this lncRNA in C. parvum may reveal novel targets for ncRNA-based therapy.
The active histone mark H3K4me3 has been monitored in peripheral blood monocytes from malaria patients. Genes related to innate immune memory response (trained immunity), such as interleukin 6 (IL6) and genes of the PI3K/AKT/mTOR pathway, showed increased levels of H3K4me3 and higher expression in monocytes primed with P. falciparum and stimulated with LPS when compared to control. This condition suggests that specific HMTs are involved in the development of immune memory in malaria [54,56]. Moreover, no differences were found in histone marks H3K27ac (open), H3K4me (open), and H3K9me3 (repressive) in innate memory genes of bone marrow monocytes from mice infected and reinfected with P. chabaudi [45]. These findings suggest that the trained immunity in Plasmodium spp. may be associated with epigenetic events, and Plasmodium species and cell lineage should be considered. Elucidating the innate immune response memory mechanism in malaria and the pathways involved in host tolerance to Plasmodium spp. will help in identifying targets for immunomodulators that can increase the host response to the parasite and avoid severe inflammation and tissue damage.
PTHM and chromatin remodeling were found to change gene expression related to processes other than immune response. In T. gondii infections of BeWo cells, the protein Ropthry 16 (ROP16) phosphorylates the ubiquitin-like containing PHD and RING finger domain (UHRF1), which is translocated to the nucleus. The UHFR1 recruits HDACs and HMTs, decreasing H3 acetylation and increasing the H3K9me3 mark in the cyclin B1 gene (CCNB1). In addition, UHFR1 recruits DNMT, increasing the CpG methylation in the CCNB1 promoter [111]. Those epigenetic events result in gene silencing, leading to cell-cycle arrest in the G2 phase (Figure 6). It has been shown that the T. gondii proliferation rate is higher when the host cell is in the G2 phase rather than in the G1 phase [140], although the reason for that remains unknown. Further investigation into the importance of cell-cycle arrest for the T. gondii infection success may identify potential drug targets for HDACi, HMTi, or DNMTi.

3.4.2. DNA Methylation

The methylation status of CpG sites is also responsible for chromatin remodeling and gene expression regulation. Usually, the hypomethylation of CpG in gene promoters is related to increased gene expression. This pattern was observed in the gene Tlr6 in the liver of mice infected with P. chabaudi. The TLR6 (toll-like receptor 6) acts with TLR2 in the recognition of pathogen-associated molecular patterns (PAMPs) by macrophages. This finding suggests the role of DNA methylation in the recognition of pathogens by Kuppfer cells in the liver [43]. The tissue damage caused by inflammation in malaria and the relation between TLR6 and malaria severity in P. falciparum and P. vivax infections [141] enforces the need for further investigation of Tlr6 and other genes of the Tlr family.
Changes in DNA methylation were also observed in the liver tissue of mice vaccinated against P. chabaudi. Some modifications observed in immune-related genes were the hypomethylations of Cx3cl1 and Gp130 genes, which are responsible for monocyte recruitment and IL-6 signal transducer, respectively. In addition, the hypermethylation of the Socs1, an inhibitor of the JAK/STAT pathway, was observed [44]. Leishmania donovani and Helicobacter pylori are two other intracellular pathogens that induce changes in the DNA methylation status of genes of the JAK/STAT pathway, downregulating or upregulating the immune response, respectively [142,143]. Taken together, these findings regarding DNA methylation in Plasmodium spp. infection suggest that DNMTi can be useful in the modulation of the host immune response and inflammation, although a deeper knowledge of the role of DNA methylation in malaria infection is needed.
The unwanted hypomethylation is important to consider in the use of DNMTi. Gupta et al. (2017) [51] demonstrated that the drug-resistance gene ABCB1 is down-methylated in severe malaria patients’ leukocytes, resulting in higher expression of the ABCB1 protein in such patients compared to that in uncomplicated malaria cases and healthy individuals. Thereby, the use of DNMTi can increase resistance to anti-malarial drugs. Therefore, future studies aiming to find targets for DNMTi drugs for this global disease should be carefully planned, considering the possible side effects of the drug and the possibility of undesired hypomethylation of the ABCB1 gene.
Epigenetic modifications driven by parasites also interfere in host reproduction and behavior. T. gondii-infected mice presented lower levels of luteinizing hormone (LH), testicular function, and sperm production than those of uninfected animals. DNA methylation analysis of testicular tissue revealed global DNA hypermethylation and hypomethylation of specific CpGs in the Crem and Hspa1 genes and the Creb1 promoter. As these genes are necessary for healthy spermatogenesis, their methylation and silencing could affect the reproductive fitness of infected animals [83].
Although T. gondii seems to interfere with testis function, testosterone levels are higher in infected mice. This hormone promotes hypomethylation and increases the expression of the arginine vasopressin (AVP) promoter gene in the poster dorsal medial amygdala (MePD), a region related to sexual behavior [84]. This effect leads to reduced fear and behavioral change in T. gondii-infected mice. Specifically, these mice become attracted to the odor of felines’ urine, favoring the perpetuation of the parasite’s life cycle [84,113].
The treatment with L-methionine, a methyl donor for DNA methylation, restored the fear of felines’ urine odor and reaffirmed the role of DNA methylation in behavior changes. L-methionine and S-adenosylmethionine, the major methyl donors, are marketed as dietary supplements in the USA, and they have been studied to treat metabolic and neurological disorders [144,145]. The medial amygdala is also part of the dopamine system. Syn et al. (2018) [92] reported that human retinal cells infected with T. gondii presented a reduced methylation and a higher expression of genes related to dopamine and amyloid pathways, which have an essential role in neurodevelopment and neurodegeneration.
Interestingly, these changes in behavior and in the neuronal pathways were not attributed to a localized effect of the T. gondii cysts, as was early suggested [146]. Whether the AVP, dopamine, and amyloid system changes are a consequence of a long-distance signaling process (i.e., testosterone-induced AVP hypomethylation and expression) or a local disturbance induced by T. gondii cysts remains unclear. Further study of the role of DNA methylation in T. gondii infection could help in identifying targets for epidrugs, with the aim of minimizing the undesirable neurological consequences of congenital toxoplasmosis.

3.4.3. ncRNA

Several miRNAs, their targets, and their regulatory pathways have already been described. It is also known that some drugs targeting miRNAs are undergoing clinical trials [147]. Some of these targeted miRNAs, such as miR155 and miR21, are involved in apicomplexan infection. The miR-155 miRNA has been investigated in many studies of Toxoplasma gondii infection [79,93,99,104,106,107,114,115,119,121]. This miRNA was upregulated in the brain of mice infected with the type-II strain of T. gondii and was related to its chronic phase during the development of cysts [79]. A recent study demonstrated that miR-155 was present in exosomes secreted by dendritic cells in T. gondii infection [121]. These exosomes are ingested by murine macrophages, and the miR-155 targets the suppressor of cytokine signaling 1 (SOCS1), which is an inhibitor of the NFκB pathway, increasing the activation of this pathway and resulting in the control of T. gondii tachyzoites proliferation.
Although miR-155 can boost the immune response and help control T. gondii infection, the activation of immune and inflammatory pathways may also be harmful. Meira-Strejevitch et al. (2020) [106] found miR-155 to be upregulated in the plasma of ocular toxoplasmosis patients, when compared to the disease’s asymptomatic form. In this scenario, the increase in immune and inflammatory pathways driven by this miRNA seemed to contribute to the retinal lesion. This suggests that this miRNA is a potential target for ncRNA-based therapy that modulates the host immune response. Nonetheless, the acute or chronic disease phase may be considered to investigate whether a miR-155 mimic or an anti-miR-155 will achieve better clinical results. In the latter case, the repurpose of MRG-106, an anti-miR-155, is in a clinical trial for lymphoma treatment [147].
The gain of miR155 expression was found in murine Kupffer cells during P. berghei genetically attenuated parasite (GAP) infection. The induced overexpression of this miRNA in GAP pre-sensitized mice raised their protection against P. berghei, suggesting that an miR-155 mimic can boost the immune response against this parasite after the GAP vaccine [39]. Cell proliferation and cancer development are other pathways related to miR-155. The miR-155 miRNA was observed to be upregulated in bovine B cells infected with T. annulata and in its exosomes [69,70]. During T. annulata infection, miR-155 targets the light-mediated developmental protein (DET1), which is involved in the ubiquitination of the c-Jun protein that works as an oncogenic transcription factor. In addition to inducing cell proliferation, c-Jun promotes positive feedback for miR-155 expression. The sustained miR-155 expression results in the maintenance of cell proliferation and the T. annulata cancer-related development. Considering the important role of miRNA in T. annulata infection, drug testing (i.e., MRG-106) should be carried out for developing new cattle treatment.
The miR-21 miRNA is also targeted by drugs that are undergoing clinical trial [147], and it seems to play a key role in apicomplexan infection. Like miR-155, miR-21 is known for its role in cancer development, characterized as an oncomiR. In colorectal cancer cells infected with C. parvum, miR-21 was found to be upregulated, suggesting that this parasite can play a critical role in oncomiR regulation, and thus in cancer development and evolution [25].
The miR-21 miRNA is also implicated in immune-response modulation as an anti-inflammatory molecule. The lower expression of this miRNA in the plasma of ocular toxoplasmosis patients, compared to that of asymptomatic individuals, may contribute to ophthalmic lesions [106]. On the contrary, this miRNA was found to be upregulated in the intestine of mice infected with C. parvum, where the miR-21 targeted the chemokine CCL20, diminishing the host defense against the parasite [21]. Moreover, these findings demonstrate that miR-21 plays a regulatory role in the immune response in apicomplexan infection, and its overexpression or silencing will vary according to the condition. The miR-21 miRNA was upregulated in the E. papillata infection of mice, but its role during the disease was not further investigated [126]. Although miR-21 may influence cell proliferation and immune response in apicomplexan infection, its target and the pathways need to be better described.
Other miRNAs have been demonstrated to have important roles in apicomplexan infection and should also be considered future drug targets. One of these miRNAs is miR-451, which is present in extracellular vesicles (EVs) secreted by red blood cells. EV secretion is increased in P. falciparum infection, and the EVs are internalized by endothelial cells and by the parasite [52]. In the endothelial cells BMEC-1, miR-451 targets CAV-1 (caveolin-1) and activates the transcription factor 2 (ATF2), downregulating their expression and resulting in endothelial barrier dysfunction [48]. In P. falciparum, miR-451 recognizes the 3′ and 5′ UTR region of var genes transcripts, blocking their translation and downregulating the expression of P. falciparum erythrocyte membrane protein-1 (PfEMP1), which can play a crucial role in host cell adhesion and invasion [52]. In addition, miR-451 targets the transcript of the regulatory subunit of the cAMP-dependent protein kinase (PKA-R) of P. falciparum, reducing its translation and suppressing parasite development. This mechanism seems to confer resistance against plasmodial infection in sickle cell disease patients [47]. Therefore, miR-451 is a great potential target for drugs that aim to control the parasite burden and endothelial cell damage.
The role of miRNAs of the let7 family has also been studied in apicomplexan diseases in C. parvum infection. Studies of cholangiocytes (H69 cells) and enterocytes (IEC4.1 cells) showed that during C. parvum infection, the toll-like receptor 4 (TLR4) is activated, leading to the stimulation of the NFκB pathway [16,22]. In H69 cells, the NFκB p50 is responsible for forming a silencer complex that deacetylases the histone H3 in the let7i promoter, inhibiting the expression of this miRNA [14]. The let7i targets several components of the immune response, such as TLR4, the cytokine-inducible Src homology 2-containing protein (CIS), and sirtuin-1 (SIRT1) [11,15,20]. Thus, the inhibition of let7i expression increases these proteins, which act as regulators of the immune response pathway. The increase in TLR4 results in an increment of NFκB, which inhibits let7i expression, showing positive feedback for TLR4 and NFκB expression [11]. The CIS protein stimulates the NFκB through the degradation of IκBα (NF-kappa-B inhibitor alpha) and inhibits inflammatory cytokines [15]. The inhibition of let7i also stimulates NFκB expression through CIS activity. However, the downregulation of let7i increases SIRT1, an inhibitor of NFκB [20]. These findings suggest that let7 family miRNAs promote a fine-tuned regulation of immune response, mainly through the NFκB pathway in cryptosporidiosis. Thus, manipulating this miRNA could increase the immune response against C. parvum.

4. Summary

Our study presented a bibliometric analysis of selected reports on apicomplexan parasite-driven epigenetic changes in hosts. It also summarized the main findings in the literature, pointing out the pathways that could be addressed by drugs targeting histone PTM, DNA methylation, and ncRNAs. The repurposing of drugs in the HDT context may be a shortcut for the implementation of new treatments for parasitic diseases. Herein, we presented putative targets for novel epidrugs and ncRNA-based therapies in apicomplexan infections. However, we understand that more detailed knowledge of the hosts’ epigenetic pathways addressed by the apicomplexan parasites is still needed before establishing a definitive drug target. Although the number of studies in this context is rather limited, the rising number of publications in the past decade reveals an increasing interest in the scientific community about the role of epigenetics in apicomplexan diseases. Moreover, we believe that the emergence of novel therapy options could be enhanced through drug repurposing. The diseases presented here cause major social and health problems globally, and we have the opportunity to improve human and animal health and welfare by consolidating international collaborative work.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/pathogens12020299/s1, Table S1: Abbreviation list; Table S2: Studies that modulate the epigenetic pathways interfered by parasites [148].

Author Contributions

Conceptualization, Y.d.O.B.; methodology, Y.d.O.B. and M.B.M.; formal analysis, Y.d.O.B. and M.B.M.; data curation, Y.d.O.B. and M.B.M.; writing—original draft preparation, Y.d.O.B.; writing—review and editing M.B.M.; supervision, M.B.M.; project administration, M.B.M.; funding acquisition, M.B.M. All authors have read and agreed to the published version of the manuscript.

Funding

Funding was provided by Coordination for the Improvement of Higher Education Personnel, CAPES 001/2018 and by the National Council of Scientific and Technological Development, CNPq 01/2019.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors thank Raphael Antônio Borges Gomes from the Food Department of the Federal University of Ouro Preto for helping with the design of Figure 5.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vilcinskas, A. The Role of Epigenetics in Host–Parasite Coevolution: Lessons from the Model Host Insects Galleria Mellonella and Tribolium Castaneum. Zoology 2016, 119, 273–280. [Google Scholar] [CrossRef]
  2. Waddington, C.H. The Epigenotype. Endeavour 1942, 1, 18–20. [Google Scholar] [CrossRef]
  3. Nicoglou, A.; Merlin, F. Epigenetics: A Way to Bridge the Gap between Biological Fields. Stud. Hist. Philos. Sci. Part C Stud. Hist. Philos. Biol. Biomed. Sci. 2017, 66, 73–82. [Google Scholar] [CrossRef]
  4. Kumar, S.; Gonzalez, E.A.; Rameshwar, P.; Etchegaray, J.P. Non-Coding RNAs as Mediators of Epigenetic Changes in Malignancies. Cancers 2020, 12, 3657. [Google Scholar] [CrossRef]
  5. Brandão, Y.d.O.; Toledo, M.B.; Chequin, A.; Cristo, T.G.; Sousa, R.S.; Ramos, E.A.S.; Klassen, G. DNA Methylation Status of the Estrogen Receptor α Gene in Canine Mammary Tumors. Vet. Pathol. 2018, 55, 510–516. [Google Scholar] [CrossRef]
  6. Pires, V.S.; de, O. Ganzella, F.A.; Minozzo, G.A.; de Castro, L.L.; Moncada, A.D.B.; Klassen, G.; Ramos, E.A.S.; Molento, M.B. Epigenetic Regulation of SLC11a1 Gene in Horses Infected with Cyathostomins. Gene Rep. 2021, 25, 101410. [Google Scholar] [CrossRef]
  7. Berdasco, M.; Esteller, M. Clinical Epigenetics: Seizing Opportunities for Translation. Nat. Rev. Genet. 2019, 20, 109–127. [Google Scholar] [CrossRef]
  8. Ganesan, A.; Arimondo, P.B.; Rots, M.G.; Jeronimo, C.; Berdasco, M. The Timeline of Epigenetic Drug Discovery: From Reality to Dreams. Clin. Epigenetics 2019, 11, 174. [Google Scholar] [CrossRef]
  9. Egea, P.F. Crossing the Vacuolar Rubicon: Structural Insights into Effector Protein Trafficking in Apicomplexan Parasites. Microorganisms 2020, 8, 865. [Google Scholar] [CrossRef]
  10. Villares, M.; Berthelet, J.; Weitzman, J.B. The Clever Strategies Used by Intracellular Parasites to Hijack Host Gene Expression. Semin. Immunopathol. 2020, 42, 215–226. [Google Scholar] [CrossRef]
  11. Chen, X.-M.; Splinter, P.L.; O’Hara, S.P.; LaRusso, N.F. A Cellular Micro-RNA, Let-7i, Regulates Toll-like Receptor 4 Expression and Contributes to Cholangiocyte Immune Responses against Cryptosporidium parvum Infection. J. Biol. Chem. 2007, 282, 28929–28938. [Google Scholar] [CrossRef]
  12. Zhou, R.; Hu, G.; Liu, J.; Gong, A.-Y.; Drescher, K.M.; Chen, X.-M. NF-KappaB P65-Dependent Transactivation of MiRNA Genes Following Cryptosporidium parvum Infection Stimulates Epithelial Cell Immune Responses. PLoS Pathog. 2009, 5, e1000681. [Google Scholar] [CrossRef]
  13. Gong, A.-Y.; Zhou, R.; Hu, G.; Liu, J.; Sosnowska, D.; Drescher, K.M.; Dong, H.; Chen, X.-M. Cryptosporidium parvum Induces B7-H1 Expression in Cholangiocytes by Down-Regulating MicroRNA-513. J. Infect. Dis. 2010, 201, 160–169. [Google Scholar] [CrossRef]
  14. O’Hara, S.P.; Splinter, P.L.; Gajdos, G.B.; Trussoni, C.E.; Fernandez-Zapico, M.E.; Chen, X.M.X.-M.; LaRusso, N.F. NFκB P50-CCAAT/Enhancer-Binding Protein β (C/EBPβ)-Mediated Transcriptional Repression of MicroRNA Let-7i Following Microbial Infection. J. Biol. Chem. 2010, 285, 216–225. [Google Scholar] [CrossRef]
  15. Hu, G.; Zhou, R.; Liu, J.; Gong, A.-Y.; Eischeid, A.N.; Dittman, J.W.; Chen, X.-M. MicroRNA-98 and Let-7 Confer Cholangiocyte Expression of Cytokine-Inducible Src Homology 2-Containing Protein in Response to Microbial Challenge. J. Immunol. 2009, 183, 1617–1624. [Google Scholar] [CrossRef]
  16. Hu, G.; Zhou, R.; Liu, J.; Gong, A.-Y.; Chen, X.-M. MicroRNA-98 and Let-7 Regulate Expression of Suppressor of Cytokine Signaling 4 in Biliary Epithelial Cells in Response to Cryptosporidium parvum Infection. J. Infect. Dis. 2010, 202, 125–135. [Google Scholar] [CrossRef]
  17. Gong, A.-Y.; Hu, G.; Zhou, R.; Liu, J.; Feng, Y.; Soukup, G.A.; Chen, X.-M. MicroRNA-221 Controls Expression of Intercellular Adhesion Molecule-1 in Epithelial Cells in Response to Cryptosporidium parvum Infection. Int. J. Parasitol. 2011, 41, 397–403. [Google Scholar] [CrossRef]
  18. Zhou, R.; Gong, A.-Y.; Eischeid, A.N.; Chen, X.-M. MiR-27b Targets KSRP to Coordinate TLR4-Mediated Epithelial Defense against Cryptosporidium parvum Infection. PLoS Pathog. 2012, 8, e1002702. [Google Scholar] [CrossRef]
  19. Hu, G.; Gong, A.-Y.; Roth, A.L.; Huang, B.Q.; Ward, H.D.; Zhu, G.; LaRusso, N.F.; Hanson, N.D.; Chen, X.-M. Release of Luminal Exosomes Contributes to TLR4-Mediated Epithelial Antimicrobial Defense. PLoS Pathog. 2013, 9, e1003261. [Google Scholar] [CrossRef]
  20. Xie, H.; Lei, N.; Gong, A.-Y.; Chen, X.-M.; Hu, G. Cryptosporidium parvum Induces SIRT1 Expression in Host Epithelial Cells through Downregulating Let-7i. Hum. Immunol. 2014, 75, 760–765. [Google Scholar] [CrossRef]
  21. Guesdon, W.; Auray, G.; Pezier, T.; Bussière, F.I.; Drouet, F.; Le Vern, Y.; Marquis, M.; Potiron, L.; Rabot, S.; Bruneau, A.; et al. CCL20 Displays Antimicrobial Activity Against Cryptosporidium parvum, but Its Expression Is Reduced During Infection in the Intestine of Neonatal Mice. J. Infect. Dis. 2015, 212, 1332–1340. [Google Scholar] [CrossRef]
  22. Wang, Y.; Gong, A.-Y.; Ma, S.; Chen, X.-M.X.; Strauss-Soukup, J.K.; Chen, X.-M.X. Delivery of Parasite Cdg7_Flc_0990 RNA Transcript into Intestinal Epithelial Cells during Cryptosporidium parvum Infection Suppresses Host Cell Gene Transcription through Epigenetic Mechanisms. Cell. Microbiol. 2017, 19, e12760. [Google Scholar] [CrossRef]
  23. Ming, Z.; Gong, A.-Y.; Wang, Y.; Zhang, X.-T.; Li, M.; Mathy, N.W.; Strauss-Soukup, J.K.; Chen, X.-M. Involvement of Cryptosporidium parvum Cdg7_FLc_1000 RNA in the Attenuation of Intestinal Epithelial Cell Migration via Trans-Suppression of Host Cell SMPD3. J. Infect. Dis. 2018, 217, 122–133. [Google Scholar] [CrossRef]
  24. Ming, Z.; Gong, A.-Y.; Wang, Y.; Zhang, X.; Li, M.; Li, Y.; Pang, J.; Dong, S.; Strauss-Soukup, J.K.; Chen, X.-M. Trans-Suppression of Host CDH3 and LOXL4 Genes during Cryptosporidium parvum Infection Involves Nuclear Delivery of Parasite Cdg7_FLc_1000 RNA. Int. J. Parasitol. 2018, 48, 423–431. [Google Scholar] [CrossRef]
  25. Jainul, M.A.; Isa, M.L.M.; Yusof, A.M.M. staining and mirna expression of H.-8 and H.-29 cell lines upon cryptosporidium infection Immunofluorescent Staining and Mirna Expression of HCT-8 and HT-29 Cell Lines upon Cryptosporidium Infection. Malays. Appl. Biol. 2018, 47, 101–112. [Google Scholar]
  26. Li, M.; Gong, A.-Y.; Zhang, X.-T.; Wang, Y.; Mathy, N.W.; Martins, G.A.; Strauss-Soukup, J.K.; Chen, X.-M. Induction of a Long Noncoding RNA Transcript, NR_045064, Promotes Defense Gene Transcription and Facilitates Intestinal Epithelial Cell Responses against Cryptosporidium Infection. J. Immunol. 2018, 201, 3630–3640. [Google Scholar] [CrossRef]
  27. Liu, T.-L.; Fan, X.-C.; Li, Y.-H.; Yuan, Y.-J.; Yin, Y.-L.; Wang, X.-T.; Zhang, L.-X.; Zhao, G.-H. Expression Profiles of MRNA and LncRNA in HCT-8 Cells Infected With Cryptosporidium parvum IId Subtype. Front. Microbiol. 2018, 9, 1409. [Google Scholar] [CrossRef]
  28. Zhao, G.-H.; Gong, A.-Y.; Wang, Y.; Zhang, X.-T.; Li, M.; Mathy, N.W.; Chen, X.-M. Nuclear Delivery of Parasite Cdg2_FLc_0220 RNA Transcript to Epithelial Cells during Cryptosporidium parvum Infection Modulates Host Gene Transcription. Vet. Parasitol. 2018, 251, 27–33. [Google Scholar] [CrossRef]
  29. Wang, C.; Liu, L.; Zhu, H.; Zhang, L.; Wang, R.; Zhang, Z.; Huang, J.; Zhang, S.; Jian, F.; Ning, C.; et al. MicroRNA Expression Profile of HCT-8 Cells in the Early Phase of Cryptosporidium parvum Infection. BMC Genom. 2019, 20, 37. [Google Scholar] [CrossRef]
  30. Zhang, G.; Zhang, Y.; Niu, Z.; Wang, C.; Xie, F.; Li, J.; Zhang, S.; Qi, M.; Jian, F.; Ning, C.; et al. Cryptosporidium parvum Upregulates MiR-942-5p Expression in HCT-8 Cells via TLR2/TLR4-NF-ΚB Signaling. Parasites Vectors 2020, 13, 4–9. [Google Scholar] [CrossRef]
  31. Gong, A.-Y.; Wang, Y.; Li, M.; Zhang, X.-T.; Deng, S.; Chen, J.M.; Lu, E.; Mathy, N.W.; Martins, G.A.; Strauss-Soukup, J.K.; et al. LncRNA XR_001779380 Primes Epithelial Cells for IFN-γ-Mediated Gene Transcription and Facilitates Age-Dependent Intestinal Antimicrobial Defense. MBio 2021, 12, e0212721. [Google Scholar] [CrossRef]
  32. Sabir, M.J.; Low, R.; Hall, N.; Kamli, M.R.; Malik, M.Z. A Bioinformatics Approach to Identifying Potential Biomarkers for Cryptosporidium parvum: A Coccidian Parasite Associated with Fetal Diarrhea. Vaccines 2021, 9, 1427. [Google Scholar] [CrossRef]
  33. Yin, Y.-L.; Liu, T.-L.; Yao, Q.; Wang, Y.-X.; Wu, X.-M.; Wang, X.-T.; Yang, X.; Song, J.-K.; Zhao, G.-H. Circular RNA CiRS-7 Affects the Propagation of Cryptosporidium parvum in HCT-8 Cells by Sponging MiR-1270 to Activate the NF-ΚB Signaling Pathway. Parasites Vectors 2021, 14, 238. [Google Scholar] [CrossRef]
  34. Mathy, N.W.; Deng, S.; Gong, A.-Y.; Li, M.; Wang, Y.; Burleigh, O.; Kochvar, A.; Whiteford, E.R.; Shibata, A.; Chen, X.-M. The Long Non-Coding RNA Nostrill Regulates Transcription of Irf7 Through Interaction With NF-ΚB P65 to Enhance Intestinal Epithelial Defense Against Cryptosporidium parvum. Front. Immunol. 2022, 13, 863957. [Google Scholar] [CrossRef]
  35. Sawant, M.; Benamrouz-Vanneste, S.; Meloni, D.; Gantois, N.; Even, G.; Guyot, K.; Creusy, C.; Duval, E.; Wintjens, R.; Weitzman, J.B.; et al. Putative SET-Domain Methyltransferases in Cryptosporidium parvum and Histone Methylation during Infection. Virulence 2022, 13, 1632–1650. [Google Scholar] [CrossRef]
  36. Ulusan Bagci, Ö.; Caner, A.; Ulusan Bağcı, Ö.; Caner, A.; Ulusan Bagci, Ö.; Caner, A.; Bağci, Ö.U.; Caner, A. miRNA Expression Profile in Ileocecal Adenocarcinoma Cells Infected with Cryptosporidium. Mikrobiyoloji Bul. 2022, 56, 449–465. [Google Scholar] [CrossRef]
  37. Xie, F.; Zhang, Y.; Li, J.; Sun, L.; Zhang, L.; Qi, M.; Zhang, S.; Jian, F.; Li, X.; Li, J.; et al. MiR-942-5p Targeting the IFI27 Gene Regulates HCT-8 Cell Apoptosis via a TRAIL-Dependent Pathway during the Early Phase of Cryptosporidium parvum Infection. Parasites Vectors 2022, 15, 291. [Google Scholar] [CrossRef]
  38. El-Assaad, F.; Hempel, C.; Combes, V.; Mitchell, A.J.; Ball, H.J.; Kurtzhals, J.A.L.L.; Hunt, N.H.; Mathys, J.-M.M.; Grau, G.E.R.R. Differential MicroRNA Expression in Experimental Cerebral and Noncerebral Malaria. Infect. Immun. 2011, 79, 2379–2384. [Google Scholar] [CrossRef]
  39. Hentzschel, F.; Hammerschmidt-Kamper, C.; Börner, K.; Heiss, K.; Knapp, B.; Sattler, J.M.; Kaderali, L.; Castoldi, M.; Bindman, J.G.; Malato, Y.; et al. AAV8-Mediated In Vivo Overexpression of MiR-155 Enhances the Protective Capacity of Genetically Attenuated Malarial Parasites. Mol. Ther. 2014, 22, 2130–2141. [Google Scholar] [CrossRef]
  40. Martin-Alonso, A.; Cohen, A.; Quispe-Ricalde, M.A.; Foronda, P.; Benito, A.; Berzosa, P.; Valladares, B.; Grau, G.E. Differentially Expressed MicroRNAs in Experimental Cerebral Malaria and Their Involvement in Endocytosis, Adherens Junctions, FoxO and TGF-β Signalling Pathways. Sci. Rep. 2018, 8, 11277. [Google Scholar] [CrossRef]
  41. Delic, D.; Dkhil, M.; Al-Quraishy, S.; Wunderlich, F.; Delić, D.; Dkhil, M.; Al-Quraishy, S.; Wunderlich, F. Hepatic MiRNA Expression Reprogrammed by Plasmodium Chabaudi Malaria. Parasitol. Res. 2011, 108, 1111–1121. [Google Scholar] [CrossRef]
  42. Al-Quraishy, S.; Dkhil, M.A.; Delic, D.; Abdel-Baki, A.A.; Wunderlich, F. Organ-Specific Testosterone-Insensitive Response of MiRNA Expression of C57BL/6 Mice to Plasmodium Chabaudi Malaria. Parasitol. Res. 2012, 111, 1093–1101. [Google Scholar] [CrossRef] [PubMed]
  43. Al-Quraishy, S.; Dkhil, M.A.; Abdel-Baki, A.A.S.; Delic, D.; Santourlidis, S.; Wunderlich, F. Genome-Wide Screening Identifies Plasmodium Chabaudi-Induced Modifications of DNA Methylation Status of Tlr1 and Tlr6 Gene Promoters in Liver, but Not Spleen, of Female C57BL/6 Mice. Parasitol. Res. 2013, 112, 3757–3770. [Google Scholar] [CrossRef]
  44. Al-Quraishy, S.; Dkhil, M.A.; Abdel-Baki, A.-A.S.; Ghanjati, F.; Erichsen, L.; Santourlidis, S.; Wunderlich, F.; Araúzo-Bravo, M.J. Protective Vaccination and Blood-Stage Malaria Modify DNA Methylation of Gene Promoters in the Liver of Balb/c Mice. Parasitol. Res. 2017, 116, 1463–1477. [Google Scholar] [CrossRef]
  45. Nahrendorf, W.; Ivens, A.; Spence, P.J. Inducible Mechanisms of Disease Tolerance Provide an Alternative Strategy of Acquired Immunity to Malaria. Elife 2021, 10, e63838. [Google Scholar] [CrossRef] [PubMed]
  46. Rathjen, T.; Nicol, C.; McConkey, G.; Dalmay, T. Analysis of Short RNAs in the Malaria Parasite and Its Red Blood Cell Host. FEBS Lett. 2006, 580, 5185–5188. [Google Scholar] [CrossRef]
  47. LaMonte, G.; Philip, N.; Reardon, J.; Lacsina, J.R.R.; Majoros, W.; Chapman, L.; Thornburg, C.D.; Telen, M.J.; Ohler, U.; Nicchitta, C.V.; et al. Translocation of Sickle Cell Erythrocyte MicroRNAs into Plasmodium Falciparum Inhibits Parasite Translation and Contributes to Malaria Resistance. Cell Host Microbe 2012, 12, 187–199. [Google Scholar] [CrossRef]
  48. Mantel, P.-Y.; Hjelmqvist, D.; Walch, M.; Kharoubi-Hess, S.; Nilsson, S.; Ravel, D.; Ribeiro, M.; Grüring, C.; Ma, S.Y.; Padmanabhan, P.; et al. Infected Erythrocyte-Derived Extracellular Vesicles Alter Vascular Function via Regulatory Ago2-MiRNA Complexes in Malaria. Nat. Commun. 2016, 7, 12727. [Google Scholar] [CrossRef]
  49. Babatunde, K.A.; Mbagwu, S.; Hernández-Castañeda, M.A.; Adapa, S.R.; Walch, M.; Filgueira, L.; Falquet, L.; Jiang, R.H.Y.; Ghiran, I.; Mantel, P.-Y. Malaria Infected Red Blood Cells Release Small Regulatory RNAs through Extracellular Vesicles. Sci. Rep. 2018, 8, 884. [Google Scholar] [CrossRef]
  50. Burel, J.G.; Apte, S.H.; Groves, P.L.; Boyle, M.J.; Langer, C.; Beeson, J.G.; McCarthy, J.S.; Doolan, D.L. Dichotomous MiR Expression and Immune Responses Following Primary Blood-Stage Malaria. JCI Insight 2017, 2, e93434. [Google Scholar] [CrossRef]
  51. Gupta, H.; Chaudhari, S.; Rai, A.; Bhat, S.; Sahu, P.K.; Hande, M.H.; D’Souza, S.C.; Shashikiran, U.; Satyamoorthy, K. Genetic and Epigenetic Changes in Host ABCB1 Influences Malaria Susceptibility to Plasmodium Falciparum. PLoS ONE 2017, 12, e0175702. [Google Scholar] [CrossRef]
  52. Wang, Z.; Xi, J.; Hao, X.; Deng, W.; Liu, J.; Wei, C.; Gao, Y.; Zhang, L.; Wang, H. Red Blood Cells Release Microparticles Containing Human Argonaute 2 and MiRNAs to Target Genes of Plasmodium Falciparum. Emerg. Microbes Infect. 2017, 6, 1–11. [Google Scholar] [CrossRef]
  53. Li, J.; Huang, M.; Li, Z.; Li, W.; Wang, F.; Wang, L.; Li, X.; Zheng, X.; Zou, Y. Identification of Potential Whole Blood MicroRNA Biomarkers for the Blood Stage of Adult Imported Falciparum Malaria through Integrated MRNA and MiRNA Expression Profiling. Biochem. Biophys. Res. Commun. 2018, 506, 471–477. [Google Scholar] [CrossRef]
  54. Schrum, J.E.; Juliet, N.C.; Dobbs, K.R.; Michael, C.K.; George, W.R.; Gazzinelli, R.T.; Netea, M.G.; Kazura, J.W.; Dent, A.E.; Katherine, A.F. Plasmodium Falciparum Induces Trained Innate Immunity. J. Immunol. 2018, 200, 1243–1248. [Google Scholar] [CrossRef]
  55. Dandewad, V.; Vindu, A.; Joseph, J.; Seshadri, V. Import of Human MiRNA-RISC Complex into Plasmodium Falciparum and Regulation of the Parasite Gene Expression. J. Biosci. 2019, 44, 50. [Google Scholar] [CrossRef]
  56. Walk, J.; Keramati, F.; de Bree, L.C.J.; Arts, R.J.W.; Blok, B.; Netea, M.G.; Stunnenberg, H.G.; Sauerwein, R.W. Controlled Human Malaria Infection Induces Long-Term Functional Changes in Monocytes. Front. Mol. Biosci. 2020, 7, 604553. [Google Scholar] [CrossRef]
  57. Chakrabarti, M.; Garg, S.; Rajagopal, A.; Pati, S.; Singh, S. Targeted Repression of Plasmodium Apicortin by Host MicroRNA Impairs Malaria Parasite Growth and Invasion. Dis. Model. Mech. 2020, 13, dmm042820. [Google Scholar] [CrossRef]
  58. Ketprasit, N.; Cheng, I.S.; Deutsch, F.; Tran, N.; Imwong, M.; Combes, V.; Palasuwan, D. The Characterization of Extracellular Vesicles-Derived MicroRNAs in Thai Malaria Patients. Malar. J. 2020, 19, 285. [Google Scholar] [CrossRef]
  59. Bertrams, W.; Griss, K.; Han, M.; Seidel, K.; Hippenstiel, S.; Suttorp, N.; Finkernagel, F.; Wilhelm, J.; Vogelmeier, C.F.; Schmeck, B. Transcriptional Analysis Identifies Potential Biomarkers and Molecular Regulators in Acute Malaria Infection. Life Sci. 2021, 270, 119158. [Google Scholar] [CrossRef] [PubMed]
  60. Gupta, H.; Rubio, M.; Sitoe, A.; Varo, R.; Cisteró, P.; Madrid, L.; Cuamba, I.; Jimenez, A.; Martiáñez-Vendrell, X.; Barrios, D.; et al. Plasma MicroRNA Profiling of Plasmodium Falciparum Biomass and Association with Severity of Malaria Disease. Emerg. Infect. Dis. 2021, 27, 430–442. [Google Scholar] [CrossRef]
  61. Hadighi, R.; Heidari, A.; Fallah, P.; Keshavarz, H.; Tavakoli, Z.; Mansouri, S.; Sadrkhanloo, M. Key Plasma MicroRNAs Variations in Patients with Plasmodium Vivax Malaria in Iran. Heliyon 2022, 8, e09018. [Google Scholar] [CrossRef] [PubMed]
  62. Oxendine Harp, K.; Bashi, A.; Botchway, F.; Dei-adomakoh, Y.; Iqbal, S.A.; Wilson, M.D.; Adjei, A.A.; Stiles, J.K.; Driss, A.; Harp, K.O.; et al. MicroRNAs MiR-451a and Let-7i-5p Profiles in Circulating Exosomes Vary among Individuals with Different Sickle Hemoglobin Genotypes and Malaria. J. Clin. Med. 2022, 11, 500. [Google Scholar] [CrossRef] [PubMed]
  63. Chamnanchanunt, S.; Kuroki, C.; Desakorn, V.; Enomoto, M.; Thanachartwet, V.; Sahassananda, D.; Sattabongkot, J.; Jenwithisuk, R.; Fucharoen, S.; Svasti, S.; et al. Downregulation of Plasma MiR-451 and MiR-16 in Plasmodium Vivax Infection. Exp. Parasitol. 2015, 155, 19–25. [Google Scholar] [CrossRef]
  64. Kaur, H.; Sehgal, R.; Kumar, A.; Sehgal, A.; Bansal, D.; Sultan, A.A. Screening and Identification of Potential Novel Biomarker for Diagnosis of Complicated Plasmodium Vivax Malaria. J. Transl. Med. 2018, 16, 272. [Google Scholar] [CrossRef] [PubMed]
  65. Santos, M.L.S.S.; Coimbra, R.S.; Sousa, T.N.; Guimarães, L.F.F.F.; Gomes, M.S.; Amaral, L.R.; Pereira, D.B.; Fontes, C.J.F.F.; Hawwari, I.; Franklin, B.S.; et al. The Interface Between Inflammatory Mediators and MicroRNAs in Plasmodium Vivax Severe Thrombocytopenia. Front. Cell. Infect. Microbiol. 2021, 11, 631333. [Google Scholar] [CrossRef] [PubMed]
  66. Cohen, A.; Zinger, A.; Tiberti, N.; Grau, G.E.R.; Combes, V. Differential Plasma Microvesicle and Brain Profiles of MicroRNA in Experimental Cerebral Malaria. Malar. J. 2018, 17, 192. [Google Scholar] [CrossRef] [PubMed]
  67. Chen, G.; Liu, S.-C.; Fan, X.-Y.; Jin, Y.-L.; Li, X.; Du, Y.-T. Plasmodium Manipulates the Expression of Host Long Non-Coding RNA during Red Blood Cell Intracellular Infection. Parasites Vectors 2022, 15, 182. [Google Scholar] [CrossRef]
  68. Cock-Rada, A.M.; Medjkane, S.; Janski, N.; Yousfi, N.; Perichon, M.; Chaussepied, M.; Chluba, J.; Langsley, G.; Weitzman, J.B. SMYD3 Promotes Cancer Invasion by Epigenetic Upregulation of the Metalloproteinase MMP-9. Cancer Res. 2012, 72, 810–820. [Google Scholar] [CrossRef]
  69. Marsolier, J.; Pineau, S.; Medjkane, S.; Perichon, M.; Yin, Q.Y.; Flemington, E.; Weitzman, M.D.; Weitzman, J.B. OncomiR Addiction Is Generated by a MiR-155 Feedback Loop in Theileria-Transformed Leukocytes. PLoS Pathog. 2013, 9, e1003222. [Google Scholar] [CrossRef]
  70. Gillan, V.; Simpson, D.M.; Kinnaird, J.; Maitland, K.; Shiels, B.; Devaney, E. Characterisation of Infection Associated MicroRNA and Protein Cargo in Extracellular Vesicles of Theileria Annulata Infected Leukocytes. Cell. Microbiol. 2019, 21, e12969. [Google Scholar] [CrossRef]
  71. Haidar, M.; Rchiad, Z.; Ansari, H.R.; Ben-Rached, F.; Tajeri, S.; Latre De Late, P.; Langsley, G.; Pain, A. MiR-126-5p by Direct Targeting of JNK-Interacting Protein-2 (JIP-2) Plays a Key Role in Theileria-Infected Macrophage Virulence. PLoS Pathog. 2018, 14, e1006942. [Google Scholar] [CrossRef] [PubMed]
  72. Leng, J.; Denkers, E.Y. Toxoplasma gondii Inhibits Covalent Modification of Histone H3 at the IL-10 Promoter in Infected Macrophages. PLoS ONE 2009, 4, e7589. [Google Scholar] [CrossRef]
  73. Leng, J.; Butcher, B.A.; Egan, C.E.; Abi Abdallah, D.S.; Denkers, E.Y. Toxoplasma gondii Prevents Chromatin Remodeling Initiated by TLR-Triggered Macrophage Activation. J. Immunol. 2009, 182, 489–497. [Google Scholar] [CrossRef]
  74. Zeiner, G.M.; Norman, K.L.; Thomson, J.M.; Hammond, S.M.; Boothroyd, J.C. Toxoplasma gondii Infection Specifically Increases the Levels of Key Host MicroRNAs. PLoS ONE 2010, 5, e8742. [Google Scholar] [CrossRef]
  75. Lang, C.; Hildebrandt, A.; Brand, F.; Opitz, L.; Dihazi, H.; Luder, C.G.K. Impaired Chromatin Remodelling at STAT1-Regulated Promoters Leads to Global Unresponsiveness of Toxoplasma gondii-Infected Macrophages to IFN-Gamma. PLoS Pathog. 2012, 8, e1002483. [Google Scholar] [CrossRef]
  76. Cai, Y.; Chen, H.; Jin, L.; You, Y.; Shen, J. STAT3-Dependent Transactivation of MiRNA Genes Following Toxoplasma gondii Infection in Macrophage. Parasites Vectors 2013, 6, 356. [Google Scholar] [CrossRef]
  77. Xu, M.-J.; Zhou, D.-H.; Nisbet, A.J.; Huang, S.-Y.; Fan, Y.-F.; Zhu, X.-Q. Characterization of Mouse Brain MicroRNAs after Infection with Cyst-Forming Toxoplasma gondii. Parasites Vectors 2013, 6, 154. [Google Scholar] [CrossRef]
  78. Cai, Y.; Chen, H.; Mo, X.; Tang, Y.; Xu, X.; Zhang, A.; Lun, Z.; Lu, F.; Wang, Y.; Shen, J. Toxoplasma gondii Inhibits Apoptosis via a Novel STAT3-MiR-17–92-Bim Pathway in Macrophages. Cell. Signal. 2014, 26, 1204–1212. [Google Scholar] [CrossRef] [PubMed]
  79. Cannella, D.; Brenier-Pinchart, M.-P.; Braun, L.; van Rooyen, J.M.; Bougdour, A.; Bastien, O.; Behnke, M.S.; Curt, R.-L.; Curt, A.; Saeij, J.P.J.; et al. MiR-146a and MiR-155 Delineate a MicroRNA Fingerprint Associated with Toxoplasma Persistence in the Host Brain. Cell Rep. 2014, 6, 928–937. [Google Scholar] [CrossRef]
  80. Jia, B.; Chang, Z.; Wei, X.; Lu, H.; Yin, J.; Jiang, N.; Chen, Q. Plasma MicroRNAs Are Promising Novel Biomarkers for the Early Detection of Toxoplasma gondii Infection. Parasites Vectors 2014, 7, 433. [Google Scholar] [CrossRef] [PubMed]
  81. Li, Y.; Kannan, G.; Pletnikov, M.V.; Yolken, R.H.; Xiao, J. Chronic Infection of Toxoplasma gondii Downregulates MiR-132 Expression in Multiple Brain Regions in a Sex-Dependent Manner. Parasitology 2014, 142, 623–632. [Google Scholar] [CrossRef]
  82. Xiao, J.; Li, Y.; Prandovszky, E.; Karuppagounder, S.S.; Talbot, C.C.; Dawson, V.L.; Dawson, T.M.; Yolken, R.H. MicroRNA-132 Dysregulation in Toxoplasma gondii Infection Has Implications for Dopamine Signaling Pathway. Neuroscience 2014, 268, 128–138. [Google Scholar] [CrossRef]
  83. Dvorakova-Hortova, K.; Sidlova, A.; Ded, L.; Hladovcova, D.; Vieweg, M.; Weidner, W.; Steger, K.; Stopka, P.; Paradowska-Dogan, A. Toxoplasma gondii Decreases the Reproductive Fitness in Mice. PLoS ONE 2014, 9, e96770. [Google Scholar] [CrossRef]
  84. Hari Dass, S.A.; Vyas, A.; Dass, S.A.H.; Vyas, A.; Hari Dass, S.A.; Vyas, A.; Dass, S.A.H.; Vyas, A.; Hari Dass, S.A.; Vyas, A.; et al. Toxoplasma gondii Infection Reduces Predator Aversion in Rats through Epigenetic Modulation in the Host Medial Amygdala. Mol. Ecol. 2014, 23, 6114–6122. [Google Scholar] [CrossRef]
  85. Rosowski, E.E.; Nguyen, Q.P.; Camejo, A.; Spooner, E.; Saeij, J.P.J. Toxoplasma gondii Inhibits Gamma Interferon (IFN-Gamma)- and IFN-Beta-Induced Host Cell STAT1 Transcriptional Activity by Increasing the Association of STAT1 with DNA. Infect. Immun. 2014, 82, 706–719. [Google Scholar] [CrossRef]
  86. He, J.-J.; Ma, J.; Wang, J.-L.; Xu, M.-J.; Zhu, X.-Q. Analysis of MiRNA Expression Profiling in Mouse Spleen Affected by Acute Toxoplasma gondii Infection. Infect. Genet. Evol. 2016, 37, 137–142. [Google Scholar] [CrossRef]
  87. Kim, M.-K.J.; Jung, B.-K.; Cho, J.; Song, H.; Pyo, K.-H.; Lee, J.M.; Kim, M.-K.J.; Chai, J.-Y. Exosomes Secreted by Toxoplasma gondii-Infected L6 Cells: Their Effects on Host Cell Proliferation and Cell Cycle Changes. Korean J. Parasitol. 2016, 54, 147–154. [Google Scholar] [CrossRef]
  88. Gay, G.; Braun, L.; Brenier-Pinchart, M.-P.P.; Vollaire, J.; Josserand, V.; Bertini, R.-L.L.; Varesano, A.; Touquet, B.; De Bock, P.-J.J.; Coute, Y.; et al. Toxoplasma gondii TgIST Co-Opts Host Chromatin Repressors Dampening STAT1-Dependent Gene Regulation and IFN-γ-Mediated Host Defenses. J. Exp. Med. 2016, 213, 1779–1798. [Google Scholar] [CrossRef]
  89. Olias, P.; Etheridge, R.D.; Zhang, Y.; Holtzman, M.J.; Sibley, L.D. Toxoplasma Effector Recruits the Mi-2/NuRD Complex to Repress STAT1 Transcription and Block IFN-γ-Dependent Gene Expression. Cell Host Microbe 2016, 20, 72–82. [Google Scholar] [CrossRef]
  90. Cong, W.; Zhang, X.-X.; He, J.-J.; Li, F.-C.; Elsheikha, H.M.; Zhu, X.-Q. Global MiRNA Expression Profiling of Domestic Cat Livers Following Acute Toxoplasma gondii Infection. Oncotarget 2017, 8, 25599–25611. [Google Scholar] [CrossRef]
  91. Ngo, H.M.; Zhou, Y.; Lorenzi, H.; Wang, K.; Kim, T.-K.; Zhou, Y.; El Bissati, K.; Mui, E.; Fraczek, L.; Rajagopala, S.V.; et al. Toxoplasma Modulates Signature Pathways of Human Epilepsy, Neurodegeneration & Cancer. Sci. Rep. 2017, 7, 11496. [Google Scholar] [CrossRef]
  92. Syn, G.; Anderson, D.; Blackwell, J.M.; Jamieson, S.E. Epigenetic Dysregulation of Host Gene Expression in Toxoplasma Infection with Specific Reference to Dopamine and Amyloid Pathways. Infect. Genet. Evol. 2018, 65, 159–162. [Google Scholar] [CrossRef]
  93. Hu, R.S.; He, J.J.; Elsheikha, H.M.; Zhang, F.K.; Zou, Y.; Zhao, G.H.; Cong, W.; Zhu, X.Q. Differential Brain MicroRNA Expression Profiles After Acute and Chronic Infection of Mice With Toxoplasma gondii Oocysts. Front. Microbiol. 2018, 9, 2316. [Google Scholar] [CrossRef]
  94. Liu, W.; Huang, L.; Wei, Q.; Zhang, Y.; Zhang, S.; Zhang, W.; Cai, L.; Liang, S. Microarray Analysis of Long Non-Coding RNA Expression Profiles Uncovers a Toxoplasma-Induced Negative Regulation of Host Immune Signaling. Parasites Vectors 2018, 11, 174. [Google Scholar] [CrossRef]
  95. Menard, K.L.; Haskins, B.E.; Colombo, A.P.; Denkers, E.Y. Toxoplasma gondii Manipulates Expression of Host Long Noncoding RNA during Intracellular Infection. Sci. Rep. 2018, 8, 15017. [Google Scholar] [CrossRef]
  96. Rezaei, F.; Daryani, A.; Sharifi, M.; Sarvi, S.; Jafari, N.; Pagheh, A.S.; Hashemi, N.; Hejazi, S.H. MiR-20a Inhibition Using Locked Nucleic Acid (LNA) Technology and Its Effects on Apoptosis of Human Macrophages Infected by Toxoplasma gondii RH Strain. Microb. Pathog. 2018, 121, 269–276. [Google Scholar] [CrossRef]
  97. Braun, L.; Brenier-Pinchart, M.-P.; Hammoudi, P.-M.; Cannella, D.; Kieffer-Jaquinod, S.; Vollaire, J.; Josserand, V.; Touquet, B.; Couté, Y.; Tardieux, I.; et al. The Toxoplasma Effector TEEGR Promotes Parasite Persistence by Modulating NF-ΚB Signalling via EZH2. Nat. Microbiol. 2019, 4, 1208–1220. [Google Scholar] [CrossRef]
  98. Detavernier, A.; Azouz, A.; Shehade, H.; Splittgerber, M.; Van Maele, L.; Nguyen, M.; Thomas, S.; Achouri, Y.; Svec, D.; Calonne, E.; et al. Monocytes Undergo Multi-Step Differentiation in Mice during Oral Infection by Toxoplasma gondii. Commun. Biol. 2019, 2, 472. [Google Scholar] [CrossRef]
  99. Hou, Z.; Liu, D.; Su, S.; Wang, L.; Zhao, Z.; Ma, Y.; Li, Q.; Jia, C.; Xu, J.; Zhou, Y.; et al. Comparison of Splenocyte MicroRNA Expression Profiles of Pigs during Acute and Chronic Toxoplasmosis. BMC Genom. 2019, 20, 97. [Google Scholar] [CrossRef]
  100. Li, D.-L.; Zou, W.-H.; Deng, S.-Q.; Peng, H.-J. Analysis of the Differential Exosomal MiRNAs of DC2.4 Dendritic Cells Induced by Toxoplasma gondii Infection. Int. J. Mol. Sci. 2019, 20, 5506. [Google Scholar] [CrossRef]
  101. Li, S.; Yang, J.; Wang, L.; Du, F.; Zhao, J.; Fang, R. Expression Profile of MicroRNAs in Porcine Alveolar Macrophages after Toxoplasma gondii Infection. Parasites Vectors 2019, 12, 65. [Google Scholar] [CrossRef]
  102. Lie, S.; Rochet, E.; Segerdell, E.; Ma, Y.; Ashander, L.M.; Shadforth, A.M.A.; Blenkinsop, T.A.; Michael, M.Z.; Appukuttan, B.; Wilmot, B.; et al. Immunological Molecular Responses of Human Retinal Pigment Epithelial Cells to Infection with Toxoplasma gondii. Front. Immunol. 2019, 10, 708. [Google Scholar] [CrossRef]
  103. Rochet, E.; Appukuttan, B.; Ma, Y.; Ashander, L.M.; Smith, J.R. Expression of Long Non-Coding RNAs by Human Retinal Muller Glial Cells Infected with Clonal and Exotic Virulent Toxoplasma gondii. Non-Coding RNA 2019, 5, 48. [Google Scholar] [CrossRef]
  104. Jiang, H.; Zhai, T.; Yu, Y.; Li, X.; Gong, P.; Zhang, X.; Li, G.; Li, J. Delayed IL-12 Production by Macrophages during Toxoplasma gondii Infection Is Regulated by MiR-187. Parasitol. Res. 2020, 119, 1023–1033. [Google Scholar] [CrossRef]
  105. Medina, L.; Castillo, C.; Liempi, A.; Guerrero-Muñoz, J.; Rojas-Pirela, M.; Maya, J.D.; Prieto, H.; Kemmerling, U.; Guerrero-Munoz, J.; Rojas-Pirela, M.; et al. Trypanosoma Cruzi and Toxoplasma gondii Induce a Differential MicroRNA Profile in Human Placental Explants. Front. Immunol. 2020, 11, 595250. [Google Scholar] [CrossRef]
  106. Meira-Strejevitch, C.S.; de Siqueira Pereira, I.; Hippólito, D.D.C.; Maia, M.M.; Cruz, A.B.; Gava, R.; Brandão de Mattos, C.C.; Frederico, F.B.; Siqueira, R.C.; Mattos, L.C.; et al. Ocular Toxoplasmosis Associated with Up-Regulation of MiR-155-5p/MiR-29c-3p and down-Regulation of MiR-21-5p/MiR-125b-5p. Cytokine 2020, 127, 154990. [Google Scholar] [CrossRef]
  107. de Siqueira Pereira, I.; Maia, M.M.; da Cruz, A.B.; Telles, J.P.M.; Vidal, J.E.; Gava, R.; Meira-Strejevitch, C.S.; Pereira-Chioccola, V.L.; De Siqueira, I.; Marta, P.; et al. Plasma Extracellular MicroRNAs Are Related to AIDS/Cerebral Toxoplasmosis Co-Infection. Parasite Immunol. 2020, 42, e12696. [Google Scholar] [CrossRef]
  108. Tyebji, S.; Hannan, A.J.; Tonkin, C.J. Pathogenic Infection in Male Mice Changes Sperm Small RNA Profiles and Transgenerationally Alters Offspring Behavior. Cell Rep. 2020, 31, 107573. [Google Scholar] [CrossRef]
  109. Zhou, C.-X.; Ai, K.; Huang, C.-Q.; Guo, J.-J.; Cong, H.; He, S.-Y.; Zhu, X.-Q. MiRNA and CircRNA Expression Patterns in Mouse Brain during Toxoplasmosis Development. BMC Genom. 2020, 21, 46. [Google Scholar] [CrossRef]
  110. Nast, R.; Choepak, T.; Lüder, C.G.K.; Lueder, C.G.K.; Lüder, C.G.K. Epigenetic Control of IFN-γ Host Responses During Infection with Toxoplasma gondii. Front. Immunol. 2020, 11, 581241. [Google Scholar] [CrossRef]
  111. Sabou, M.; Doderer-Lang, C.C.; Leyer, C.; Konjic, A.; Kubina, S.; Lennon, S.; Rohr, O.; Viville, S.S.; Cianférani, S.; Candolfi, E.; et al. Toxoplasma gondii ROP16 Kinase Silences the Cyclin B1 Gene Promoter by Hijacking Host Cell UHRF1-Dependent Epigenetic Pathways. Cell. Mol. Life Sci. 2020, 77, 2141–2156. [Google Scholar] [CrossRef]
  112. Sciume, G.; Mikami, Y.; Jankovic, D.; Nagashima, H.; Villarino, A.V.; Morrison, T.; Yao, C.; Signorella, S.; Sun, H.-W.W.; Brooks, S.R.; et al. Rapid Enhancer Remodeling and Transcription Factor Repurposing Enable High Magnitude Gene Induction upon Acute Activation of NK Cells. Immunity 2020, 53, 745–758.e4. [Google Scholar] [CrossRef]
  113. Singh, D.K.; Hari Dass, S.A.; Abdulai-Saiku, S.; Vyas, A. Testosterone Acts Within the Medial Amygdala of Rats to Reduce Innate Fear to Predator Odor Akin to the Effects of Toxoplasma gondii Infection. Front. Psychiatry 2020, 11, 630. [Google Scholar] [CrossRef]
  114. El-Sayad, M.; Abdel Rahman, M.; Hussein, N.; Abdel Aziz, R.; El-Taweel, H.A.; Abd El-Latif, N. MicroRNA-155 Expression and Butyrylcholinesterase Activity in the Liver Tissue of Mice Infected with Toxoplasma gondii (Avirulent and Virulent Strains). Acta Parasitol. 2021, 66, 1167–1176. [Google Scholar] [CrossRef]
  115. Maia, M.M.; Cruz, A.B.; de Siqueira Pereira, I.; Taniwaki, N.N.; Namiyama, G.M.; Pereira-Chioccola, V.L. Characterization of Murine Extracellular Vesicles and Toxoplasma gondii Infection. Parasite Immunol. 2021, 43, e12869. [Google Scholar] [CrossRef]
  116. Menard, K.L.; Bu, L.; Denkers, E.Y. Transcriptomics Analysis of Toxoplasma gondii-Infected Mouse Macrophages Reveals Coding and Noncoding Signatures in the Presence and Absence of MyD88. BMC Genom. 2021, 22, 130. [Google Scholar] [CrossRef]
  117. Quiarim, T.M.; Maia, M.M.; da Cruz, A.B.; Taniwaki, N.N.; Namiyama, G.M.; Pereira-Chioccola, V.L. Characterization of Extracellular Vesicles Isolated from Types I, II and III Strains of Toxoplasma gondii. Acta Trop. 2021, 219, 105915. [Google Scholar] [CrossRef]
  118. Yang, J.; He, Z.; Chen, C.; Li, S.; Qian, J.; Zhao, J.; Fang, R. Toxoplasma gondii Infection Inhibits Histone Crotonylation to Regulate Immune Response of Porcine Alveolar Macrophages. Front. Immunol. 2021, 12, 696061. [Google Scholar] [CrossRef]
  119. Hou, Z.; Wang, L.; Su, D.; Cai, W.; Zhu, Y.; Liu, D.; Huang, S.; Xu, J.; Pan, Z.; Tao, J. Global MicroRNAs Expression Profile Analysis Reveals Possible Regulatory Mechanisms of Brain Injury Induced by Toxoplasma gondii Infection. Front. Neurosci. 2022, 16, 827570. [Google Scholar] [CrossRef]
  120. Hou, Z.; Zhang, H.; Xu, K.; Zhu, S.; Wang, L.; Su, D.; Liu, J.; Su, S.; Liu, D.; Huang, S.; et al. Cluster Analysis of Splenocyte MicroRNAs in the Pig Reveals Key Signal Regulators of Immunomodulation in the Host during Acute and Chronic Toxoplasma gondii Infection. Parasites Vectors 2022, 15, 58. [Google Scholar] [CrossRef]
  121. Jiang, D.; Wu, S.; Xu, L.; Xie, G.; Li, D.; Peng, H. Anti-Infection Roles of MiR-155-5p Packaged in Exosomes Secreted by Dendritic Cells Infected with Toxoplasma gondii. Parasites Vectors 2022, 15, 3. [Google Scholar] [CrossRef] [PubMed]
  122. Jung, B.-K.; Song, H.; Shin, H.; Chai, J.-Y. Exosomal MiRNA-21 from Toxoplasma gondii-Infected Microglial Cells Induces the Growth of U87 Glioma Cells by Inhibiting Tumor Suppressor Genes. Sci. Rep. 2022, 12, 16450. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, S.-S.; Zhou, C.-X.; Elsheikha, H.M.; He, J.-J.; Zou, F.-C.; Zheng, W.-B.; Zhu, X.-Q.; Zhao, G.-H. Temporal Transcriptomic Changes in Long Non-Coding RNAs and Messenger RNAs Involved in the Host Immune and Metabolic Response during Toxoplasma gondii Lytic Cycle. Parasites Vectors 2022, 15, 22. [Google Scholar] [CrossRef]
  124. Guo, X.-D.; Zhou, C.-X.; Li, L.-Y.; Ai, K.; Wang, Y.-L.; Zhou, D.-H. Comprehensive Analysis of MRNA-LncRNA Co-Expression Profiles in Mouse Brain during Infection with Toxoplasma gondii. Acta Trop. 2023, 237, 106722. [Google Scholar] [CrossRef]
  125. Al-Quraishy, S.; Delic, D.; Sies, H.; Wunderlich, F.; Abdel-Baki, A.A.S.; Dkhil, M.A.M. Differential MiRNA Expression in the Mouse Jejunum during Garlic Treatment of Eimeria Papillata Infections. Parasitol. Res. 2011, 109, 387–394. [Google Scholar] [CrossRef]
  126. Dkhil, M.; Abdel-Baki, A.A.; Delić, D.; Wunderlich, F.; Sies, H.; Al-Quraishy, S. Eimeria papillata: Upregulation of Specific miRNA-Species in the Mouse Jejunum. Exp. Parasitol. 2011, 127, 581–586. [Google Scholar] [CrossRef]
  127. Bigna, J.J.; Tochie, J.N.; Tounouga, D.N.; Bekolo, A.O.; Ymele, N.S.; Youda, E.L.; Sime, P.S.; Nansseu, J.R. Global, Regional, and Country Seroprevalence of Toxoplasma gondii in Pregnant Women: A Systematic Review, Modelling and Meta-Analysis. Sci. Rep. 2020, 10, 12102. [Google Scholar] [CrossRef]
  128. WHO. World Malaria Report 2019; WHO: Geneva, Switzerland, 2019; ISBN 9789241565721. [Google Scholar]
  129. King, P.; Tyler, K.M.; Hunter, P.R. Anthroponotic Transmission of Cryptosporidium parvum Predominates in Countries with Poorer Sanitation: A Systematic Review and Meta-Analysis. Parasites Vectors 2019, 12, 16. [Google Scholar] [CrossRef]
  130. Aguirre, A.A.; Longcore, T.; Barbieri, M.; Dabritz, H.; Hill, D.; Klein, P.N.; Lepczyk, C.; Lilly, E.L.; McLeod, R.; Milcarsky, J.; et al. The One Health Approach to Toxoplasmosis: Epidemiology, Control, and Prevention Strategies. Ecohealth 2019, 16, 378–390. [Google Scholar] [CrossRef]
  131. Lodoen, M.B.; Smith, N.C.; Soldati-Favre, D.; Ferguson, D.J.P.; van Dooren, G.G. Nanos Gigantium Humeris Insidentes: Old Papers Informing New Research into Toxoplasma gondii. Int. J. Parasitol. 2021, 51, 1193–1212. [Google Scholar] [CrossRef]
  132. Christensen, P.; Racklyeft, A.; Ward, K.E.; Matheson, J.; Suwanarusk, R.; Chua, A.C.Y.; Kaneko, O.; Aung, H.L.; Rénia, L.; Amanzougaghene, N.; et al. Improving in Vitro Continuous Cultivation of Plasmodium cynomolgi, a Model for P. Vivax. Parasitol. Int. 2022, 89, 102589. [Google Scholar] [CrossRef]
  133. Certad, G.; Viscogliosi, E.; Chabé, M.; Cacciò, S.M. Pathogenic Mechanisms of Cryptosporidium and Giardia. Trends Parasitol. 2017, 33, 561–576. [Google Scholar] [CrossRef] [PubMed]
  134. Weinberg, M.S.; Morris, K.V. Transcriptional Gene Silencing in Humans. Nucleic Acids Res. 2016, 44, 6505–6517. [Google Scholar] [CrossRef] [PubMed]
  135. Sun, B.; Liu, C.; Li, H.; Zhang, L.; Luo, G.; Liang, S.; Lü, M. Research Progress on the Interactions between Long Non-Coding RNAs and MicroRNAs in Human Cancer (Review). Oncol. Lett. 2020, 19, 595–605. [Google Scholar] [CrossRef]
  136. Singh, B.; Varikuti, S.; Halsey, G.; Volpedo, G.; Hamza, O.M.; Satoskar, A.R. Host-Directed Therapies for Parasitic Diseases. Future Med. Chem. 2019, 11, 1999–2018. [Google Scholar] [CrossRef]
  137. Coetzee, N.; von Grüning, H.; Opperman, D.; van der Watt, M.; Reader, J.; Birkholtz, L.-M. Epigenetic Inhibitors Target Multiple Stages of Plasmodium Falciparum Parasites. Sci. Rep. 2020, 10, 2355. [Google Scholar] [CrossRef]
  138. Pennini, M.E.; Pai, R.K.; Schultz, D.C.; Boom, W.H.; Harding, C.V. Mycobacterium tuberculosis 19-KDa Lipoprotein Inhibits IFN-γ-Induced Chromatin Remodeling of MHC2TA by TLR2 and MAPK Signaling. J. Immunol. 2006, 176, 4323–4330. [Google Scholar] [CrossRef]
  139. Parmar, N.; Chandrakar, P.; Kar, S. Leishmania Donovani Subverts Host Immune Response by Epigenetic Reprogramming of Macrophage M(Lipopolysaccharides + IFN-γ)/M(IL-10) Polarization. J. Immunol. 2020, 204, 2762–2778. [Google Scholar] [CrossRef]
  140. Lima, T.S.; Lodoen, M.B. Mechanisms of Human Innate Immune Evasion by Toxoplasma gondii. Front. Cell. Infect. Microbiol. 2019, 9, 103. [Google Scholar] [CrossRef]
  141. Ramirez Ramirez, A.D.; de Jesus, M.C.S.; Rossit, J.; Reis, N.F.; Santos-Filho, M.C.; Sudré, A.P.; de Oliveira-Ferreira, J.; de Souza Baptista, A.R.; Storti-Melo, L.M.; Machado, R.L.D. Association of Toll-like Receptors in Malaria Susceptibility and Immunopathogenesis: A Meta-Analysis. Heliyon 2022, 8, e09318. [Google Scholar] [CrossRef]
  142. Jan, I.; Rather, R.A.; Mushtaq, I.; Malik, A.A.; Besina, S.; Baba, A.B.; Farooq, M.; Yousuf, T.; Rah, B.; Afroze, D. Helicobacter Pylori Subdues Cytokine Signaling to Alter Mucosal Inflammation via Hypermethylation of Suppressor of Cytokine Signaling 1 Gene During Gastric Carcinogenesis. Front. Oncol. 2021, 10, 604747. [Google Scholar] [CrossRef] [PubMed]
  143. Marr, A.K.; MacIsaac, J.L.; Jiang, R.; Airo, A.M.; Kobor, M.S.; McMaster, W.R. Leishmania donovani Infection Causes Distinct Epigenetic DNA Methylation Changes in Host Macrophages. PLoS Pathog. 2014, 10, e1004419. [Google Scholar] [CrossRef]
  144. Ullah, H.; Di Minno, A.; Esposito, C.; El-Seedi, H.R.; Khalifa, S.A.M.; Baldi, A.; Greco, A.; Santonastaso, S.; Cioffi, V.; Sperandeo, R.; et al. Efficacy of a Food Supplement Based on S-Adenosyl Methionine and Probiotic Strains in Subjects with Subthreshold Depression and Mild-to-Moderate Depression: A Monocentric, Randomized, Cross-over, Double-Blind, Placebo-Controlled Clinical Trial. Biomed. Pharmacother. 2022, 156, 113930. [Google Scholar] [CrossRef]
  145. Navik, U.; Sheth, V.G.; Kabeer, S.W.; Tikoo, K. Dietary Supplementation of Methyl Donor L-Methionine Alters Epigenetic Modification in Type 2 Diabetes. Mol. Nutr. Food Res. 2019, 63, 1801401. [Google Scholar] [CrossRef]
  146. McConkey, G.A.; Martin, H.L.; Bristow, G.C.; Webster, J.P. Toxoplasma gondii Infection and Behaviour—Location, Location, Location? J. Exp. Biol. 2013, 216, 113–119. [Google Scholar] [CrossRef]
  147. Ratti, M.; Lampis, A.; Ghidini, M.; Salati, M.; Mirchev, M.B.; Valeri, N.; Hahne, J.C. MicroRNAs (MiRNAs) and Long Non-Coding RNAs (LncRNAs) as New Tools for Cancer Therapy: First Steps from Bench to Bedside. Target. Oncol. 2020, 15, 261–278. [Google Scholar] [CrossRef]
  148. Jung, T.W.; Park, J.; Sun, J.L.; Ahn, S.H.; Abd El-Aty, A.M.; Hacimuftuoglu, A.; Kim, H.-C.; Shim, J.-H.; Shin, S.; Jeong, J.H. Administration of Kynurenic Acid Reduces Hyperlipidemia-Induced Inflammation and Insulin Resistance in Skeletal Muscle and Adipocytes. Mol. Cell. Endocrinol. 2020, 518, 110928. [Google Scholar] [CrossRef]
Figure 1. Flow chart of returned results from the present database search.
Figure 1. Flow chart of returned results from the present database search.
Pathogens 12 00299 g001
Figure 2. Map of the distribution of publications by country. The color intensity varies according to the number of publications.
Figure 2. Map of the distribution of publications by country. The color intensity varies according to the number of publications.
Pathogens 12 00299 g002
Figure 3. Co-occurrence network by country. The cluster (different colors) is formed by the frequency of the country’s names co-occurring, as the node size is proportional to the number of pub-lished articles. The blue cluster displays the countries most cooperated with Germany (Saudi Arabia, Egypt, Denmark, Brazil, and the Netherlands – node not labeled). The green cluster is composed of countries that most collaborated with France, and in the yellow cluster, the collaborations with Australia. In the red cluster, it is displayed the countries that cooperated with USA and China. The line thickness is proportional to the number of articles published in collaboration between countries.
Figure 3. Co-occurrence network by country. The cluster (different colors) is formed by the frequency of the country’s names co-occurring, as the node size is proportional to the number of pub-lished articles. The blue cluster displays the countries most cooperated with Germany (Saudi Arabia, Egypt, Denmark, Brazil, and the Netherlands – node not labeled). The green cluster is composed of countries that most collaborated with France, and in the yellow cluster, the collaborations with Australia. In the red cluster, it is displayed the countries that cooperated with USA and China. The line thickness is proportional to the number of articles published in collaboration between countries.
Pathogens 12 00299 g003
Figure 4. Co-occurrence network by keyword segregated according to their occurrence in colored clusters (red: words related to T. gondii; blue: C. parvum; yellow: Plasmodium spp.). The node size is proportional to the word occurrence among the studies. Linked nodes represent the co-occurrence of the keyword in articles.
Figure 4. Co-occurrence network by keyword segregated according to their occurrence in colored clusters (red: words related to T. gondii; blue: C. parvum; yellow: Plasmodium spp.). The node size is proportional to the word occurrence among the studies. Linked nodes represent the co-occurrence of the keyword in articles.
Pathogens 12 00299 g004
Figure 5. Modulation of IFNGγ response genes in macrophages upon T. gondii infection (A) and in infected macrophage treated with HDACi (B). (A) T. gondii tachyzoite replicates in the parasitophorous vacuole (PV), releasing the T. gondii inhibitor of STAT1 transcriptional activity (TgIST) (pink), which is translocated to the nucleus. TgIST sequesters STAT1 and recruits the Mi2/NurD complex to the promoter of IFNγ response genes, remodeling the chromatin to a repressive state, thus silencing the gene. (B) The HDACi inhibits the action of the histone deacetylase (HDAC) present in the Mi2/NurD complex. The inhibition of HDAC prevents the histone deacetylation and leads to an open chromatin state, restoring the Ciita gene expression (an IFNγ response gene).
Figure 5. Modulation of IFNGγ response genes in macrophages upon T. gondii infection (A) and in infected macrophage treated with HDACi (B). (A) T. gondii tachyzoite replicates in the parasitophorous vacuole (PV), releasing the T. gondii inhibitor of STAT1 transcriptional activity (TgIST) (pink), which is translocated to the nucleus. TgIST sequesters STAT1 and recruits the Mi2/NurD complex to the promoter of IFNγ response genes, remodeling the chromatin to a repressive state, thus silencing the gene. (B) The HDACi inhibits the action of the histone deacetylase (HDAC) present in the Mi2/NurD complex. The inhibition of HDAC prevents the histone deacetylation and leads to an open chromatin state, restoring the Ciita gene expression (an IFNγ response gene).
Pathogens 12 00299 g005
Figure 6. Epigenetic regulation of CCNB gene in BeWo cells upon Toxoplasma gondii infection and the mechanism of inhibition by putative drug targets. T. gondii secretes the effector Ropthry 16 (ROP16) in the cytoplasm of the infected cell that phosphorylates the ubiquitin-like containing PHD and RING finger domain (UHRF1) that translocates to the nucleus. The UHRF1 recruits the histone deacetylase (HDAC), histone methyltransferase (HMT), and DNA methyltransferase (DNMT) enzymes to the CCNB promoter, silencing its transcription and arresting the cell cycle in the G2 phase. We hypothesize that treatment with histone deacetylase inhibitor (HDACi), histone methyltransferase inhibitor (HMTi), or DNA methyltransferase inhibitor (DNMTi) can prevent histone H3 deacetylation, H3K9 methylation, and DNA methylation, respectively. It would restore the cyclin B expression and the continuity of the cell cycle.
Figure 6. Epigenetic regulation of CCNB gene in BeWo cells upon Toxoplasma gondii infection and the mechanism of inhibition by putative drug targets. T. gondii secretes the effector Ropthry 16 (ROP16) in the cytoplasm of the infected cell that phosphorylates the ubiquitin-like containing PHD and RING finger domain (UHRF1) that translocates to the nucleus. The UHRF1 recruits the histone deacetylase (HDAC), histone methyltransferase (HMT), and DNA methyltransferase (DNMT) enzymes to the CCNB promoter, silencing its transcription and arresting the cell cycle in the G2 phase. We hypothesize that treatment with histone deacetylase inhibitor (HDACi), histone methyltransferase inhibitor (HMTi), or DNA methyltransferase inhibitor (DNMTi) can prevent histone H3 deacetylation, H3K9 methylation, and DNA methylation, respectively. It would restore the cyclin B expression and the continuity of the cell cycle.
Pathogens 12 00299 g006
Table 1. List of the articles included in this review related to the parasite, regulatory mechanism, target process, research model, and country of publication.
Table 1. List of the articles included in this review related to the parasite, regulatory mechanism, target process, research model, and country of publication.
ParasiteRegulatory MechanismTarget ProcessResearch ModelCountryRef.
In Vitro/In SilicoIn Vivo
C. parvummiRNAHost defenseH69 cells-USA[11,12,13,14]
C. parvummiRNAHost defenseH69 and HIBEpiC cells-USA[15,16]
C. parvummiRNACell proliferationH69 and HIBEpiC cell lines-USA, China[17]
C. parvummiRNAHost defenseH69, HIBEpiC, 603B, IEC4.1, SW480 cellsC57BL/6J miceUSA[18]
C. parvummiRNAHost defenseH69, 603B, IEC4.1C57BL/6J miceUSA[19]
C. parvummiRNAHost defenseH69 cells-USA, China[20]
C. parvummiRNAHost defensemICcl2 cellsC57BL/6 miceFrance[21]
C. parvumPTHMHost defenseFHs 74 INT, HCT-8, IEC4.1 cellsMice in vivo infectionUSA[22]
C. parvumPTHMCell migrationFHs 74 INT, HCT-8 cells-USA, China[23,24]
C. parvummiRNAApoptosis; cancerHT-29 and HCT-8 cells-Malaysia[25]
C. parvumlncRNA; PTHMHost defenseIEC4.1, muINTEPI cellsC57BL/6J miceUSA[26]
C. parvumlncRNACell adhesionHCT-8 cells-China[27]
C. parvumPTHMHost defenseFHs 74 INT, HCT-8 cells-USA, China[28]
C. parvummiRNAHost defenseHCT-8 cells-China[29]
C. parvummiRNAHost defense; apoptosisHCT-8 cells-China[30]
C. parvumlncRNAHost defenseIEC4.1, RAW264.7 cellsC57BL/6J miceUSA[31]
C. parvummiRNACell cycle regulation; cancerBioinformatics (GEO)/in silico-UK, Saudi Arabia, India[32]
C. parvumcircRNAApoptosisHCT-8 cells-China[33]
C. parvumlncRNA; PTHMHost defenseHCT-8, IEC4.1 cellsC57BL/6J miceUSA[34]
C. parvumPTHMHost defense; cancerHCT-8 cellsCB17-SCID miceFrance, Belgium[35]
C. parvummiRNAHost defense; cell proliferation; apoptosisHCT-8 cells-Turkey[36]
C. parvummiRNAApoptosisHCT-8 cells-China[37]
P. bergheimiRNAHost defense-C57BL/6 and CBA miceAustralia, Denmark[38]
P. bergheimiRNAHost defense; cell proliferation; apoptosisHEK293T cellsC57BL/6J and NMRI miceGermany, USA[39]
P. berghei and P. yoellimiRNAHost defense; cell adhesion; endocytosis; apoptosis-CBA miceSpain, Australia, Peru[40]
P. chabaudimiRNAHost defense; apoptosis-C57BL/6 miceGermany, Saudi Arabia[41]
P. chabaudimiRNAHost defense; cell cycle regulation; apoptosis; cancer-C57BL/6 miceSaudi Arabia, Germany, Egypt[42]
P. chabaudiDNAmHost defense-C57BL/6 miceSaudi Arabia, Germany, Egypt[43]
P. chabaudiDNAmHost defense; erythropoiesis-Balb/c miceSaudi Arabia, Germany, Egypt, Spain[44]
P. chabaudiPTHMHost defense-C57BL/6J miceUK[45]
P. falciparummiRNAHost defenseHuman RBC, J2E, F4N, BW5147, J558L and 416B cells-UK[46]
P. falciparummiRNAHost defenseHuman RBC -USA, UK[47]
P. falciparummiRNAHost defenseHuman RBC -USA, Switzerland, UK[48]
P. falciparummiRNAHost defense; cell communicationHuman RBC -USA, Switzerland[49]
P. falciparummiRNAHost defense-Artificial human infectionAustralia[50]
P. falciparumDNAmDrug resistance-Malaria patientsIndia[51]
P. falciparummiRNAHost defenseHuman RBC -China[52]
P. falciparum and P. vivaxmiRNAHost defense; cell adhesion-Malaria patientsChina[53]
P. falciparumPTHMHost defense-Malaria patientsUSA, Brazil, Netherlands[54]
P. falciparummiRNAHost defenseHuman RBC -India[55]
P. falciparumPTHMHost defense Artificial human infectionNetherlands, Denmark, Germany[56]
P. falciparummiRNAHost defenseHuman RBC and HEK 293T cells-India, Belgium[57]
P. falciparum and P. vivaxmiRNAHost defense; cell adhesion-Malaria patientsThailand, Australia[58]
P. falciparummiRNAHost defense-Malaria patientsGermany[59]
P. falciparummiRNAHost defense; cell proliferation; neural pathwaysHBE cellsMalaria patientsSpain, Mozambique, USA[60]
P. falciparummiRNAHost defense; cell adhesion; erythropoiesis-Malaria patientsIran[61]
P. falciparummiRNAHost defense-Malaria patientsUSA, Ghana[62]
P. vivaxmiRNAHost defense-Malaria patientsThailand, Japan[63]
P. vivaxmiRNAHost defense; erythropoiesis; apoptosis-Malaria patientsIndia, Qatar[64]
P. vivaxmiRNAHost defense-Malaria patientsBrazil, Germany[65]
P. yoellimiRNAHost defense; neural pathways-CBA miceAustralia, USA[66]
P. yoellilncRNAHost defense-BALB/c miceChina[67]
T. annulataPTHMCancerBL3 cells-France[68]
T. annulatamiRNACancerBL3 cells-France, USA[69]
T. annulatamiRNAHost defense; cancerBL20 cells-UK[70]
T. annulatamiRNACancerBL3,BL20 cells-Saudi Arabia, France, Japan[71]
T. gondiiPTHMHost defense-C57BL/6 miceUSA[72]
T. gondiiPTHMHost defense-C57BL/6 miceUSA[73]
T. gondiimiRNAApoptosisHFF, VERO cells-USA[74]
T. gondiiPTHMHost defenseRAW264.7, BMDM (BALB/c mice) cells-Germany[75]
T. gondiimiRNAApoptosisHuman macrophage, THP-1 cells-China, Australia[76]
T. gondiimiRNAHost defense-Kunming miceChina, UK[77]
T. gondiimiRNAApoptosisHuman macrophage, THP-1 cells-China[78]
T. gondiimiRNAHost defenseHFF cellsSwiss OF1, BALB/cJRj, C57BL/6JRj miceFrance, USA[79]
T. gondiimiRNAHost defense; cell cycle regulation-BALB/c miceChina[80]
T. gondiimiRNANeural pathways-BALB/c miceUSA, China[81]
T. gondiimiRNANeural pathwaysSK-N-MC cellsCD-1 miceUSA, China[82]
T. gondiiDNAmReproductive pathways-C57Bl/6 miceCzech Republic, Germany[83]
T. gondiiDNAmNeural pathways-Wistar ratsSingapore[84]
T. gondiiPTHMHost defenseHFF, 293FT, HEK293 cells-USA[85]
T. gondiimiRNAHost defense; apoptosis; cancer-Kunming miceChina[86]
T. gondiimiRNACell cycle regulation; cell proliferationL6 cells-Korea[87]
T. gondiiPTHMHost defenseHFF, RAW264.7, 2fTGH, U3A, HEK-Blue IFN-γ cellsBALB/cJRj miceFrance[88]
T. gondiiPTHMHost defenseRAW 264.7, U3A, U3A-STAT1, HeLa cellsCD-1, C57BL/6 miceUSA[89]
T. gondiimiRNAHost defense; apoptosis-Chinese Li Hua domestic cats (Felis catus)China[90]
T. gondiimiRNANeural pathwaysL-NSC, S-NSC, human monocytic cellsToxoplasmosis patientsUSA, UK, Canada, Australia[91]
T. gondiiDNAmNeural pathwaysWERI-Rb-1 cells-Australia[92]
T. gondiimiRNAHost defense; cancer-BALB/c miceChina, UK[93]
T. gondiilncRNAHost defenseHFF, THP-1 cells-China[94]
T. gondiilncRNAHost defenseBMDM (C57BL/6 mice)-USA[95]
T. gondiimiRNAApoptosisHuman macrophage -Iran[96]
T. gondiiPTHMHost defenseBMDM (C57BL/6 mice), HFF, 293-T-Rex cellsBALB/cJRj miceFrance[97]
T. gondiiPTHMHost defenseHFF cellsC57BL/6 miceBelgium, Czech Republic[98]
T. gondiimiRNAHost defense; apoptosis-Pigs (Sus scrofa)China[99]
T. gondiimiRNAHost defense; apoptosisDC2.4, HFF cells-China[100]
T. gondiimiRNAHost defense; cell proliferation; apoptosisHFF, porcine alveolar macrophages cells-China[101]
T. gondiimiRNAHost defensePrimary human retinal pigment epithelial cells, ARPE-19 cells-Australia, USA[102]
T. gondiilncRNAHost defense; cell proliferation; cancerPrimary retinal Müller cells and MIO-M1 cells-Australia[103]
T. gondiimiRNAHost defenseMouse macrophage (C57BL/6J mice)-China[104]
T. gondiimiRNAHost defense; apoptosis; cell cycle regulationHuman placental explants -Chile[105]
T. gondiimiRNAHost defense-Ocular toxoplasmosis patientsBrazil[106]
T. gondiimiRNAHost defense-Toxoplasmosis and AIDS patientsBrazil[107]
T. gondiimiRNANeural pathways-C57BL/6J miceAustralia[108]
T. gondiicircRNA; miRNAHost defense; cell proliferation; erythropoiesis; cancer-BALB/c miceChina[109]
T. gondiiPTHM; DNAmHost defenseRAW264.7 cells-Germany[110]
T. gondiiPTHM; DNAmCell cycle regulationBeWo, U-118MG cells-France[111]
T. gondiiPTHMHost defense-C57BL/6J miceUSA, Italy[112]
T. gondiiDNAmNeural pathways-Wistar ratsSingapore[113]
T. gondiimiRNAHost defense-Swiss Albin miceEgypt[114]
T. gondiimiRNAHost defense-A/Sn miceBrazil[115]
T. gondiilncRNAApoptosisBMDM (C57BL/6 mice)-USA[116]
T. gondiimiRNAHost defenseVERO cells-Brazil[117]
T. gondiiPTHMHost defense; cell proliferation3D4/21, PK-15 cells-China[118]
T. gondiimiRNAHost defense; cell proliferation; apoptosis; cancer-Pigs (Sus scrofa)China[119]
T. gondiimiRNAHost defense-Pigs (Sus scrofa)China[120]
T. gondiimiRNAHost defenseDC2.4, HFF, RAW264.7 cells-China[121]
T. gondiimiRNACell proliferationBV2, U87, U118 cellsC57BL/6 miceKorea[122]
T. gondiilncRNAHost defenseHFF cells-China, UK[123]
T. gondiilncRNAHost defense-BALB/c, Kunming miceChina[124]
E. papillatamiRNACancer; host defense; apoptosis-Balb/c miceSaudi Arabia, Germany, Egypt[125,126]
circRNA: circular RNA; PTHM: post-translational histone modification; DNAm: DNA methylation. The cell line names are listed in Supplemental Table S1.
Table 2. Percentage of studies and the relative number of each parasite species or genus, regulatory mechanism, target process, and research model.
Table 2. Percentage of studies and the relative number of each parasite species or genus, regulatory mechanism, target process, and research model.
ParasitesRegulatory MechanismTargeted ProcessResearch Model
Specie or GenusOccurrenceMechanismOccurrencePathwayOccurrenceModelOccurrence
T. gondii53/116; 45.7%ncRNA88/116; 75.9%Host defense87/116; 75%in vitro72/116; 62.1%
Plasmodium spp.30/116; 25.8%miRNA76/88; 86.4%Apoptosis23/116; 19.8%in vivo63/116; 54.3%
C. parvum27/116;23.3%lncRNA11/88; 12.5%Cancer14/116; 12%
T. annulata4/116; 3.5%circRNA2/88; 2.3%Cell proliferation11/116; 9.5%
E. papillata2/116; 1.8%PTHM23/116; 19.8%Neural pathways9/116;7.8%
DNAm9/116; 7.8%Cell cycle regulation6/116; 5.2%
Cell adhesion5/116; 4.3%
Erythropoiesis4/116; 3.4%
Cell migration2/116; 1.7%
Cell comunication1/116; 0.9%
Reproductive pathway1/116; 0.9%
circRNA: circular RNA; PTHM: post-translational histone modification; DNAm: DNA methylation.
Table 3. Epidrugs that were FDA-approved or tested in clinical trials in the selected studies.
Table 3. Epidrugs that were FDA-approved or tested in clinical trials in the selected studies.
ParasitePathwayCompoundCompound Analogous in Clinical TrialRef.
CompoundPhaseDiseaseNumberStatus
T. gondiimiRNAanti-miR21RG0121ASNCT03373786Completed[122]
T. gondiiPTHMMS-275Entinostat
(MS-275)
3BCNCT02115282Active[75]
T. gondiiDNAmL-methionineSAMe4RMDNCT04832178Active[84]
T. gondiiPTHMHDAC2- SiRNAVorinostat;
romidepsin
FDA approvedCTCL--[118]
T. annulatamiRNAanti-miR- 155Cobomarsen
(MRG-106)
2CTCLNCT02580552Complete[69]
PTHM: post-translational histone modification; DNAm: DNA methylation; AP: Alport syndrome; BC: breast cancer; CTCL: cutaneous T cell lymphoma; RMD: resistant major depression; SAMe: S-adenosylmethionine.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Brandão, Y.d.O.; Molento, M.B. A Systematic Review of Apicomplexa Looking into Epigenetic Pathways and the Opportunity for Novel Therapies. Pathogens 2023, 12, 299. https://doi.org/10.3390/pathogens12020299

AMA Style

Brandão YdO, Molento MB. A Systematic Review of Apicomplexa Looking into Epigenetic Pathways and the Opportunity for Novel Therapies. Pathogens. 2023; 12(2):299. https://doi.org/10.3390/pathogens12020299

Chicago/Turabian Style

Brandão, Yara de Oliveira, and Marcelo Beltrão Molento. 2023. "A Systematic Review of Apicomplexa Looking into Epigenetic Pathways and the Opportunity for Novel Therapies" Pathogens 12, no. 2: 299. https://doi.org/10.3390/pathogens12020299

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop