首页 > 最新文献

Inflammation and Regeneration最新文献

英文 中文
Autoinflammatory disease: clinical perspectives and therapeutic strategies. 自身炎症性疾病:临床观点和治疗策略。
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-12-02 DOI: 10.1186/s41232-022-00217-7
Atsushi Kawakami, Endo Yushiro, Koga Tomohiro, Yoshiura Koh-Ichiro, Migita Kiyoshi

The molecular platforms of the innate immune system are essential to recognize pathologic external factors that are crucial to differentiate these danger signals from host motifs. A set of sensors recognizing pathologic factors is present and defined as a membrane-bound family of Toll-like receptors as well as the cytosolic ones including the family of nucleotide-binding domain leucine-rich repeat proteins. In this regard, the inflammasomes have been identified as an innate immune sensor toward pathologic external factors as well as endogenous damage-associated molecular pattern signals transducing from the above-mentioned receptors to gene expressions. Recent research has shown novel findings in inflammasome biology and genetics which lead to the alteration of diagnosis and management in autoinflammatory diseases as well as developing novel treatments, including the examples of nucleotide-binding domain leucine-rich repeat proteins-inflammasomes and pyrin-inflammasomes. The pyrin protein is encoded by the Mediterranean Fever gene on chromosome 16 that acts as a major regulatory component of the inflammasome, and is responsible for familial Mediterranean fever. We have recently examined the whole nucleotide sequence of the Mediterranean Fever gene in Japanese familial Mediterranean fever patients and revealed single nucleotide variants associated with the susceptibility of familial Mediterranean fever from a nation-wide survey by the next-generation sequencing. In a cytokine profile analysis of familial Mediterranean fever patients, we have found that interleukin-6 is considered to be one of the most crucial cytokines in familial Mediterranean fever attack since interleukin-6 had the best performance for distinguishing familial Mediterranean fever in attack from healthy controls or familial Mediterranean fever in remission, and in vitro interleukin-6 production is regulated by microRNAs-204-3p/phosphoinositide 3-kinase g pathway. Accordingly, we have been investigating the efficacy and safety of anti-human interleukin-6 receptor monoclonal antibody, tocilizumab, in patients with familial Mediterranean fever refractory or intolerant to colchicine through an investigator-initiated clinical trial supported by the Japan Agency for Medical Research and Development. Like interleukin-1b, interleukin-18 can be processed by caspase-1 and proteinase-3 to be activated within the inflammasomes. We have also found the importance of interleukin-18 in several autoinflammatory conditions. Recently, the concept of autoinflammation is widely distributed into many common diseases; thus, the attention to a wide spectrum of diseases MEFV gene deeply involved is required.

先天免疫系统的分子平台对于识别病理外部因素至关重要,这些外部因素对于区分这些危险信号与宿主基序至关重要。一组识别病理因子的传感器存在,并被定义为膜结合的toll样受体家族以及包括核苷酸结合结构域富含亮氨酸重复蛋白家族的细胞质受体。在这方面,炎性小体已被确定为对病理性外部因素的先天免疫传感器,以及从上述受体到基因表达的内源性损伤相关分子模式信号转导。最近的研究显示了炎症小体生物学和遗传学的新发现,这些发现导致了自身炎症疾病的诊断和管理的改变,以及开发新的治疗方法,包括核苷酸结合结构域富含亮氨酸的重复蛋白-炎症小体和pyrin-炎症小体的例子。pyrin蛋白由16号染色体上的地中海热基因编码,该基因是炎症小体的主要调节成分,并负责家族性地中海热。我们最近检查了日本家族性地中海热患者的地中海热基因的全核苷酸序列,并通过下一代测序在全国范围内调查发现了与家族性地中海热易感性相关的单核苷酸变异。在对家族性地中海热患者的细胞因子谱分析中,我们发现白细胞介素-6被认为是家族性地中海热发作中最重要的细胞因子之一,因为白细胞介素-6在区分家族性发作型地中海热与健康对照或缓解型家族性地中海热方面的表现最好,并且体外白细胞介素-6的产生受micrornas - 2043p /磷酸肌肽3-激酶g途径的调控。因此,我们一直在通过一项由日本医学研究与发展机构支持的研究者发起的临床试验,研究抗人白细胞介素-6受体单克隆抗体tocilizumab对秋水仙碱难治性或不耐受的家族性地中海热患者的疗效和安全性。与白细胞介素-1b一样,白细胞介素-18也可通过caspase-1和蛋白酶-3在炎性小体内活化。我们还发现白细胞介素-18在几种自身炎症条件下的重要性。近年来,自体炎症的概念被广泛应用于许多常见疾病;因此,需要关注MEFV基因深涉的广谱疾病。
{"title":"Autoinflammatory disease: clinical perspectives and therapeutic strategies.","authors":"Atsushi Kawakami,&nbsp;Endo Yushiro,&nbsp;Koga Tomohiro,&nbsp;Yoshiura Koh-Ichiro,&nbsp;Migita Kiyoshi","doi":"10.1186/s41232-022-00217-7","DOIUrl":"https://doi.org/10.1186/s41232-022-00217-7","url":null,"abstract":"<p><p>The molecular platforms of the innate immune system are essential to recognize pathologic external factors that are crucial to differentiate these danger signals from host motifs. A set of sensors recognizing pathologic factors is present and defined as a membrane-bound family of Toll-like receptors as well as the cytosolic ones including the family of nucleotide-binding domain leucine-rich repeat proteins. In this regard, the inflammasomes have been identified as an innate immune sensor toward pathologic external factors as well as endogenous damage-associated molecular pattern signals transducing from the above-mentioned receptors to gene expressions. Recent research has shown novel findings in inflammasome biology and genetics which lead to the alteration of diagnosis and management in autoinflammatory diseases as well as developing novel treatments, including the examples of nucleotide-binding domain leucine-rich repeat proteins-inflammasomes and pyrin-inflammasomes. The pyrin protein is encoded by the Mediterranean Fever gene on chromosome 16 that acts as a major regulatory component of the inflammasome, and is responsible for familial Mediterranean fever. We have recently examined the whole nucleotide sequence of the Mediterranean Fever gene in Japanese familial Mediterranean fever patients and revealed single nucleotide variants associated with the susceptibility of familial Mediterranean fever from a nation-wide survey by the next-generation sequencing. In a cytokine profile analysis of familial Mediterranean fever patients, we have found that interleukin-6 is considered to be one of the most crucial cytokines in familial Mediterranean fever attack since interleukin-6 had the best performance for distinguishing familial Mediterranean fever in attack from healthy controls or familial Mediterranean fever in remission, and in vitro interleukin-6 production is regulated by microRNAs-204-3p/phosphoinositide 3-kinase g pathway. Accordingly, we have been investigating the efficacy and safety of anti-human interleukin-6 receptor monoclonal antibody, tocilizumab, in patients with familial Mediterranean fever refractory or intolerant to colchicine through an investigator-initiated clinical trial supported by the Japan Agency for Medical Research and Development. Like interleukin-1b, interleukin-18 can be processed by caspase-1 and proteinase-3 to be activated within the inflammasomes. We have also found the importance of interleukin-18 in several autoinflammatory conditions. Recently, the concept of autoinflammation is widely distributed into many common diseases; thus, the attention to a wide spectrum of diseases MEFV gene deeply involved is required.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9717532/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10814234","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 5
Cytokines and cytokine receptors as targets of immune-mediated inflammatory diseases-RA as a role model. 细胞因子和细胞因子受体作为免疫介导的炎症性疾病的靶标--以 RA 为榜样。
IF 5 3区 医学 Q2 IMMUNOLOGY Pub Date : 2022-12-01 DOI: 10.1186/s41232-022-00221-x
Tsutomu Takeuchi

Recent advances in our understanding in the immune-mediated inflammatory diseases (IMID) are explored and promoted by the targeted treatment. Among these targets, cytokines and cytokine receptors have become the good candidates for the drug development. In this review, the cytokine and cytokine receptors, which are approved in IMID, are overviewed, and modalities of the treatment, the role of cytokines and cytokine receptors in each disease, and the updated molecular information by modern technologies in rheumatoid arthritis as a role model are shown and discussed for the future perspectives.

最近,我们对免疫介导的炎症性疾病(IMID)的认识取得了进展,并通过靶向治疗进行了探索和推广。在这些靶点中,细胞因子和细胞因子受体已成为药物开发的最佳候选靶点。在这篇综述中,综述了在免疫介导的炎症性疾病(IMID)中被批准使用的细胞因子和细胞因子受体,并展示了治疗方式、细胞因子和细胞因子受体在每种疾病中的作用,以及以类风湿性关节炎为例,通过现代技术更新的分子信息,并对未来前景进行了讨论。
{"title":"Cytokines and cytokine receptors as targets of immune-mediated inflammatory diseases-RA as a role model.","authors":"Tsutomu Takeuchi","doi":"10.1186/s41232-022-00221-x","DOIUrl":"10.1186/s41232-022-00221-x","url":null,"abstract":"<p><p>Recent advances in our understanding in the immune-mediated inflammatory diseases (IMID) are explored and promoted by the targeted treatment. Among these targets, cytokines and cytokine receptors have become the good candidates for the drug development. In this review, the cytokine and cytokine receptors, which are approved in IMID, are overviewed, and modalities of the treatment, the role of cytokines and cytokine receptors in each disease, and the updated molecular information by modern technologies in rheumatoid arthritis as a role model are shown and discussed for the future perspectives.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":5.0,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9713106/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10446860","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Quercetin enhances survival and axonal regeneration of motoneurons after spinal root avulsion and reimplantation: experiments in a rat model of brachial plexus avulsion. 槲皮素促进脊髓根撕脱和再植后运动神经元的存活和轴突再生:臂丛撕脱大鼠模型的实验。
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-12-01 DOI: 10.1186/s41232-022-00245-3
Yanfeng Huang, Xie Zhang, Qionghui Huang, Yaoxing Dou, Chang Qu, Qingqing Xu, Qiuju Yuan, Yan-Fang Xian, Zhi-Xiu Lin

Background: Brachial plexus avulsion (BPA) physically involves the detachment of spinal nerve roots themselves and the associated spinal cord segment, leading to permanent paralysis of motor function of the upper limb. Root avulsion induces severe pathological changes, including inflammatory reaction, oxidative damage, and finally massive motoneuron apoptosis. Quercetin (QCN), a polyphenolic flavonoid found in abundance in fruit and vegetables, has been reported to possess anti-oxidative, anti-inflammatory, and neuroprotective effects in many experimental models of both central nervous system (CNS) and peripheral nervous system (PNS) disorders. The purpose of this study was to investigate whether QCN could improve motor function recovery after C5-7 ventral root avulsion and C6 reimplantation in a rat model of BPA.

Methods: The right fifth cervical (C5) to C7 ventral roots were avulsed followed by re-implantation of only C6 to establish the spinal root avulsion plus re-implantation model in rats. After surgery, rats were treated with QCN (25, 50, and 100 mg/kg) by gavage for 2 or 8 consecutive weeks. The effects of QCN were assessed using behavior test (Terzis grooming test, TGT) and histological evaluation. The molecular mechanisms were determined by immunohistochemistry analysis and western blotting.

Results: Our results demonstrated that QCN significantly expedited motor function recovery in the forelimb as shown by the increased Terzis grooming test score, and accelerated motor axon regeneration as evidenced by the ascending number of Fluoro-Ruby-labeled and P75-positive regenerative motoneurons. The raised ChAT-immunopositive and cresyl violet-stained neurons indicated the enhanced survival of motoneurons by QCN administration. Furthermore, QCN treatment markedly alleviated muscle atrophy, restored functional motor endplates in biceps and inhibited the microglial and astroglia activation via modulating Nrf2/HO-1 and neurotrophin/Akt/MAPK signaling pathway.

Conclusions: Taken together, these findings have for the first time unequivocally indicated that QCN has promising potential for further development into a novel therapeutic in conjunction with reimplantation surgery for the treatment of BPA. .

背景:臂丛撕脱症(Brachial plexus avulsion, BPA)在物理上涉及脊神经根本身和相关脊髓段的脱离,导致上肢运动功能的永久性瘫痪。根撕脱引起严重的病理改变,包括炎症反应、氧化损伤,最终导致大量运动神经元凋亡。槲皮素(QCN)是一种在水果和蔬菜中大量发现的多酚类黄酮,据报道,槲皮素在中枢神经系统(CNS)和周围神经系统(PNS)疾病的许多实验模型中具有抗氧化、抗炎和神经保护作用。本研究的目的是探讨QCN是否能改善大鼠双酚a模型C5-7腹根撕脱和C6再植后的运动功能恢复。方法:取大鼠右侧第五颈椎(C5) ~ C7椎弓根撕脱后仅C6椎弓根再植入术,建立脊髓根撕脱+再植入术模型。术后大鼠分别给予QCN(25、50、100 mg/kg)灌胃,连续2、8周。采用行为学测试(Terzis grooming test, TGT)和组织学评估QCN的效果。通过免疫组织化学和免疫印迹分析确定其分子机制。结果:我们的研究结果表明,QCN显著加速了前肢运动功能的恢复,这可以从Terzis修饰测试分数的增加中看出;QCN显著加速了前肢运动轴突的再生,这可以从氟红宝石标记和p75阳性再生运动神经元的数量增加中看出。chat免疫阳性和甲酚紫染色神经元升高,表明给药后运动神经元存活增强。此外,QCN通过调节Nrf2/HO-1和神经营养因子/Akt/MAPK信号通路,显著缓解肌肉萎缩,恢复二头肌功能运动终板,抑制小胶质细胞和星形胶质细胞的激活。结论:综上所述,这些发现首次明确表明,QCN有希望进一步发展成为一种新的治疗方法,与再植手术一起治疗BPA。
{"title":"Quercetin enhances survival and axonal regeneration of motoneurons after spinal root avulsion and reimplantation: experiments in a rat model of brachial plexus avulsion.","authors":"Yanfeng Huang,&nbsp;Xie Zhang,&nbsp;Qionghui Huang,&nbsp;Yaoxing Dou,&nbsp;Chang Qu,&nbsp;Qingqing Xu,&nbsp;Qiuju Yuan,&nbsp;Yan-Fang Xian,&nbsp;Zhi-Xiu Lin","doi":"10.1186/s41232-022-00245-3","DOIUrl":"https://doi.org/10.1186/s41232-022-00245-3","url":null,"abstract":"<p><strong>Background: </strong>Brachial plexus avulsion (BPA) physically involves the detachment of spinal nerve roots themselves and the associated spinal cord segment, leading to permanent paralysis of motor function of the upper limb. Root avulsion induces severe pathological changes, including inflammatory reaction, oxidative damage, and finally massive motoneuron apoptosis. Quercetin (QCN), a polyphenolic flavonoid found in abundance in fruit and vegetables, has been reported to possess anti-oxidative, anti-inflammatory, and neuroprotective effects in many experimental models of both central nervous system (CNS) and peripheral nervous system (PNS) disorders. The purpose of this study was to investigate whether QCN could improve motor function recovery after C5-7 ventral root avulsion and C6 reimplantation in a rat model of BPA.</p><p><strong>Methods: </strong>The right fifth cervical (C5) to C7 ventral roots were avulsed followed by re-implantation of only C6 to establish the spinal root avulsion plus re-implantation model in rats. After surgery, rats were treated with QCN (25, 50, and 100 mg/kg) by gavage for 2 or 8 consecutive weeks. The effects of QCN were assessed using behavior test (Terzis grooming test, TGT) and histological evaluation. The molecular mechanisms were determined by immunohistochemistry analysis and western blotting.</p><p><strong>Results: </strong>Our results demonstrated that QCN significantly expedited motor function recovery in the forelimb as shown by the increased Terzis grooming test score, and accelerated motor axon regeneration as evidenced by the ascending number of Fluoro-Ruby-labeled and P75-positive regenerative motoneurons. The raised ChAT-immunopositive and cresyl violet-stained neurons indicated the enhanced survival of motoneurons by QCN administration. Furthermore, QCN treatment markedly alleviated muscle atrophy, restored functional motor endplates in biceps and inhibited the microglial and astroglia activation via modulating Nrf2/HO-1 and neurotrophin/Akt/MAPK signaling pathway.</p><p><strong>Conclusions: </strong>Taken together, these findings have for the first time unequivocally indicated that QCN has promising potential for further development into a novel therapeutic in conjunction with reimplantation surgery for the treatment of BPA. .</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9714227/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10327247","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
2D and 3D cultured human umbilical cord-derived mesenchymal stem cell-conditioned medium has a dual effect in type 1 diabetes model in rats: immunomodulation and beta-cell regeneration. 2D和3D培养人脐带间充质干细胞条件培养基对大鼠1型糖尿病模型具有双重作用:免疫调节和β细胞再生。
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-11-30 DOI: 10.1186/s41232-022-00241-7
Basak Isildar, Serbay Ozkan, Merve Ercin, Selda Gezginci-Oktayoglu, Mahmut Oncul, Meral Koyuturk

Background: Type 1 diabetes (T1D) is a T-cell-mediated autoimmune disease characterized by the irreversible destruction of insulin-producing β-cells in pancreatic islets. Helper and cytotoxic T-cells and cytokine production, which is impaired by this process, take a synergetic role in β-cell destruction, and hyperglycemia develops due to insulin deficiency in the body. Mesenchymal stem cells (MSCs) appear like an excellent therapeutic tool for autoimmune diseases with pluripotent, regenerative, and immunosuppressive properties. Paracrine factors released from MSCs play a role in immunomodulation by increasing angiogenesis and proliferation and suppressing apoptosis. In this context, the study aims to investigate the therapeutic effects of MSC's secretomes by conditioned medium (CM) obtained from human umbilical cord-derived MSCs cultured in 2-dimensional (2D) and 3-dimensional (3D) environments in the T1D model.

Methods: First, MSCs were isolated from the human umbilical cord, and the cells were characterized. Then, two different CMs were prepared by culturing MSCs in 2D and 3D environments. The CM contents were analyzed in terms of total protein, IL-4, IL-10, IL-17, and IFN-λ. In vivo studies were performed in Sprague-Dawley-type rats with an autoimmune T1D model, and twelve doses of CM were administered intraperitoneally for 4 weeks within the framework of a particular treatment model. In order to evaluate immunomodulation, the Treg population was determined in lymphocytes isolated from the spleen after sacrification, and IL-4, IL-10, IL-17, and IFN-λ cytokines were analyzed in serum. Finally, β-cell regeneration was evaluated immunohistochemically by labeling Pdx1, Nkx6.1, and insulin markers, which are critical for the formation of β-cells.

Results: Total protein and IL-4 levels were higher in 3D-CM compared to 2D-CM. In vivo results showed that CMs induce the Treg population and regulate cytokine release. When the immunohistochemical results were evaluated together, it was determined that CM application significantly increased the rate of β-cells in the islets. This increase was at the highest level in the 3D-CM applied group.

Conclusion: The dual therapeutic effect of MSC-CM on immunomodulation and homeostasis/regeneration of β-cells in the T1D model has been demonstrated. Furthermore, this effect could be improved by using 3D scaffolds for culturing MSCs while preparing CM.

背景:1型糖尿病(T1D)是一种t细胞介导的自身免疫性疾病,其特征是胰岛中产生胰岛素的β细胞的不可逆破坏。辅助性和细胞毒性t细胞以及细胞因子的产生在这一过程中受损,在β细胞破坏中起协同作用,并且由于体内胰岛素缺乏而产生高血糖。间充质干细胞(MSCs)具有多能性、再生性和免疫抑制特性,是治疗自身免疫性疾病的良好工具。MSCs释放的旁分泌因子通过增加血管生成和增殖,抑制细胞凋亡,发挥免疫调节作用。在此背景下,本研究旨在研究在T1D模型中,在二维(2D)和三维(3D)环境中培养的人脐带源性间充质干细胞获得的条件培养基(CM)对MSC分泌组的治疗作用。方法:首先从人脐带分离骨髓间充质干细胞,并对其进行表征。然后,通过在二维和三维环境中培养MSCs制备两种不同的CMs。以总蛋白、IL-4、IL-10、IL-17和IFN-λ为指标分析CM的含量。在sprague - dawley型自身免疫性T1D模型大鼠中进行体内研究,在特定治疗模型框架内,腹腔注射12剂CM,持续4周。为了评估免疫调节作用,我们测定了牺牲后脾脏分离淋巴细胞中的Treg群体,并分析了血清中的IL-4、IL-10、IL-17和IFN-λ细胞因子。最后,通过标记Pdx1、Nkx6.1和胰岛素标记物来评估β细胞再生,这些标记物对β细胞的形成至关重要。结果:3D-CM的总蛋白和IL-4水平高于2D-CM。体内实验结果表明,CMs可诱导Treg种群,调节细胞因子的释放。将免疫组化结果结合评估,可以确定CM的应用显著增加了胰岛中β-细胞的比例。这种增加在3D-CM应用组中达到最高水平。结论:mscs - cm在T1D模型中具有免疫调节和β-细胞稳态/再生的双重治疗作用。此外,在制备CM的同时使用3D支架培养MSCs可以改善这种效果。
{"title":"2D and 3D cultured human umbilical cord-derived mesenchymal stem cell-conditioned medium has a dual effect in type 1 diabetes model in rats: immunomodulation and beta-cell regeneration.","authors":"Basak Isildar,&nbsp;Serbay Ozkan,&nbsp;Merve Ercin,&nbsp;Selda Gezginci-Oktayoglu,&nbsp;Mahmut Oncul,&nbsp;Meral Koyuturk","doi":"10.1186/s41232-022-00241-7","DOIUrl":"https://doi.org/10.1186/s41232-022-00241-7","url":null,"abstract":"<p><strong>Background: </strong>Type 1 diabetes (T1D) is a T-cell-mediated autoimmune disease characterized by the irreversible destruction of insulin-producing β-cells in pancreatic islets. Helper and cytotoxic T-cells and cytokine production, which is impaired by this process, take a synergetic role in β-cell destruction, and hyperglycemia develops due to insulin deficiency in the body. Mesenchymal stem cells (MSCs) appear like an excellent therapeutic tool for autoimmune diseases with pluripotent, regenerative, and immunosuppressive properties. Paracrine factors released from MSCs play a role in immunomodulation by increasing angiogenesis and proliferation and suppressing apoptosis. In this context, the study aims to investigate the therapeutic effects of MSC's secretomes by conditioned medium (CM) obtained from human umbilical cord-derived MSCs cultured in 2-dimensional (2D) and 3-dimensional (3D) environments in the T1D model.</p><p><strong>Methods: </strong>First, MSCs were isolated from the human umbilical cord, and the cells were characterized. Then, two different CMs were prepared by culturing MSCs in 2D and 3D environments. The CM contents were analyzed in terms of total protein, IL-4, IL-10, IL-17, and IFN-λ. In vivo studies were performed in Sprague-Dawley-type rats with an autoimmune T1D model, and twelve doses of CM were administered intraperitoneally for 4 weeks within the framework of a particular treatment model. In order to evaluate immunomodulation, the Treg population was determined in lymphocytes isolated from the spleen after sacrification, and IL-4, IL-10, IL-17, and IFN-λ cytokines were analyzed in serum. Finally, β-cell regeneration was evaluated immunohistochemically by labeling Pdx1, Nkx6.1, and insulin markers, which are critical for the formation of β-cells.</p><p><strong>Results: </strong>Total protein and IL-4 levels were higher in 3D-CM compared to 2D-CM. In vivo results showed that CMs induce the Treg population and regulate cytokine release. When the immunohistochemical results were evaluated together, it was determined that CM application significantly increased the rate of β-cells in the islets. This increase was at the highest level in the 3D-CM applied group.</p><p><strong>Conclusion: </strong>The dual therapeutic effect of MSC-CM on immunomodulation and homeostasis/regeneration of β-cells in the T1D model has been demonstrated. Furthermore, this effect could be improved by using 3D scaffolds for culturing MSCs while preparing CM.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9710085/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10333098","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 6
Cancer ego-system in glioma: an iron-replenishing niche network systemically self-organized by cancer stem cells. 胶质瘤中的癌症自我系统:一个由癌症干细胞系统自组织的补铁生态位网络。
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-11-30 DOI: 10.1186/s41232-022-00240-8
Kouichi Tabu, Tetsuya Taga

For all living organisms, the adaptation to outside environments is an essential determinant to survive natural and artificial selections and to sustain the whole ecosystem intact with functional biodiversity. Likewise, cancer cells have similar characteristics that evade not only stresses from the host-internal innate and adaptive immune systems but also those from host-externally administered therapeutic interventions. Such selfish characteristics of cancer cells lead to the formation of cancerous ecosystem with a wide variety of phenotypic heterogeneity, which should be called cancer "egosystem" from the host point of view. Recently increasing evidence demonstrates that cancer stem cells (CSCs) are responsible for this cancer egosystem by effectively exploiting host inflammatory and hematopoietic cells and thereby reconstructing their own advantageous niches, which may well be a driving force in cancer recurrence. CSCs are further likely to render multiple niches mutually interconnected and cooperating as a network to support back CSCs themselves. Here, we summarize a recently identified iron-replenishing niche network self-organized by glioma CSCs (GSCs) through remote regulation of host myeloid and erythroid lineage cells. GSCs recruit bone marrow (BM)-derived inflammatory monocytes into tumor parenchyma, facilitate their differentiation into macrophages (Mφs) and skew their polarization into pro-tumoral phenotype, i.e., tumor-associated Mφs (TAMs). Meanwhile, GSCs distantly enhance erythropoiesis in host hematopoietic organs like BM and spleen potentially by secreting some soluble mediators that maintain continuous supply of erythrocytes within tumors. In addition, as normal red pulp Mφs (RPMs) under steady state conditions in spleen recycle iron by phagocytosing the aged or damaged erythrocytes (a/dECs) and release it in time of need, TAMs at least in gliomas phagocytose the hemorrhaged erythrocytes within tumors and potentially serve as a source of iron, an important nutrient indispensable to GSC survival and glioma progression. Taken together, these studies provide the substantial evidence that CSCs have a unique strategy to orchestrate multiple niches as an ecosystem that threatens the host living, which in this sense must be an egosystem. Targeting such an adaptive subpopulation of CSCs could achieve drastic disturbance of the CSC niches and subsequent extinction of malignant neoplasms.

对于所有生物来说,适应外部环境是生存于自然和人为选择以及维持整个生态系统完整和功能性生物多样性的基本决定因素。同样,癌细胞也具有类似的特征,不仅可以逃避宿主内部固有免疫系统和适应性免疫系统的压力,还可以逃避宿主外部给予的治疗干预。癌细胞的这种自私自利的特性导致形成了具有多种表型异质性的癌变生态系统,从宿主的角度来看,这应该被称为癌症的“自我系统”。最近越来越多的证据表明,癌症干细胞(CSCs)通过有效地利用宿主的炎症细胞和造血细胞,从而重建自己的优势生态位,从而负责这种癌症自我系统,这很可能是癌症复发的驱动力。csc进一步有可能使多个利基相互联系并作为一个网络合作,以支持csc本身。在这里,我们总结了最近发现的一个由胶质瘤CSCs (GSCs)通过远程调节宿主髓系和红系细胞自我组织的补铁生态位网络。GSCs将骨髓(BM)来源的炎症单核细胞招募到肿瘤实质,促进其分化为巨噬细胞(m - φs),并使其极化为肿瘤前表型,即肿瘤相关的m - φs (tam)。同时,GSCs通过分泌一些可溶性介质,维持肿瘤内红细胞的持续供应,潜在地远距离促进宿主造血器官如骨髓和脾脏的红细胞生成。此外,在稳态条件下,正常的红浆Mφs (rpm)通过吞噬老化或受损的红细胞(a/dECs)在脾脏循环铁,并在需要时释放出来,tam至少在胶质瘤中吞噬肿瘤内的出血红细胞,并可能作为铁的来源,铁是GSC存活和胶质瘤进展所不可缺少的重要营养物质。综上所述,这些研究提供了大量证据,证明csc有一种独特的策略来协调多个生态位,作为一个威胁宿主生活的生态系统,从这个意义上说,宿主必须是一个自我系统。针对这样一个适应性的CSC亚群可以实现CSC生态位的剧烈干扰和随后的恶性肿瘤的灭绝。
{"title":"Cancer ego-system in glioma: an iron-replenishing niche network systemically self-organized by cancer stem cells.","authors":"Kouichi Tabu,&nbsp;Tetsuya Taga","doi":"10.1186/s41232-022-00240-8","DOIUrl":"https://doi.org/10.1186/s41232-022-00240-8","url":null,"abstract":"<p><p>For all living organisms, the adaptation to outside environments is an essential determinant to survive natural and artificial selections and to sustain the whole ecosystem intact with functional biodiversity. Likewise, cancer cells have similar characteristics that evade not only stresses from the host-internal innate and adaptive immune systems but also those from host-externally administered therapeutic interventions. Such selfish characteristics of cancer cells lead to the formation of cancerous ecosystem with a wide variety of phenotypic heterogeneity, which should be called cancer \"egosystem\" from the host point of view. Recently increasing evidence demonstrates that cancer stem cells (CSCs) are responsible for this cancer egosystem by effectively exploiting host inflammatory and hematopoietic cells and thereby reconstructing their own advantageous niches, which may well be a driving force in cancer recurrence. CSCs are further likely to render multiple niches mutually interconnected and cooperating as a network to support back CSCs themselves. Here, we summarize a recently identified iron-replenishing niche network self-organized by glioma CSCs (GSCs) through remote regulation of host myeloid and erythroid lineage cells. GSCs recruit bone marrow (BM)-derived inflammatory monocytes into tumor parenchyma, facilitate their differentiation into macrophages (Mφs) and skew their polarization into pro-tumoral phenotype, i.e., tumor-associated Mφs (TAMs). Meanwhile, GSCs distantly enhance erythropoiesis in host hematopoietic organs like BM and spleen potentially by secreting some soluble mediators that maintain continuous supply of erythrocytes within tumors. In addition, as normal red pulp Mφs (RPMs) under steady state conditions in spleen recycle iron by phagocytosing the aged or damaged erythrocytes (a/dECs) and release it in time of need, TAMs at least in gliomas phagocytose the hemorrhaged erythrocytes within tumors and potentially serve as a source of iron, an important nutrient indispensable to GSC survival and glioma progression. Taken together, these studies provide the substantial evidence that CSCs have a unique strategy to orchestrate multiple niches as an ecosystem that threatens the host living, which in this sense must be an egosystem. Targeting such an adaptive subpopulation of CSCs could achieve drastic disturbance of the CSC niches and subsequent extinction of malignant neoplasms.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9710158/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10678638","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 3
Next-generation proteomics of serum extracellular vesicles combined with single-cell RNA sequencing identifies MACROH2A1 associated with refractory COVID-19. 新一代血清细胞外囊泡蛋白质组学结合单细胞RNA测序鉴定出与难治性COVID-19相关的MACROH2A1
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-11-30 DOI: 10.1186/s41232-022-00243-5
Takahiro Kawasaki, Yoshito Takeda, Ryuya Edahiro, Yuya Shirai, Mari Nogami-Itoh, Takanori Matsuki, Hiroshi Kida, Takatoshi Enomoto, Reina Hara, Yoshimi Noda, Yuichi Adachi, Takayuki Niitsu, Saori Amiya, Yuta Yamaguchi, Teruaki Murakami, Yasuhiro Kato, Takayoshi Morita, Hanako Yoshimura, Makoto Yamamoto, Daisuke Nakatsubo, Kotaro Miyake, Takayuki Shiroyama, Haruhiko Hirata, Jun Adachi, Yukinori Okada, Atsushi Kumanogoh

Background: The coronavirus disease 2019 (COVID-19) pandemic is widespread; however, accurate predictors of refractory cases have not yet been established. Circulating extracellular vesicles, involved in many pathological processes, are ideal resources for biomarker exploration.

Methods: To identify potential serum biomarkers and examine the proteins associated with the pathogenesis of refractory COVID-19, we conducted high-coverage proteomics on serum extracellular vesicles collected from 12 patients with COVID-19 at different disease severity levels and 4 healthy controls. Furthermore, single-cell RNA sequencing of peripheral blood mononuclear cells collected from 10 patients with COVID-19 and 5 healthy controls was performed.

Results: Among the 3046 extracellular vesicle proteins that were identified, expression of MACROH2A1 was significantly elevated in refractory cases compared to non-refractory cases; moreover, its expression was increased according to disease severity. In single-cell RNA sequencing of peripheral blood mononuclear cells, the expression of MACROH2A1 was localized to monocytes and elevated in critical cases. Consistently, single-nucleus RNA sequencing of lung tissues revealed that MACROH2A1 was highly expressed in monocytes and macrophages and was significantly elevated in fatal COVID-19. Moreover, molecular network analysis showed that pathways such as "estrogen signaling pathway," "p160 steroid receptor coactivator (SRC) signaling pathway," and "transcriptional regulation by STAT" were enriched in the transcriptome of monocytes in the peripheral blood mononuclear cells and lungs, and they were also commonly enriched in extracellular vesicle proteomics.

Conclusions: Our findings highlight that MACROH2A1 in extracellular vesicles is a potential biomarker of refractory COVID-19 and may reflect the pathogenesis of COVID-19 in monocytes.

背景:2019冠状病毒病(COVID-19)大流行广泛存在;然而,对难治性病例的准确预测尚未建立。循环细胞外囊泡参与了许多病理过程,是探索生物标志物的理想资源。方法:为了鉴定难治性COVID-19的潜在血清生物标志物并检测与发病机制相关的蛋白质,我们对12例不同疾病严重程度的COVID-19患者和4名健康对照者的血清细胞外囊泡进行了高覆盖率的蛋白质组学分析。此外,对10例COVID-19患者和5名健康对照者的外周血单个核细胞进行单细胞RNA测序。结果:在鉴定的3046个细胞外囊泡蛋白中,与非难治性病例相比,难治性病例中MACROH2A1的表达显著升高;其表达随疾病严重程度的增加而增加。在外周血单核细胞的单细胞RNA测序中,MACROH2A1的表达定位于单核细胞,在危重病例中表达升高。与此一致,肺组织单核RNA测序结果显示,MACROH2A1在单核细胞和巨噬细胞中高表达,在致死性COVID-19中显著升高。此外,分子网络分析显示,外周血单核细胞和肺的转录组中富集了“雌激素信号通路”、“p160类固醇受体共激活因子(SRC)信号通路”和“STAT转录调控”等通路,细胞外囊泡蛋白质组学中也普遍富集。结论:我们的研究结果表明,细胞外囊泡中的MACROH2A1是难治性COVID-19的潜在生物标志物,可能反映了COVID-19在单核细胞中的发病机制。
{"title":"Next-generation proteomics of serum extracellular vesicles combined with single-cell RNA sequencing identifies MACROH2A1 associated with refractory COVID-19.","authors":"Takahiro Kawasaki,&nbsp;Yoshito Takeda,&nbsp;Ryuya Edahiro,&nbsp;Yuya Shirai,&nbsp;Mari Nogami-Itoh,&nbsp;Takanori Matsuki,&nbsp;Hiroshi Kida,&nbsp;Takatoshi Enomoto,&nbsp;Reina Hara,&nbsp;Yoshimi Noda,&nbsp;Yuichi Adachi,&nbsp;Takayuki Niitsu,&nbsp;Saori Amiya,&nbsp;Yuta Yamaguchi,&nbsp;Teruaki Murakami,&nbsp;Yasuhiro Kato,&nbsp;Takayoshi Morita,&nbsp;Hanako Yoshimura,&nbsp;Makoto Yamamoto,&nbsp;Daisuke Nakatsubo,&nbsp;Kotaro Miyake,&nbsp;Takayuki Shiroyama,&nbsp;Haruhiko Hirata,&nbsp;Jun Adachi,&nbsp;Yukinori Okada,&nbsp;Atsushi Kumanogoh","doi":"10.1186/s41232-022-00243-5","DOIUrl":"https://doi.org/10.1186/s41232-022-00243-5","url":null,"abstract":"<p><strong>Background: </strong>The coronavirus disease 2019 (COVID-19) pandemic is widespread; however, accurate predictors of refractory cases have not yet been established. Circulating extracellular vesicles, involved in many pathological processes, are ideal resources for biomarker exploration.</p><p><strong>Methods: </strong>To identify potential serum biomarkers and examine the proteins associated with the pathogenesis of refractory COVID-19, we conducted high-coverage proteomics on serum extracellular vesicles collected from 12 patients with COVID-19 at different disease severity levels and 4 healthy controls. Furthermore, single-cell RNA sequencing of peripheral blood mononuclear cells collected from 10 patients with COVID-19 and 5 healthy controls was performed.</p><p><strong>Results: </strong>Among the 3046 extracellular vesicle proteins that were identified, expression of MACROH2A1 was significantly elevated in refractory cases compared to non-refractory cases; moreover, its expression was increased according to disease severity. In single-cell RNA sequencing of peripheral blood mononuclear cells, the expression of MACROH2A1 was localized to monocytes and elevated in critical cases. Consistently, single-nucleus RNA sequencing of lung tissues revealed that MACROH2A1 was highly expressed in monocytes and macrophages and was significantly elevated in fatal COVID-19. Moreover, molecular network analysis showed that pathways such as \"estrogen signaling pathway,\" \"p160 steroid receptor coactivator (SRC) signaling pathway,\" and \"transcriptional regulation by STAT\" were enriched in the transcriptome of monocytes in the peripheral blood mononuclear cells and lungs, and they were also commonly enriched in extracellular vesicle proteomics.</p><p><strong>Conclusions: </strong>Our findings highlight that MACROH2A1 in extracellular vesicles is a potential biomarker of refractory COVID-19 and may reflect the pathogenesis of COVID-19 in monocytes.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-11-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9709739/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10678641","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Current understanding of T cell immunity against SARS-CoV-2. 目前对SARS-CoV-2的T细胞免疫的认识。
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-11-29 DOI: 10.1186/s41232-022-00242-6
Xiuyuan Lu, Sho Yamasaki

As an important part of adaptive immunity, T cells are indispensable in the defense against pathogens including viruses. SARS-CoV-2 is a new human coronavirus that occurred at the end of 2019 and has caused the COVID-19 pandemic. Nevertheless, most of the infected patients recovered without any antiviral therapies, suggesting an effective immunity developed in the bodies. T cell immunity responds upon SARS-CoV-2 infection or vaccination and plays crucial roles in eliminating the viruses and generating T cell memory. Specifically, a subpopulation of CD4+ T cells could support the production of anti-SARS-CoV-2 antibodies, and cytotoxic CD8+ T cells are also protective against the infection. SARS-CoV-2-recognizing T cells could be detected in SARS-CoV-2-unexposed donors, but the role of these cross-reactive T cells is still in debate. T cell responses could be diverse across individuals, mainly due to the polymorphism of HLAs. Thus, compared to antibodies, T cell responses are generally less affected by the mutations of SARS-CoV-2 variants. Up to now, a huge number of studies on SARS-CoV-2-responsive T cells have been published. In this review, we introduced some major findings addressing the questions in the main aspects about T cell responses elicited by SARS-CoV-2, to summarize the current understanding of COVID-19.

T细胞作为适应性免疫的重要组成部分,在抵御包括病毒在内的病原体方面发挥着不可缺少的作用。SARS-CoV-2是一种新的人类冠状病毒,于2019年底发生,并引起了COVID-19大流行。然而,大多数受感染的患者在没有任何抗病毒治疗的情况下康复,这表明体内形成了有效的免疫力。T细胞免疫对SARS-CoV-2感染或疫苗接种作出反应,并在消除病毒和产生T细胞记忆方面发挥关键作用。具体来说,CD4+ T细胞亚群可以支持抗sars - cov -2抗体的产生,细胞毒性CD8+ T细胞也可以防止感染。在未暴露于sars - cov -2的供体中可以检测到识别sars - cov -2的T细胞,但这些交叉反应性T细胞的作用仍存在争议。不同个体的T细胞反应可能不同,这主要是由于hla的多态性。因此,与抗体相比,T细胞反应通常受SARS-CoV-2变体突变的影响较小。到目前为止,已经发表了大量关于sars - cov -2反应性T细胞的研究。本文综述了SARS-CoV-2诱导T细胞应答的主要方面的一些主要发现,以总结目前对COVID-19的认识。
{"title":"Current understanding of T cell immunity against SARS-CoV-2.","authors":"Xiuyuan Lu,&nbsp;Sho Yamasaki","doi":"10.1186/s41232-022-00242-6","DOIUrl":"https://doi.org/10.1186/s41232-022-00242-6","url":null,"abstract":"<p><p>As an important part of adaptive immunity, T cells are indispensable in the defense against pathogens including viruses. SARS-CoV-2 is a new human coronavirus that occurred at the end of 2019 and has caused the COVID-19 pandemic. Nevertheless, most of the infected patients recovered without any antiviral therapies, suggesting an effective immunity developed in the bodies. T cell immunity responds upon SARS-CoV-2 infection or vaccination and plays crucial roles in eliminating the viruses and generating T cell memory. Specifically, a subpopulation of CD4<sup>+</sup> T cells could support the production of anti-SARS-CoV-2 antibodies, and cytotoxic CD8<sup>+</sup> T cells are also protective against the infection. SARS-CoV-2-recognizing T cells could be detected in SARS-CoV-2-unexposed donors, but the role of these cross-reactive T cells is still in debate. T cell responses could be diverse across individuals, mainly due to the polymorphism of HLAs. Thus, compared to antibodies, T cell responses are generally less affected by the mutations of SARS-CoV-2 variants. Up to now, a huge number of studies on SARS-CoV-2-responsive T cells have been published. In this review, we introduced some major findings addressing the questions in the main aspects about T cell responses elicited by SARS-CoV-2, to summarize the current understanding of COVID-19.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9706904/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10694571","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 10
Resolving the conflicts around Par2 opposing roles in regeneration by comparing immune-mediated and toxic-induced injuries. 通过比较免疫介导和毒性诱导的损伤,解决Par2在再生中对立作用的冲突。
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-11-29 DOI: 10.1186/s41232-022-00238-2
Gal Reches, Netta R Blondheim Shraga, Florent Carrette, Assaf Malka, Natalia Saleev, Yehuda Gubbay, Offir Ertracht, Izhak Haviv, Linda M Bradley, Fred Levine, Ron Piran

Background: Different factors may lead to hepatitis. Among which are liver inflammation and poisoning. We chose two hepatitis models, typical for these two underlying causes. Thus, we aimed to characterize the role of protease-activated receptor 2 (Par2) in liver regeneration and inflammation to reconcile Par2 conflicting role in many damage models, which sometimes aggravates the induced damage and sometimes alleviates it.

Methods: WT and knockout (Par2KO) mice were injected with concanavalin A (ConA) to induce immune-mediated hepatitis or with carbon tetrachloride (CCl4) to elicit direct hepatic damage. To distinguish the immune component from the liver regenerative response, we conducted bone marrow (BM) replacements of WT and Par2KO mice and repeated the damage models.

Results: ConA injection caused limited damage in Par2KO mice livers, while in the WT mice severe damage followed by leukocyte infiltration was evident. Reciprocal BM replacement of WT and Par2KO showed that WT BM-reconstituted Par2KO mice displayed marked liver damage, while in Par2KO BM-reconstituted WT mice, the tissue was generally protected. In the CCl4 direct damage model, hepatocytes regenerated in WT mice, whereas Par2KO mice failed to recover. Reciprocal BM replacement did not show significant differences in hepatic regeneration. In Par2KO mice, hepatitis was more apparent, while WT recovered regardless of the BM origin.

Conclusions: We conclude that Par2 activation in the immune system aggravates hepatitis and that Par2 activation in the damaged tissue promotes liver regeneration. When we incorporate this finding and revisit the literature reports, we reconciled the conflicts surrounding Par2's role in injury, recovery, and inflammation.

背景:不同的因素可能导致肝炎。其中包括肝脏炎症和中毒。我们选择了两种肝炎模型,典型的这两个潜在的原因。因此,我们的目的是表征蛋白酶激活受体2 (Par2)在肝脏再生和炎症中的作用,以调和Par2在许多损伤模型中的冲突作用,这种冲突有时会加重诱导损伤,有时会减轻损伤。方法:给WT和敲除型(Par2KO)小鼠注射ConA诱导免疫介导性肝炎,或给四氯化碳(CCl4)直接肝损伤。为了区分免疫成分和肝脏再生反应,我们对WT和Par2KO小鼠进行了骨髓(BM)置换,并重复了损伤模型。结果:ConA注射对Par2KO小鼠肝脏损伤有限,而对WT小鼠肝脏损伤严重,并伴有白细胞浸润。对WT和Par2KO进行BM互换,WT - BM重组的Par2KO小鼠肝脏损伤明显,而Par2KO - BM重组的WT小鼠肝脏组织基本得到保护。在CCl4直接损伤模型中,WT小鼠的肝细胞再生,而Par2KO小鼠则无法恢复。相互替代脑基在肝再生方面没有显着差异。在Par2KO小鼠中,肝炎更为明显,而与BM来源无关的WT恢复。结论:免疫系统中的Par2激活可加重肝炎,而受损组织中的Par2激活可促进肝脏再生。当我们结合这一发现并重新审视文献报道时,我们调和了围绕Par2在损伤、恢复和炎症中的作用的冲突。
{"title":"Resolving the conflicts around Par2 opposing roles in regeneration by comparing immune-mediated and toxic-induced injuries.","authors":"Gal Reches,&nbsp;Netta R Blondheim Shraga,&nbsp;Florent Carrette,&nbsp;Assaf Malka,&nbsp;Natalia Saleev,&nbsp;Yehuda Gubbay,&nbsp;Offir Ertracht,&nbsp;Izhak Haviv,&nbsp;Linda M Bradley,&nbsp;Fred Levine,&nbsp;Ron Piran","doi":"10.1186/s41232-022-00238-2","DOIUrl":"https://doi.org/10.1186/s41232-022-00238-2","url":null,"abstract":"<p><strong>Background: </strong>Different factors may lead to hepatitis. Among which are liver inflammation and poisoning. We chose two hepatitis models, typical for these two underlying causes. Thus, we aimed to characterize the role of protease-activated receptor 2 (Par2) in liver regeneration and inflammation to reconcile Par2 conflicting role in many damage models, which sometimes aggravates the induced damage and sometimes alleviates it.</p><p><strong>Methods: </strong>WT and knockout (Par2KO) mice were injected with concanavalin A (ConA) to induce immune-mediated hepatitis or with carbon tetrachloride (CCl<sub>4</sub>) to elicit direct hepatic damage. To distinguish the immune component from the liver regenerative response, we conducted bone marrow (BM) replacements of WT and Par2KO mice and repeated the damage models.</p><p><strong>Results: </strong>ConA injection caused limited damage in Par2KO mice livers, while in the WT mice severe damage followed by leukocyte infiltration was evident. Reciprocal BM replacement of WT and Par2KO showed that WT BM-reconstituted Par2KO mice displayed marked liver damage, while in Par2KO BM-reconstituted WT mice, the tissue was generally protected. In the CCl<sub>4</sub> direct damage model, hepatocytes regenerated in WT mice, whereas Par2KO mice failed to recover. Reciprocal BM replacement did not show significant differences in hepatic regeneration. In Par2KO mice, hepatitis was more apparent, while WT recovered regardless of the BM origin.</p><p><strong>Conclusions: </strong>We conclude that Par2 activation in the immune system aggravates hepatitis and that Par2 activation in the damaged tissue promotes liver regeneration. When we incorporate this finding and revisit the literature reports, we reconciled the conflicts surrounding Par2's role in injury, recovery, and inflammation.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9706915/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40723006","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Association of cellular immunity with severity of COVID-19 from the perspective of antigen-specific memory T cell responses and cross-reactivity. 从抗原特异性记忆T细胞反应和交叉反应性的角度看细胞免疫与COVID-19严重程度的关联
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-11-29 DOI: 10.1186/s41232-022-00239-1
Shin-Ichiro Fujii, Satoru Yamasaki, Tomonori Iyoda, Kanako Shimizu

Coronaviruses regularly cause outbreaks of zoonotic diseases characterized by severe pneumonia. The new coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused the global pandemic disease COVID-19 that began at the end of 2019 and spread rapidly owing to its infectious nature and rapidly progressing pneumonia. Although the infectivity of SARS-CoV-2 is high, indicated by the worldwide spread of the disease in a very short period, many individuals displayed only subclinical infection, and some of them transmitted the disease to individuals who then developed a severe symptomatic infection. Furthermore, there are differences in the severity of infection across countries, which can be attributed to factors such as the emergence of viral mutations in a short period of time as well as to the immune responses to viral factors. Anti-viral immunity generally consists of neutralizing antibodies that block viral infection and cytotoxic CD8+ T cells that eliminate the virus-infected cells. There is compelling evidence for the role of neutralizing antibodies in protective immunity in SARS-CoV-2 infection. However, the role of CD4+ and CD8+ T cells after the viral entry is complex and warrants a comprehensive discussion. Here, we discuss the protection afforded by cellular immunity against initial infection and development of severe disease. The initial failure of cellular immunity to control the infection worsens the clinical outcomes and functional profiles that inflict tissue damage without effectively eliminating viral reservoirs, while robust T cell responses are associated with mild outcomes. We also discuss persistent long-lasting memory T cell-mediated protection after infection or vaccination, which is rather complicated as it may involve SARS-CoV-2-specific cytotoxic T lymphocytes or cross-reactivity with previously infected seasonal coronaviruses, which are largely related to HLA genotypes. In addition, cross-reactivity with mutant strains is also discussed. Lastly, we discuss appropriate measures to be taken against the disease for immunocompromised patients. In conclusion, we provide evidence and discuss the causal relationship between natural infection- or vaccine-mediated memory T cell immunity and severity of COVID-19. This review is expected to provide a basis to develop strategies for the next generation of T cell-focused vaccines and aid in ending the current pandemic.

冠状病毒经常引起以严重肺炎为特征的人畜共患疾病暴发。新型冠状病毒,即严重急性呼吸综合征冠状病毒2 (SARS-CoV-2),引发了2019年底开始的全球大流行疾病COVID-19,由于其传染性和迅速发展的肺炎而迅速传播。虽然SARS-CoV-2的传染性很高,表明该疾病在很短的时间内在全球范围内传播,但许多个体仅表现出亚临床感染,其中一些个体将该疾病传播给个体,然后发展为严重的症状感染。此外,各国感染的严重程度存在差异,这可归因于诸如在短时间内出现病毒突变以及对病毒因素的免疫反应等因素。抗病毒免疫通常由阻断病毒感染的中和抗体和消除病毒感染细胞的细胞毒性CD8+ T细胞组成。有令人信服的证据表明,中和抗体在SARS-CoV-2感染的保护性免疫中起作用。然而,CD4+和CD8+ T细胞在病毒进入后的作用是复杂的,值得全面讨论。在这里,我们讨论了细胞免疫对初始感染和严重疾病发展的保护作用。细胞免疫控制感染的初始失败恶化了临床结果和造成组织损伤的功能特征,而没有有效地消除病毒库,而强大的T细胞反应与轻度结果相关。我们还讨论了感染或接种疫苗后持续持久记忆T细胞介导的保护,这是相当复杂的,因为它可能涉及sars - cov -2特异性细胞毒性T淋巴细胞或与先前感染的季节性冠状病毒的交叉反应,这在很大程度上与HLA基因型有关。此外,还讨论了与突变株的交叉反应性。最后,我们讨论了免疫功能低下患者应采取的适当措施。总之,我们提供证据并讨论了自然感染或疫苗介导的记忆T细胞免疫与COVID-19严重程度之间的因果关系。这项审查预计将为制定下一代T细胞聚焦疫苗的战略提供基础,并有助于结束当前的大流行。
{"title":"Association of cellular immunity with severity of COVID-19 from the perspective of antigen-specific memory T cell responses and cross-reactivity.","authors":"Shin-Ichiro Fujii,&nbsp;Satoru Yamasaki,&nbsp;Tomonori Iyoda,&nbsp;Kanako Shimizu","doi":"10.1186/s41232-022-00239-1","DOIUrl":"https://doi.org/10.1186/s41232-022-00239-1","url":null,"abstract":"<p><p>Coronaviruses regularly cause outbreaks of zoonotic diseases characterized by severe pneumonia. The new coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused the global pandemic disease COVID-19 that began at the end of 2019 and spread rapidly owing to its infectious nature and rapidly progressing pneumonia. Although the infectivity of SARS-CoV-2 is high, indicated by the worldwide spread of the disease in a very short period, many individuals displayed only subclinical infection, and some of them transmitted the disease to individuals who then developed a severe symptomatic infection. Furthermore, there are differences in the severity of infection across countries, which can be attributed to factors such as the emergence of viral mutations in a short period of time as well as to the immune responses to viral factors. Anti-viral immunity generally consists of neutralizing antibodies that block viral infection and cytotoxic CD8<sup>+</sup> T cells that eliminate the virus-infected cells. There is compelling evidence for the role of neutralizing antibodies in protective immunity in SARS-CoV-2 infection. However, the role of CD4<sup>+</sup> and CD8<sup>+</sup> T cells after the viral entry is complex and warrants a comprehensive discussion. Here, we discuss the protection afforded by cellular immunity against initial infection and development of severe disease. The initial failure of cellular immunity to control the infection worsens the clinical outcomes and functional profiles that inflict tissue damage without effectively eliminating viral reservoirs, while robust T cell responses are associated with mild outcomes. We also discuss persistent long-lasting memory T cell-mediated protection after infection or vaccination, which is rather complicated as it may involve SARS-CoV-2-specific cytotoxic T lymphocytes or cross-reactivity with previously infected seasonal coronaviruses, which are largely related to HLA genotypes. In addition, cross-reactivity with mutant strains is also discussed. Lastly, we discuss appropriate measures to be taken against the disease for immunocompromised patients. In conclusion, we provide evidence and discuss the causal relationship between natural infection- or vaccine-mediated memory T cell immunity and severity of COVID-19. This review is expected to provide a basis to develop strategies for the next generation of T cell-focused vaccines and aid in ending the current pandemic.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9706959/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"40709539","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 8
Collagen type I-mediated mechanotransduction controls epithelial cell fate conversion during intestinal inflammation. i型胶原介导的机械转导控制肠道炎症期间上皮细胞命运的转化。
IF 8.1 3区 医学 Q1 Medicine Pub Date : 2022-11-28 DOI: 10.1186/s41232-022-00237-3
Sakurako Kobayashi, Nobuhiko Ogasawara, Satoshi Watanabe, Yosuke Yoneyama, Sakura Kirino, Yui Hiraguri, Masami Inoue, Sayaka Nagata, Yoshimi Okamoto-Uchida, Satoshi Kofuji, Hiromichi Shimizu, Go Ito, Tomohiro Mizutani, Shinichi Yamauchi, Yusuke Kinugasa, Yoshihito Kano, Yasuhiro Nemoto, Mamoru Watanabe, Kiichiro Tsuchiya, Hiroshi Nishina, Ryuichi Okamoto, Shiro Yui

Background: The emerging concepts of fetal-like reprogramming following tissue injury have been well recognized as an important cue for resolving regenerative mechanisms of intestinal epithelium during inflammation. We previously revealed that the remodeling of mesenchyme with collagen fibril induces YAP/TAZ-dependent fate conversion of intestinal/colonic epithelial cells covering the wound bed towards fetal-like progenitors. To fully elucidate the mechanisms underlying the link between extracellular matrix (ECM) remodeling of mesenchyme and fetal-like reprogramming of epithelial cells, it is critical to understand how collagen type I influence the phenotype of epithelial cells. In this study, we utilize collagen sphere, which is the epithelial organoids cultured in purified collagen type I, to understand the mechanisms of the inflammatory associated reprogramming. Resolving the entire landscape of regulatory networks of the collagen sphere is useful to dissect the reprogrammed signature of the intestinal epithelium.

Methods: We performed microarray, RNA-seq, and ATAC-seq analyses of the murine collagen sphere in comparison with Matrigel organoid and fetal enterosphere (FEnS). We subsequently cultured human colon epithelium in collagen type I and performed RNA-seq analysis. The enriched genes were validated by gene expression comparison between published gene sets and immunofluorescence in pathological specimens of ulcerative colitis (UC).

Results: The murine collagen sphere was confirmed to have inflammatory and regenerative signatures from RNA-seq analysis. ATAC-seq analysis confirmed that the YAP/TAZ-TEAD axis plays a central role in the induction of the distinctive signature. Among them, TAZ has implied its relevant role in the process of reprogramming and the ATAC-based motif analysis demonstrated not only Tead proteins, but also Fra1 and Runx2, which are highly enriched in the collagen sphere. Additionally, the human collagen sphere also showed a highly significant enrichment of both inflammatory and fetal-like signatures. Immunofluorescence staining confirmed that the representative genes in the human collagen sphere were highly expressed in the inflammatory region of ulcerative colitis.

Conclusions: Collagen type I showed a significant influence in the acquisition of the reprogrammed inflammatory signature in both mice and humans. Dissection of the cell fate conversion and its mechanisms shown in this study can enhance our understanding of how the epithelial signature of inflammation is influenced by the ECM niche.

背景:组织损伤后胎儿样重编程的新概念已被公认为解决炎症期间肠上皮再生机制的重要线索。我们之前揭示了胶原原纤维间质重塑诱导覆盖在伤口床上的肠/结肠上皮细胞向胎儿样祖细胞的YAP/ taz依赖性命运转化。为了充分阐明间质细胞外基质(ECM)重塑与上皮细胞胎儿样重编程之间联系的机制,了解I型胶原如何影响上皮细胞的表型至关重要。在本研究中,我们利用胶原球,即纯化型胶原中培养的上皮类器官,来了解炎症相关重编程的机制。解决胶原球调控网络的整个景观有助于解剖肠上皮的重编程特征。方法:我们对小鼠胶原球进行微阵列、RNA-seq和ATAC-seq分析,并与Matrigel类器官和胎儿肠球(FEnS)进行比较。随后,我们用I型胶原培养人结肠上皮,并进行RNA-seq分析。通过已发表的基因集和溃疡性结肠炎(UC)病理标本的免疫荧光基因表达比较,证实了富集基因的有效性。结果:通过RNA-seq分析证实小鼠胶原球具有炎症和再生特征。ATAC-seq分析证实,YAP/TAZ-TEAD轴在诱导独特特征中起核心作用。其中,TAZ暗示了其在重编程过程中的相关作用,基于atac的基序分析不仅发现了Tead蛋白,还发现了在胶原球中高度富集的Fra1和Runx2蛋白。此外,人胶原球也显示出高度显著的炎症和胎儿样特征的富集。免疫荧光染色证实,人胶原球的代表性基因在溃疡性结肠炎炎症区高度表达。结论:I型胶原蛋白对小鼠和人类重编程炎症特征的获得有显著影响。本研究中对细胞命运转化及其机制的解剖可以增强我们对炎症的上皮特征如何受到ECM生态位的影响的理解。
{"title":"Collagen type I-mediated mechanotransduction controls epithelial cell fate conversion during intestinal inflammation.","authors":"Sakurako Kobayashi,&nbsp;Nobuhiko Ogasawara,&nbsp;Satoshi Watanabe,&nbsp;Yosuke Yoneyama,&nbsp;Sakura Kirino,&nbsp;Yui Hiraguri,&nbsp;Masami Inoue,&nbsp;Sayaka Nagata,&nbsp;Yoshimi Okamoto-Uchida,&nbsp;Satoshi Kofuji,&nbsp;Hiromichi Shimizu,&nbsp;Go Ito,&nbsp;Tomohiro Mizutani,&nbsp;Shinichi Yamauchi,&nbsp;Yusuke Kinugasa,&nbsp;Yoshihito Kano,&nbsp;Yasuhiro Nemoto,&nbsp;Mamoru Watanabe,&nbsp;Kiichiro Tsuchiya,&nbsp;Hiroshi Nishina,&nbsp;Ryuichi Okamoto,&nbsp;Shiro Yui","doi":"10.1186/s41232-022-00237-3","DOIUrl":"https://doi.org/10.1186/s41232-022-00237-3","url":null,"abstract":"<p><strong>Background: </strong>The emerging concepts of fetal-like reprogramming following tissue injury have been well recognized as an important cue for resolving regenerative mechanisms of intestinal epithelium during inflammation. We previously revealed that the remodeling of mesenchyme with collagen fibril induces YAP/TAZ-dependent fate conversion of intestinal/colonic epithelial cells covering the wound bed towards fetal-like progenitors. To fully elucidate the mechanisms underlying the link between extracellular matrix (ECM) remodeling of mesenchyme and fetal-like reprogramming of epithelial cells, it is critical to understand how collagen type I influence the phenotype of epithelial cells. In this study, we utilize collagen sphere, which is the epithelial organoids cultured in purified collagen type I, to understand the mechanisms of the inflammatory associated reprogramming. Resolving the entire landscape of regulatory networks of the collagen sphere is useful to dissect the reprogrammed signature of the intestinal epithelium.</p><p><strong>Methods: </strong>We performed microarray, RNA-seq, and ATAC-seq analyses of the murine collagen sphere in comparison with Matrigel organoid and fetal enterosphere (FEnS). We subsequently cultured human colon epithelium in collagen type I and performed RNA-seq analysis. The enriched genes were validated by gene expression comparison between published gene sets and immunofluorescence in pathological specimens of ulcerative colitis (UC).</p><p><strong>Results: </strong>The murine collagen sphere was confirmed to have inflammatory and regenerative signatures from RNA-seq analysis. ATAC-seq analysis confirmed that the YAP/TAZ-TEAD axis plays a central role in the induction of the distinctive signature. Among them, TAZ has implied its relevant role in the process of reprogramming and the ATAC-based motif analysis demonstrated not only Tead proteins, but also Fra1 and Runx2, which are highly enriched in the collagen sphere. Additionally, the human collagen sphere also showed a highly significant enrichment of both inflammatory and fetal-like signatures. Immunofluorescence staining confirmed that the representative genes in the human collagen sphere were highly expressed in the inflammatory region of ulcerative colitis.</p><p><strong>Conclusions: </strong>Collagen type I showed a significant influence in the acquisition of the reprogrammed inflammatory signature in both mice and humans. Dissection of the cell fate conversion and its mechanisms shown in this study can enhance our understanding of how the epithelial signature of inflammation is influenced by the ECM niche.</p>","PeriodicalId":13588,"journal":{"name":"Inflammation and Regeneration","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2022-11-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9703763/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10388668","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 2
期刊
Inflammation and Regeneration
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