首页 > 最新文献

Journal of Advanced Research最新文献

英文 中文
Cardiac ATP production and contractility are favorably regulated by short-term S100A9 blockade after myocardial infarction
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-25 DOI: 10.1016/j.jare.2025.01.041
Raluca M. Boteanu, Viorel I. Suica, Elena Uyy, Luminita Ivan, Diana V. Uta, Razvan G. Mares, Maya Simionescu, Alexandru Schiopu, Felicia Antohe

Introduction

The infarcted heart is energetically compromised exhibiting a deficient production of adenosine triphosphate (ATP) and the ensuing impaired contractile function. Short-term blockade of the protein S100A9 improves cardiac performance in mice after myocardial infarction (MI). The implications upon ATP production during this process are not known.

Objectives

This study evaluates whether S100A9 blockade effects ATP synthesis and cardiac contractility in C57BL/6 mice at seven days post-MI.

Methods

Three experimental groups were used: (i) mice with MI, induced by permanent left coronary ligation, (ii) mice with MI, short-term treated with the S100A9 blocker ABR-238901, and (iii) sham (control) mice. After removing the left ventricle, mass spectrometry, pathway enrichment analysis, Western blot, RT-PCR and pharmacological network analysis were performed.

Results

A number of 600 differential abundance proteins (DAPs) was significantly altered by the S100A9 blocker in MI-treated mice compared with MI mice. Some of these proteins were associated with oxidative phosphorylation, citrate cycle (TCA), mitochondrial fatty acid beta-oxidation, glycolysis and cardiac muscle contraction pathways. In the ischemic ventricle, ABR-238901 treatment increased (1.8- to 38-fold) the abundance of proteins NDUFAB1, UQCRC1, HADHA, ACAA2, ALDOA, PKM1, DLD, DLAT, PDHX, ACO2, IDH3A, FH1, CKM, CKMT2, TNNC1, crucial for early cellular metabolic changes, ATP distribution and contractility. The cardiac level of ATP increased (1.8-fold, p < 0.05) in MI mice treated with ABR-238901 compared to MI mice. The network pharmacology analysis uncovered potential pharmacologic targets of ABR-238901 that may interact with DAPs related to ATP production and contractility.

Conclusion

Short-term S100A9 blockade effectively regulates the proteins implicated in ATP production and cardiac contractility post-MI, providing a framework for future cardiac energy metabolism studies.
{"title":"Cardiac ATP production and contractility are favorably regulated by short-term S100A9 blockade after myocardial infarction","authors":"Raluca M. Boteanu, Viorel I. Suica, Elena Uyy, Luminita Ivan, Diana V. Uta, Razvan G. Mares, Maya Simionescu, Alexandru Schiopu, Felicia Antohe","doi":"10.1016/j.jare.2025.01.041","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.041","url":null,"abstract":"<h3>Introduction</h3>The infarcted heart is energetically compromised exhibiting a deficient production of adenosine triphosphate (ATP) and the ensuing impaired contractile function. Short-term blockade of the protein S100A9 improves cardiac performance in mice after myocardial infarction (MI). The implications upon ATP production during this process are not known.<h3>Objectives</h3>This study evaluates whether S100A9 blockade effects ATP synthesis and cardiac contractility in C57BL/6 mice at seven days post-MI.<h3>Methods</h3>Three experimental groups were used: (i) mice with MI, induced by permanent left coronary ligation, (ii) mice with MI, short-term treated with the S100A9 blocker ABR-238901, and (iii) sham (control) mice. After removing the left ventricle, mass spectrometry, pathway enrichment analysis, Western blot, RT-PCR and pharmacological network analysis were performed.<h3>Results</h3>A number of 600 differential abundance proteins (DAPs) was significantly altered by the S100A9 blocker in MI-treated mice compared with MI mice. Some of these proteins were associated with oxidative phosphorylation, citrate cycle (TCA), mitochondrial fatty acid beta-oxidation, glycolysis and cardiac muscle contraction pathways. In the ischemic ventricle, ABR-238901 treatment increased (1.8- to 38-fold) the abundance of proteins NDUFAB1, UQCRC1, HADHA, ACAA2, ALDOA, PKM1, DLD, DLAT, PDHX, ACO2, IDH3A, FH1, CKM, CKMT2, TNNC1, crucial for early cellular metabolic changes, ATP distribution and contractility. The cardiac level of ATP increased (1.8-fold, p &lt; 0.05) in MI mice treated with ABR-238901 compared to MI mice. The network pharmacology analysis uncovered potential pharmacologic targets of ABR-238901 that may interact with DAPs related to ATP production and contractility.<h3>Conclusion</h3>Short-term S100A9 blockade effectively regulates the proteins implicated in ATP production and cardiac contractility post-MI, providing a framework for future cardiac energy metabolism studies.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"35 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143035071","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrative multi-omics analysis of autism spectrum disorder reveals unique microbial macromolecules interactions
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-25 DOI: 10.1016/j.jare.2025.01.036
Aya Osama, Ali Mostafa Anwar, Shahd Ezzeldin, Eman Ali Ahmed, Sebaey Mahgoub, Omneya Ibrahim, Sherif Abdelaziz Ibrahim, Ismail Abdelshafy Abdelhamid, Usama Bakry, Aya A. Diab, Ahmed A.Sayed, Sameh Magdeldin

Introduction

Gut microbiota alterations have been implicated in Autism Spectrum Disorder (ASD), yet the mechanisms linking these changes to ASD pathophysiology remain unclear.

Objectives

This study utilized a multi-omics approach to uncover mechanisms linking gut microbiota to ASD by examining microbial diversity, bacterial metaproteins, associated metabolic pathways and host proteome.

Methods

The gut microbiota of 30 children with severe ASD and 30 healthy controls was analyzed. Microbial diversity was assessed using 16S rRNA V3 and V4 sequencing. A novel metaproteomics pipeline identified bacterial proteins, while untargeted metabolomics explored altered metabolic pathways. Finally, multi-omics integration was employed to connect macromolecular changes to neurodevelopmental deficits.

Results

Children with ASD exhibited significant alterations in gut microbiota, including lower diversity and richness compared to controls. Tyzzerella was uniquely associated with the ASD group. Microbial network analysis revealed rewiring and reduced stability in ASD. Major metaproteins identified were produced by Bifidobacterium and Klebsiella (e.g., xylose isomerase and NADH peroxidase). Metabolomics profiling identified neurotransmitters (e.g., glutamate, DOPAC), lipids, and amino acids capable of crossing the blood–brain barrier, potentially contributing to neurodevelopmental and immune dysregulation. Host proteome analysis revealed altered proteins, including kallikrein (KLK1) and transthyretin (TTR), involved in neuroinflammation and immune regulation. Finally, multi-omics integration supported single-omics findings and reinforced the hypothesis that gut microbiota and their macromolecular products may contribute to ASD-associated symptoms.

Conclusions

The integration of multi-omics data provided critical evidence that alteration in gut microbiota and associated macromolecule production may play a role in ASD-related symptoms and co-morbidities. Key bacterial metaproteins and metabolites were identified as potential contributors to neurological and immune dysregulation in ASD, underscoring possible novel targets for therapeutic intervention.
{"title":"Integrative multi-omics analysis of autism spectrum disorder reveals unique microbial macromolecules interactions","authors":"Aya Osama, Ali Mostafa Anwar, Shahd Ezzeldin, Eman Ali Ahmed, Sebaey Mahgoub, Omneya Ibrahim, Sherif Abdelaziz Ibrahim, Ismail Abdelshafy Abdelhamid, Usama Bakry, Aya A. Diab, Ahmed A.Sayed, Sameh Magdeldin","doi":"10.1016/j.jare.2025.01.036","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.036","url":null,"abstract":"<h3>Introduction</h3>Gut microbiota alterations have been implicated in Autism Spectrum Disorder (ASD), yet the mechanisms linking these changes to ASD pathophysiology remain unclear.<h3>Objectives</h3>This study utilized a multi-omics approach to uncover mechanisms linking gut microbiota to ASD by examining microbial diversity, bacterial metaproteins, associated metabolic pathways and host proteome.<h3>Methods</h3>The gut microbiota of 30 children with severe ASD and 30 healthy controls was analyzed. Microbial diversity was assessed using 16S rRNA V3 and V4 sequencing. A novel metaproteomics pipeline identified bacterial proteins, while untargeted metabolomics explored altered metabolic pathways. Finally, multi-omics integration was employed to connect macromolecular changes to neurodevelopmental deficits.<h3>Results</h3>Children with ASD exhibited significant alterations in gut microbiota, including lower diversity and richness compared to controls. <em>Tyzzerella</em> was uniquely associated with the ASD group. Microbial network analysis revealed rewiring and reduced stability in ASD. Major metaproteins identified were produced by <em>Bifidobacterium</em> and <em>Klebsiella</em> (e.g., xylose isomerase and NADH peroxidase). Metabolomics profiling identified neurotransmitters (e.g., glutamate, DOPAC), lipids, and amino acids capable of crossing the blood–brain barrier, potentially contributing to neurodevelopmental and immune dysregulation. Host proteome analysis revealed altered proteins, including kallikrein (KLK1) and transthyretin (TTR), involved in neuroinflammation and immune regulation. Finally, multi-omics integration supported single-omics findings and reinforced the hypothesis that gut microbiota and their macromolecular products may contribute to ASD-associated symptoms.<h3>Conclusions</h3>The integration of multi-omics data provided critical evidence that alteration in gut microbiota and associated macromolecule production may play a role in ASD-related symptoms and co-morbidities. Key bacterial metaproteins and metabolites were identified as potential contributors to neurological and immune dysregulation in ASD, underscoring possible novel targets for therapeutic intervention.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"113 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031104","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
FOXS1, frequently inactivated by promoter methylation, inhibited colorectal cancer cell growth by promoting TGFBI degradation through autophagy-lysosome pathway
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-24 DOI: 10.1016/j.jare.2025.01.037
Yeye Kuang, Yijian Yu, Chan Wang, Hui Li, Yiru Zhou, Lijuan Pan, Yi Zhang, Xiaoqing Cheng, Zhinong Jiang, Xiaotong Hu

Introduction

Tumor suppressor gene (TSG) inactivation by epigenetic modifications contributes to the carcinogenesis and progression of colorectal cancer (CRC). Expression profiling and CpG methylomics revealed that a forkhead-box transcriptional factor, FOXS1, is downregulated and methylated in CRC.

Objectives

To assess the biological functions and underlying mechanisms of FOXS1 in colorectal cancer.

Methods

Public databases, semi-quantitative RT-PCR, immunohistochemistry, MSP, and BGS were used to analyze FOXS1 expression and promoter methylation in CRC. Stable FOXS1-overexpressing or knockdown cell lines were established. Cell growth, colony formation, flow cytometry, GFP-LC3 puncta detection, Ad-mCherry-GFP-LC3B, qPCR, in vivo subcutaneous tumor model, RNA-seq, western blotting, immunofluorescence, Co-IP assays, and protein stability analysis were performed to investigate the underlying molecular mechanisms of FOXS1.

Results

In CRC, FOXS1 was frequently downregulated due to promoter CpG methylation, acting as an independent prognostic marker. Moreover, FOXS1 exerts inhibitory effects on the growth of CRC cells in vitro and in vivo, while concurrently promoting CRC cell autophagy. Intriguingly, we found that FOXS1 interacted with transforming growth factor beta induced (TGFBI) and FOXS1 promoted TGFBI degradation through the autophagy–lysosome pathway rather than the ubiquitin–proteasome system. FOXS1 was also found to facilitate the interaction between TGFBI and lysosomal associated membrane protein 2A (LAMP2A), leading to the translocation of TGFBI into lysosomes for degradation. Additionally, FOXS1 regulates AKT phosphorylation and FOXO3a nuclear translocation, promoting the transcription of autophagy-related genes downstream of FOXO3a. Restoration of TGFBI expression reversed the suppressive effect exerted by FOXS1 on the growth of colorectal cancer cells.

Conclusion

FOXS1 functions as a tumor suppressor that is methylated in CRC and promotes the lysosomal degradation of TGFBI, regulates cell growth and promotes autophagy in CRC through the TGFBI/AKT/FOXO3a signaling pathway. These findings indicate that FOXS1 exhibits potential as a promising biomarker and therapeutic target for colorectal cancer.
{"title":"FOXS1, frequently inactivated by promoter methylation, inhibited colorectal cancer cell growth by promoting TGFBI degradation through autophagy-lysosome pathway","authors":"Yeye Kuang, Yijian Yu, Chan Wang, Hui Li, Yiru Zhou, Lijuan Pan, Yi Zhang, Xiaoqing Cheng, Zhinong Jiang, Xiaotong Hu","doi":"10.1016/j.jare.2025.01.037","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.037","url":null,"abstract":"<h3>Introduction</h3>Tumor suppressor gene (TSG) inactivation by epigenetic modifications contributes to the carcinogenesis and progression of colorectal cancer (CRC). Expression profiling and CpG methylomics revealed that a forkhead-box transcriptional factor, FOXS1, is downregulated and methylated in CRC.<h3>Objectives</h3>To assess the biological functions and underlying mechanisms of FOXS1 in colorectal cancer<strong>.</strong><h3>Methods</h3>Public databases, semi-quantitative RT-PCR, immunohistochemistry, MSP, and BGS were used to analyze FOXS1 expression and promoter methylation in CRC. Stable FOXS1-overexpressing or knockdown cell lines were established. Cell growth, colony formation, flow cytometry, GFP-LC3 puncta detection, Ad-mCherry-GFP-LC3B, qPCR, <em>in vivo</em> subcutaneous tumor model, RNA-seq, western blotting, immunofluorescence, Co-IP assays, and protein stability analysis were performed to investigate the underlying molecular mechanisms of FOXS1.<h3>Results</h3>In CRC, FOXS1 was frequently downregulated due to promoter CpG methylation, acting as an independent prognostic marker. Moreover, FOXS1 exerts inhibitory effects on the growth of CRC cells <em>in vitro</em> and <em>in vivo</em>, while concurrently promoting CRC cell autophagy. Intriguingly, we found that FOXS1 interacted with transforming growth factor beta induced (TGFBI) and FOXS1 promoted TGFBI degradation through the autophagy–lysosome pathway rather than the ubiquitin–proteasome system. FOXS1 was also found to facilitate the interaction between TGFBI and lysosomal associated membrane protein 2A (LAMP2A), leading to the translocation of TGFBI into lysosomes for degradation. Additionally, FOXS1 regulates AKT phosphorylation and FOXO3a nuclear translocation, promoting the transcription of autophagy-related genes downstream of FOXO3a. Restoration of TGFBI expression reversed the suppressive effect exerted by FOXS1 on the growth of colorectal cancer cells.<h3>Conclusion</h3>FOXS1 functions as a tumor suppressor that is methylated in CRC and promotes the lysosomal degradation of TGFBI, regulates cell growth and promotes autophagy in CRC through the TGFBI/AKT/FOXO3a signaling pathway. These findings indicate that FOXS1 exhibits potential as a promising biomarker and therapeutic target for colorectal cancer.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"58 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143031110","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Astragaloside IV ameliorates autism-like behaviors in BTBR mice by modulating Camk2n2-dependent OXPHOS and neurotransmission in the mPFC 黄芪甲苷通过调节camk2n2依赖性OXPHOS和mPFC中的神经传递改善BTBR小鼠的自闭症样行为
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-23 DOI: 10.1016/j.jare.2025.01.030
Mei Chen, Jiahui Shi, Tianyao Liu, Jiayin Liu, Yulong Liu, Jianghui Li, Yi Luo, Jing Luo, Xin Li, Hong Gong, Xiaotang Fan

Introduction

Autism spectrum disorder (ASD) represents a multifaceted set of neurodevelopmental conditions marked by social deficits and repetitive behaviors. Astragaloside IV (ASIV), a natural compound derived from the traditional Chinese herb Astragali Radix, exhibits robust neuroprotective effects. However, whether ASIV can ameliorate behavioral deficits in ASD remains unknown.

Objectives

This work aimed to determine the efficacy and molecular mechanisms of ASIV in ASD.

Methods

The autistic BTBR T + tf/J (BTBR) mice were used in this study. Behavioral tests were performed to assess the ASD-like phenotypes. The neurotransmitter levels and synaptic transmission were evaluated by high-performance liquid chromatography and whole-cell patch-clamp recordings, respectively. Molecular biological techniques, immunostaining, and RNA-sequencing (RNA-seq) were combined to uncover the underlying molecular mechanisms.

Results

Our study showed that both social impairment and repetitive behaviors were significantly improved after ASIV treatment in a dose-dependent manner in BTBR mice. The ASIV treatment normalized the neurotransmitter levels (GABA and glutamate) and their corresponding vesicular transporters (vGAT, vGLUT1) in the medial prefrontal cortex (mPFC). Furthermore, the excitation-inhibition imbalance in layer V of mPFC was reversed after ASIV administration. Mechanistically, bulk RNA-seq and PPI network analysis identified Camk2n2 as the crucial bridging gene regulating oxidative phosphorylation and neurotransmission. Camk2n2 overexpression in the mPFC abolished the beneficial effects of ASIV on autistic symptoms in BTBR mice via the Camk2/CREB pathway.

Conclusion

The evidence demonstrates that ASIV may become a promising treatment option for ASD and implies that targeting the Camk2n2/Camk2/CREB axis is warranted to investigate these ASD individuals further.
自闭症谱系障碍(ASD)代表了以社会缺陷和重复行为为特征的多方面的神经发育状况。黄芪甲苷(Astragaloside IV, ASIV)是一种从传统中药黄芪中提取的天然化合物,具有强大的神经保护作用。然而,asv是否可以改善ASD的行为缺陷仍然未知。目的探讨asv治疗ASD的疗效及分子机制。方法采用自闭BTBR T + tf/J (BTBR)小鼠。行为测试评估asd样表型。分别用高效液相色谱法和全细胞膜片钳法测定神经递质水平和突触传递。结合分子生物学技术,免疫染色和rna测序(RNA-seq)来揭示潜在的分子机制。结果BTBR小鼠经asv治疗后,社交障碍和重复行为均有剂量依赖性改善。asv治疗使内侧前额叶皮层(mPFC)的神经递质(GABA和谷氨酸)及其相应的囊泡转运蛋白(vGAT, vGLUT1)水平正常化。此外,注射asv后,mPFC第V层的兴奋-抑制不平衡被逆转。在机制上,大量RNA-seq和PPI网络分析发现Camk2n2是调节氧化磷酸化和神经传递的关键桥接基因。mPFC中的Camk2n2过表达通过Camk2/CREB途径消除了asv对BTBR小鼠自闭症症状的有益作用。结论有证据表明,asv可能成为ASD的一种有前景的治疗选择,并表明靶向Camk2n2/Camk2/CREB轴是进一步研究这些ASD个体的必要条件。
{"title":"Astragaloside IV ameliorates autism-like behaviors in BTBR mice by modulating Camk2n2-dependent OXPHOS and neurotransmission in the mPFC","authors":"Mei Chen, Jiahui Shi, Tianyao Liu, Jiayin Liu, Yulong Liu, Jianghui Li, Yi Luo, Jing Luo, Xin Li, Hong Gong, Xiaotang Fan","doi":"10.1016/j.jare.2025.01.030","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.030","url":null,"abstract":"<h3>Introduction</h3>Autism spectrum disorder (ASD) represents a multifaceted set of neurodevelopmental conditions marked by social deficits and repetitive behaviors. Astragaloside IV (ASIV), a natural compound derived from the traditional Chinese herb Astragali Radix, exhibits robust neuroprotective effects. However, whether ASIV can ameliorate behavioral deficits in ASD remains unknown.<h3>Objectives</h3>This work aimed to determine the efficacy and molecular mechanisms of ASIV in ASD.<h3>Methods</h3>The autistic BTBR T + tf/J (BTBR) mice were used in this study. Behavioral tests were performed to assess the ASD-like phenotypes. The neurotransmitter levels and synaptic transmission were evaluated by high-performance liquid chromatography and whole-cell patch-clamp recordings, respectively. Molecular biological techniques, immunostaining, and RNA-sequencing (RNA-seq) were combined to uncover the underlying molecular mechanisms.<h3>Results</h3>Our study showed that both social impairment and repetitive behaviors were significantly improved after ASIV treatment in a dose-dependent manner in BTBR mice. The ASIV treatment normalized the neurotransmitter levels (GABA and glutamate) and their corresponding vesicular transporters (vGAT, vGLUT1) in the medial prefrontal cortex (mPFC). Furthermore, the excitation-inhibition imbalance in layer V of mPFC was reversed after ASIV administration. Mechanistically, bulk RNA-seq and PPI network analysis identified Camk2n2 as the crucial bridging gene regulating oxidative phosphorylation and neurotransmission. Camk2n2 overexpression in the mPFC abolished the beneficial effects of ASIV on autistic symptoms in BTBR mice via the Camk2/CREB pathway.<h3>Conclusion</h3>The evidence demonstrates that ASIV may become a promising treatment option for ASD and implies that targeting the Camk2n2/Camk2/CREB axis is warranted to investigate these ASD individuals further.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"27 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143020922","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PIK3R3 regulates differentiation and senescence of periodontal ligament stem cells and mitigates age-related alveolar bone loss by modulating FOXO1 expression
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-23 DOI: 10.1016/j.jare.2025.01.031
Xuenan Liu, Donghao Wei, Feilong Wang, Fanyu Yan, Xiao Zhang, Yongsheng Zhou, Ping Zhang, Yunsong Liu

Introduction

Periodontal diseases are prevalent among middle-aged and elderly individuals. There’s still no satisfactory solution for tooth loss caused by periodontal diseases. Human periodontal ligament stem cells (hPDLSCs) is a distinctive subgroup of mesenchymal stem cells, which play a crucial role in periodontal supportive tissues, but their application value hasn’t been fully explored yet. As a regulatory subunit of PI3K, PIK3R3′s role in stem cell regulation remains poorly comprehended.

Objectives

This study aims to explore the regulatory effect of PIK3R3 on differentiation and senescence of hPDLSCs and the underlying mechanism, as well as whether overexpression of PIK3R3 mitigate alveolar bone loss in aged rats.

Methods

Human PDLSC lines with both PIK3R3 knockdown and overexpression are established. Osteogenic, adipogenic, chondrogenic and senescent induction are used to test the effect of PIK3R3 on senescence in vitro. Model of alveolar bone loss in aged mice is used to reveal the effect of PIK3R3 in vivo. FOXO1 siRNA is used for mechanism exploration.

Results

Knockdown of PIK3R3 inhibits the mRNA and protein expression of markers in osteogenic, adipogenic, and chondrogenic differentiation of hPDLSCs but promotes in vitro senescence of hPDLSCs, including cell proliferation, senescence markers expression, telomerase density and reactive oxygen species. Overexpression of PIK3R3 has the opposite effect. Furthermore, the result of Micro-CT and tissue section shows that overexpression of PIK3R3 in elder rats mitigates alveolar bone loss. Mechanistically, PIK3R3 regulates senescence of hPDLSCs through modulating FOXO1 expression. Expression of FOXO1 is altered when PIK3R3 is knocked down or overexpressed in senescent medium. Knockdown of FOXO1 promotes senescence of hPDLSCs and the senescence promoting effect of knocking down PIK3R3 is weakened when FOXO1 is highly expressed.

Conclusion

These findings indicate that PIK3R3 modulates senescence of MSCs by regulating FOXO1 expression and shows promise as a therapeutic target for mitigating age-related alveolar bone loss.
{"title":"PIK3R3 regulates differentiation and senescence of periodontal ligament stem cells and mitigates age-related alveolar bone loss by modulating FOXO1 expression","authors":"Xuenan Liu, Donghao Wei, Feilong Wang, Fanyu Yan, Xiao Zhang, Yongsheng Zhou, Ping Zhang, Yunsong Liu","doi":"10.1016/j.jare.2025.01.031","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.031","url":null,"abstract":"<h3>Introduction</h3>Periodontal diseases are prevalent among middle-aged and elderly individuals. There’s still no satisfactory solution for tooth loss caused by periodontal diseases. Human periodontal ligament stem cells (hPDLSCs) is a distinctive subgroup of mesenchymal stem cells, which play a crucial role in periodontal supportive tissues, but their application value hasn’t been fully explored yet. As a regulatory subunit of PI3K, PIK3R3′s role in stem cell regulation remains poorly comprehended.<h3>Objectives</h3>This study aims to explore the regulatory effect of PIK3R3 on differentiation and senescence of hPDLSCs and the underlying mechanism, as well as whether overexpression of PIK3R3 mitigate alveolar bone loss in aged rats.<h3>Methods</h3>Human PDLSC lines with both <em>PIK3R3</em> knockdown and overexpression are established. Osteogenic, adipogenic, chondrogenic and senescent induction are used to test the effect of PIK3R3 on senescence <em>in vitro</em>. Model of alveolar bone loss in aged mice is used to reveal the effect of PIK3R3 <em>in vivo</em>. FOXO1 siRNA is used for mechanism exploration.<h3>Results</h3>Knockdown of <em>PIK3R3</em> inhibits the mRNA and protein expression of markers in osteogenic, adipogenic, and chondrogenic differentiation of hPDLSCs but promotes <em>in vitro</em> senescence of hPDLSCs, including cell proliferation, senescence markers expression, telomerase density and reactive oxygen species. Overexpression of <em>PIK3R3</em> has the opposite effect. Furthermore, the result of Micro-CT and tissue section shows that overexpression of <em>PIK3R3</em> in elder rats mitigates alveolar bone loss. Mechanistically, PIK3R3 regulates senescence of hPDLSCs through modulating FOXO1 expression. Expression of FOXO1 is altered when <em>PIK3R3</em> is knocked down or overexpressed in senescent medium. Knockdown of FOXO1 promotes senescence of hPDLSCs and the senescence promoting effect of knocking down <em>PIK3R3</em> is weakened when FOXO1 is highly expressed.<h3>Conclusion</h3>These findings indicate that PIK3R3 modulates senescence of MSCs by regulating FOXO1 expression and shows promise as a therapeutic target for mitigating age-related alveolar bone loss.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"13 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143026617","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phospholipase C epsilon 1 as a therapeutic target in cardiovascular diseases 磷脂酶cepsilon 1作为心血管疾病的治疗靶点
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-22 DOI: 10.1016/j.jare.2025.01.032
Jie Wang, Ting Gao, Dongmei Zhang, Yufeng Tang, Junlian Gu

Background

Phospholipase C epsilon 1 (PLCε1) can hydrolyze phosphatidylinositol-4,5-bisphosphate and phosphatidylinositol-4-phosphate at the plasma membrane and perinuclear membrane in the cardiovascular system, producing lipid-derived second messengers. These messengers are considered prominent triggers for various signal transduction processes. Notably, diverse cardiac phenotypes have been observed in cardiac-specific and global Plce1 knockout mice under conditions of pathological stress. It is well established that the cardiac-specific Plce1 knockout confers cardioprotective benefits. Therefore, the development of tissue/cell-specific targeting approaches is critical for advancing therapeutic interventions.

Aim of Review:

This review aims to distill the foundational biology and functional significance of PLCε1 in cardiovascular diseases, as well as to explore potential avenues for research and the development of novel therapeutic strategies targeting PLCε1.

Key Scientific Concepts of Review:

Cardiovascular diseases remain the leading cause of morbidity and mortality worldwide, with incidence rates escalating annually. A comprehensive understanding of the multifaceted role of PLCε1 is essential for enhancing the diagnosis, management, and prognostic assessment of patients suffering from cardiovascular diseases.
磷脂酶Cε1 (PLCε1)在心血管系统的质膜和核周膜上水解磷脂酰肌醇-4,5-二磷酸和磷脂酰肌醇-4-磷酸,产生脂质衍生的第二信使。这些信使被认为是各种信号转导过程的突出触发器。值得注意的是,在病理应激条件下,在心脏特异性和全局Plce1敲除小鼠中观察到不同的心脏表型。众所周知,心脏特异性Plce1敲除具有心脏保护作用。因此,组织/细胞特异性靶向方法的发展对于推进治疗干预至关重要。综述目的:本文旨在总结PLCε1在心血管疾病中的基础生物学和功能意义,并探索针对PLCε1的研究和开发新的治疗策略的潜在途径。综述的主要科学概念:心血管疾病仍然是世界范围内发病率和死亡率的主要原因,发病率每年都在上升。全面了解PLCε1的多方面作用对于加强心血管疾病患者的诊断、管理和预后评估至关重要。
{"title":"Phospholipase C epsilon 1 as a therapeutic target in cardiovascular diseases","authors":"Jie Wang, Ting Gao, Dongmei Zhang, Yufeng Tang, Junlian Gu","doi":"10.1016/j.jare.2025.01.032","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.032","url":null,"abstract":"<h3>Background</h3>Phospholipase C epsilon 1 (PLCε1) can hydrolyze phosphatidylinositol-4,5-bisphosphate and phosphatidylinositol-4-phosphate at the plasma membrane and perinuclear membrane in the cardiovascular system, producing lipid-derived second messengers. These messengers are considered prominent triggers for various signal transduction processes. Notably, diverse cardiac phenotypes have been observed in cardiac-specific and global <em>Plce1</em> knockout mice under conditions of pathological stress. It is well established that the cardiac-specific <em>Plce1</em> knockout confers cardioprotective benefits. Therefore, the development of tissue/cell-specific targeting approaches is critical for advancing therapeutic interventions.<h3>Aim of Review:</h3>This review aims to distill the foundational biology and functional significance of PLCε1 in cardiovascular diseases, as well as to explore potential avenues for research and the development of novel therapeutic strategies targeting PLCε1.<h3>Key Scientific Concepts of Review:</h3>Cardiovascular diseases remain the leading cause of morbidity and mortality worldwide, with incidence rates escalating annually. A comprehensive understanding of the multifaceted role of PLCε1 is essential for enhancing the diagnosis, management, and prognostic assessment of patients suffering from cardiovascular diseases.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"18 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142992625","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Integrated multi-omics reveals the Bacillus amyloliquefaciens BA40 against Clostridium perfringens infection in weaned piglets 综合多组学研究发现解淀粉芽孢杆菌BA40对断奶仔猪产气荚膜梭菌感染具有抑制作用
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-22 DOI: 10.1016/j.jare.2025.01.033
Zipeng Jiang, Weifa Su, Mingzhi Yang, Jie Fu, Tao Gong, Wentao Li, Chaoyue Wen, Xinxia Wang, Fengqin Wang, Mingliang Jin, Yizhen Wang, Zeqing Lu

Introduction

Clostridium perfringens (C. perfringens) can cause necrotic enteritis and higher mortality rates in piglets, by impairing the intestinal barrier function. Bacillus amyloliquefaciens 40 (BA40) has showed potential ability to reduce C. perfringens infections, but the mechanisms responsible for its effectiveness remain unclear.

Objectives

This study aims to evaluate the impact of BA40 on inflammation induced by C. perfringens and to explain the mechanisms underlying its therapeutic effects. We aim to show how BA40 can bolster piglet health by strengthening the intestinal barrier and regulating immune responses.

Methods

We used piglets and cellular models, alongside microbiomics, metabolomic, and transcriptomic analyses, to investigate BA40′s impact on C. perfringens-induced inflammation. A model of C. perfringens infection was constructed using piglets and cells to investigate the effect of BA40 on its phenotype. Microbiomics, metabolomics, and transcriptomics analyses were subsequently used to investigate the mechanisms of protection and immune response to BA40 on the intestinal barrier of piglets.

Results

Our study revealed significant improvements in piglet health following BA40 administration. Notably, BA40 strengthened the intestinal mucosal barrier and mitigated the inflammatory response triggered by C. perfringens BA40 decreased harmful bacteria and increased beneficial bacteria. Metabolite profiles improved, showing a reduction in harmful substances. Transscriptomics analysis indicated BA40′s role in TNF/NF-κB signaling pathway, hinting at its ability to regulate immune responses and reduce intestinal inflammation. Cellular assays further confirmed BA40′s capacity to diminish inflammatory cytokine release and encourage the differentiation of anti-inflammatory macrophages.

Conclusion

Datasets from the present study demonstrate that BA40 modulates gut microbes and metabolites, inhibits inflammation-related signaling pathways, and maintains gut barrier function. Our findings not only deepen our understanding of the therapeutic capacity of BA40 but also provide a theoretical foundation for the development of probiotics and alternative therapies aimed at improving piglet gut health.
产气荚膜梭菌(C. perfringens)可通过损害肠道屏障功能引起仔猪坏死性肠炎和较高的死亡率。解淀粉芽孢杆菌40 (BA40)已显示出减少产气荚膜杆菌感染的潜在能力,但其有效性的机制尚不清楚。目的探讨BA40对产气荚膜原梭菌所致炎症的影响,并探讨其治疗作用的机制。我们的目标是展示BA40如何通过加强肠道屏障和调节免疫反应来促进仔猪健康。方法采用仔猪和细胞模型,结合微生物组学、代谢组学和转录组学分析,研究BA40对产气荚膜梭菌诱导炎症的影响。采用仔猪和细胞构建产气荚膜原梭菌感染模型,探讨BA40对其表型的影响。随后采用微生物组学、代谢组学和转录组学分析来研究BA40对仔猪肠道屏障的保护和免疫反应机制。结果本研究显示,饲喂BA40后仔猪健康状况有显著改善。值得注意的是,BA40增强了肠黏膜屏障,减轻了产气荚膜原梭菌引发的炎症反应,减少了有害菌,增加了有益菌。代谢物谱得到改善,有害物质减少。转录组学分析表明BA40参与TNF/NF-κB信号通路,提示其具有调节免疫反应和减轻肠道炎症的能力。细胞实验进一步证实BA40具有减少炎症细胞因子释放和促进抗炎巨噬细胞分化的能力。结论本研究数据表明,BA40可调节肠道微生物和代谢物,抑制炎症相关信号通路,维持肠道屏障功能。我们的研究结果不仅加深了我们对BA40治疗能力的认识,而且为开发益生菌和旨在改善仔猪肠道健康的替代疗法提供了理论基础。
{"title":"Integrated multi-omics reveals the Bacillus amyloliquefaciens BA40 against Clostridium perfringens infection in weaned piglets","authors":"Zipeng Jiang, Weifa Su, Mingzhi Yang, Jie Fu, Tao Gong, Wentao Li, Chaoyue Wen, Xinxia Wang, Fengqin Wang, Mingliang Jin, Yizhen Wang, Zeqing Lu","doi":"10.1016/j.jare.2025.01.033","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.033","url":null,"abstract":"<h3>Introduction</h3><em>Clostridium perfringens</em> (<em>C. perfringens</em>) can cause necrotic enteritis and higher mortality rates in piglets, by impairing the intestinal barrier function. <em>Bacillus amyloliquefaciens</em> 40 (BA40) has showed potential ability to reduce <em>C. perfringens</em> infections, but the mechanisms responsible for its effectiveness remain unclear.<h3>Objectives</h3>This study aims to evaluate the impact of BA40 on inflammation induced by <em>C. perfringens</em> and to explain the mechanisms underlying its therapeutic effects. We aim to show how BA40 can bolster piglet health by strengthening the intestinal barrier and regulating immune responses.<h3>Methods</h3>We used piglets and cellular models, alongside microbiomics, metabolomic, and transcriptomic analyses, to investigate BA40′s impact on <em>C. perfringens</em>-induced inflammation. A model of <em>C. perfringens</em> infection was constructed using piglets and cells to investigate the effect of BA40 on its phenotype. Microbiomics, metabolomics, and transcriptomics analyses were subsequently used to investigate the mechanisms of protection and immune response to BA40 on the intestinal barrier of piglets.<h3>Results</h3>Our study revealed significant improvements in piglet health following BA40 administration. Notably, BA40 strengthened the intestinal mucosal barrier and mitigated the inflammatory response triggered by <em>C. perfringens</em> BA40 decreased harmful bacteria and increased beneficial bacteria. Metabolite profiles improved, showing a reduction in harmful substances. Transscriptomics analysis indicated BA40′s role in TNF/NF-κB signaling pathway, hinting at its ability to regulate immune responses and reduce intestinal inflammation. Cellular assays further confirmed BA40′s capacity to diminish inflammatory cytokine release and encourage the differentiation of anti-inflammatory macrophages.<h3>Conclusion</h3>Datasets from the present study demonstrate that BA40 modulates gut microbes and metabolites, inhibits inflammation-related signaling pathways, and maintains gut barrier function. Our findings not only deepen our understanding of the therapeutic capacity of BA40 but also provide a theoretical foundation for the development of probiotics and alternative therapies aimed at improving piglet gut health.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"33 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143020923","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Molecular and cellular morphology of placenta unveils new mechanisms of reproductive immunology 胎盘的分子和细胞形态学揭示了生殖免疫学的新机制
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-20 DOI: 10.1016/j.jare.2025.01.025
Penghao Li, Liting Zeng, Xiaomiao Yan, Ziqi Zhu, Qiaoxiu Gu, Xuqing He, Sujuan Zhang, Rurong Mao, Jingliang Xu, Fengshan Xie, Hui Wang, Ziteng Li, Jing Shu, Weifeng Zhang, Yulin Sha, Jin Huang, Meng Su, Qu Zheng, Jian Ma, Xiaolin Zhou, Jiang Gu

Introduction

Despite of numerous studies of the placenta, some molecular and cellular characteristics, particularly the relationship among different cell types, have not been well understood. We aim to investigate the basic and intricate details of cellular and molecular elements in early and late phase placentas to gain better understanding of the immune regulation of human reproductive process.

Methods

A novel combination of techniques of spatial transcriptomics(ST), multiple immunohistochemistry, and a dual labeling combining immunohistochemistry and (fluorescence in situ hybridization) FISH on normal and ectopic pregnancy and animal models was employed to investigate the placenta at tissue, cell, protein and molecular levels and to trace the fetal and maternal origin of every cell in early and late placentas.

Results

Original discoveries include early expression of immune checkpoint proteins in embryo trophoblasts even before implantation. The detailed distributional relationships among different cell types of fetal and maternal origins in placenta and decidua indicate an immune rejection of the mother towards the fetus and this was counterbalanced by immune inhibitory proteins and blocking antibody Immunoglobulin G4 (IgG4) at the junction between the fetus and the mother. In contrary to common believe, we found that vascular endothelial and glandular epithelial cells in the decidua remain maternal in origin and were not replaced by fetal cells. At term placenta, fetal immune cells infiltrated into the maternal side of the decidus and vice versa indicating a possible immune reaction between fetal and maternal immune systems and suggesting a possible immune mechanism for trigger of parturition. The ability of trophoblasts to create an immune suppressed environment was also supported by findings in ectopic pregnancy and the animal models.

Conclusion

The findings indicate a fetus-driven mechanism of immune balance involving both cellular and humoral immunity in human reproduction.
尽管对胎盘进行了大量的研究,但一些分子和细胞特征,特别是不同细胞类型之间的关系,尚未得到很好的理解。我们的目的是研究早期和晚期胎盘细胞和分子元件的基本和复杂的细节,以更好地了解人类生殖过程的免疫调节。方法采用空间转录组学(ST)、多重免疫组织化学技术、免疫组织化学和荧光原位杂交(FISH)双标记技术结合正常妊娠、异位妊娠和动物模型,从组织、细胞、蛋白和分子水平对胎盘进行研究,追踪胎盘早期和晚期各细胞的胎母来源。结果在胚胎着床前,免疫检查点蛋白在胚胎滋养细胞中有早期表达。胎盘和蜕膜中胎儿和母体不同类型细胞之间的详细分布关系表明,母体对胎儿存在免疫排斥,而这种排斥被胎儿和母体交界处的免疫抑制蛋白和阻断抗体免疫球蛋白G4 (IgG4)所抵消。与通常的看法相反,我们发现蜕膜中的血管内皮细胞和腺上皮细胞仍然来自母体,而不是被胎儿细胞所取代。足月胎盘时,胎儿免疫细胞浸润到母体的蜕膜中,反之亦然,这表明胎儿和母体免疫系统之间可能存在免疫反应,可能存在触发分娩的免疫机制。滋养层细胞创造免疫抑制环境的能力也得到异位妊娠和动物模型研究结果的支持。结论胎儿驱动的免疫平衡机制涉及人体生殖过程中的细胞免疫和体液免疫。
{"title":"Molecular and cellular morphology of placenta unveils new mechanisms of reproductive immunology","authors":"Penghao Li, Liting Zeng, Xiaomiao Yan, Ziqi Zhu, Qiaoxiu Gu, Xuqing He, Sujuan Zhang, Rurong Mao, Jingliang Xu, Fengshan Xie, Hui Wang, Ziteng Li, Jing Shu, Weifeng Zhang, Yulin Sha, Jin Huang, Meng Su, Qu Zheng, Jian Ma, Xiaolin Zhou, Jiang Gu","doi":"10.1016/j.jare.2025.01.025","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.025","url":null,"abstract":"<h3>Introduction</h3>Despite of numerous studies of the placenta, some molecular and cellular characteristics, particularly the relationship among different cell types, have not been well understood. We aim to investigate the basic and intricate details of cellular and molecular elements in early and late phase placentas to gain better understanding of the immune regulation of human reproductive process.<h3>Methods</h3>A novel combination of techniques of spatial transcriptomics(ST), multiple immunohistochemistry, and a dual labeling combining immunohistochemistry and (fluorescence in situ hybridization) FISH on normal and ectopic pregnancy and animal models was employed to investigate the placenta at tissue, cell, protein and molecular levels and to trace the fetal and maternal origin of every cell in early and late placentas.<h3>Results</h3>Original discoveries include early expression of immune checkpoint proteins in embryo trophoblasts even before implantation. The detailed distributional relationships among different cell types of fetal and maternal origins in placenta and decidua indicate an immune rejection of the mother towards the fetus and this was counterbalanced by immune inhibitory proteins and blocking antibody Immunoglobulin G4 (IgG4) at the junction between the fetus and the mother. In contrary to common believe, we found that vascular endothelial and glandular epithelial cells in the decidua remain maternal in origin and were not replaced by fetal cells. At term placenta, fetal immune cells infiltrated into the maternal side of the decidus and vice versa indicating a possible immune reaction between fetal and maternal immune systems and suggesting a possible immune mechanism for trigger of parturition. The ability of trophoblasts to create an immune suppressed environment was also supported by findings in ectopic pregnancy and the animal models.<h3>Conclusion</h3>The findings indicate a fetus-driven mechanism of immune balance involving both cellular and humoral immunity in human reproduction.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"49 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142991052","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Development of novel broad-spectrum amphipathic antimicrobial peptides against multidrug-resistant bacteria through a rational combination strategy 通过合理组合策略开发抗多药耐药细菌的新型广谱两性抗菌肽
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-18 DOI: 10.1016/j.jare.2025.01.029
Jing Zhang, Liang Luan, Youdong Xu, Shuyuan Jiang, Wenpeng Zhang, Long Tian, Weifeng Ye, Jiaqi Han, Changhao Zhang, Taoran Wang, Qingbing Meng

Introduction

In recent years, cationic amphipathic antimicrobial peptides (AMPs) have shown great promise in combating antibiotic resistance on account of their distinctive membrane-disruptive mechanism. However, the clinical application of AMPs is restricted by their unsatisfactory stability and safety. Although attempts have been made to improve the stability and safety of AMPs, many of them are accompanied by a decline in their antimicrobial activity and bacterial selectivity.

Objectives

To develop AMPs with excellent and balanced antimicrobial activity, stability, and safety using a combination strategy.

Methods

A series of sC184b-derived peptide analogues were designed by a combination strategy of subtly adjusting the charges, hydrophobic properties, and introducing specific unnatural amino acids in a well-balanced manner. The antimicrobial activity, cytotoxicity, hemolytic activity, stability, anti-biofilm activity, mechanism of action, synergistic effects, in vivo efficacy, and pharmacokinetics of the analogues were evaluated.

Results

Among these analogues, P-α-02-B stood out for its broad-spectrum and potent antimicrobial activity, anti-biofilm activity, desirable bacterial selectivity, high plasma stability, and synergistic effect with antibiotic levofloxacin. P-α-02-B exhibited strong membrane disturbance effect, which could be explained by its rigid α-helical structure revealed by molecular dynamics simulations. More importantly, P-α-02-B showed favorable therapeutic efficacy in vivo, whether used alone or in combination with levofloxacin.

Conclusion

P-α-02-B is a promising antimicrobial agent for MDR bacterial infections, demonstrating the effectiveness of the combination strategy for AMP development.
近年来,阳离子两性抗菌肽(AMPs)由于其独特的膜破坏机制在对抗抗生素耐药性方面显示出巨大的前景。然而,由于其稳定性和安全性不理想,抗菌肽的临床应用受到限制。尽管人们已经尝试提高抗菌肽的稳定性和安全性,但它们中的许多都伴随着抗菌活性和细菌选择性的下降。目的采用联合用药策略开发抗菌活性、稳定性和安全性均良好的抗菌肽。方法采用巧妙调整电荷、疏水性和平衡引入特定非天然氨基酸的组合策略,设计了一系列源自sc184b的肽类似物。对其抑菌活性、细胞毒性、溶血活性、稳定性、抗生物膜活性、作用机制、协同效应、体内药效和药代动力学进行了评价。结果P-α-02-B具有广谱有效的抗菌活性、抗生物膜活性、良好的细菌选择性、较高的血浆稳定性和与抗生素左氧氟沙星的协同作用。P-α-02-B表现出较强的膜扰动效应,分子动力学模拟表明其具有刚性的α-螺旋结构。更重要的是,无论是单独使用还是与左氧氟沙星联合使用,P-α-02-B在体内均显示出良好的治疗效果。结论p -α-02-B是一种很有前景的耐多药细菌感染抗菌药物,表明联合策略开发AMP是有效的。
{"title":"Development of novel broad-spectrum amphipathic antimicrobial peptides against multidrug-resistant bacteria through a rational combination strategy","authors":"Jing Zhang, Liang Luan, Youdong Xu, Shuyuan Jiang, Wenpeng Zhang, Long Tian, Weifeng Ye, Jiaqi Han, Changhao Zhang, Taoran Wang, Qingbing Meng","doi":"10.1016/j.jare.2025.01.029","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.029","url":null,"abstract":"<h3>Introduction</h3>In recent years, cationic amphipathic antimicrobial peptides (AMPs) have shown great promise in combating antibiotic resistance on account of their distinctive membrane-disruptive mechanism. However, the clinical application of AMPs is restricted by their unsatisfactory stability and safety. Although attempts have been made to improve the stability and safety of AMPs, many of them are accompanied by a decline in their antimicrobial activity and bacterial selectivity.<h3>Objectives</h3>To develop AMPs with excellent and balanced antimicrobial activity, stability, and safety using a combination strategy.<h3>Methods</h3>A series of sC18<sub>4b</sub>-derived peptide analogues were designed by a combination strategy of subtly adjusting the charges, hydrophobic properties, and introducing specific unnatural amino acids in a well-balanced manner. The antimicrobial activity, cytotoxicity, hemolytic activity, stability, anti-biofilm activity, mechanism of action, synergistic effects, <em>in vivo</em> efficacy, and pharmacokinetics of the analogues were evaluated.<h3>Results</h3>Among these analogues, P-α-02-B stood out for its broad-spectrum and potent antimicrobial activity, anti-biofilm activity, desirable bacterial selectivity, high plasma stability, and synergistic effect with antibiotic levofloxacin. P-α-02-B exhibited strong membrane disturbance effect, which could be explained by its rigid α-helical structure revealed by molecular dynamics simulations. More importantly, P-α-02-B showed favorable therapeutic efficacy <em>in vivo</em>, whether used alone or in combination with levofloxacin.<h3>Conclusion</h3>P-α-02-B is a promising antimicrobial agent for MDR bacterial infections, demonstrating the effectiveness of the combination strategy for AMP development.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"44 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142988994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SCARB1 links cholesterol metabolism-mediated ferroptosis inhibition to radioresistance in tumor cells SCARB1将胆固醇代谢介导的铁垂症抑制与肿瘤细胞的放射抗性联系起来
IF 10.7 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Pub Date : 2025-01-18 DOI: 10.1016/j.jare.2025.01.026
Xiaojuan Mao, Jingwen Xiong, Mengjiao Cai, Chao Wang, Qian He, Binxian Wang, Jing Chen, Zhengtao Xiao, Baofeng Wang, Suxia Han, Yilei Zhang

Introduction

Ferroptosis is an iron-dependent form of cell death triggered by the excessive accumulation of lipid peroxides. Understanding the regulatory mechanisms of ferroptosis and developing strategies to target this process hold significant clinical applications in tumor therapy.

Objective

Our study aims to search for novel candidate genes involved in the regulation of ferroptosis and to investigate their mechanism of action in ferroptosis and tumor therapy.

Methods

We employed a CRISPR-Cas9 library to perform a genome-wide screen under ferroptosis inducer treatment conditions, revealing Scavenger Receptor Class B Member 1(SCARB1) as a novel candidate gene involved in ferroptosis regulation. Subsequently, lipidomic analyses, metabolic interventions, and relevant cellular experimental analyses were performed to elucidate the role of SCARB1 in ferroptosis, lipid peroxidation, and tumor therapy.

Results

Our study confirmed that SCARB1 significantly inhibits ferroptosis and lipid peroxidation induced by ferroptosis inducers. Mechanistically, SCARB1 inhibits ferroptosis through the regulation of cholesterol metabolism, and the upregulation of CoQ10 level is demonstrated to mediate the suppression of ferroptosis by SCARB1 after lipidomic analysis and metabolic intervention. Interestingly, SCARB1 exerts a tumor suppressive effect regarding tumor growth, migration and invasion, which is possibly independent of ferroptosis regulation. However, SCARB1 promotes radioresistance through the upregulation of cholesterol metabolism and inhibition of ferroptosis, while the combination of ferroptosis inducers can overcome radioresistance in tumor cells with high SCARB1 expression.

Conclusion

This study establishes a theoretical foundation for the regulation of ferroptosis by SCARB1 and highlights the potential of targeting lipid metabolism to overcome radioresistance in cancer therapy. The identification of SCARB1 as a key player in ferroptosis and its dual role in tumor suppression and radioresistance provides new avenues for therapeutic intervention in cancer treatment.
铁下垂是一种铁依赖性的细胞死亡形式,由脂质过氧化物的过度积累引发。了解铁下垂的调节机制并制定针对这一过程的策略在肿瘤治疗中具有重要的临床应用价值。目的寻找参与铁下垂调控的新候选基因,探讨其在铁下垂和肿瘤治疗中的作用机制。方法利用CRISPR-Cas9文库在铁衰亡诱变剂处理条件下进行全基因组筛选,发现清道夫受体B类成员1(SCARB1)是参与铁衰亡调控的新候选基因。随后,脂质组学分析、代谢干预和相关的细胞实验分析来阐明SCARB1在铁死亡、脂质过氧化和肿瘤治疗中的作用。结果我们的研究证实SCARB1显著抑制铁下垂诱导剂诱导的铁下垂和脂质过氧化。机制上,SCARB1通过调节胆固醇代谢抑制铁下沉,脂质组学分析和代谢干预证实,CoQ10水平上调介导SCARB1对铁下沉的抑制。有趣的是,SCARB1对肿瘤的生长、迁移和侵袭有抑制作用,可能不依赖于对铁下沉的调节。然而,SCARB1通过上调胆固醇代谢和抑制铁下沉来促进辐射耐药,而铁下沉诱导剂联合使用可以克服SCARB1高表达的肿瘤细胞的辐射耐药。结论本研究为SCARB1调控铁下沉奠定了理论基础,突出了靶向脂质代谢克服肿瘤放疗耐药的潜力。SCARB1在铁吊中起着关键作用,其在肿瘤抑制和放射耐药中的双重作用为癌症治疗的治疗干预提供了新的途径。
{"title":"SCARB1 links cholesterol metabolism-mediated ferroptosis inhibition to radioresistance in tumor cells","authors":"Xiaojuan Mao, Jingwen Xiong, Mengjiao Cai, Chao Wang, Qian He, Binxian Wang, Jing Chen, Zhengtao Xiao, Baofeng Wang, Suxia Han, Yilei Zhang","doi":"10.1016/j.jare.2025.01.026","DOIUrl":"https://doi.org/10.1016/j.jare.2025.01.026","url":null,"abstract":"<h3>Introduction</h3>Ferroptosis is an iron-dependent form of cell death triggered by the excessive accumulation of lipid peroxides. Understanding the regulatory mechanisms of ferroptosis and developing strategies to target this process hold significant clinical applications in tumor therapy.<h3>Objective</h3>Our study aims to search for novel candidate genes involved in the regulation of ferroptosis and to investigate their mechanism of action in ferroptosis and tumor therapy.<h3>Methods</h3>We employed a CRISPR-Cas9 library to perform a genome-wide screen under ferroptosis inducer treatment conditions, revealing Scavenger Receptor Class B Member 1(SCARB1) as a novel candidate gene involved in ferroptosis regulation. Subsequently, lipidomic analyses, metabolic interventions, and relevant cellular experimental analyses were performed to elucidate the role of SCARB1 in ferroptosis, lipid peroxidation, and tumor therapy.<h3>Results</h3>Our study confirmed that SCARB1 significantly inhibits ferroptosis and lipid peroxidation induced by ferroptosis inducers. Mechanistically, SCARB1 inhibits ferroptosis through the regulation of cholesterol metabolism, and the upregulation of CoQ10 level is demonstrated to mediate the suppression of ferroptosis by SCARB1 after lipidomic analysis and metabolic intervention. Interestingly, SCARB1 exerts a tumor suppressive effect regarding tumor growth, migration and invasion, which is possibly independent of ferroptosis regulation. However, SCARB1 promotes radioresistance through the upregulation of cholesterol metabolism and inhibition of ferroptosis, while the combination of ferroptosis inducers can overcome radioresistance in tumor cells with high SCARB1 expression.<h3>Conclusion</h3>This study establishes a theoretical foundation for the regulation of ferroptosis by SCARB1 and highlights the potential of targeting lipid metabolism to overcome radioresistance in cancer therapy. The identification of SCARB1 as a key player in ferroptosis and its dual role in tumor suppression and radioresistance provides new avenues for therapeutic intervention in cancer treatment.","PeriodicalId":14952,"journal":{"name":"Journal of Advanced Research","volume":"16 1","pages":""},"PeriodicalIF":10.7,"publicationDate":"2025-01-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142988996","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Advanced Research
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:481959085
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