Journal home Browse Most accessed

Most accessed

  • Select all
  • LETTER
    Yun Hong, Zheng Tian, Liangjie Jia, Yiguo Wang
    Protein & Cell, 2023, 14(6): 459-463. https://doi.org/10.1093/procel/pwac034
  • REVIEW
    Tao Wen, Guoqing Niu, Tong Chen, Qirong Shen, Jun Yuan, Yong-Xin Liu
    Protein & Cell, 2023, 14(10): 713-725. https://doi.org/10.1093/procel/pwad024

    With the gradual maturity of sequencing technology, many microbiome studies have published, driving the emergence and advance of related analysis tools. R language is the widely used platform for microbiome data analysis for powerful functions. However, tens of thousands of R packages and numerous similar analysis tools have brought major challenges for many researchers to explore microbiome data. How to choose suitable, efficient, convenient, and easy-to-learn tools from the numerous R packages has become a problem for many microbiome researchers. We have organized 324 common R packages for microbiome analysis and classified them according to application categories (diversity, difference, biomarker, correlation and network, functional prediction, and others), which could help researchers quickly find relevant R packages for microbiome analysis. Furthermore, we systematically sorted the integrated R packages (phyloseq, microbiome, MicrobiomeAnalystR, Animalcules, microeco, and amplicon) for microbiome analysis, and summarized the advantages and limitations, which will help researchers choose the appropriate tools. Finally, we thoroughly reviewed the R packages for microbiome analysis, summarized most of the common analysis content in the microbiome, and formed the most suitable pipeline for microbiome analysis. This paper is accompanied by hundreds of examples with 10,000 lines codes in GitHub, which can help beginners to learn, also help analysts compare and test different tools. This paper systematically sorts the application of R in microbiome, providing an important theoretical basis and practical reference for the development of better microbiome tools in the future. All the code is available at GitHub github.com/taowenmicro/EasyMicrobiomeR.

  • RECOLLECTION
    Huan Liu, Zhaoqi Liu, Xue Gong, Hao Cheng
    Protein & Cell, 2023, 14(5): 315-317. https://doi.org/10.1093/procel/pwac049
  • RECOLLECTION
    Huan Liu, Yifan Wei, Wanying Gao, Hao Cheng
    Protein & Cell, 2023, 14(6): 393-397. https://doi.org/10.1093/procel/pwac046
  • LETTER
    Hainan Zhang, Xiangfeng Kong, Mingxing Xue, Jing Hu, Zikang Wang, Yinghui Wei, Haoqiang Wang, Jingxing Zhou, Weihong Zhang, Mengqiu Xu, Xiaowen Shen, Fengcai Yin, Zhiyuan Ai, Guangyan Huang, Junhui Xia, Xueqiong Song, Hengbin Li, Yuan Yuan, Jinhui Li, Na Zhong, Meiling Zhang, Yingsi Zhou, Hui Yang
    Protein & Cell, 2023, 14(7): 538-543. https://doi.org/10.1093/procel/pwac052
  • RESEARCH ARTICLE
    Zekai Wu, Yuan Shi, Yueli Cui, Xin Xing, Liya Zhang, Da Liu, Yutian Zhang, Ji Dong, Li Jin, Meijun Pang, Rui-Ping Xiao, Zuoyan Zhu, Jing-Wei Xiong, Xiangjun Tong, Yan Zhang, Shiqiang Wang, Fuchou Tang, Bo Zhang
    Protein & Cell, 2023, 14(5): 350-368. https://doi.org/10.1093/procel/pwac010

    Mammals exhibit limited heart regeneration ability, which can lead to heart failure after myocardial infarction. In contrast, zebrafish exhibit remarkable cardiac regeneration capacity. Several cell types and signaling pathways have been reported to participate in this process. However, a comprehensive analysis of how different cells and signals interact and coordinate to regulate cardiac regeneration is unavailable. We collected major cardiac cell types from zebrafish and performed high-precision single-cell transcriptome analyses during both development and post-injury regeneration. We revealed the cellular heterogeneity as well as the molecular progress of cardiomyocytes during these processes, and identified a subtype of atrial cardiomyocyte exhibiting a stem-like state which may transdifferentiate into ventricular cardiomyocytes during regeneration. Furthermore, we identified a regeneration-induced cell (RIC) population in the epicardium-derived cells (EPDC), and demonstrated Angiopoietin 4 (Angpt4) as a specific regulator of heart regeneration. angpt4 expression is specifically and transiently activated in RIC, which initiates a signaling cascade from EPDC to endocardium through the Tie2-MAPK pathway, and further induces activation of cathepsin K in cardiomyocytes through RA signaling. Loss of angpt4 leads to defects in scar tissue resolution and cardiomyocyte proliferation, while overexpression of angpt4 accelerates regeneration. Furthermore, we found that ANGPT4 could enhance proliferation of neonatal rat cardiomyocytes, and promote cardiac repair in mice after myocardial infarction, indicating that the function of Angpt4 is conserved in mammals. Our study provides a mechanistic understanding of heart regeneration at single-cell precision, identifies Angpt4 as a key regulator of cardiomyocyte proliferation and regeneration, and offers a novel therapeutic target for improved recovery after human heart injuries.

  • RECOLLECTION
    Shunping He, Yiyu Chen
    Protein & Cell, 2023, 14(7): 471-473. https://doi.org/10.1093/procel/pwac054
  • RESEARCH ARTICLE
    Qian Zhao, Yandong Zheng, Dongxin Zhao, Liyun Zhao, Lingling Geng, Shuai Ma, Yusheng Cai, Chengyu Liu, Yupeng Yan, Juan Carlos Izpisua Belmonte, Si Wang, Weiqi Zhang, Guang-Hui Liu, Jing Qu
    Protein & Cell, 2023, 14(6): 398-415. https://doi.org/10.1093/procel/pwac062

    Hair loss affects millions of people at some time in their life, and safe and efficient treatments for hair loss are a significant unmet medical need. We report that topical delivery of quercetin (Que) stimulates resting hair follicles to grow with rapid follicular keratinocyte proliferation and replenishes perifollicular microvasculature in mice. We construct dynamic single-cell transcriptome landscape over the course of hair regrowth and find that Que treatment stimulates the differentiation trajectory in the hair follicles and induces an angiogenic signature in dermal endothelial cells by activating HIF-1α in endothelial cells. Skin administration of a HIF-1α agonist partially recapitulates the pro-angiogenesis and hair-growing effects of Que. Together, these findings provide a molecular understanding for the efficacy of Que in hair regrowth, which underscores the translational potential of targeting the hair follicle niche as a strategy for regenerative medicine, and suggest a route of pharmacological intervention that may promote hair regrowth.

  • LETTER
    Yuxiang Chen, Bo Chen, Tingting Wu, Fangfang Zhou, Fei Xu
    Protein & Cell, 2023, 14(6): 464-468. https://doi.org/10.1093/procel/pwac033
  • LETTER
    Wen Cui, Haojun Huang, Yinkai Duan, Zhi Luo, Haofeng Wang, Tenan Zhang, Henry C. Nguyen, Wei Shen, Dan Su, Xi Li, Xiaoyun Ji, Haitao Yang, Wei Wang
    Protein & Cell, 2023, 14(6): 469-472. https://doi.org/10.1093/procel/pwac051
  • Research Article
    Zhifen Tu, Yan Bi, Xuehao Zhu, Wenqiang Liu, Jindian Hu, Li Wu, Tengyan Mao, Jianfeng Zhou, Hanwei Wang, Hong Wang, Shaorong Gao, Yixuan Wang
    Protein & Cell, 2023, 14(5): 337-349. https://doi.org/10.1093/procel/pwac041

    Human pluripotent stem cells provide an inexhaustible model to study human embryogenesis in vitro. Recent studies have provided diverse models to generate human blastoids by self-organization of different pluripotent stem cells or somatic reprogramming intermediates. However, whether blastoids can be generated from other cell types or whether they can recapitulate postimplantation development in vitro is unknown. Here, we develop a strategy to generate human blastoids from heterogeneous intermediates with epiblast, trophectoderm, and primitive endoderm signatures of the primed-to-naïve conversion process, which resemble natural blastocysts in morphological architecture, composition of cell lineages, transcriptome, and lineage differentiation potential. In addition, these blastoids reflect many features of human peri-implantation and pregastrulation development when further cultured in an in vitro 3D culture system. In summary, our study provides an alternative strategy to generate human blastoids and offers insights into human early embryogenesis by modeling peri- and postimplantation development in vitro.

  • RESEARCH ARTICLE
    Ying Jing, Yuesheng Zuo, Yang Yu, Liang Sun, Zhengrong Yu, Shuai Ma, Qian Zhao, Guoqiang Sun, Huifang Hu, Jingyi Li, Daoyuan Huang, Lixiao Liu, Jiaming Li, Zijuan Xin, Haoyan Huang, Juan Carlos Izpisua Belmonte, Weiqi Zhang, Si Wang, Jing Qu, Guang-Hui Liu
    Protein & Cell, 2023, 14(7): 497-512. https://doi.org/10.1093/procel/pwac061

    Age-dependent loss of skeletal muscle mass and function is a feature of sarcopenia, and increases the risk of many aging-related metabolic diseases. Here, we report phenotypic and single-nucleus transcriptomic analyses of non-human primate skeletal muscle aging. A higher transcriptional fluctuation was observed in myonuclei relative to other interstitial cell types, indicating a higher susceptibility of skeletal muscle fiber to aging. We found a downregulation of FOXO3 in aged primate skeletal muscle, and identified FOXO3 as a hub transcription factor maintaining skeletal muscle homeostasis. Through the establishment of a complementary experimental pipeline based on a human pluripotent stem cell-derived myotube model, we revealed that silence of FOXO3 accelerates human myotube senescence, whereas genetic activation of endogenous FOXO3 alleviates human myotube aging. Altogether, based on a combination of monkey skeletal muscle and human myotube aging research models, we unraveled the pivotal role of the FOXO3 in safeguarding primate skeletal muscle from aging, providing a comprehensive resource for the development of clinical diagnosis and targeted therapeutic interventions against human skeletal muscle aging and the onset of sarcopenia along with aging-related disorders.

  • LETTER
    Lan-Zhu Li, Kuan Yang, Yaobin Jing, Yanling Fan, Xiaoyu Jiang, Si Wang, Guang-Hui Liu, Jing Qu, Shuai Ma, Weiqi Zhang
    Protein & Cell, 2023, 14(8): 623-628. https://doi.org/10.1093/procel/pwad012
  • LETTER
    Beibei Li, Shuo Han, Mu Wang, Yu Yu, Limin Ma, Xiaojing Chu, Qiuxiang Tan, Qiang Zhao, Beili Wu
    Protein & Cell, 2023, 14(5): 382-386. https://doi.org/10.1093/procel/pwac025
  • COMMENTARY
    Gaoxiang Zhao, Qian Lin, Zhaoyuan Meng, Xinlei Sheng, Leina Ma, Yingming Zhao
    Protein & Cell, 2023, 14(7): 474-476. https://doi.org/10.1093/procel/pwad006
  • RESEARCH ARTICLE
    Ji Dong, Xinglong Wu, Xin Zhou, Yuan Gao, Changliang Wang, Wendong Wang, Weiya He, Jingyun Li, Wenjun Deng, Jiayu Liao, Xiaotian Wu, Yongqu Lu, Antony K. Chen, Lu Wen, Wei Fu, Fuchou Tang
    Protein & Cell, 2023, 14(6): 433-447. https://doi.org/10.1093/procel/pwac059

    Molecular knowledge of human gastric corpus epithelium remains incomplete. Here, by integrated analyses using single-cell RNA sequencing (scRNA-seq), spatial transcriptomics, and single-cell assay for transposase accessible chromatin sequencing (scATAC-seq) techniques, we uncovered the spatially resolved expression landscape and gene-regulatory network of human gastric corpus epithelium. Specifically, we identified a stem/progenitor cell population in the isthmus of human gastric corpus, where EGF and WNT signaling pathways were activated. Meanwhile, LGR4, but not LGR5, was responsible for the activation of WNT signaling pathway. Importantly, FABP5 and NME1 were identified and validated as crucial for both normal gastric stem/progenitor cells and gastric cancer cells. Finally, we explored the epigenetic regulation of critical genes for gastric corpus epithelium at chromatin state level, and identified several important cell-type-specific transcription factors. In summary, our work provides novel insights to systematically understand the cellular diversity and homeostasis of human gastric corpus epithelium in vivo.

  • RESEARCH ARTICLE
    Shan Sun, Yan Gao, Xiaolin Yang, Xiuna Yang, Tianyu Hu, Jingxi Liang, Zhiqi Xiong, Yuting Ran, Pengxuan Ren, Fang Bai, Luke W. Guddat, Haitao Yang, Zihe Rao, Bing Zhang
    Protein & Cell, 2023, 14(6): 448-458. https://doi.org/10.1093/procel/pwac060

    The adenosine 5′-triphosphate (ATP)-binding cassette (ABC) transporter, IrtAB, plays a vital role in the replication and viability of Mycobacterium tuberculosis (Mtb), where its function is to import iron-loaded siderophores. Unusually, it adopts the canonical type IV exporter fold. Herein, we report the structure of unliganded Mtb IrtAB and its structure in complex with ATP, ADP, or ATP analogue (AMP-PNP) at resolutions ranging from 2.8 to 3.5 Å. The structure of IrtAB bound ATP-Mg2+ shows a “head-to-tail” dimer of nucleo-tide-binding domains (NBDs), a closed amphipathic cavity within the transmembrane domains (TMDs), and a metal ion liganded to three histidine residues of IrtA in the cavity. Cryo-electron microscopy (Cryo-EM) structures and ATP hydrolysis assays show that the NBD of IrtA has a higher affinity for nucleotides and increased ATPase activity compared with IrtB. Moreover, the metal ion located in the TM region of IrtA is critical for the stabilization of the conformation of IrtAB during the transport cycle. This study provides a structural basis to explain the ATP-driven conformational changes that occur in IrtAB.

  • RESEARCH ARTICLE
    Yinghui Wei, Meiling Zhang, Jing Hu, Yingsi Zhou, Mingxing Xue, Jianhang Yin, Yuanhua Liu, Hu Feng, Ling Zhou, Zhifang Li, Dongshuang Wang, Zhiguo Zhang, Yin Zhou, Hongbin Liu, Ning Yao, Erwei Zuo, Jiazhi Hu, Yanzhi Du, Wen Li, Chunlong Xu, Hui Yang
    Protein & Cell, 2023, 14(6): 416-432. https://doi.org/10.1093/procel/pwac043

    Approximately 140 million people worldwide are homozygous carriers of APOE4 (ϵ4), a strong genetic risk factor for late onset familial and sporadic Alzheimer’s disease (AD), 91% of whom will develop AD at earlier age than heterozygous carriers and noncarriers. Susceptibility to AD could be reduced by targeted editing of APOE4, but a technical basis for controlling the off-target effects of base editors is necessary to develop low-risk personalized gene therapies. Here, we first screened eight cytosine base editor variants at four injection stages (from 1- to 8-cell stage), and found that FNLS-YE1 variant in 8-cell embryos achieved the comparable base conversion rate (up to 100%) with the lowest bystander effects. In particular, 80% of AD-susceptible ϵ4 allele copies were converted to the AD-neutral ϵ3 allele in human ϵ4-carrying embryos. Stringent control measures combined with targeted deep sequencing, whole genome sequencing, and RNA sequencing showed no DNA or RNA off-target events in FNLS-YE1-treated human embryos or their derived stem cells. Furthermore, base editing with FNLS-YE1 showed no effects on embryo development to the blastocyst stage. Finally, we also demonstrated FNLS-YE1 could introduce known protective variants in human embryos to potentially reduce human susceptivity to systemic lupus erythematosus and familial hypercholesterolemia. Our study therefore suggests that base editing with FNLS-YE1 can efficiently and safely introduce known preventive variants in 8-cell human embryos, a potential approach for reducing human susceptibility to AD or other genetic diseases.

  • LETTER
    Alexander Stevens, Yanxiang Cui, Sakar Shivakoti, Z. Hong Zhou
    Protein & Cell, 2023, 14(7): 544-548. https://doi.org/10.1093/procel/pwad002
  • LETTER
    Yu Chen, Zhen Cui, Zhixi Chen, Ying Jiang, Zhiyong Mao
    Protein & Cell, 2023, 14(5): 369-375. https://doi.org/10.1093/procel/pwac001
  • RESEARCH ARTICLE
    Yuhan Chen, Jiansen Lu, Yanwen Xu, Yaping Huang, Dazhuang Wang, Peiling Liang, Shaofang Ren, Xuesong Hu, Yewen Qin, Wei Ke, Ralf Jauch, Andrew Paul Hutchins, Mei Wang, Fuchou Tang, Xiao-Yang Zhao
    Protein & Cell, 2023, 14(7): 477-496. https://doi.org/10.1093/procel/pwac044

    Although somatic cells can be reprogrammed to pluripotent stem cells (PSCs) with pure chemicals, authentic pluripotency of chemically induced pluripotent stem cells (CiPSCs) has never been achieved through tetraploid complementation assay. Spontaneous reprogramming of spermatogonial stem cells (SSCs) was another non-transgenic way to obtain PSCs, but this process lacks mechanistic explanation. Here, we reconstructed the trajectory of mouse SSC reprogramming and developed a five-chemical combination, boosting the reprogramming efficiency by nearly 80- to 100-folds. More importantly, chemical induced germline-derived PSCs (5C-gPSCs), but not gPSCs and chemical induced pluripotent stem cells, had authentic pluripotency, as determined by tetraploid complementation. Mechanistically, SSCs traversed through an inverted pathway of in vivo germ cell development, exhibiting the expression signatures and DNA methylation dynamics from spermatogonia to primordial germ cells and further to epiblasts. Besides, SSC-specific imprinting control regions switched from biallelic methylated states to monoallelic methylated states by imprinting demethylation and then re-methylation on one of the two alleles in 5C-gPSCs, which was apparently distinct with the imprinting reprogramming in vivo as DNA methylation simultaneously occurred on both alleles. Our work sheds light on the unique regulatory network underpinning SSC reprogramming, providing insights to understand generic mechanisms for cell-fate decision and epigenetic-related disorders in regenerative medicine.

  • LETTER
    Fei Liu, Ningning Pang, Rui-Ming Xu, Na Yang
    Protein & Cell, 2023, 14(5): 387-392. https://doi.org/10.1093/procel/pwac039
  • RESEARCH ARTICLE
    Dawei Meng, Qian Zheng, Xue Zhang, Xuejiao Piao, Li Luo, Yichang Jia
    Protein & Cell, 2023, 14(5): 318-336. https://doi.org/10.1093/procel/pwac008

    Emerging evidence suggests that intron-detaining transcripts (IDTs) are a nucleus-detained and polyadenylated mRNA pool for cell to quickly and effectively respond to environmental stimuli and stress. However, the underlying mechanisms of detained intron (DI) splicing are still largely unknown. Here, we suggest that post-transcriptional DI splicing is paused at the Bact state, an active spliceosome but not catalytically primed, which depends on Smad Nuclear Interacting Protein 1 (SNIP1) and RNPS1 (a serine-rich RNA binding protein) interaction. RNPS1 and Bact components preferentially dock at DIs and the RNPS1 docking is sufficient to trigger spliceosome pausing. Haploinsufficiency of Snip1 attenuates neurodegeneration and globally rescues IDT accumulation caused by a previously reported mutant U2 snRNA, a basal spliceosomal component. Snip1 conditional knockout in the cerebellum decreases DI splicing efficiency and causes neurodegeneration. Therefore, we suggest that SNIP1 and RNPS1 form a molecular brake to promote spliceosome pausing, and that its misregulation contributes to neurodegeneration.

  • LETTER
    Ya-Hui Zhao, Wei Jiang, Hai Gao, Guo-Zheng Pang, Yu-Shuang Wu, Yuan-Xian Wang, Meng-Yao Sheng, Jia-Ying Xie, Wan-Ling Wu, Zhi-Jian Ji, Ya-Rui Du, Lei Zhang, Xiao-Qin Wang, Colum P. Walsh, Hai Jiang, Guo-Liang Xu, Dan Zhou
    Protein & Cell, 2023, 14(7): 532-537. https://doi.org/10.1093/procel/pwac028
  • RESEARCH ARTICLE
    Pingping Nie, Weihong Zhang, Yan Meng, Moubin Lin, Fenghua Guo, Hui Zhang, Zhenzhu Tong, Meng Wang, Fan Chen, Liwei An, Yang Tang, Yi Han, Ruixian Yu, Wenjia Wang, Yuanzhi Xu, Linxin Wei, Zhaocai Zhou, Shi Jiao
    Protein & Cell, 2023, 14(7): 513-531. https://doi.org/10.1093/procel/pwac045

    As an important part of tumor microenvironment, neutrophils are poorly understood due to their spatiotemporal heterogeneity in tumorigenesis. Here we defined, at single-cell resolution, CD44CXCR2 neutrophils as tumor-specific neutrophils (tsNeus) in both mouse and human gastric cancer (GC). We uncovered a Hippo regulon in neutrophils with unique YAP signature genes (e.g., ICAM1, CD14, EGR1) distinct from those identified in epithelial and/or cancer cells. Importantly, knockout of YAP/TAZ in neutrophils impaired their differentiation into CD54+ tsNeus and reduced their antitumor activity, leading to accelerated GC progression. Moreover, the relative amounts of CD54+ tsNeus were found to be negatively associated with GC progression and positively associated with patient survival. Interestingly, GC patients receiving neoadjuvant chemotherapy had increased numbers of CD54+ tsNeus. Furthermore, pharmacologically enhancing YAP activity selectively activated neutrophils to suppress refractory GC, with no significant inflammation-related side effects. Thus, our work characterized tumor-specific neutrophils in GC and revealed an essential role of YAP/TAZ-CD54 axis in tsNeus, opening a new possibility to develop neutrophil-based antitumor therapeutics.

  • RESEARCH ARTICLE
    Yingrui Wang, Qianru Zhu, Rui Sun, Xiao Yi, Lingling Huang, Yifan Hu, Weigang Ge, Huanhuan Gao, Xinfu Ye, Yu Song, Li Shao, Yantao Li, Jie Li, Tiannan Guo, Junping Shi
    Protein & Cell, 2023, 14(9): 668-682. https://doi.org/10.1093/procel/pwad004

    Although the development of COVID-19 vaccines has been a remarkable success, the heterogeneous individual antibody generation and decline over time are unknown and still hard to predict. In this study, blood samples were collected from 163 participants who next received two doses of an inactivated COVID-19 vaccine (CoronaVac®) at a 28-day interval. Using TMT-based proteomics, we identified 1,715 serum and 7,342 peripheral blood mononuclear cells (PBMCs) proteins. We proposed two sets of potential biomarkers (seven from serum, five from PBMCs) at baseline using machine learning, and predicted the individual seropositivity 57 days after vaccination (AUC = 0.87). Based on the four PBMC’s potential biomarkers, we predicted the antibody persistence until 180 days after vaccination (AUC = 0.79). Our data highlighted characteristic hematological host responses, including altered lymphocyte migration regulation, neutrophil degranulation, and humoral immune response. This study proposed potential blood-derived protein biomarkers before vaccination for predicting heterogeneous antibody generation and decline after COVID-19 vaccination, shedding light on immunization mechanisms and individual booster shot planning.

  • EDITORIAL
    Yunyun Gao, Danyi Li, Yong-Xin Liu
    Protein & Cell, 2023, 14(10): 709-712. https://doi.org/10.1093/procel/pwad031
  • RECOLLECTION
    Yujing Qian, Xiazhao Yu, Yangqing Sun, Qun Xie, Ge Liang, Gaiping Bai
    Protein & Cell, 2023, 14(9): 629-631. https://doi.org/10.1093/procel/pwac055
  • LETTER
    Chen Zhu, Yanfeng Shi, Jing Yu, Wenhao Zhao, Lingqiao Li, Jingxi Liang, Xiaolin Yang, Bing Zhang, Yao Zhao, Yan Gao, Xiaobo Chen, Xiuna Yang, Lu Zhang, Luke W. Guddat, Lei Liu, Haitao Yang, Zihe Rao, Jun Li
    Protein & Cell, 2023, 14(12): 919-923. https://doi.org/10.1093/procel/pwad022
  • LETTER
    Jiani Cao, Meng Li, Kun Liu, Xingxing Shi, Ning Sui, Yuchen Yao, Xiaojing Wang, Shiyu Li, Yuchang Tian, Shaojing Tan, Qian Zhao, Liang Wang, Xiahua Chai, Lin Zhang, Chong Liu, Xing Li, Zhijie Chang, Dong Li, Tongbiao Zhao
    Protein & Cell, 2023, 14(5): 376-381. https://doi.org/10.1093/procel/pwac009