Journal home Browse Most Down Articles

Most Down Articles

  • Select all
  • RESEARCH ARTICLE
    Kai Jiang, Yue Xu, Dandan Wang, Feng Chen, Zizhuo Tu, Jie Qian, Sheng Xu, Yixiang Xu, John Hwa, Jian Li, Hongcai Shang, Yaozu Xiang
    Protein & Cell, 2022, 13(5): 336-359. https://doi.org/10.1007/s13238-020-00809-4

    Sodium-glucose cotransporter 2 (SGLT2) inhibitors reduce cardiovascular mortality in patients with diabetes mellitus but the protective mechanism remains elusive. Here we demonstrated that the SGLT2 inhibitor, Empagliflozin (EMPA), suppresses cardiomyocytes autosis (autophagic cell death) to confer cardioprotective effects. Using myocardial infarction (MI) mouse models with and without diabetes mellitus, EMPA treatment significantly reduced infarct size, and myocardial fibrosis, thereby leading to improved cardiac function and survival. In the context of ischemia and nutritional glucose deprivation where autosis is already highly stimulated, EMPA directly inhibits the activity of the Na+/H+ exchanger 1 (NHE1) in the cardiomyocytes to regulate excessive autophagy. Knockdown of NHE1 significantly rescued glucose deprivation-induced autosis. In contrast, overexpression of NHE1 aggravated the cardiomyocytes death in response to starvation, which was effectively rescued by EMPA treatment. Furthermore, in vitro and in vivo analysis of NHE1 and Beclin 1 knockout mice validated that EMPA’s cardioprotective effects are at least in part through downregulation of autophagic flux. These findings provide new insights for drug development, specifically targeting NHE1 and autosis for ventricular remodeling and heart failure after MI in both diabetic and non-diabetic patients.

  • REVIEW
    Tzu-Lung Lin, Chia-Chen Lu, Wei-Fan Lai, Ting-Shu Wu, Jang-Jih Lu, Young-Mao Chen, Chi-Meng Tzeng, Hong-Tao Liu, Hong Wei, Hsin-Chih Lai
    Protein & Cell, 2021, 12(5): 394-410. https://doi.org/10.1007/s13238-020-00784-w

    Traditional Chinese Medicine (TCM) has been extensively used to ameliorate diseases in Asia for over thousands of years. However, owing to a lack of formal scientific validation, the absence of information regarding the mechanisms underlying TCMs restricts their application. After oral administration, TCM herbal ingredients frequently are not directly absorbed by the host, but rather enter the intestine to be transformed by gut microbiota. The gut microbiota is a microbial community living in animal intestines, and functions to maintain host homeostasis and health. Increasing evidences indicate that TCM herbs closely affect gut microbiota composition, which is associated with the conversion of herbal components into active metabolites. These may significantly affect the therapeutic activity of TCMs. Microbiota analyses, in conjunction with modern multiomics platforms, can together identify novel functional metabolites and form the basis of future TCM research.

  • RECOLLECTION
    Yuan Chen, Alan W. Adame, Ge-Zhi Chen, Yuanyuan Meng
    Protein & Cell, 2022, 13(3): 157-162. https://doi.org/10.1007/s13238-020-00815-6
  • RESEARCH ARTICLE
    Xiang Li, Chuan-Qi Zhong, Rui Wu, Xiaozheng Xu, Zhang-Hua Yang, Shaowei Cai, Xiurong Wu, Xin Chen, Zhiyong Yin, Qingzu He, Dianjie Li, Fei Xu, Yihua Yan, Hong Qi, Changchuan Xie, Jianwei Shuai, Jiahuai Han
    Protein & Cell, 2021, 12(11): 858-876. https://doi.org/10.1007/s13238-020-00810-x

    There remains a significant gap in our quantitative understanding of crosstalk between apoptosis and necroptosis pathways. By employing the SWATH-MS technique, we quantified absolute amounts of up to thousands of proteins in dynamic assembling/deassembling of TNF signaling complexes. Combining SWATH-MS-based network modeling and experimental validation, we found that when RIP1 level is below ∼1000 molecules/cell (mpc), the cell solely undergoes TRADDdependent apoptosis. When RIP1 is above ∼1000 mpc, pro-caspase-8 and RIP3 are recruited to necrosome respectively with linear and nonlinear dependence on RIP1 amount, which well explains the co-occurrence of apoptosis and necroptosis and the paradoxical observations that RIP1 is required for necroptosis but its increase down-regulates necroptosis. Higher amount of RIP1 (>∼46,000 mpc) suppresses apoptosis, leading to necroptosis alone. The relation between RIP1 level and occurrence of necroptosis or total cell death is biphasic. Our study provides a resource for encoding the complexity of TNF signaling and a quantitative picture how distinct dynamic interplay among proteins function as basis sets in signaling complexes, enabling RIP1 to play diverse roles in governing cell fate decisions.

  • RESEARCH ARTICLE
    Weili Yang, Xiangyu Guo, Zhuchi Tu, Xiusheng Chen, Rui Han, Yanting Liu, Sen Yan, Qi Wang, Zhifu Wang, Xianxian Zhao, Yunpeng Zhang, Xin Xiong, Huiming Yang, Peng Yin, Huida Wan, Xingxing Chen, Jifeng Guo, Xiao-Xin Yan, Lujian Liao, Shihua Li, Xiao-Jiang Li
    Protein & Cell, 2022, 13(1): 26-46. https://doi.org/10.1007/s13238-021-00888-x

    In vitro studies have established the prevalent theory that the mitochondrial kinase PINK1 protects neurodegeneration by removing damaged mitochondria in Parkinson's disease (PD). However, difficulty in detecting endogenous PINK1 protein in rodent brains and cell lines has prevented the rigorous investigation of the in vivo role of PINK1. Here we report that PINK1 kinase form is selectively expressed in the human and monkey brains. CRISPR/Cas9-mediated deficiency of PINK1 causes similar neurodegeneration in the brains of fetal and adult monkeys as well as cultured monkey neurons without affecting mitochondrial protein expression and morphology. Importantly, PINK1 mutations in the primate brain and human cells reduce protein phosphorylation that is important for neuronal function and survival. Our findings suggest that PINK1 kinase activity rather than its mitochondrial function is essential for the neuronal survival in the primate brains and that its kinase dysfunction could be involved in the pathogenesis of PD.

  • RESEARCH ARTICLE
    He Li, Lei Zhu, Rong Wang, Lihui Xie, Jie Ren, Shuai Ma, Weiqi Zhang, Xiuxing Liu, Zhaohao Huang, Binyao Chen, Zhaohuai Li, Huyi Feng, Guang-Hui Liu, Si Wang, Jing Qu, Wenru Su
    Protein & Cell, 0: 422-445. https://doi.org/10.1007/s13238-021-00882-3

    Aging-induced changes in the immune system are associated with a higher incidence of infection and vaccination failure. Lymph nodes, which filter the lymph to identify and fight infections, play a central role in this process. However, careful characterization of the impact of aging on lymph nodes and associated autoimmune diseases is lacking. We combined single-cell RNA sequencing (scRNA-seq) with flow cytometry to delineate the immune cell atlas of cervical draining lymph nodes (CDLNs) of both young and old mice with or without experimental autoimmune uveitis (EAU). We found extensive and complicated changes in the cellular constituents of CDLNs during aging. When confronted with autoimmune challenges, old mice developed milder EAU compared to young mice. Within this EAU process, we highlighted that the pathogenicity of T helper 17 cells (Th17) was dampened, as shown by reduced GM-CSF secretion in old mice. The mitigated secretion of GM-CSF contributed to alleviation of IL-23 secretion by antigen-presenting cells (APCs) and may, in turn, weaken APCs’ effects on facilitating the pathogenicity of Th17 cells. Meanwhile, our study further unveiled that aging downregulated GM-CSF secretion through reducing both the transcript and protein levels of IL-23R in Th17 cells from CDLNs. Overall, aging altered immune cell responses, especially through toning down Th17 cells, counteracting EAU challenge in old mice.

  • RESEARCH ARTICLE
    Yichen Li, Shuaiyao Lu, Jinge Gu, Wencheng Xia, Shengnan Zhang, Shenqing Zhang, Yan Wang, Chong Zhang, Yunpeng Sun, Jian Lei, Cong Liu, Zhaoming Su, Juntao Yang, Xiaozhong Peng, Dan Li
    Protein & Cell, 2022, 13(8): 602-614. https://doi.org/10.1007/s13238-022-00905-7

    The nucleocapsid (N) protein of SARS-CoV-2 has been reported to have a high ability of liquid-liquid phase separation, which enables its incorporation into stress granules (SGs) of host cells. However, whether SG invasion by N protein occurs in the scenario of SARS-CoV-2 infection is unknow, neither do we know its consequence. Here, we used SARS-CoV-2 to infect mammalian cells and observed the incorporation of N protein into SGs, which resulted in markedly impaired self-disassembly but stimulated cell cellular clearance of SGs. NMR experiments further showed that N protein binds to the SG-related amyloid proteins via non-specific transient interactions, which not only expedites the phase transition of these proteins to aberrant amyloid aggregation in vitro, but also promotes the aggregation of FUS with ALS-associated P525L mutation in cells. In addition, we found that ACE2 is not necessary for the infection of SARS-CoV-2 to mammalian cells. Our work indicates that SARS-CoV-2 infection can impair the disassembly of host SGs and promote the aggregation of SG-related amyloid proteins, which may lead to an increased risk of neurodegeneration.

  • REVIEW
    Jiayu Wu, Kai Wang, Xuemei Wang, Yanli Pang, Changtao Jiang
    Protein & Cell, 2021, 12(5): 360-373. https://doi.org/10.1007/s13238-020-00814-7

    It is well known that an unhealthy lifestyle is a major risk factor for metabolic diseases, while in recent years, accumulating evidence has demonstrated that the gut microbiome and its metabolites also play a crucial role in the onset and development of many metabolic diseases, including obesity, type 2 diabetes, nonalcoholic fatty liver disease, cardiovascular disease and so on. Numerous microorganisms dwell in the gastrointestinal tract, which is a key interface for energy acquisition and can metabolize dietary nutrients into many bioactive substances, thus acting as a link between the gut microbiome and its host. The gut microbiome is shaped by host genetics, immune responses and dietary factors. The metabolic and immune potential of the gut microbiome determines its significance in host health and diseases. Therefore, targeting the gut microbiome and relevant metabolic pathways would be effective therapeutic treatments for many metabolic diseases in the near future. This review will summarize information about the role of the gut microbiome in organism metabolism and the relationship between gut microbiome-derived metabolites and the pathogenesis of many metabolic diseases. Furthermore, recent advances in improving metabolic diseases by regulating the gut microbiome will be discussed.

  • RESEARCH ARTICLE
    Chenglei Tian, Linlin Liu, Ming Zeng, Xiaoyan Sheng, Dai Heng, Lingling Wang, Xiaoying Ye, David L. Keefe, Lin Liu
    Protein & Cell, 2021, 12(12): 947-964. https://doi.org/10.1007/s13238-021-00865-4

    Parthenogenetic embryos, created by activation and diploidization of oocytes, arrest at mid-gestation for defective paternal imprints, which impair placental development. Also, viable offspring has not been obtained without genetic manipulation from parthenogenetic embryonic stem cells (pESCs) derived from parthenogenetic embryos, presumably attributable to their aberrant imprinting. We show that an unlimited number of oocytes can be derived from pESCs and produce healthy offspring. Moreover, normal expression of imprinted genes is found in the germ cells and the mice. pESCs exhibited imprinting consistent with exclusively maternal lineage, and higher X-chromosome activation compared to female ESCs derived from the same mouse genetic background. pESCs differentiated into primordial germ cell-like cells (PGCLCs) and formed oocytes following in vivo transplantation into kidney capsule that produced fertile pups and reconstituted ovarian endocrine function. The transcriptome and methylation of imprinted and X-linked genes in pESC-PGCLCs closely resembled those of in vivo produced PGCs, consistent with efficient reprogramming of methylation and genomic imprinting. These results demonstrate that amplification of germ cells through parthenogenesis faithfully maintains maternal imprinting, offering a promising route for deriving functional oocytes and having potential in rebuilding ovarian endocrine function.

  • LETTER
    Rujin Huang, Jinyang Liu, Xi Chen, Ying Zhi, Shuangyuan Ding, Jia Ming, Yulin Li, Yangming Wang, Jie Na
    Protein & Cell, 2023, 14(2): 153-157. https://doi.org/10.1093/procel/pwac019
  • RESEARCH ARTICLE
    Chun-Chun Gao, Man Li, Wei Deng, Chun-Hui Ma, Yu-Sheng Chen, Yong-Qiao Sun, Tingfu Du, Qian-Lan Liu, Wen-Jie Li, Bing Zhang, Lihong Sun, Si-Meng Liu, Fengli Li, Feifei Qi, Yajin Qu, Xinyang Ge, Jiangning Liu, Peng Wang, Yamei Niu, Zhiyong Liang, Yong-Liang Zhao, Bo Huang, Xiao-Zhong Peng, Ying Yang, Chuan Qin, Wei-Min Tong, Yun-Gui Yang
    Protein & Cell, 2022, 13(12): 920-939. https://doi.org/10.1007/s13238-022-00915-5

    SARS-CoV-2 infection causes complicated clinical manifestations with variable multi-organ injuries, however, the underlying mechanism, in particular immune responses in different organs, remains elusive. In this study, comprehensive transcriptomic alterations of 14 tissues from rhesus macaque infected with SARS-CoV-2 were analyzed. Compared to normal controls, SARS-CoV-2 infection resulted in dysregulation of genes involving diverse functions in various examined tissues/organs, with drastic transcriptomic changes in cerebral cortex and right ventricle. Intriguingly, cerebral cortex exhibited a hyperinflammatory state evidenced by significant upregulation of inflammation response-related genes. Meanwhile, expressions of coagulation, angiogenesis and fibrosis factors were also up-regulated in cerebral cortex. Based on our findings, neuropilin 1 (NRP1), a receptor of SARS-CoV-2, was significantly elevated in cerebral cortex post infection, accompanied by active immune response releasing inflammatory factors and signal transmission among tissues, which enhanced infection of the central nervous system (CNS) in a positive feedback way, leading to viral encephalitis. Overall, our study depicts a multi-tissue/organ transcriptomic landscapes of rhesus macaque with early infection of SARS-CoV-2, and provides important insights into the mechanistic basis for COVID-19-associated clinical complications.

  • REVIEW
    Yuliang Feng, Xingguo Liu, Siim Pauklin
    Protein & Cell, 2021, 12(6): 440-454. https://doi.org/10.1007/s13238-020-00819-2

    Dedifferentiation of cell identity to a progenitor-like or stem cell-like state with increased cellular plasticity is frequently observed in cancer formation. During this process, a subpopulation of cells in tumours acquires a stem cell-like state partially resembling to naturally occurring pluripotent stem cells that are temporarily present during early embryogenesis. Such characteristics allow these cancer stem cells (CSCs) to give rise to the whole tumour with its entire cellular heterogeneity and thereby support metastases formation while being resistant to current cancer therapeutics. Cancer development and progression are demarcated by transcriptional dysregulation. In this article, we explore the epigenetic mechanisms shaping gene expression during tumorigenesis and cancer stem cell formation, with an emphasis on 3D chromatin architecture. Comparing the pluripotent stem cell state and epigenetic reprogramming to dedifferentiation in cellular transformation provides intriguing insight to chromatin dynamics. We suggest that the 3D chromatin architecture could be used as a target for re-sensitizing cancer stem cells to therapeutics.

  • CORRECTION
    Xin Huang, Nazym Bashkenova, Jihong Yang, Dan Li, Jianlong Wang
    Protein & Cell, 2021, 12(6): 511-513. https://doi.org/10.1007/s13238-020-00817-4
  • LETTER
    Mingyue Ma, Zhenxing Zhong, Yuwen Zhu, Yuan Gu, Ruxin Jin, Zhipeng Meng, Yu Wang, Fa-Xing Yu
    Protein & Cell, 2023, 14(2): 137-142. https://doi.org/10.1093/procel/pwac005
  • RESEARCH ARTICLE
    Shi Wei, Miaomiao Dai, Chi Zhang, Kai Teng, Fengwei Wang, Hongbo Li, Weipeng Sun, Zihao Feng, Tiebang Kang, Xinyuan Guan, Ruihua Xu, Muyan Cai, Dan Xie
    Protein & Cell, 2021, 12(10): 788-809. https://doi.org/10.1007/s13238-020-00766-y

    Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and is the fourth-leading cause of cancer-related deaths worldwide. HCC is refractory to many standard cancer treatments and the prognosis is often poor, highlighting a pressing need to identify biomarkers of aggressiveness and potential targets for future treatments. Kinesin family member 2C (KIF2C) is reported to be highly expressed in several human tumors. Nevertheless, the molecular mechanisms underlying the role of KIF2C in tumor development and progression have not been investigated. In this study, we found that KIF2C expression was significantly upregulated in HCC, and that KIF2C up-regulation was associated with a poor prognosis. Utilizing both gain and loss of function assays, we showed that KIF2C promoted HCC cell proliferation, migration, invasion, and metastasis both in vitro and in vivo. Mechanistically, we identified TBC1D7 as a binding partner of KIF2C, and this interaction disrupts the formation of the TSC complex, resulting in the enhancement of mammalian target of rapamycin complex1 (mTORC1) signal transduction. Additionally, we found that KIF2C is a direct target of the Wnt/β-catenin pathway, and acts as a key factor in mediating the crosstalk between Wnt/β-catenin and mTORC1 signaling. Thus, the results of our study establish a link between Wnt/β-catenin and mTORC1 signaling, which highlights the potential of KIF2C as a therapeutic target for the treatment of HCC.

  • RESEARCH ARTICLE
    Kuisheng Liu, Xiaocui Xu, Dandan Bai, Yanhe Li, Yalin Zhang, Yanping Jia, Mingyue Guo, Xiaoxiao Han, Yingdong Liu, Yifan Sheng, Xiaochen Kou, Yanhong Zhao, Jiqing Yin, Sheng Liu, Jiayu Chen, Hong Wang, Yixuan Wang, Wenqiang Liu, Shaorong Gao
    Protein & Cell, 2023, 14(4): 262-278. https://doi.org/10.1093/procel/pwac029

    Self-organized blastoids from extended pluripotent stem (EPS) cells possess enormous potential for investigating postimplantation embryo development and related diseases. However, the limited ability of postimplantation development of EPS-blastoids hinders its further application. In this study, single-cell transcriptomic analysis indicated that the “trophectoderm (TE)-like structure” of EPS-blastoids was primarily composed of primitive endoderm (PrE)-related cells instead of TE-related cells. We further identified PrE-like cells in EPS cell culture that contribute to the blastoid formation with TE-like structure. Inhibition of PrE cell differentiation by inhibiting MEK signaling or knockout of Gata6 in EPS cells markedly suppressed EPS-blastoid formation. Furthermore, we demonstrated that blastocyst-like structures reconstituted by combining the EPS-derived bilineage embryo-like structure (BLES) with either tetraploid embryos or tetraploid TE cells could implant normally and develop into live fetuses. In summary, our study reveals that TE improvement is critical for constructing a functional embryo using stem cells in vitro.

  • RESEARCH ARTICLE
    Xiaojuan Liang, Cong Tao, Jianfei Pan, Lilan Zhang, Lulu Liu, Ying Zhao, Yiping Fan, Chunwei Cao, Jiali Liu, Jin Zhang, Sin Man Lam, Guanghou Shui, Wanzhu Jin, Wei Li, Jianguo Zhao, Kui Li, Yanfang Wang
    Protein & Cell, 2021, 12(6): 475-492. https://doi.org/10.1007/s13238-020-00770-2

    RNF20, an E3 ligase critical for monoubiquitination of histone H2B at lysine 120 (H2Bub), has been implicated in the regulation of various cellar processes; however, its physiological roles in adipocytes remain poorly characterized. Here, we report that the adipocyte-specific knockout of Rnf20 (ASKO) in mice led to progressive fat loss, organomegaly and hyperinsulinemia. Despite signs of hyperinsulinemia, normal insulin sensitivity and improved glucose tolerance were observed in the young and aged CD-fed ASKO mice. In addition, high-fat dietfed ASKO mice developed severe liver steatosis. Moreover, we observed that the ASKO mice were extremely sensitive to a cold environment due to decreased expression levels of brown adipose tissue (BAT) selective genes, including uncoupling protein 1 (Ucp1), and impaired mitochondrial functions. Significantly decreased levels of peroxisome proliferator-activated receptor gamma (Pparγ) were observed in the gonadal white adipose tissues (gWAT) from the ASKO mice, suggesting that Rnf20 regulates adipogenesis, at least in part, through Pparγ. Rosiglitazone-treated ASKO mice exhibited increased fat mass compared to that of the non-treated ASKO mice. Collectively, our results illustrate the critical role of RNF20 in control of white and brown adipose tissue development and physiological function.

  • RESEARCH ARTICLE
    Yulin Xu, Xiangjun Zeng, Mingming Zhang, Binsheng Wang, Xin Guo, Wei Shan, Shuyang Cai, Qian Luo, Honghu Li, Xia Li, Xue Li, Hao Zhang, Limengmeng Wang, Yu Lin, Lizhen Liu, Yanwei Li, Meng Zhang, Xiaohong Yu, Pengxu Qian, He Huang
    Protein & Cell, 2022, 13(11): 808-824. https://doi.org/10.1007/s13238-021-00900-4

    Although widely applied in treating hematopoietic malignancies, transplantation of hematopoietic stem/progenitor cells (HSPCs) is impeded by HSPC shortage. Whether circulating HSPCs (cHSPCs) in steady-state blood could be used as an alternative source remains largely elusive. Here we develop a three-dimensional culture system (3DCS) including arginine, glycine, aspartate, and a series of factors. Fourteen-day culture of peripheral blood mononuclear cells (PBMNCs) in 3DCS led to 125- and 70-fold increase of the frequency and number of CD34+ cells. Further, 3DCS-expanded cHSPCs exhibited the similar reconstitution rate compared to CD34+ HSPCs in bone marrow. Mechanistically, 3DCS fabricated an immunomodulatory niche, secreting cytokines as TNF to support cHSPC survival and proliferation. Finally, 3DCS could also promote the expansion of cHSPCs in patients who failed in HSPC mobilization. Our 3DCS successfully expands rare cHSPCs, providing an alternative source for the HSPC therapy, particularly for the patients/donors who have failed in HSPC mobilization.

  • RESEARCH ARTICLE
    Yirui Cheng, Xin Lu, Fan Li, Zhuo Chen, Yanshuang Zhang, Qing Han, Qingyu Zeng, Tingyu Wu, Ziming Li, Shun Lu, Cecilia Williams, Weiliang Xia
    Protein & Cell, 2023, 14(2): 123-136. https://doi.org/10.1093/procel/pwac017

    NDFIP1 has been previously reported as a tumor suppressor in multiple solid tumors, but the function of NDFIP1 in NSCLC and the underlying mechanism are still unknown. Besides, the WW domain containing proteins can be recognized by NDFIP1, resulted in the loading of the target proteins into exosomes. However, whether WW domain-containing transcription regulator 1 (WWTR1, also known as TAZ) can be packaged into exosomes by NDFIP1 and if so, whether the release of this oncogenic protein via exosomes has an effect on tumor development has not been investigated to any extent. Here, we first found that NDFIP1 was low expressed in NSCLC samples and cell lines, which is associated with shorter OS. Then, we confirmed the interaction between TAZ and NDFIP1, and the existence of TAZ in exosomes, which requires NDFIP1. Critically, knockout of NDFIP1 led to TAZ accumulation with no change in its mRNA level and degradation rate. And the cellular TAZ level could be altered by exosome secretion. Furthermore, NDFIP1 inhibited proliferation in vitro and in vivo, and silencing TAZ eliminated the increase of proliferation caused by NDFIP1 knockout. Moreover, TAZ was negatively correlated with NDFIP1 in subcutaneous xenograft model and clinical samples, and the serum exosomal TAZ level was lower in NSCLC patients. In summary, our data uncover a new tumor suppressor, NDFIP1 in NSCLC, and a new exosome-related regulatory mechanism of TAZ.

  • LETTER
    Sheng Zhang, Zeming Wu, Yue Shi, Si Wang, Jie Ren, Zihui Yu, Daoyuan Huang, Kaowen Yan, Yifang He, Xiaoqian Liu, Qianzhao Ji, Beibei Liu, Zunpeng Liu, Jing Qu, Guang-Hui Liu, Weimin Ci, Xiaoqun Wang, Weiqi Zhang
    Protein & Cell, 2022, 13(12): 954-960. https://doi.org/10.1007/s13238-022-00914-6
  • REVIEW
    Linsen Li, Mindan Tong, Yuhui Fu, Fang Chen, Shen Zhang, Hanmo Chen, Xi Ma, Defa Li, Xiaoxia Liu, Qing Zhong
    Protein & Cell, 2021, 12(7): 520-544. https://doi.org/10.1007/s13238-020-00793-9

    Autophagy is essential for the maintenance of cellular homeostasis and its dysfunction has been linked to various diseases. Autophagy is a membrane driven process and tightly regulated by membrane-associated proteins. Here, we summarized membrane lipid composition, and membrane-associated proteins relevant to autophagy from a spatiotemporal perspective. In particular, we focused on three important membrane remodeling processes in autophagy, lipid transfer for phagophore elongation, membrane scission for phagophore closure, and autophagosome-lysosome membrane fusion. We discussed the significance of the discoveries in this field and possible avenues to follow for future studies. Finally, we summarized the membrane-associated biochemical techniques and assays used to study membrane properties, with a discussion of their applications in autophagy.

  • RESEARCH ARTICLE
    Shuofeng Yuan, Xiaopan Gao, Kaiming Tang, Jian-Piao Cai, Menglong Hu, Peng Luo, Lei Wen, Zi-Wei Ye, Cuiting Luo, Jessica Oi-Ling Tsang, Chris Chun-Yiu Chan, Yaoqiang Huang, Jianli Cao, Ronghui Liang, Zhenzhi Qin, Bo Qin, Feifei Yin, Hin Chu, Dong-Yan Jin, Ren Sun, Jasper Fuk-Woo Chan, Sheng Cui, Kwok-Yung Yuen
    Protein & Cell, 2022, 13(12): 940-953. https://doi.org/10.1007/s13238-022-00909-3

    The emergence of SARS-CoV-2 variants of concern and repeated outbreaks of coronavirus epidemics in the past two decades emphasize the need for next-generation pan-coronaviral therapeutics. Drugging the multi-functional papain-like protease (PLpro) domain of the viral nsp3 holds promise. However, none of the known coronavirus PLpro inhibitors has been shown to be in vivo active. Herein, we screened a structurally diverse library of 50,080 compounds for potential coronavirus PLpro inhibitors and identified a noncovalent lead inhibitor F0213 that has broad-spectrum anti-coronaviral activity, including against the Sarbecoviruses (SARS-CoV-1 and SARS-CoV-2), Merbecovirus (MERS-CoV), as well as the Alphacoronavirus (hCoV-229E and hCoV-OC43). Importantly, F0213 confers protection in both SARS-CoV-2-infected hamsters and MERS-CoV-infected human DPP4-knockin mice. F0213 possesses a dual therapeutic functionality that suppresses coronavirus replication via blocking viral polyprotein cleavage, as well as promoting antiviral immunity by antagonizing the PLpro deubiquitinase activity. Despite the significant difference of substrate recognition, mode of inhibition studies suggest that F0213 is a competitive inhibitor against SARS2-PLpro via binding with the 157K amino acid residue, whereas an allosteric inhibitor of MERS-PLpro interacting with its 271E position. Our proof-ofconcept findings demonstrated that PLpro is a valid target for the development of broad-spectrum anti-coronavirus agents. The orally administered F0213 may serve as a promising lead compound for combating the ongoing COVID-19 pandemic and future coronavirus outbreaks.

  • RECOLLECTION
    Xunfeng Xu, Lei Fu
    Protein & Cell, 2021, 12(12): 903-905. https://doi.org/10.1007/s13238-020-00758-y
  • RECOLLECTION
    Jia-huai Wang
    Protein & Cell, 2022, 13(12): 869-872. https://doi.org/10.1007/s13238-022-00916-4
  • RECOLLECTION
    Huan Liu, Kaijing Huang, Xuefan Yuan, Hao Cheng
    Protein & Cell, 2023, 14(2): 79-83. https://doi.org/10.1093/procel/pwac047
  • RESEARCH ARTICLE
    Zheng Li, Fang Yao, Peng Yu, Dandan Li, Mingzhi Zhang, Lin Mao, Xiaomeng Shen, Zongna Ren, Li Wang, Bingying Zhou
    Protein & Cell, 2022, 13(11): 842-862. https://doi.org/10.1007/s13238-022-00908-4

    Postnatal heart maturation is the basis of normal cardiac function and provides critical insights into heart repair and regenerative medicine. While static snapshots of the maturing heart have provided much insight into its molecular signatures, few key events during postnatal cardiomyocyte maturation have been uncovered. Here, we report that cardiomyocytes (CMs) experience epigenetic and transcriptional decline of cardiac gene expression immediately after birth, leading to a transition state of CMs at postnatal day 7 (P7) that was essential for CM subtype specification during heart maturation. Large-scale single-cell analysis and genetic lineage tracing confirm the presence of transition state CMs at P7 bridging immature state and mature states. Silencing of key transcription factor JUN in P1-hearts significantly repressed CM transition, resulting in perturbed CM subtype proportions and reduced cardiac function in mature hearts. In addition, transplantation of P7-CMs into infarcted hearts exhibited cardiac repair potential superior to P1-CMs. Collectively, our data uncover CM state transition as a key event in postnatal heart maturation, which not only provides insights into molecular foundations of heart maturation, but also opens an avenue for manipulation of cardiomyocyte fate in disease and regenerative medicine.

  • REVIEW
    Yong-Xin Liu, Yuan Qin, Tong Chen, Meiping Lu, Xubo Qian, Xiaoxuan Guo, Yang Bai
    Protein & Cell, 2021, 12(5): 315-330. https://doi.org/10.1007/s13238-020-00724-8

    Advances in high-throughput sequencing (HTS) have fostered rapid developments in the field of microbiome research, and massive microbiome datasets are now being generated. However, the diversity of software tools and the complexity of analysis pipelines make it difficult to access this field. Here, we systematically summarize the advantages and limitations of microbiome methods. Then, we recommend specific pipelines for amplicon and metagenomic analyses, and describe commonly-used software and databases, to help researchers select the appropriate tools. Furthermore, we introduce statistical and visualization methods suitable for microbiome analysis, including alpha- and betadiversity, taxonomic composition, difference comparisons, correlation, networks, machine learning, evolution, source tracing, and common visualization styles to help researchers make informed choices. Finally, a stepby-step reproducible analysis guide is introduced. We hope this review will allow researchers to carry out data analysis more effectively and to quickly select the appropriate tools in order to efficiently mine the biological significance behind the data.

  • RECOLLECTION
    Ting Shi, Lei Fu
    Protein & Cell, 2022, 13(7): 473-475. https://doi.org/10.1007/s13238-020-00759-x
  • REVIEW
    Peng Yin, Shihua Li, Xiao-Jiang Li, Weili Yang
    Protein & Cell, 2022, 13(10): 707-720. https://doi.org/10.1007/s13238-022-00912-8

    Animal models are essential for investigating the pathogenesis and developing the treatment of human diseases. Identification of genetic mutations responsible for neurodegenerative diseases has enabled the creation of a large number of small animal models that mimic genetic defects found in the affected individuals. Of the current animal models, rodents with genetic modifications are the most commonly used animal models and provided important insights into pathogenesis. However, most of genetically modified rodent models lack overt neurodegeneration, imposing challenges and obstacles in utilizing them to rigorously test the therapeutic effects on neurodegeneration. Recent studies that used CRISPR/Cas9-targeted large animal (pigs and monkeys) have uncovered important pathological events that resemble neurodegeneration in the patient’s brain but could not be produced in small animal models. Here we highlight the unique nature of large animals to model neurodegenerative diseases as well as the limitations and challenges in establishing large animal models of neurodegenerative diseases, with focus on Huntington disease, Amyotrophic lateral sclerosis, and Parkinson diseases. We also discuss how to use the important pathogenic insights from large animal models to make rodent models more capable of recapitulating important pathological features of neurodegenerative diseases.

  • CORRECTION
    Qun Chu, Feifei Liu, Yifang He, Xiaoyu Jiang, Yusheng Cai, Zeming Wu, Kaowen Yan, Lingling Geng, Yichen Zhang, Huyi Feng, Kaixin Zhou, Si Wang, Weiqi Zhang, Guang-Hui Liu, Shuai Ma, Jing Qu, Moshi Song
    Protein & Cell, 2022, 13(12): 961. https://doi.org/10.1007/s13238-022-00917-3