首页 > 最新文献

ACS Infectious Diseases最新文献

英文 中文
Yet to SET: Plasmodium falciparum Histone Lysine Methyltransferases 尚未确定:恶性疟原虫组蛋白赖氨酸甲基转移酶。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-13 DOI: 10.1021/acsinfecdis.5c00578
Disha Shah,  and , Krishanpal Karmodiya*, 

Malaria caused by Plasmodium falciparum continues to remain a global health challenge. Its prevention, treatment and elimination efforts are threatened by the inevitable emergence of drug resistance to currently effective treatment regimes. New antimalarials with distinct modes of action and multistage and multispecies activity will be an important addition to the arms race against the malarial parasite. P. falciparum’s epigenome represents a promising target in this battle and offers exciting opportunities for targeted intervention. With an unusually AT-rich genome, a relative paucity of specific transcription factors and limited heterochromatin, epigenetic control has emerged as an important contributor to P. falciparum’s survival and virulence. P. falciparum histones are marked dynamically with a vast array of post translational modifications. These include several well studied and some novel marks. The parasite has an epigenetic signature distinct from its host and shows several parasite-specific adaptations. The regulators of these marks remain understudied, however. While histone acetylation and its regulators have been more extensively studied in the field, research on other epigenetic effectors is also catching up. This review highlights the research efforts aimed at understanding the role of the parasite’s histone lysine methyltransferases in shaping transcriptional regulation and the histone modification landscape.

恶性疟原虫引起的疟疾仍然是一项全球卫生挑战。它的预防、治疗和消除努力受到对目前有效治疗方案不可避免地出现耐药性的威胁。具有不同作用方式和多阶段、多物种活性的新型抗疟药将是对抗疟原虫军备竞赛的重要补充。恶性疟原虫的表观基因组在这场战斗中代表了一个有希望的目标,并为有针对性的干预提供了令人兴奋的机会。具有异常丰富的at基因组,特异性转录因子的相对缺乏和有限的异染色质,表观遗传控制已成为恶性疟原虫生存和毒力的重要贡献者。恶性疟原虫组蛋白被动态标记为大量的翻译后修饰。其中包括一些研究得很好的标记和一些新的标记。寄生虫具有与其宿主不同的表观遗传特征,并表现出几种寄生虫特有的适应性。然而,这些标志的监管机构仍未得到充分研究。虽然组蛋白乙酰化及其调控因子已经在该领域得到了更广泛的研究,但对其他表观遗传效应的研究也在追赶。本文综述了旨在了解寄生虫组蛋白赖氨酸甲基转移酶在形成转录调控和组蛋白修饰景观中的作用的研究工作。
{"title":"Yet to SET: Plasmodium falciparum Histone Lysine Methyltransferases","authors":"Disha Shah,&nbsp; and ,&nbsp;Krishanpal Karmodiya*,&nbsp;","doi":"10.1021/acsinfecdis.5c00578","DOIUrl":"10.1021/acsinfecdis.5c00578","url":null,"abstract":"<p >Malaria caused by <i>Plasmodium falciparum</i> continues to remain a global health challenge. Its prevention, treatment and elimination efforts are threatened by the inevitable emergence of drug resistance to currently effective treatment regimes. New antimalarials with distinct modes of action and multistage and multispecies activity will be an important addition to the arms race against the malarial parasite. <i>P. falciparum</i>’s epigenome represents a promising target in this battle and offers exciting opportunities for targeted intervention. With an unusually AT-rich genome, a relative paucity of specific transcription factors and limited heterochromatin, epigenetic control has emerged as an important contributor to <i>P. falciparum</i>’s survival and virulence. <i>P. falciparum</i> histones are marked dynamically with a vast array of post translational modifications. These include several well studied and some novel marks. The parasite has an epigenetic signature distinct from its host and shows several parasite-specific adaptations. The regulators of these marks remain understudied, however. While histone acetylation and its regulators have been more extensively studied in the field, research on other epigenetic effectors is also catching up. This review highlights the research efforts aimed at understanding the role of the parasite’s histone lysine methyltransferases in shaping transcriptional regulation and the histone modification landscape.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"2979–2992"},"PeriodicalIF":3.8,"publicationDate":"2025-10-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145285084","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hantavirus GnH Nanoparticle Immunogen Elicits a Cross-Neutralizing Antibody Response in Mice 汉坦病毒GnH纳米颗粒免疫原在小鼠体内引发交叉中和抗体反应。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-12 DOI: 10.1021/acsinfecdis.5c00415
Kevin E. Ramos, , , Margarette C. Mariano, , , Eva Mittler, , , Romina Pardo, , , Vanessa Zylberman, , , Pablo Guardado-Calvo, , , Kartik Chandran, , and , Jonathan R. Lai*, 

Hantaviruses are zoonotic pathogens that are spread by rodents and can cause severe and fatal disease in humans. “New World” hantaviruses (endemic to North and South America) include the human pathogenic Andes orthohantavirus (ANDV), Choclo orthohantavirus (CHOV), and Sin Nombre orthohantavirus (SNV). Human infections can lead to hantavirus cardiopulmonary syndrome (HCPS), which is associated with ∼40% mortality. Currently, there are no FDA-approved hantavirus vaccines or treatments, but neutralizing antibodies targeting the glycoproteins Gn and Gc have been shown to be protective in animals. Here, we develop nanoparticle immunogens bearing the Gn head domain (GnH) from ANDV, CHOV, or SNV. Initial immunization studies with the ANDV-GnH monomer indicated that this antigen elicited a reactive but non-neutralizing antibody response in mice. To bolster the immune response, we developed a strategy to link GnHs to mi3 nanoparticles using the SpyCatcher/SpyTag bioconjugation technology. We found that ANDV-GnH-mi3 nanoparticles elicited a cross-reactive antibody response that neutralized pseudotyped viruses containing ANDV and CHOV glycoproteins but not SNV. In contrast, CHOV-GnH-mi3 nanoparticles elicited only a homotypic neutralizing response. Finally, the reactivity of sera from mice immunized with a cocktail of ANDV-GnH-mi3 and SNV-GnH-mi3 nanoparticles was similar to sera from mice immunized with ANDV-GnH-mi3, only indicating that the SNV-GnH-mi3 antibody response was not even homotypically neutralizing. These results suggest that there are differences in immunodominance that may contribute to the breadth of the hantavirus-targeting neutralizing response elicited by GnH-based immunogens. Nonetheless, the cross-neutralizing sera obtained by ANDV-GnH-mi3 immunization suggest that developing broad immunogens may be possible with appropriate engineering.

汉坦病毒是由啮齿动物传播的人畜共患病原体,可引起人类严重和致命的疾病。“新世界”汉坦病毒(北美和南美特有)包括人类致病性安第斯正汉坦病毒(ANDV)、巧克力正汉坦病毒(CHOV)和Sin Nombre正汉坦病毒(SNV)。人类感染可导致汉坦病毒心肺综合征(HCPS),死亡率约为40%。目前,还没有fda批准的汉坦病毒疫苗或治疗方法,但针对糖蛋白Gn和Gc的中和抗体已被证明对动物有保护作用。在这里,我们从ANDV, CHOV或SNV中开发了带有Gn头部结构域(GnH)的纳米颗粒免疫原。ANDV-GnH单体的初步免疫研究表明,该抗原在小鼠中引起反应性但非中和性抗体反应。为了增强免疫反应,我们开发了一种使用SpyCatcher/SpyTag生物偶联技术将GnHs与mi3纳米颗粒连接的策略。我们发现ANDV- gnh -mi3纳米颗粒引发了一种交叉反应性抗体反应,可以中和含有ANDV和CHOV糖蛋白的假型病毒,但不能中和SNV。相比之下,CHOV-GnH-mi3纳米颗粒只引起同型中和反应。最后,用ANDV-GnH-mi3和SNV-GnH-mi3纳米颗粒混合免疫小鼠血清的反应性与用ANDV-GnH-mi3免疫小鼠血清的反应性相似,只是表明SNV-GnH-mi3抗体反应甚至没有同型中和。这些结果表明,免疫优势的差异可能有助于gnh免疫原引起的汉坦病毒靶向中和反应的广度。尽管如此,ANDV-GnH-mi3免疫获得的交叉中和血清表明,通过适当的工程设计,开发广泛的免疫原是可能的。
{"title":"Hantavirus GnH Nanoparticle Immunogen Elicits a Cross-Neutralizing Antibody Response in Mice","authors":"Kevin E. Ramos,&nbsp;, ,&nbsp;Margarette C. Mariano,&nbsp;, ,&nbsp;Eva Mittler,&nbsp;, ,&nbsp;Romina Pardo,&nbsp;, ,&nbsp;Vanessa Zylberman,&nbsp;, ,&nbsp;Pablo Guardado-Calvo,&nbsp;, ,&nbsp;Kartik Chandran,&nbsp;, and ,&nbsp;Jonathan R. Lai*,&nbsp;","doi":"10.1021/acsinfecdis.5c00415","DOIUrl":"10.1021/acsinfecdis.5c00415","url":null,"abstract":"<p >Hantaviruses are zoonotic pathogens that are spread by rodents and can cause severe and fatal disease in humans. “New World” hantaviruses (endemic to North and South America) include the human pathogenic <i>Andes orthohantavirus</i> (ANDV), <i>Choclo orthohantavirus</i> (CHOV), and <i>Sin Nombre orthohantavirus</i> (SNV). Human infections can lead to hantavirus cardiopulmonary syndrome (HCPS), which is associated with ∼40% mortality. Currently, there are no FDA-approved hantavirus vaccines or treatments, but neutralizing antibodies targeting the glycoproteins Gn and Gc have been shown to be protective in animals. Here, we develop nanoparticle immunogens bearing the Gn head domain (Gn<sup>H</sup>) from ANDV, CHOV, or SNV. Initial immunization studies with the ANDV-Gn<sup>H</sup> monomer indicated that this antigen elicited a reactive but non-neutralizing antibody response in mice. To bolster the immune response, we developed a strategy to link Gn<sup>H</sup>s to mi3 nanoparticles using the SpyCatcher/SpyTag bioconjugation technology. We found that ANDV-Gn<sup>H</sup>-mi3 nanoparticles elicited a cross-reactive antibody response that neutralized pseudotyped viruses containing ANDV and CHOV glycoproteins but not SNV. In contrast, CHOV-Gn<sup>H</sup>-mi3 nanoparticles elicited only a homotypic neutralizing response. Finally, the reactivity of sera from mice immunized with a cocktail of ANDV-Gn<sup>H</sup>-mi3 and SNV-Gn<sup>H</sup>-mi3 nanoparticles was similar to sera from mice immunized with ANDV-Gn<sup>H</sup>-mi3, only indicating that the SNV-Gn<sup>H</sup>-mi3 antibody response was not even homotypically neutralizing. These results suggest that there are differences in immunodominance that may contribute to the breadth of the hantavirus-targeting neutralizing response elicited by Gn<sup>H</sup>-based immunogens. Nonetheless, the cross-neutralizing sera obtained by ANDV-Gn<sup>H</sup>-mi3 immunization suggest that developing broad immunogens may be possible with appropriate engineering.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"3061–3070"},"PeriodicalIF":3.8,"publicationDate":"2025-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145278487","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Effects of Sexual Dimorphism and the Estrous Cycle on Clostridioides difficile Infection Prophylaxis in Two Rodent Models 两性异形和发情周期对两种啮齿类动物难辨梭菌感染预防的影响。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-10 DOI: 10.1021/acsinfecdis.5c00511
Jacqueline R. Phan, , , McKenzie Washington, , , Dung M. Do, , , Tiffany V. Mata, , , Maria Niamba, , , Efren Heredia, , , Robert Soriano, , , Chandler Hassan, , , Chad L. Cross, , and , Ernesto Abel-Santos*, 

Clostridioides difficile infection (CDI) is responsible for the majority of identifiable antibiotic-associated diarrhea. Epidemiological studies have consistently shown that women are more at risk for CDI than men. C. difficile is spread by spores that germinate in the antibiotic-altered gut of patients to generate toxin-producing vegetative cells. Since germination is required for CDI, we have shown that cholan-24-amides containing m-sulfanilic acid (CamSA) or aniline (CaPA) inhibit C. difficile spore germination and prevent CDI in rodents. In this study, we found that CDI prophylaxis showed clear sexual dimorphism. Male mice developed less severe CDI but were also more refractory to treatment. On the other hand, anti-germinants protected female mice from developing CDI during most stages of their estrous cycle. Interestingly, infection sexual dimorphism was reversed in hamsters, with male hamsters developing more severe CDI signs than females. Furthermore, anti-germinant compounds protected female hamsters more strongly than male hamsters.

艰难梭菌感染(CDI)是大多数可识别的抗生素相关性腹泻的原因。流行病学研究一致表明,女性患CDI的风险高于男性。艰难梭菌通过孢子传播,这些孢子在抗生素改变的患者肠道中萌发,产生产生毒素的营养细胞。由于CDI需要萌发,我们已经证明含有间磺胺酸(CamSA)或苯胺(CaPA)的胆兰-24酰胺可抑制艰难梭菌孢子萌发并预防啮齿动物的CDI。在本研究中,我们发现CDI预防表现出明显的性别二态性。雄性小鼠CDI的严重程度较轻,但对治疗也更难治。另一方面,抗生发剂保护雌性小鼠在其发情周期的大多数阶段不发生CDI。有趣的是,感染的两性二态性在仓鼠中被逆转,雄性仓鼠比雌性出现更严重的CDI症状。此外,抗发芽化合物对雌性仓鼠的保护作用强于雄性仓鼠。
{"title":"Effects of Sexual Dimorphism and the Estrous Cycle on Clostridioides difficile Infection Prophylaxis in Two Rodent Models","authors":"Jacqueline R. Phan,&nbsp;, ,&nbsp;McKenzie Washington,&nbsp;, ,&nbsp;Dung M. Do,&nbsp;, ,&nbsp;Tiffany V. Mata,&nbsp;, ,&nbsp;Maria Niamba,&nbsp;, ,&nbsp;Efren Heredia,&nbsp;, ,&nbsp;Robert Soriano,&nbsp;, ,&nbsp;Chandler Hassan,&nbsp;, ,&nbsp;Chad L. Cross,&nbsp;, and ,&nbsp;Ernesto Abel-Santos*,&nbsp;","doi":"10.1021/acsinfecdis.5c00511","DOIUrl":"10.1021/acsinfecdis.5c00511","url":null,"abstract":"<p ><i>Clostridioides difficile</i> infection (CDI) is responsible for the majority of identifiable antibiotic-associated diarrhea. Epidemiological studies have consistently shown that women are more at risk for CDI than men. <i>C. difficile</i> is spread by spores that germinate in the antibiotic-altered gut of patients to generate toxin-producing vegetative cells. Since germination is required for CDI, we have shown that cholan-24-amides containing <i>m</i>-sulfanilic acid (CamSA) or aniline (CaPA) inhibit <i>C. difficile</i> spore germination and prevent CDI in rodents. In this study, we found that CDI prophylaxis showed clear sexual dimorphism. Male mice developed less severe CDI but were also more refractory to treatment. On the other hand, anti-germinants protected female mice from developing CDI during most stages of their estrous cycle. Interestingly, infection sexual dimorphism was reversed in hamsters, with male hamsters developing more severe CDI signs than females. Furthermore, anti-germinant compounds protected female hamsters more strongly than male hamsters.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"3030–3035"},"PeriodicalIF":3.8,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145273158","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mn(III) Porphyrin MnTE-2-PyP5+ Associated with Ascorbate: A Redox-Active Therapeutic Strategy against Leishmaniasis Mn(III)卟啉MnTE-2-PyP5+与抗坏血酸相关:抗利什曼病的氧化还原活性治疗策略。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-10 DOI: 10.1021/acsinfecdis.5c00520
Tiago H. S. Souza*, , , Jacqueline C. Bueno-Janice*, , , Letícia S. Vasconcelos, , , Paulo E. Cabral Filho, , , Julio S. Reboucas, , , Regina C. B. Q. Figueiredo, , and , Adriana Fontes, 

Toxicity and rising resistance to current leishmaniasis drugs highlight the need for alternative therapies. Manganese porphyrins (MnPs) have demonstrated therapeutic potential in various oxidative stress-based diseases/ailments due to their redox-modulating properties. Thus, this study aimed to evaluate the redox-active effects of MnTE-2-PyP5+ (BMX-010, AEOL10113, MnP ethyl) combined with ascorbate (Asc, vitamin C) on Leishmania amazonensis, Leishmania braziliensis, and Leishmania chagasi in vitro. The effects on promastigote growth were assessed, and the mechanism of action was studied by quantifying reactive oxygen species (ROS) and using catalase to evaluate H2O2 involvement. The effects on intracellular amastigotes and the mitochondrial membrane potential (ΔΨm) of promastigotes from the most susceptible species were evaluated. Cytotoxicity assays were carried out on mammalian cells. MnP ethyl alone had no impact on parasite growth; however, MnP ethyl/Asc treatment led to a significant reduction in the promastigote growth: 88% for L. amazonensis, 43% for L. chagasi, and 37% for L. braziliensis after 48 h. MnP ethyl/Asc generated about 300% more ROS than the untreated control and induced ΔΨm depolarization. Catalase addition restored parasite survival, confirming H2O2 as the primary mediator of the MnP ethyl/Asc effect. Moreover, MnP ethyl/Asc exhibited minimal cytotoxicity on mammalian cells. The MnP ethyl/Asc treatment reduced the infection index by about 58% and the number of amastigotes per macrophage by 42% in L. amazonensis after 24 h. These findings demonstrated that MnP ethyl/Asc exerted an antileishmanial effect through oxidative stress, providing a promising alternative for preclinical evaluation.

目前利什曼病药物的毒性和不断增加的耐药性突出表明需要替代疗法。锰卟啉(MnPs)由于其氧化还原调节特性,在各种基于氧化应激的疾病/疾病中显示出治疗潜力。因此,本研究旨在评价MnTE-2-PyP5+ (BMX-010、AEOL10113、MnP乙基)联合抗坏血酸(Asc、维生素C)对亚马逊利什曼原虫、巴西利什曼原虫和查加西利什曼原虫的体外氧化还原活性。通过定量测定活性氧(ROS)和过氧化氢酶(过氧化氢酶)对H2O2参与的影响,研究了其对promastigote生长的影响。评价了对最敏感种原毛菌胞内无尾线虫和线粒体膜电位(ΔΨm)的影响。对哺乳动物细胞进行了细胞毒性试验。MnP乙酯对寄生虫生长无明显影响;然而,MnP乙基/Asc处理导致48 h后L. amazonensis的promastigote生长显著降低,L. chagasi的生长降低了88%,L. brasiliensis的生长降低了43%,MnP乙基/Asc的生长降低了37%。MnP乙基/Asc产生的ROS比未处理的对照多300%,并诱导ΔΨm去极化。过氧化氢酶的添加恢复了寄生虫的存活,证实H2O2是MnP乙基/Asc效应的主要媒介。此外,MnP乙基/Asc对哺乳动物细胞的细胞毒性很小。MnP乙基/Asc处理24 h后,使亚马逊河蛭感染指数降低约58%,每个巨噬细胞的无尾线虫数量降低42%。这些结果表明MnP乙基/Asc通过氧化应激发挥抗利什曼原虫的作用,为临床前评价提供了有希望的替代方法。
{"title":"Mn(III) Porphyrin MnTE-2-PyP5+ Associated with Ascorbate: A Redox-Active Therapeutic Strategy against Leishmaniasis","authors":"Tiago H. S. Souza*,&nbsp;, ,&nbsp;Jacqueline C. Bueno-Janice*,&nbsp;, ,&nbsp;Letícia S. Vasconcelos,&nbsp;, ,&nbsp;Paulo E. Cabral Filho,&nbsp;, ,&nbsp;Julio S. Reboucas,&nbsp;, ,&nbsp;Regina C. B. Q. Figueiredo,&nbsp;, and ,&nbsp;Adriana Fontes,&nbsp;","doi":"10.1021/acsinfecdis.5c00520","DOIUrl":"10.1021/acsinfecdis.5c00520","url":null,"abstract":"<p >Toxicity and rising resistance to current leishmaniasis drugs highlight the need for alternative therapies. Manganese porphyrins (MnPs) have demonstrated therapeutic potential in various oxidative stress-based diseases/ailments due to their redox-modulating properties. Thus, this study aimed to evaluate the redox-active effects of MnTE-2-PyP<sup>5+</sup> (BMX-010, AEOL10113, MnP ethyl) combined with ascorbate (Asc, vitamin C) on <i>Leishmania amazonensis</i>, <i>Leishmania braziliensis</i>, and <i>Leishmania chagasi</i> <i>in vitro</i>. The effects on promastigote growth were assessed, and the mechanism of action was studied by quantifying reactive oxygen species (ROS) and using catalase to evaluate H<sub>2</sub>O<sub>2</sub> involvement. The effects on intracellular amastigotes and the mitochondrial membrane potential (ΔΨm) of promastigotes from the most susceptible species were evaluated. Cytotoxicity assays were carried out on mammalian cells. MnP ethyl alone had no impact on parasite growth; however, MnP ethyl/Asc treatment led to a significant reduction in the promastigote growth: 88% for <i>L. amazonensis</i>, 43% for <i>L. chagasi</i>, and 37% for <i>L. braziliensis</i> after 48 h. MnP ethyl/Asc generated about 300% more ROS than the untreated control and induced ΔΨm depolarization. Catalase addition restored parasite survival, confirming H<sub>2</sub>O<sub>2</sub> as the primary mediator of the MnP ethyl/Asc effect. Moreover, MnP ethyl/Asc exhibited minimal cytotoxicity on mammalian cells. The MnP ethyl/Asc treatment reduced the infection index by about 58% and the number of amastigotes per macrophage by 42% in <i>L. amazonensis</i> after 24 h. These findings demonstrated that MnP ethyl/Asc exerted an antileishmanial effect through oxidative stress, providing a promising alternative for preclinical evaluation.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"3128–3137"},"PeriodicalIF":3.8,"publicationDate":"2025-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsinfecdis.5c00520","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145273180","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Platelet-Derived Transforming Growth Factor-β1: A New Hope for Cerebral Malaria Treatment 血小板衍生转化生长因子-β1:脑型疟疾治疗的新希望。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-09 DOI: 10.1021/acsinfecdis.5c00710
Shuangchun Liu, , , Bingjing Guo, , , Liguo Song, , , Guang Chen*, , , Tao Zhang*, , and , Yunting Du*, 

Cerebral malaria (CM), a fatal neurological complication arising from Plasmodium falciparum (P. falciparum) infection, remains a significant global health challenge due to the inadequacy of current drugs and vaccines. Consequently, novel therapeutic strategies for CM are urgently needed. Recent research identifies platelets as pivotal in CM pathogenesis, significantly contributing to immunopathological damage and vascular blockage. Platelet-derived transforming growth factor (TGF)-β1 induces apoptosis in endothelial cells, fostering microangiopathy and potentially compromising blood–brain barrier integrity, thus provoking brain edema and inflammation. Notably, TGF-β1 concentrations vary markedly between systemic and local levels, with reduced TGF-β1 levels in mouse/human tissue and peripheral circulation correlating with CM severity. The primary regulatory mechanism involves isolated platelets interacting with infected red blood cells and brain endothelium, elevating local TGF-β1 production, and possibly harming brain endothelial cells. Future CM prevention or treatment strategies should focus on targeting TGF-β1, with an emphasis on brain-targeted drug delivery methods. Exosomes, as natural drug carriers, are extensively utilized for brain-specific delivery. Exosomes loaded with TGF-β1 antibodies, which were surface to enhancing brain-targeting ability, offer a promising therapeutic approach for CM.

脑型疟疾是由恶性疟原虫感染引起的一种致命的神经系统并发症,由于目前药物和疫苗不足,脑型疟疾仍然是一项重大的全球卫生挑战。因此,迫切需要新的CM治疗策略。最近的研究发现血小板在CM发病机制中起关键作用,在免疫病理损伤和血管阻塞中起重要作用。血小板衍生的转化生长因子(TGF)-β1诱导内皮细胞凋亡,促进微血管病变,并可能损害血脑屏障的完整性,从而引发脑水肿和炎症。值得注意的是,TGF-β1浓度在全身和局部水平之间存在显著差异,小鼠/人体组织和外周循环中TGF-β1水平的降低与CM严重程度相关。其主要调控机制涉及分离的血小板与受感染的红细胞和脑内皮相互作用,提高局部TGF-β1的产生,并可能损害脑内皮细胞。未来CM的预防或治疗策略应以TGF-β1为靶点,重点研究脑靶向给药方法。外泌体作为天然药物载体,被广泛用于脑特异性给药。负载TGF-β1抗体的外泌体可增强脑靶向能力,为CM的治疗提供了一种很有前景的方法。
{"title":"Platelet-Derived Transforming Growth Factor-β1: A New Hope for Cerebral Malaria Treatment","authors":"Shuangchun Liu,&nbsp;, ,&nbsp;Bingjing Guo,&nbsp;, ,&nbsp;Liguo Song,&nbsp;, ,&nbsp;Guang Chen*,&nbsp;, ,&nbsp;Tao Zhang*,&nbsp;, and ,&nbsp;Yunting Du*,&nbsp;","doi":"10.1021/acsinfecdis.5c00710","DOIUrl":"10.1021/acsinfecdis.5c00710","url":null,"abstract":"<p >Cerebral malaria (CM), a fatal neurological complication arising from <i><i>Plasmodium falciparum</i></i> (<i><i>P. falciparum</i></i>) infection, remains a significant global health challenge due to the inadequacy of current drugs and vaccines. Consequently, novel therapeutic strategies for CM are urgently needed. Recent research identifies platelets as pivotal in CM pathogenesis, significantly contributing to immunopathological damage and vascular blockage. Platelet-derived transforming growth factor (TGF)-β1 induces apoptosis in endothelial cells, fostering microangiopathy and potentially compromising blood–brain barrier integrity, thus provoking brain edema and inflammation. Notably, TGF-β1 concentrations vary markedly between systemic and local levels, with reduced TGF-β1 levels in mouse/human tissue and peripheral circulation correlating with CM severity. The primary regulatory mechanism involves isolated platelets interacting with infected red blood cells and brain endothelium, elevating local TGF-β1 production, and possibly harming brain endothelial cells. Future CM prevention or treatment strategies should focus on targeting TGF-β1, with an emphasis on brain-targeted drug delivery methods. Exosomes, as natural drug carriers, are extensively utilized for brain-specific delivery. Exosomes loaded with TGF-β1 antibodies, which were surface to enhancing brain-targeting ability, offer a promising therapeutic approach for CM.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"3021–3029"},"PeriodicalIF":3.8,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsinfecdis.5c00710","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145256890","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Mechanism of Action of Gepotidacin: Well-Balanced Dual-Targeting against Neisseria gonorrhoeae Gyrase and Topoisomerase IV in Cells and In Vitro Gepotidacin的作用机制:细胞内和体外对淋病奈瑟菌旋切酶和拓扑异构酶IV的平衡双重靶向。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-09 DOI: 10.1021/acsinfecdis.5c00497
Alexandria A. Oviatt, , , Jessica A. Collins, , , Chelsea A. Mann, , , Jianzhong Huang, , , Karen Mattern, , , Pan F. Chan*, , and , Neil Osheroff*, 

Gonorrhea, a sexually transmitted infection caused by Neisseria gonorrhoeae, remains a major public health concern. Fluoroquinolones, which target gyrase and topoisomerase IV, once served as first-line therapy for gonorrhea. However, rising target-mediated resistance led to their removal from treatment guidelines. In response to growing antibacterial resistance, gepotidacin, a first-in-class triazaacenaphthylene, offers a promising new treatment strategy. Gepotidacin targets gyrase and topoisomerase IV but is structurally and mechanistically different from fluoroquinolones. A phase III clinical trial of gepotidacin in the treatment of uncomplicated urogenital gonorrhea demonstrated a positive outcome. However, interactions of the drug with N. gonorrhoeae gyrase and topoisomerase IV have not been reported. Consequently, we determined the targeting of gepotidacin in N. gonorrhoeae cells and its effects on purified N. gonorrhoeae gyrase and topoisomerase IV. Although fluoroquinolones primarily target gyrase in Gram-negative bacteria, gepotidacin displayed well-balanced dual-targeting of both gyrase and topoisomerase IV in cultured cells. Reduced gepotidacin susceptibility required concurrent target-specific mutations in both enzymes, predicting a low propensity for developing target-mediated resistance. Consistent with this cellular dual-targeting, gepotidacin inhibited gyrase-catalyzed DNA supercoiling and topoisomerase IV-catalyzed DNA decatenation at similar low micromolar concentrations. Gepotidacin also induced primarily single-stranded DNA breaks mediated by both enzymes at comparable concentrations. Finally, mutations in aspartic acid residues predicted to mediate important gepotidacin-protein interactions in N. gonorrhoeae gyrase (GyrAD90) and topoisomerase IV (ParCD86) markedly diminished the activity of gepotidacin against the respective enzymes. Our findings differentiate gepotidacin targeting and mechanism from those of fluoroquinolones and highlight its potential to combat drug-resistant gonorrhea.

淋病是一种由淋病奈瑟菌引起的性传播感染,仍然是一个主要的公共卫生问题。以回转酶和拓扑异构酶IV为靶点的氟喹诺酮类药物曾作为淋病的一线治疗药物。然而,靶标介导耐药性的上升导致它们从治疗指南中被删除。为了应对日益增长的抗菌素耐药性,gepotidacin,一种一流的三氮杂苊,提供了一种有前途的新治疗策略。Gepotidacin以回转酶和拓扑异构酶IV为靶点,但在结构和机制上与氟喹诺酮类药物不同。gepotidacin治疗无并发症的泌尿生殖器淋病的III期临床试验显示出积极的结果。然而,药物与淋病奈瑟菌旋切酶和拓扑异构酶IV的相互作用尚未报道。因此,我们确定了gepotidacin在淋病奈瑟菌细胞中的靶向性及其对纯化淋病奈瑟菌旋转酶和拓扑异构酶IV的影响。尽管氟喹诺酮类药物主要靶向革兰氏阴性菌中的旋转酶,但在培养细胞中,gepotidacin表现出良好的旋转酶和拓扑异构酶IV的双重靶向性。降低gepotidacin敏感性需要两种酶同时发生靶标特异性突变,这预示着发生靶标介导的耐药的低倾向。与这种细胞双重靶向一致,gepotidacin在相似的低微摩尔浓度下抑制了回转酶催化的DNA超缠绕和拓扑异构酶iv催化的DNA十癸烯化。Gepotidacin也诱导了两种酶在相当浓度下介导的主要单链DNA断裂。最后,预测介导淋病奈瑟菌旋切酶(GyrAD90)和拓扑异构酶IV (ParCD86)中重要的gepotidacin-蛋白相互作用的天冬氨酸残基突变显著降低了gepotidacin对各自酶的活性。我们的研究结果区分了gepotidacin与氟喹诺酮类药物的靶向和机制,并强调了其对抗耐药淋病的潜力。
{"title":"Mechanism of Action of Gepotidacin: Well-Balanced Dual-Targeting against Neisseria gonorrhoeae Gyrase and Topoisomerase IV in Cells and In Vitro","authors":"Alexandria A. Oviatt,&nbsp;, ,&nbsp;Jessica A. Collins,&nbsp;, ,&nbsp;Chelsea A. Mann,&nbsp;, ,&nbsp;Jianzhong Huang,&nbsp;, ,&nbsp;Karen Mattern,&nbsp;, ,&nbsp;Pan F. Chan*,&nbsp;, and ,&nbsp;Neil Osheroff*,&nbsp;","doi":"10.1021/acsinfecdis.5c00497","DOIUrl":"10.1021/acsinfecdis.5c00497","url":null,"abstract":"<p >Gonorrhea, a sexually transmitted infection caused by <i>Neisseria gonorrhoeae</i>, remains a major public health concern. Fluoroquinolones, which target gyrase and topoisomerase IV, once served as first-line therapy for gonorrhea. However, rising target-mediated resistance led to their removal from treatment guidelines. In response to growing antibacterial resistance, gepotidacin, a first-in-class triazaacenaphthylene, offers a promising new treatment strategy. Gepotidacin targets gyrase and topoisomerase IV but is structurally and mechanistically different from fluoroquinolones. A phase III clinical trial of gepotidacin in the treatment of uncomplicated urogenital gonorrhea demonstrated a positive outcome. However, interactions of the drug with <i>N. gonorrhoeae</i> gyrase and topoisomerase IV have not been reported. Consequently, we determined the targeting of gepotidacin in <i>N. gonorrhoeae</i> cells and its effects on purified <i>N. gonorrhoeae</i> gyrase and topoisomerase IV. Although fluoroquinolones primarily target gyrase in Gram-negative bacteria, gepotidacin displayed well-balanced dual-targeting of both gyrase and topoisomerase IV in cultured cells. Reduced gepotidacin susceptibility required concurrent target-specific mutations in both enzymes, predicting a low propensity for developing target-mediated resistance. Consistent with this cellular dual-targeting, gepotidacin inhibited gyrase-catalyzed DNA supercoiling and topoisomerase IV-catalyzed DNA decatenation at similar low micromolar concentrations. Gepotidacin also induced primarily single-stranded DNA breaks mediated by both enzymes at comparable concentrations. Finally, mutations in aspartic acid residues predicted to mediate important gepotidacin-protein interactions in <i>N. gonorrhoeae</i> gyrase (GyrA<sup>D90</sup>) and topoisomerase IV (ParC<sup>D86</sup>) markedly diminished the activity of gepotidacin against the respective enzymes. Our findings differentiate gepotidacin targeting and mechanism from those of fluoroquinolones and highlight its potential to combat drug-resistant gonorrhea.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"3093–3105"},"PeriodicalIF":3.8,"publicationDate":"2025-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsinfecdis.5c00497","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145248947","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Benzodeazaoxaflavin Sirtuin Inhibitors Inhibit Schistosoma mansoni Sirt2 and Cause Phenotypic Changes and Lethality in Schistosomula and Adult Worm Stages 苯并地氮杂黄素Sirtuin抑制剂抑制曼氏血吸虫Sirt2并引起血吸虫和成虫期表型变化和致死性
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-07 DOI: 10.1021/acsinfecdis.5c00515
Roberto Gimmelli, , , Giuliana Papoff, , , Emanuele Fabbrizi, , , Michela Guida, , , Cristiana Lalli, , , Fulvio Saccoccia, , , Cécile Häberli, , , Jennifer Keiser, , , Daria Monaldi, , , Manfred Jung, , , Christophe Romier, , , Dante Rotili*, , , Antonello Mai*, , and , Giovina Ruberti*, 

Schistosomiasis, a neglected tropical disease caused by trematodes of Schistosoma genus, urgently requires new treatments due to praziquantel’s limited efficacy against juvenile worms as well as the threat of drug resistance. In this study, we evaluated a series of benzodeazaoxaflavin (BDF4)-based compounds as inhibitors of the parasite’s epigenetic enzyme SmSirt2. Three compounds, 79 (MC2346, MC2141, and MC2345), showed activity against both Liberian and Puerto Rican strains of Schistosoma mansoni. The compounds reduced schistosomula and adult worm pair viability, pairing, and egg production, with low cytotoxicity in mammalian cells. These effects were linked to histone H3 hyperacetylation and cytochrome c-mediated apoptosis, confirming SmSirt2 as a functional target. These findings support the development of SmSirt2 inhibitors as novel antischistosomal agents with therapeutic potential for both curative and preventive applications. Further in vivo studies are warranted to assess their pharmacokinetic and safety profiles.

血吸虫病是由血吸虫属吸虫引起的一种被忽视的热带病,由于吡喹酮对血吸虫幼虫的疗效有限以及存在耐药性威胁,迫切需要新的治疗方法。在这项研究中,我们评估了一系列基于苯并二氮杂黄素(BDF4)的化合物作为寄生虫表观遗传酶SmSirt2的抑制剂。其中化合物7-9 (MC2346、MC2141和MC2345)对利比里亚和波多黎各的曼氏血吸虫均有抑制作用。该化合物降低了血吸虫和成虫对活力、配对和产卵,对哺乳动物细胞具有低细胞毒性。这些作用与组蛋白H3超乙酰化和细胞色素c介导的细胞凋亡有关,证实SmSirt2是一个功能靶点。这些发现支持SmSirt2抑制剂作为具有治疗和预防应用潜力的新型抗血吸虫药物的开发。需要进一步的体内研究来评估它们的药代动力学和安全性。
{"title":"Benzodeazaoxaflavin Sirtuin Inhibitors Inhibit Schistosoma mansoni Sirt2 and Cause Phenotypic Changes and Lethality in Schistosomula and Adult Worm Stages","authors":"Roberto Gimmelli,&nbsp;, ,&nbsp;Giuliana Papoff,&nbsp;, ,&nbsp;Emanuele Fabbrizi,&nbsp;, ,&nbsp;Michela Guida,&nbsp;, ,&nbsp;Cristiana Lalli,&nbsp;, ,&nbsp;Fulvio Saccoccia,&nbsp;, ,&nbsp;Cécile Häberli,&nbsp;, ,&nbsp;Jennifer Keiser,&nbsp;, ,&nbsp;Daria Monaldi,&nbsp;, ,&nbsp;Manfred Jung,&nbsp;, ,&nbsp;Christophe Romier,&nbsp;, ,&nbsp;Dante Rotili*,&nbsp;, ,&nbsp;Antonello Mai*,&nbsp;, and ,&nbsp;Giovina Ruberti*,&nbsp;","doi":"10.1021/acsinfecdis.5c00515","DOIUrl":"10.1021/acsinfecdis.5c00515","url":null,"abstract":"<p >Schistosomiasis, a neglected tropical disease caused by trematodes of <i>Schistosoma</i> genus, urgently requires new treatments due to praziquantel’s limited efficacy against juvenile worms as well as the threat of drug resistance. In this study, we evaluated a series of benzodeazaoxaflavin (BDF4)-based compounds as inhibitors of the parasite’s epigenetic enzyme <i>Sm</i>Sirt2. Three compounds, <b>7</b>–<b>9</b> (MC2346, MC2141, and MC2345), showed activity against both Liberian and Puerto Rican strains of <i>Schistosoma mansoni</i>. The compounds reduced schistosomula and adult worm pair viability, pairing, and egg production, with low cytotoxicity in mammalian cells. These effects were linked to histone H3 hyperacetylation and cytochrome c-mediated apoptosis, confirming <i>Sm</i>Sirt2 as a functional target. These findings support the development of <i>Sm</i>Sirt2 inhibitors as novel antischistosomal agents with therapeutic potential for both curative and preventive applications. Further <i>in vivo</i> studies are warranted to assess their pharmacokinetic and safety profiles.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"3115–3127"},"PeriodicalIF":3.8,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsinfecdis.5c00515","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145243305","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting Mycolic Acid Biosynthesis with Cyclic Sulfamates: A New Strategy against Mycobacterium tuberculosis 环氨基甲酸酯靶向霉菌酸生物合成:抗结核分枝杆菌的新策略。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-07 DOI: 10.1021/acsinfecdis.5c00419
Priscila Cristina Bartolomeu Halicki, , , Jed H. Kim, , , Amory Francis Griffin, , , Daniel S. Rampon, , , Kindra L. Becker, , , Jessica C. Seeliger, , , Jennifer M. Schomaker*, , and , Kyle H. Rohde*, 

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is the deadliest infectious disease globally. Current TB regimens involving multidrug cocktails for ≥4 months with significant side effects leave much to be desired, with the first- and second-line drugs inhibiting only a limited number of bacterial targets. Thus, potent antimycobacterial agents with novel targets and mechanisms of action are urgently needed to overcome these limitations and the emergence of multidrug-resistant strains. To address this need, we tested a panel of cyclic sulfamate (CS) compounds and identified novel chemotypes that exhibit potent and highly selective activity against Mtb. Most importantly, multiple lines of evidence that include whole genome sequencing of spontaneous resistant mutants, cell-wall damage reporter assays, modeling of drug–target interactions, and cell wall lipid profiling support the hypothesis that these compounds kill Mtb by inhibiting KasA. KasA encodes a β-ketoacyl synthase, whose role in elongation of acyl-AcpM chains is required for the biosynthesis of mycolic acids. Despite being well validated as an essential enzyme, KasA is still an underexploited drug target in Mtb. In our work, the unchanged susceptibility of CS-resistant mutants to front-line TB drugs provides further evidence that the CS series of compounds acts via a novel mechanism of action. The knowledge gained in this study about structure–activity relationships will guide future medicinal chemistry optimization of the CS scaffold and evaluation of the in vivo efficacy of this chemical series. If successful, this novel chemotype may serve as the starting point for the development of alternative treatment options for TB.

由结核分枝杆菌(Mtb)引起的结核病是全球最致命的传染病。目前的结核病治疗方案包括持续≥4个月的多药鸡尾酒治疗,副作用显著,一线和二线药物仅能抑制有限数量的细菌靶点。因此,迫切需要具有新靶点和作用机制的强效抗真菌药物来克服这些限制和多药耐药菌株的出现。为了满足这一需求,我们测试了一组环氨基甲酸酯(CS)化合物,并确定了对结核分枝杆菌具有强效和高选择性活性的新型化学型。最重要的是,包括自发耐药突变体的全基因组测序、细胞壁损伤报告分析、药物-靶点相互作用建模和细胞壁脂质分析在内的多种证据支持这些化合物通过抑制KasA杀死Mtb的假设。KasA编码β-酮酰基合成酶,其在酰基- acpm链的延伸中的作用是霉菌酸生物合成所必需的。尽管KasA作为一种必需酶得到了很好的证实,但它仍然是结核分枝杆菌中一个未被充分利用的药物靶点。在我们的工作中,CS耐药突变体对一线结核病药物的易感性不变,进一步证明CS系列化合物通过一种新的作用机制起作用。本研究获得的结构-活性关系知识将指导未来CS支架的药物化学优化和该化学系列的体内疗效评估。如果成功,这种新的化学型可以作为开发结核病替代治疗方案的起点。
{"title":"Targeting Mycolic Acid Biosynthesis with Cyclic Sulfamates: A New Strategy against Mycobacterium tuberculosis","authors":"Priscila Cristina Bartolomeu Halicki,&nbsp;, ,&nbsp;Jed H. Kim,&nbsp;, ,&nbsp;Amory Francis Griffin,&nbsp;, ,&nbsp;Daniel S. Rampon,&nbsp;, ,&nbsp;Kindra L. Becker,&nbsp;, ,&nbsp;Jessica C. Seeliger,&nbsp;, ,&nbsp;Jennifer M. Schomaker*,&nbsp;, and ,&nbsp;Kyle H. Rohde*,&nbsp;","doi":"10.1021/acsinfecdis.5c00419","DOIUrl":"10.1021/acsinfecdis.5c00419","url":null,"abstract":"<p >Tuberculosis (TB), caused by <i>Mycobacterium tuberculosis</i> (<i>Mtb</i>), is the deadliest infectious disease globally. Current TB regimens involving multidrug cocktails for ≥4 months with significant side effects leave much to be desired, with the first- and second-line drugs inhibiting only a limited number of bacterial targets. Thus, potent antimycobacterial agents with novel targets and mechanisms of action are urgently needed to overcome these limitations and the emergence of multidrug-resistant strains. To address this need, we tested a panel of cyclic sulfamate (CS) compounds and identified novel chemotypes that exhibit potent and highly selective activity against <i>Mtb</i>. Most importantly, multiple lines of evidence that include whole genome sequencing of spontaneous resistant mutants, cell-wall damage reporter assays, modeling of drug–target interactions, and cell wall lipid profiling support the hypothesis that these compounds kill <i>Mtb</i> by inhibiting KasA. KasA encodes a β-ketoacyl synthase, whose role in elongation of acyl-AcpM chains is required for the biosynthesis of mycolic acids. Despite being well validated as an essential enzyme, KasA is still an underexploited drug target in <i>Mtb</i>. In our work, the unchanged susceptibility of CS-resistant mutants to front-line TB drugs provides further evidence that the CS series of compounds acts via a novel mechanism of action. The knowledge gained in this study about structure–activity relationships will guide future medicinal chemistry optimization of the CS scaffold and evaluation of the in vivo efficacy of this chemical series. If successful, this novel chemotype may serve as the starting point for the development of alternative treatment options for TB.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"3071–3084"},"PeriodicalIF":3.8,"publicationDate":"2025-10-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145237511","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Membrane-Permeable 5-Fluorodeoxyuridine Triphosphate Derivatives Inhibit the Proliferation of Plasmodium falciparum 膜透性5-氟脱氧尿苷三磷酸衍生物抑制恶性疟原虫的增殖。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-03 DOI: 10.1021/acsinfecdis.5c00544
Vella Nikolova, , , Karen Linnemannstöns, , , Marie-Elise Bendel, , , Marta Machado, , , Benedikt Ganter, , , Patricia Budimir, , , Michelle Vogts, , , Celine Fischer, , , Markus Ganter, , , Chris Meier, , and , Matthias Dobbelstein*, 

Malaria tropica remains a major global health challenge, raising the need for new therapeutic strategies against Plasmodium falciparum. While nucleoside analogues are effective against viruses and cancer, their use against P. falciparum is limited by the lack of nucleoside kinases in this species. To overcome this, we generated and tested cell-permeable derivatives of 5-fluorodeoxyuridine triphosphate (cpFdUTP) for antiparasitic activity in infected human red blood cells. cpFdUTP rapidly and potently inhibited the proliferation of P. falciparum, arresting development at the trophozoite-to-schizont transition by stalling DNA replication, as observed in a P. falciparum nuclear cycle sensor line. Although cpFdUTP also impaired the growth of human cells, supplementation with thymidine or cell-permeable deoxythymidine triphosphate (cpdTTP) selectively rescued human cells while maintaining parasite inhibition. This identifies a potential therapeutic window for cpFdUTP in combination with thymidine, outlining a novel approach for malaria treatment.

热带疟疾仍然是一项重大的全球卫生挑战,需要针对恶性疟原虫制定新的治疗策略。虽然核苷类似物对病毒和癌症有效,但由于该物种缺乏核苷激酶,它们对恶性疟原虫的使用受到限制。为了克服这个问题,我们生成并测试了5-氟脱氧尿苷三磷酸(cpFdUTP)的细胞渗透性衍生物在感染的人红细胞中的抗寄生虫活性。cpFdUTP快速有效地抑制恶性疟原虫的增殖,通过阻止DNA复制,阻止滋养体向分裂体转变的发育,在恶性疟原虫核周期传感器系中观察到。虽然cpFdUTP也会损害人类细胞的生长,但补充胸腺嘧啶或细胞渗透性脱氧胸腺嘧啶三磷酸(cpdTTP)选择性地拯救了人类细胞,同时保持了寄生虫的抑制作用。这确定了cpFdUTP与胸苷联合的潜在治疗窗口,概述了一种治疗疟疾的新方法。
{"title":"Membrane-Permeable 5-Fluorodeoxyuridine Triphosphate Derivatives Inhibit the Proliferation of Plasmodium falciparum","authors":"Vella Nikolova,&nbsp;, ,&nbsp;Karen Linnemannstöns,&nbsp;, ,&nbsp;Marie-Elise Bendel,&nbsp;, ,&nbsp;Marta Machado,&nbsp;, ,&nbsp;Benedikt Ganter,&nbsp;, ,&nbsp;Patricia Budimir,&nbsp;, ,&nbsp;Michelle Vogts,&nbsp;, ,&nbsp;Celine Fischer,&nbsp;, ,&nbsp;Markus Ganter,&nbsp;, ,&nbsp;Chris Meier,&nbsp;, and ,&nbsp;Matthias Dobbelstein*,&nbsp;","doi":"10.1021/acsinfecdis.5c00544","DOIUrl":"10.1021/acsinfecdis.5c00544","url":null,"abstract":"<p >Malaria tropica remains a major global health challenge, raising the need for new therapeutic strategies against <i>Plasmodium falciparum</i>. While nucleoside analogues are effective against viruses and cancer, their use against <i>P. falciparum</i> is limited by the lack of nucleoside kinases in this species. To overcome this, we generated and tested cell-permeable derivatives of 5-fluorodeoxyuridine triphosphate (cpFdUTP) for antiparasitic activity in infected human red blood cells. cpFdUTP rapidly and potently inhibited the proliferation of <i>P. falciparum</i>, arresting development at the trophozoite-to-schizont transition by stalling DNA replication, as observed in a <i>P. falciparum</i> nuclear cycle sensor line. Although cpFdUTP also impaired the growth of human cells, supplementation with thymidine or cell-permeable deoxythymidine triphosphate (cpdTTP) selectively rescued human cells while maintaining parasite inhibition. This identifies a potential therapeutic window for cpFdUTP in combination with thymidine, outlining a novel approach for malaria treatment.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 11","pages":"3138–3151"},"PeriodicalIF":3.8,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145211074","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Structural Exploitation of Cinnarizine Identified Novel Drug-Like Anthelmintic Agents Against Angiostrongylus cantonensis Cinnarizine鉴定的新型广州管圆线虫类药物驱虫剂的结构开发。
IF 3.8 2区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-10-01 DOI: 10.1021/acsinfecdis.5c00634
Bruna L. Lemes, , , Mariana A. Siegl-Breno, , , Mikaelly K. Silva-Nunes, , , Flavia B. Lopes, , , Aline S. Silva, , , Natalia E. P. Motta, , , Josué de Moraes*, , and , João Paulo S. Fernandes*, 

The impact of helminthiases on global health for both humans and animals and the limited efficacy of existing drugs against these infections reinforces the urgent need for novel anthelmintic agents. On this background, in previous work we identified cinnarizine, a first-generation antihistamine, as effective anthelmintic agent against Angiostrongylus cantonensis first-stage larvae (L1) in vitro. A. cantonensis worm is the causative agent of neuroangiostrongyliasis, a condition that leads to eosinophilic meningitis with no effective treatment to date. In the present work, modifications on cinnarizine structure were designed to improve its efficacy against the larvae but keeping its ability to cross the blood brain barrier allied to improvement in the drug-like and solubility profile. A set of 11 compounds were synthesized and tested against L1 larvae, showing EC50 values ranging from 9.3 to 4.2 μM. The most effective were also tested against infective third-stage larvae (L3), with EC50 18.1–8.6 μM. None of the compounds showed toxicity to both HaCat mammalian cells (at 500 μM) and Caenorhabditis elegans (at 1000 μM), indicating their high selective toxicity toward A. cantonensis. Structure–activity relationship analysis using molecular descriptors indicated that presence of two basic nitrogen atoms and balanced lipophilicity of compound 2b (EC50 L1 9.3 μM; L3 8.8 μM) played the role in the anthelmintic activity, and simplified compound 3a (EC50 L1 8.7 μM; L3 18.1 μM) represent a novel prototype for further modifications.

蠕虫病对全球人类和动物健康的影响以及现有药物对这些感染的有限疗效,加强了对新型驱虫药的迫切需要。在此背景下,在之前的工作中,我们发现了第一代抗组胺药肉桂嗪是一种有效的体外抗广州管圆线虫第一阶段幼虫(L1)的驱虫剂。广东蛔虫是神经血管圆线虫病的病原体,这种疾病可导致嗜酸性脑膜炎,迄今尚无有效治疗方法。在本研究中,通过对肉桂碱结构的修饰来提高其对幼虫的作用,同时保持其通过血脑屏障的能力,从而改善其药物样和溶解度。合成的11个化合物对L1幼虫的EC50值在9.3 ~ 4.2 μM之间。对感染的第三期幼虫(L3)的EC50为18.1 ~ 8.6 μM,效果最好。这些化合物对500 μM的HaCat哺乳动物细胞和1000 μM的秀丽隐杆线虫细胞均无毒性,表明它们对广东线虫具有高选择性毒性。基于分子描述符的构效关系分析表明,化合物2b (EC50 L1 9.3 μM; L3 8.8 μM)的两个碱性氮原子的存在和平衡的亲脂性对其驱虫活性起作用,简化后的化合物3a (EC50 L1 8.7 μM; L3 18.1 μM)为进一步修饰提供了新的原型。
{"title":"Structural Exploitation of Cinnarizine Identified Novel Drug-Like Anthelmintic Agents Against Angiostrongylus cantonensis","authors":"Bruna L. Lemes,&nbsp;, ,&nbsp;Mariana A. Siegl-Breno,&nbsp;, ,&nbsp;Mikaelly K. Silva-Nunes,&nbsp;, ,&nbsp;Flavia B. Lopes,&nbsp;, ,&nbsp;Aline S. Silva,&nbsp;, ,&nbsp;Natalia E. P. Motta,&nbsp;, ,&nbsp;Josué de Moraes*,&nbsp;, and ,&nbsp;João Paulo S. Fernandes*,&nbsp;","doi":"10.1021/acsinfecdis.5c00634","DOIUrl":"10.1021/acsinfecdis.5c00634","url":null,"abstract":"<p >The impact of helminthiases on global health for both humans and animals and the limited efficacy of existing drugs against these infections reinforces the urgent need for novel anthelmintic agents. On this background, in previous work we identified cinnarizine, a first-generation antihistamine, as effective anthelmintic agent against <i>Angiostrongylus cantonensis</i> first-stage larvae (L1) in vitro. <i>A. cantonensis</i> worm is the causative agent of neuroangiostrongyliasis, a condition that leads to eosinophilic meningitis with no effective treatment to date. In the present work, modifications on cinnarizine structure were designed to improve its efficacy against the larvae but keeping its ability to cross the blood brain barrier allied to improvement in the drug-like and solubility profile. A set of 11 compounds were synthesized and tested against L1 larvae, showing EC<sub>50</sub> values ranging from 9.3 to 4.2 μM. The most effective were also tested against infective third-stage larvae (L3), with EC<sub>50</sub> 18.1–8.6 μM. None of the compounds showed toxicity to both HaCat mammalian cells (at 500 μM) and <i>Caenorhabditis elegans</i> (at 1000 μM), indicating their high selective toxicity toward <i>A. cantonensis</i>. Structure–activity relationship analysis using molecular descriptors indicated that presence of two basic nitrogen atoms and balanced lipophilicity of compound <b>2b</b> (EC<sub>50</sub> L1 9.3 μM; L3 8.8 μM) played the role in the anthelmintic activity, and simplified compound <b>3a</b> (EC<sub>50</sub> L1 8.7 μM; L3 18.1 μM) represent a novel prototype for further modifications.</p>","PeriodicalId":17,"journal":{"name":"ACS Infectious Diseases","volume":"11 10","pages":"2871–2881"},"PeriodicalIF":3.8,"publicationDate":"2025-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.acs.org/doi/pdf/10.1021/acsinfecdis.5c00634","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145197389","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
ACS Infectious Diseases
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1