Toxoplasmosis, Animal

弓形虫病,动物
  • 文章类型: Journal Article
    Toxoplasma gondii (T. gondii) is an obligate intracellular, zoonotic protozoan parasite of interest to physicians and veterinarians with its highly complex structure. It is known to infect about one-third of the world\'s population. Since it is a zoonotic disease, it is necessary to keep the animal population under control in order to prevent human exposure. Many studies have been conducted on the detection of T. gondii and it has been determined that there are three clonal groups consisting of types 1, 2, 3. Developments in molecular studies have led to changes in the taxonomy and new developments in parasitic diseases. It has helped in diagnosis, treatment, development of antiparasitic drugs and research on resistance. They also provided research on vaccine studies, genetic typing and phylogenetics of parasitic diseases. Conventional polymerase chain reaction (PCR), real-time PCR and genotyping studies conducted today increase our knowledge about T. gondii. Methods such as B1, SAG1, SAG2, GRA1, 529-bp repeat element, OWP genes and 18S rRNAs are mostly used in PCR, and methods such as MS, MLST, PCR-RFLP, RAPD-PCR and HRM are used in genotyping. Toxoplasmosis is a disease that is within the framework of the concept of one health and must attract attention, has not yet been eradicated in the world and needs joint studies for humans, animals and ecosystems to be eradicated. This can only be possible by establishing interdisciplinary groups, conducting surveys and training.
    Toxoplasma gondii (T. gondii) oldukça karışık olan yapısı ile hekimleri ve veteriner hekimleri ilgilendiren, zorunlu hücre içinde bulunan, zoonotik protozoan bir parazittir. Dünya nüfusunun yaklaşık üçte birini enfekte ettiği bilinmektedir. Zoonoz bir hastalık olması nedeniyle insan maruziyetini önlemek için, hayvan popülasyonunu da kontrol altında tutmak gerekir. T. gondii tespiti ile ilgili birçok çalışma yapılmış ve tip 1, 2, 3’ten oluşan üç klonal grubu olduğu tespit edilmiştir. Moleküler çalışmalarda oluşan gelişmeler paraziter hastalıklarda da taksonominin değişmesini ve yeni gelişmelerin oluşumunu sağlamıştır. Tanı, tedavi, antiparaziter ilaçların geliştirilmesi ve direncinin araştırılmasına yardımcı olmuştur. Ayrıca paraziter hastalıkların aşı çalışmalarının, genetik tiplendirmesinin ve filogenetiğin araştırılmasını da sağlamıştır. Bugün yapılan konvasiyonel polimeraz zincir reaksiyonu (PZR), gerçek zamanlı PZR ve genotiplendirme çalışmaları T. gondii hakkındaki bilgimizi artırmaktadır. PZR’de en fazla B1, SAG1, SAG2, GRA1, 529-bp repeat element, OWP genleri ve 18S rRNA’lar ve genotiplendirmede ise MS, MLST, PZR-RFLP, RAPD-PZR ve HRM gibi yöntemler kullanılmaktadır. Toxoplasmosis tek sağlık kavramı çerçevesinde yer alan ve ilgi çekmesi zorunlu, Dünya’da halen eradike edilememiş ve edilmesi için insan, hayvan ve ekosistem için ortak çalışmalara ihtiyaç duyan bir hastalıktır. Bu ancak disiplinlerarası gruplar kurup, sürveyans ve eğitim çalışmaları yaparak mümkün olabilir.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    OBJECTIVE: To investigate the development and dynamic changes of cysts in the brain of mice following infection with different forms of Toxoplasma gondii, so as to provide insights into for toxoplasmosis prevention and control.
    METHODS: ICR mice at ages of 6 to 8 weeks, each weighing 20 to 25 g, were intraperitoneally injected with tachyzoites of the T. gondii PRU strain at a dose of 1 × 105 tachyzoites per mouse, orally administered with cysts at a dose of 20 oocysts per mouse or oocysts at a dose of 200 oocysts per mouse for modeling chronic T. gondii infection in mice, and the clinical symptoms and survival of mice were observed post-infection. Mice were orally infected with T. gondii cysts at doses of 10 (low-dose group), 20 (medium-dose group), 40 cysts per mouse (high-dose group), and the effect of different doses of T. gondii infections on the number of cysts was examined in the mouse brain. Mice were orally administered with T. gondii cysts at a dose of 20 cysts per mouse, and grouped according to gender (female and male) and time points of infections (20, 30, 60, 90, 120, 150, 180 days post-infection), and the effects of gender and time points of infections on the number of cysts was examined in the mouse brain. In addition, mice were divided into the tachyzoite group (Group T), the first-generation cyst group (Group C1), the second-generation cyst group (Group C2), the third-generation cyst (Group C3) and the fourth-generation cyst group (Group C4). Mice in the Group T were intraperitoneally injected with T. gondii tachyzoites at a dose of 1 × 105 tachyzoites per mouse, and the cysts were collected from the mouse brain tissues 30 days post-infection, while mice in the Group C1 were orally infected with the collected cysts at a dose of 30 cysts per mouse. Continuous passage was performed by oral administration with cysts produced by the previous generation in mice, and the effect of continuous passage on the number of cysts was examined in the mouse brain.
    RESULTS: Following infection with T. gondii tachyzoites, cysts and oocysts in mice, obvious clinical symptoms were observed on days 6 to 13 and mice frequently died on days 7 to 12. The survival rates of mice were 67.0%, 87.0% and 53.0%, and the mean numbers of cysts were (516.0 ± 257.2), (1 203.0 ± 502.0) and (581.0 ± 183.1) in the mouse brain (F = 11.94, P < 0.01) on day 30 post-infection with T. gondii tachyzoites, cysts and oocysts, respectively, and the numbers of cysts in the brain tissues were significantly lower in mice infected with T. gondii tachyzoites and oocysts than in those infected with cysts (all P values < 0.01). The survival rates of mice were 87.0%, 87.0% and 60.0%, and the mean numbers of cysts were (953.0 ± 355.5), (1 084.0 ± 474.3) and (1 113.0 ± 546.0) in the mouse brain in the low-, medium- and high-dose groups on day 30 post-infection, respectively (F = 0.42, P > 0.05). The survival rates of male and female mice were 73.0% and 80.0%, and the mean numbers of cysts were (946.4 ± 411.4) and (932.1 ± 322.4) in the brain tissues of male and female mice, respectively (F = 1.63, P > 0.05). Following continuous passage, the mean numbers of cysts were (516.0 ± 257.2), (1 203.0 ± 502.0), (896.8 ± 332.3), (782.5 ± 423.9) and (829.2 ± 306.0) in the brain tissues of mice in the T, C1, C2, C3 and C4 groups, respectively (F = 4.82, P < 0.01), and the number of cysts was higher in the mouse brain in Group 1 than in Group T (P < 0.01). Following oral administration of 20 T. gondii cysts in mice, cysts were found in the moues brain for the first time on day 20 post-infection, and the number of cysts gradually increased over time, peaked on days 30 and 90 post-infection and then gradually decreased; however, the cysts were still found in the mouse brain on day 180 post-infection.
    CONCLUSIONS: There is a higher possibility of developing chronic T. gondii infection in mice following infection with cysts than with oocysts or tachyzoites and the most severe chronic infection is seen following infection with cysts. The number of cysts does not correlate with the severity of chronic T. gondii infection, and the number of cysts peaks in the mouse brain on days 30 and 90 post-infection.
    [摘要] 目的 观察不同形态刚地弓形虫感染后小鼠脑内包囊形成及其动态变化, 为弓形虫病防控提供依据。方法 取 6~8周龄ICR小鼠 (20~25 g) 建立慢性弓形虫感染模型, 其中弓形虫PRU株速殖子按1 × 105个/只剂量腹腔注射感染小鼠, 包囊和卵囊分别按20、200个/只剂量通过灌胃针口服感染小鼠, 观察感染后小鼠临床症状和存活情况。分别以10 (低剂 量组) 、20 (中剂量组) 、40个包囊/只 (高剂量组) 剂量感染小鼠, 观察弓形虫不同感染剂量对小鼠脑内包囊数量的影响。 将小鼠按性别 (雌、雄性) 、感染时间 (感染后20、30、60、90、120、150、180 d) 分组, 按20个/只剂量口服弓形虫包囊后, 分别 观察性别和感染时间对小鼠脑内包囊数量的影响。将小鼠分成速殖子组 (T组) 、包囊1代组 (C1组) 、包囊2代组 (C2 组) 、包囊3代组 (C3组) 、包囊4代组 (C4组); T组小鼠按1 × 105个/只剂量腹腔注射弓形虫速殖子, 感染后第30天处死小 鼠并收集其脑组织内包囊, 再按20个/只感染C1组小鼠。此后每一代小鼠均采用上一代所产生包囊进行口服连续传代, 观察连续传代对小鼠脑内弓形虫包囊数量的影响。结果 以弓形虫速殖子、包囊、卵囊分别感染小鼠, 感染第6~13天 小鼠出现明显临床症状、感染第 7~12 天小鼠出现集中死亡。感染第 30 天时, 感染速殖子、包囊、卵囊的小鼠存活率分 别为67.0%、87.0%、53.0%, 平均脑内包囊数量分别为 (516.0 ± 257.2) 、 (1 203.0 ± 502.0) 、 (581.0 ± 183.1) 个, 差异有统计 学意义 (F = 11.94, P < 0.01), 感染速殖子、卵囊的小鼠脑内包囊数低于感染包囊的小鼠 (P 均< 0.01) 。感染后第30天, 低、中、高剂量组小鼠存活率分别为87.0%、87.0%、60.0%, 平均脑内包囊数量分别为 (953.0 ± 355.5) 、 (1 084.0 ± 474.3) 、 (1 113.0 ± 546.0) 个, 差异无统计学意义 (F = 0.42, P > 0.05); 雄、雌性组小鼠存活率分别为73.0%和80.0%, 平均脑内包 囊数量分别为 (946.4 ± 411.4) 、 (932.1 ± 322.4) 个, 差异无统计学意义 (F = 1.63, P > 0.05) 。通过连续传代感染后, T、C1、 C2、C3、C4组小鼠平均脑内包囊数量分别为 (516.0 ± 257.2) 、 (1 203.0 ± 502.0) 、 (896.8 ± 332.3) 、 (782.5 ± 423.9) 、 (829.2 ± 306.0) 个, 差异有统计学意义 (F = 4.82, P < 0.01); C1组小鼠脑内包囊数高于速殖子组, 差异有统计学意义 (P < 0.01) 。 小鼠口服20个包囊后, 感染第20天首次查见脑内弓形虫包囊, 随感染时间延长脑内包囊数量逐渐增加; 至感染第30、90 天时, 脑内包囊数量分别达峰值, 此后逐步下降, 至感染第180天时仍能查见脑内包囊。结论 刚地弓形虫包囊较速殖 子、卵囊感染后形成慢性感染的可能性更高, 且慢性感染程度亦最严重; 感染弓形虫包囊数量与慢性感染严重程度无关; 脑内包囊形成数量于感染第30天和90天时达高峰。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    共感染是一个常见的现实,但了解免疫系统在这种情况下的反应是复杂的,并且可能是不可预测的。Heligmosomoidesbakeri(寄生虫,以前的多回螺旋体)和弓形虫(原生动物寄生虫)是经过充分研究的生物体,它们刺激特征性的Th2和Th1反应,分别。若干研究已经证明,在与这些生物体共感染的动物中,炎性细胞因子应答降低。然而,虽然已经检查了一般的细胞因子特征,不同细胞因子产生淋巴细胞对寄生虫控制/清除的影响尚不完全清楚.我们调查了五种不同的淋巴细胞群体(NK,NKT,γδT,CD4+T和CD8+T细胞),五个器官(小肠,Peyer的补丁,肠系膜淋巴结,脾脏和肝脏),和4种细胞因子(IFN©,IL-4,IL-10和IL-13)在两个不同的时间点(弓形虫感染后第5天和第10天)。我们发现共感染的动物的死亡率明显高于任一单一感染。这伴随着寄生虫负荷和细胞因子谱的瞬时和局部变化。尽管淋巴细胞和细胞因子谱的早期变化,共感染小鼠的严重肠道病理可能导致早期死亡,这是由于小肠中两种寄生虫的严重损伤。我们的工作证明了在感染研究期间采取广泛观点的重要性,研究多种细胞类型,器官/组织和时间点将免疫学与病理发现联系起来和/或分离。我们的结果提供了与刺激免疫系统不同臂的寄生虫共同感染如何导致感染动力学的急剧变化的见解。
    Co-infections are a common reality but understanding how the immune system responds in this context is complex and can be unpredictable. Heligmosomoides bakeri (parasitic roundworm, previously Heligmosomoides polygyrus) and Toxoplasma gondii (protozoan parasite) are well studied organisms that stimulate a characteristic Th2 and Th1 response, respectively. Several studies have demonstrated reduced inflammatory cytokine responses in animals co-infected with such organisms. However, while general cytokine signatures have been examined, the impact of the different cytokine producing lymphocytes on parasite control/clearance is not fully understood. We investigated five different lymphocyte populations (NK, NKT, γδ T, CD4+ T and CD8+ T cells), five organs (small intestine, Peyer\'s patches, mesenteric lymph nodes, spleen and liver), and 4 cytokines (IFN©, IL-4, IL-10 and IL-13) at two different time points (days 5 and 10 post T. gondii infection). We found that co-infected animals had significantly higher mortality than either single infection. This was accompanied by transient and local changes in parasite loads and cytokine profiles. Despite the early changes in lymphocyte and cytokine profiles, severe intestinal pathology in co-infected mice likely contributed to early mortality due to significant damage by both parasites in the small intestine. Our work demonstrates the importance of taking a broad view during infection research, studying multiple cell types, organs/tissues and time points to link and/or uncouple immunological from pathological findings. Our results provide insights into how co-infection with parasites stimulating different arms of the immune system can lead to drastic changes in infection dynamics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:弓形虫是人类和温血动物中广泛流行的人畜共患原生动物寄生虫。这种寄生虫感染人类会导致严重的临床症状,特别是在患有先天性弓形虫病或免疫功能低下的患者中。污染主要通过食源性途径发生,特别是食用动物的生肉或未煮熟的肉。
    目的:本研究的目的是使用PCR检测大不里士屠宰场屠宰的水牛和牛的组织和器官中的弓形虫,在伊朗。
    方法:50克心脏,大腿,选择在大不里士工业屠宰场屠宰的50只水牛和100头牛的隔膜和舌头进行采样,并结合方便采样。使用先前公开的PCR方法测试样品。
    结果:在150个动物样本中,在10人中检测到弓形虫(6.7%,95CI:3.2-11.9),包括一头水牛(2%,95CI:0.1-10.6)和9头牛(9%,95CI:4.2-16.4)。不同年龄和性别的牛弓形虫感染率差异无统计学意义(p>0.05)。
    结论:结果表明弓形虫通过食用受感染的肉类传播给人类的潜在风险。因此,应采取适当有效的预防措施,限制这种寄生虫向人类的传播,应劝阻食用生肉和未煮熟的肉。
    BACKGROUND: Toxoplasma gondii is a widely prevalent zoonotic protozoan parasite in humans and warm-blooded animals worldwide. Infection of humans by this parasite can result in severe clinical symptoms, particularly in individuals with congenital toxoplasmosis or immunocompromised patients. Contamination mainly occurs through foodborne routes, especially the consumption of raw or undercooked meat from animals.
    OBJECTIVE: The aim of this study was to use PCR to detect T. gondii in tissues and organs of buffaloes and cattle slaughtered at Tabriz slaughterhouse, in Iran.
    METHODS: Fifty grams of heart, thigh, diaphragm and tongue from 50 buffaloes and 100 cattle slaughtered at the Tabriz industrial slaughterhouse were selected for sampling using a combination of convenience sampling. The samples were tested using a previously published PCR method.
    RESULTS: Out of the 150 animal samples, T. gondii was detected in 10 (6.7%, 95%CI: 3.2-11.9), including one buffalo (2%, 95%CI: 0.1-10.6) and nine cattle (9%, 95%CI: 4.2-16.4). There was no statistically significant difference in the rate of T. gondii infection among cattle based on age and sex (p > 0.05).
    CONCLUSIONS: The results indicated a potential risk of T. gondii transmission to humans through the consumption of infected meat. Therefore, appropriate and effective preventive measures should be taken to limit the transmission of this parasite to humans, and the consumption of raw and undercooked meat should be discouraged.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    弓形虫是一种对人类和兽医健康至关重要的细胞内寄生虫。弓形虫基因型种群的结构和多样性在地理上有很大差异,但是三个血统,I型,II和III,分布在全球。谱系III基因型在生物学方面特征最差,宿主免疫力和毒力。一旦宿主感染了T.gondii,参与先天免疫机制以减少组织中的寄生虫负担,并创造促炎环境,在该环境中,TH1应答发展以确保存活.这项研究调查了Swiss-Webster小鼠腹膜内感染后的早期细胞免疫反应,该小鼠具有四种不同的非克隆基因型III和局部分离株ToxoDB#1的10个速殖子。毒力表型,ROP5,ROP16,ROP18和GRA15的累积死亡率(CM)和等位基因谱先前已发表。
    通过实时PCR和IFNγ的相对表达水平分析寄生虫在不同组织中的传播,颈淋巴结(CLN)中的IL12-p40,IL-10和TBX21,使用ΔΔCt方法计算脑和脾。通过检测脑中的BAG1转录物确定阶段转化。
    组织播散取决于毒力表型,但不取决于CM,而TBX21和细胞因子水平和动力学与CM的相关性比毒力表型更好。BAG1的最早检测是感染后7天。只有高CM基因型(69.4%)的感染与CLN24h中的高T-bet水平和在第一周内持续的高全身IFNγ表达有关,而感染基因型较低的CM(38.8%,10.7%和6.8%)的特征在于IFNγ的下调和/或系统水平低。响应强度,通过细胞因子水平评估,随着时间的推移,高CM的基因型逐渐减弱,而逐渐增加到低CM的基因型。
    结果表明,免疫应答与毒力表型和/或等位基因谱无关,但是早期发作,强烈的促炎反应是高CM基因型的特征。此外,大脑中的高IFNγ水平可能会阻碍阶段转换。
    UNASSIGNED: Toxoplasma gondii is an intracellular parasite of importance to human and veterinary health. The structure and diversity of the genotype population of T. gondii varies considerably with respect to geography, but three lineages, type I, II and III, are distributed globally. Lineage III genotypes are the least well characterized in terms of biology, host immunity and virulence. Once a host is infected with T.gondii, innate immune mechanisms are engaged to reduce the parasite burden in tissues and create a pro-inflammatory environment in which the TH1 response develops to ensure survival. This study investigated the early cellular immune response of Swiss-Webster mice post intraperitoneal infection with 10 tachyzoites of four distinct non-clonal genotypes of lineage III and a local isolate of ToxoDB#1. The virulence phenotype, cumulative mortality (CM) and allele profiles of ROP5, ROP16, ROP18 and GRA15 were published previously.
    UNASSIGNED: Parasite dissemination in different tissues was analyzed by real-time PCR and relative expression levels of IFNγ, IL12-p40, IL-10 and TBX21 in the cervical lymph nodes (CLN), brain and spleen were calculated using the ΔΔCt method. Stage conversion was determined by detection of the BAG1 transcript in the brain.
    UNASSIGNED: Tissue dissemination depends on the virulence phenotype but not CM, while the TBX21 and cytokine levels and kinetics correlate better with CM than virulence phenotype. The earliest detection of BAG1 was seven days post infection. Only infection with the genotype of high CM (69.4%) was associated with high T-bet levels in the CLN 24 h and high systemic IFNγ expression which was sustained over the first week, while infection with genotypes of lower CM (38.8%, 10.7% and 6.8%) is characterized by down-regulation and/or low systemic levels of IFNγ. The response intensity, as assessed by cytokine levels, to the genotype of high CM wanes over time, while it increases gradually to genotypes of lower CM.
    UNASSIGNED: The results point to the conclusion that the immune response is not correlated with the virulence phenotype and/or allele profile, but an early onset, intense pro-inflammatory response is characteristic of genotypes with high CM. Additionally, high IFNγ level in the brain may hamper stage conversion.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:病原体可以影响宿主RNA修饰机制,为其复制建立有利的细胞环境。在本研究中,我们研究了弓形虫感染对宿主RNA修饰谱的影响,并探讨了这些修饰如何影响宿主-寄生虫的相互作用.
    结果:我们分析了小鼠肝脏中~80nttRNA和17-50ntsncRNAs的修饰水平,脾,脾和血清使用液相色谱和串联质谱分析。结果揭示了RNA修饰谱的改变,特别是在急性感染期间。肝脏比脾脏表现出更多的差异丰富的RNA修饰。与对照小鼠相比,急性感染期间血清中的RNA修饰水平大部分下调。在感染期间肝脏和脾脏中的不同RNA修饰之间以及几种RNA修饰与许多细胞因子之间检测到相关性。RNA修饰的改变影响tRNA稳定性和蛋白质翻译。
    结论:这些发现为RNA修饰在介导鼠宿主对弓形虫感染的反应中的作用提供了新的见解。
    BACKGROUND: Pathogens can impact host RNA modification machinery to establish a favorable cellular environment for their replication. In the present study, we investigated the effect of Toxoplasma gondii infection on host RNA modification profiles and explored how these modifications may influence the host-parasite interaction.
    RESULTS: We analyzed the modification levels of ∼ 80 nt tRNA and 17-50 nt sncRNAs in mouse liver, spleen, and serum using liquid chromatography and tandem mass spectrometry analysis. The results revealed alterations in RNA modification profiles, particularly during acute infection. The liver exhibited more differentially abundant RNA modifications than the spleen. RNA modification levels in serum were mostly downregulated during acute infection compared to control mice. Correlations were detected between different RNA modifications in the liver and spleen during infection and between several RNA modifications and many cytokines. Alterations in RNA modifications affected tRNA stability and protein translation.
    CONCLUSIONS: These findings provide new insight into the role of RNA modifications in mediating the murine host response to T. gondii infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    弓形虫病是由弓形虫原虫引起的食源性疾病,并通过食用生肉或未煮熟的肉传播给人类,主要是。家禽,牛肉,猪肉是秘鲁消费的主要肉类;尽管如此,豚鼠肉也被广泛食用。出于这个原因,这项研究的目的是对库斯科马兰加尼地区的家养和野生豚鼠中的弓形虫进行分子检测,秘鲁,并确定与该病原体相关的一些危险因素。从豚鼠(30只国产和30只野生)的脑组织样本中提取DNA,和PCR方案用于从弓形虫基因组中扩增内部转录间隔区(ITS-1)和529bp片段。在14只(23.3%)豚鼠中检测到弓形虫DNA。家养豚鼠的弓形虫频率为33.3%,野生豚鼠为13.3%。我们的结果表明,豚鼠是该地区人群中弓形虫感染的重要来源。
    Toxoplasmosis is a foodborne disease caused by the protozoan Toxoplasma gondii, and transmitted to humans by eating raw or undercooked meat, mainly. Poultry, beef, and pork are the main meats consumed in Peru; despite this, guinea pig meat is also widely consumed. For this reason, the objective of this study was to molecularly detect T. gondii in domestic and wild guinea pigs from the Marangani district in Cuzco, Peru, and identify some risk factors associated with this pathogen. DNA was extracted from the brain tissue samples of guinea pigs (30 domestic and 30 wild), and PCR protocols were used to amplify the internal transcribed spacer (ITS-1) region and a 529 bp fragment from the T. gondii genome. T. gondii DNA was detected in 14 (23.3%) guinea pigs. T. gondii frequency was 33.3% in domestic guinea pigs and 13.3% in wild guinea pigs. Our results demonstrated that guinea pigs represent an important source for T. gondii infection in human populations in this locality.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:弓形虫是一种细胞内原生动物寄生虫,广泛分布于人类和温血动物中。弓形虫慢性感染可引起弓形虫脑病,不良妊娠,和男性生殖障碍。在男性繁殖中,睾丸的主要功能是为精子发生和免疫保护提供稳定的场所。影响睾丸组织的疾病包括生殖细胞周期的异常,生精迟缓,或完全停止精子发育。然而,弓形虫与生殖系统相互作用的机制尚不清楚。目的是研究精子发生相关基因的表达水平,弓形虫感染后,在小鼠睾丸组织中。
    方法:对感染或未感染弓形虫II型Prugniaud(PRU)菌株的小鼠睾丸组织进行RNA-seq测序,并结合实时定量PCR和免疫荧光分析进行验证。
    结果:结果表明,有250个显着的差异表达基因(DEGs)(P<0.05,|log2倍变化|≥1)。生物信息学剖析显示,101DEGs注释为1696基因本体论(GO)术语。虽然整个生物过程分类中的DEGs数量较多,GO富集显示DEGs在细胞组分分类中的显著存在。弓形虫感染后,Arhgap18和Syne1基因发生调节变化,两者都参与了形成血睾丸屏障(BTB)的细胞骨架。MAPK信号通路中富含DEGs的数量,ERK1/2信号通路,和JNK信号通路显著。PTGDS基因位于花生四烯酸代谢通路,在睾丸中BTB的形成和维持中起着重要作用。弓形虫感染后,PTGDS的表达下调,可能对睾丸内BTB的完整性和生精微环境产生有害影响。
    结论:总体而言,我们的研究深入了解了慢性弓形虫感染可能如何影响睾丸组织并可能影响男性生育能力.这些发现为弓形虫感染对男性生殖系统的影响提供了新的视角。
    BACKGROUND: Toxoplasma gondii is an intracellular protozoan parasite that is widely distributed in humans and warm-blooded animals. T. gondii chronic infections can cause toxoplasmic encephalopathy, adverse pregnancy, and male reproductive disorders. In male reproduction, the main function of the testis is to provide a stable place for spermatogenesis and immunological protection. The disorders affecting testis tissue encompass abnormalities in the germ cell cycle, spermatogenic retardation, or complete cessation of sperm development. However, the mechanisms of interaction between T. gondii and the reproductive system is unclear. The aims were to study the expression levels of genes related to spermatogenesis, following T. gondii infection, in mouse testicular tissue.
    METHODS: RNA-seq sequencing was carried out on mouse testicular tissues from mice infected or uninfected with the T. gondii type II Prugniaud (PRU) strain and validated in combination with real-time quantitative PCR and immunofluorescence assays.
    RESULTS: The results showed that there were 250 significant differentially expressed genes (DEGs) (P < 0.05, |log2fold change| ≧ 1). Bioinformatics analysis showed that 101 DEGs were annotated to the 1696 gene ontology (GO) term. While there was a higher number of DEGs in the biological process classification as a whole, the GO enrichment revealed a significant presence of DEGs in the cellular component classification. The Arhgap18 and Syne1 genes undergo regulatory changes following T. gondii infection, and both were involved in shaping the cytoskeleton of the blood-testis barrier (BTB). The number of DEGs enriched in the MAPK signaling pathway, the ERK1/2 signaling pathway, and the JNK signaling pathway were significant. The PTGDS gene is located in the Arachidonic acid metabolism pathway, which plays an important role in the formation and maintenance of BTB in the testis. The expression of PTGDS is downregulated subsequent to T. gondii infection, potentially exerting deleterious effects on the integrity of the BTB and the spermatogenic microenvironment within the testes.
    CONCLUSIONS: Overall, our research provides in-depth insights into how chronic T. gondii infection might affect testicular tissue and potentially impact male fertility. These findings offer a new perspective on the impact of T. gondii infection on the male reproductive system.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    当弓形虫通过其宿主传播时,寄生虫必须感知并适应其环境并清除营养。氧气(O2)是这样的环境因素之一,胞质脯氨酸4-羟化酶(PHD)是进化上保守的O2细胞传感蛋白,可调节对O2可用性变化的反应。弓形虫表达2个PHDs。其中一个,TgPHYa羟基化SCF-E3泛素连接酶复合物的亚基SKP1。体外,TgPHYa对于在低O2水平下的生长是重要的。然而,研究尚未检查TgPHYa或任何其他病原体编码的PHD在毒力和疾病中的作用。使用II型ME49弓形虫TgPHYa基因敲除,我们报道TgPHYa对小鼠弓形虫毒力和脑囊肿形成有重要作用。我们进一步发现,虽然TgPHYa突变寄生虫可以在肠道中建立感染,它们不能有效地传播到外周组织,因为突变的寄生虫不能在募集的免疫细胞中存活。由于这种表型在IFNγ敲除小鼠中被废除,我们研究了TgPHYa如何在IFNγ处理的细胞中介导存活。我们发现,将寄生虫编码的效应子释放到中和IFNγ诱导的抗寄生虫过程的宿主细胞中不需要TgPHYa。相比之下,我们发现TgPHYa是寄生虫清除色氨酸所必需的,这是一种氨基酸,其水平在IFNγ上调色氨酸分解代谢酶后降低,吲哚胺双加氧酶(IDO)。我们进一步发现,相对于野生型小鼠,当感染TgPHYa基因敲除寄生虫时,IDO基因敲除小鼠的发病率增加。一起,这些数据确定了逃避IFNγ诱导的营养免疫的第一个寄生虫机制,并强调了氧敏感蛋白在病原体生长和毒力中发挥的新作用。
    As Toxoplasma gondii disseminates through its host, the parasite must sense and adapt to its environment and scavenge nutrients. Oxygen (O2) is one such environmental factor and cytoplasmic prolyl 4-hydroxylases (PHDs) are evolutionarily conserved O2 cellular sensing proteins that regulate responses to changes in O2 availability. Toxoplasma expresses 2 PHDs. One of them, TgPHYa hydroxylates SKP1, a subunit of the SCF-E3 ubiquitin ligase complex. In vitro, TgPHYa is important for growth at low O2 levels. However, studies have yet to examine the role that TgPHYa or any other pathogen-encoded PHD plays in virulence and disease. Using a type II ME49 Toxoplasma TgPHYa knockout, we report that TgPHYa is important for Toxoplasma virulence and brain cyst formation in mice. We further find that while TgPHYa mutant parasites can establish an infection in the gut, they are unable to efficiently disseminate to peripheral tissues because the mutant parasites are unable to survive within recruited immune cells. Since this phenotype was abrogated in IFNγ knockout mice, we studied how TgPHYa mediates survival in IFNγ-treated cells. We find that TgPHYa is not required for release of parasite-encoded effectors into host cells that neutralize anti-parasitic processes induced by IFNγ. In contrast, we find that TgPHYa is required for the parasite to scavenge tryptophan, which is an amino acid whose levels are decreased after IFNγ up-regulates the tryptophan-catabolizing enzyme, indoleamine dioxygenase (IDO). We further find, relative to wild-type mice, that IDO knockout mice display increased morbidity when infected with TgPHYa knockout parasites. Together, these data identify the first parasite mechanism for evading IFNγ-induced nutritional immunity and highlight a novel role that oxygen-sensing proteins play in pathogen growth and virulence.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    本研究调查了两种主要的血液传播病原体的分子流行和系统发育特征,弓形虫(T.刚地)和疟原虫。,在四个地区(Layyah,德拉·加齐·汗,拉合尔,和木尔坦)在旁遮普邦,巴基斯坦,在2021年至2023年的狩猎季节。此外,检查这些病原体对宿主全血细胞计数(CBC)的影响。在测试的433只鹌鹑中,25(5.8%)显示弓形虫的内部转录间隔区(ITS-1)基因扩增,而15(3.5%)显示疟原虫的细胞色素b基因扩增。危险因素分析表明,两种病原体的患病率不仅限于特定的采样地点或鸟类性别(P>0.05)。区域分析强调,母鸡更容易感染弓形虫和疟原虫。感染比公鸡。与养殖鸟类相比,野生鹌鹑对弓形虫的易感性更高。与未感染的禽类相比,感染的禽类中记录了显着的CBC变化。产生的序列的BLAST分析已经证实了回收的PCR扩增子作为弓形虫和恶性疟原虫的身份。系统发育分析显示,巴基斯坦分离株与全球各国报告的分离株聚集在一起。这项研究提供了弓形虫和疟原虫的第一份文件。巴基斯坦鹌鹑感染,强调需要在不同地区进行详细调查,以增强我们对感染率和这些寄生虫的人畜共患潜力的了解。
    This study investigates the molecular prevalence and phylogenetic characteristics of two prominent blood-borne pathogens, Toxoplasma gondii (T. gondii) and Plasmodium spp., in common quails (Coturnix coturnix) sampled from both wild (N = 236) and farmed (N = 197) populations across four districts (Layyah, Dera Ghazi Khan, Lahore, and Multan) in Punjab, Pakistan, during the hunting seasons from 2021 to 2023. Additionally, the impact of these pathogens on the complete blood count (CBC) of the hosts is examined. Out of 433 quails tested, 25 (5.8%) exhibited amplification of the internal transcribed spacer (ITS-1) gene for T. gondii, while 15 (3.5%) showed amplification of the Cytochrome b gene for Plasmodium spp. A risk factor analysis indicated that the prevalence of both pathogens was not confined to specific sampling sites or bird sexes (P > 0.05). District-wise analysis highlighted that hens were more susceptible to both T. gondii and Plasmodium spp. infections than cocks. Wild quails exhibited a higher susceptibility to T. gondii compared to farmed birds. Significant CBC variations were recorded in infected birds as compared to uninfected ones. BLAST analysis of generated sequences has confirmed the identity of recovered PCR amplicons as T. gondii and Plasmodium relictum. Phylogenetic analysis revealed that Pakistani isolates clustered with those reported from various countries globally. This study provides the first documentation of T. gondii and Plasmodium sp. infections in Pakistani quails, underscoring the need for detailed investigations across different regions to enhance our understanding of infection rates and the zoonotic potential of these parasites.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号