Cellular reprogramming

细胞重编程
  • 文章类型: Journal Article
    代谢重编程为肿瘤提供了能量来源和生物燃料,以支持其在恶性微环境中的生存。对肿瘤微环境(TME)的内在致癌机制的广泛研究已经确定,癌症相关成纤维细胞(CAFs)和代谢重编程通过许多生物学活性调节肿瘤进展。包括肿瘤免疫抑制,慢性炎症,生态位重塑。具体来说,通过共同支持慢性炎症的CAFs和多种免疫细胞促进免疫抑制性TME形成,释放介质,从而诱导转移前生态位的形成,并最终推动肿瘤增殖和转移的恶性循环。本文综述了肿瘤适应性TME动态演化的CAFs和代谢调控过程,特别关注CAFs促进免疫抑制微环境形成和支持转移的机制。现有的发现证实了TME的多个组成部分协同作用以加速肿瘤事件的进展。在推进与CAF相关的研究的背景下,进一步讨论了基于CAF的靶向治疗在临床环境中的潜在应用和挑战。
    Metabolic reprogramming provides tumors with an energy source and biofuel to support their survival in the malignant microenvironment. Extensive research into the intrinsic oncogenic mechanisms of the tumor microenvironment (TME) has established that cancer-associated fibroblast (CAFs) and metabolic reprogramming regulates tumor progression through numerous biological activities, including tumor immunosuppression, chronic inflammation, and ecological niche remodeling. Specifically, immunosuppressive TME formation is promoted and mediators released via CAFs and multiple immune cells that collectively support chronic inflammation, thereby inducing pre-metastatic ecological niche formation, and ultimately driving a vicious cycle of tumor proliferation and metastasis. This review comprehensively explores the process of CAFs and metabolic regulation of the dynamic evolution of tumor-adapted TME, with particular focus on the mechanisms by which CAFs promote the formation of an immunosuppressive microenvironment and support metastasis. Existing findings confirm that multiple components of the TME act cooperatively to accelerate the progression of tumor events. The potential applications and challenges of targeted therapies based on CAFs in the clinical setting are further discussed in the context of advancing research related to CAFs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    星形胶质细胞在维持脑能量稳态中起着关键作用,通过糖酵解和脂质代谢支持神经元功能。这篇综述探讨了星形胶质细胞在生理和病理条件下的代谢复杂性,突出了它们的适应性可塑性和多样化的功能。在正常情况下,星形胶质细胞调节突触活动,回收神经递质,维持血脑屏障,确保平衡的能源供应和防止氧化应激。然而,作为对中枢神经系统病变如神经创伤的反应,中风,感染,和神经退行性疾病,如阿尔茨海默病和亨廷顿病,星形胶质细胞经历显著的形态学,分子,和代谢变化。反应性星形胶质细胞上调糖酵解和脂肪酸氧化,以满足增加的能量需求,这可以在急性环境中保护,但可能加剧慢性炎症和疾病进展。这篇综述强调了对先进分子的需求,遗传,和生理工具,以进一步了解星形胶质细胞异质性及其在疾病状态下的代谢重编程。
    Astrocytes play a pivotal role in maintaining brain energy homeostasis, supporting neuronal function through glycolysis and lipid metabolism. This review explores the metabolic intricacies of astrocytes in both physiological and pathological conditions, highlighting their adaptive plasticity and diverse functions. Under normal conditions, astrocytes modulate synaptic activity, recycle neurotransmitters, and maintain the blood-brain barrier, ensuring a balanced energy supply and protection against oxidative stress. However, in response to central nervous system pathologies such as neurotrauma, stroke, infections, and neurodegenerative diseases like Alzheimer\'s and Huntington\'s disease, astrocytes undergo significant morphological, molecular, and metabolic changes. Reactive astrocytes upregulate glycolysis and fatty acid oxidation to meet increased energy demands, which can be protective in acute settings but may exacerbate chronic inflammation and disease progression. This review emphasizes the need for advanced molecular, genetic, and physiological tools to further understand astrocyte heterogeneity and their metabolic reprogramming in disease states.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    尽管现代医学和外科疗法取得了重要进展,但心血管疾病仍然是世界范围内死亡的主要原因。由于人类成年心肌细胞的再生能力有限,心肌梗死后丢失的心肌细胞被纤维化瘢痕组织取代,导致心脏功能障碍和心力衰竭。为了取代丢失的心肌细胞,一个有前途的方法是直接心脏重新编程,其中心脏成纤维细胞转分化为诱导的心肌细胞样细胞(iCM)。在这里,我们回顾心脏重编程鸡尾酒(包括转录因子,微RNA和小分子)介导iCM产生。我们还强调机械研究,探索这一过程的障碍和促进者。然后,我们回顾了iCM重新编程的最新进展,专注于单细胞组学研究。最后,我们讨论了临床应用的障碍。
    Cardiovascular disease remains a leading cause of death worldwide despite important advances in modern medical and surgical therapies. As human adult cardiomyocytes have limited regenerative ability, cardiomyocytes lost after myocardial infarction are replaced by fibrotic scar tissue, leading to cardiac dysfunction and heart failure. To replace lost cardiomyocytes, a promising approach is direct cardiac reprogramming, in which cardiac fibroblasts are transdifferentiated into induced cardiomyocyte-like cells (iCMs). Here we review cardiac reprogramming cocktails (including transcription factors, microRNAs and small molecules) that mediate iCM generation. We also highlight mechanistic studies exploring the barriers to and facilitators of this process. We then review recent progress in iCM reprogramming, with a focus on single-cell \'-omics\' research. Finally, we discuss obstacles to clinical application.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    三阴性乳腺癌(TNBC)因其高侵袭性而成为乳腺癌治疗中的棘手问题,转移和复发。尽管免疫疗法在TNBC中取得了重要进展,多种因素引起的免疫逃逸,尤其是代谢重编程,仍是TNBC免疫治疗的瓶颈。遗憾的是,导致免疫逃逸的机制仍然知之甚少。在代谢水平探索TNBC免疫逃逸的机制为后续靶向或免疫治疗提供了靶点和方向。在这次审查中,我们关注TNBC通过缺氧影响免疫细胞和间质细胞的机制,葡萄糖代谢,脂质代谢和氨基酸代谢,改变肿瘤代谢和肿瘤微环境。这将有助于为TNBC免疫治疗寻找新的靶点和策略。
    Triple-negative breast cancer (TNBC) has become a thorny problem in the treatment of breast cancer because of its high invasiveness, metastasis and recurrence. Although immunotherapy has made important progress in TNBC, immune escape caused by many factors, especially metabolic reprogramming, is still the bottleneck of TNBC immunotherapy. Regrettably, the mechanisms responsible for immune escape remain poorly understood. Exploring the mechanism of TNBC immune escape at the metabolic level provides a target and direction for follow-up targeting or immunotherapy. In this review, we focus on the mechanism that TNBC affects immune cells and interstitial cells through hypoxia, glucose metabolism, lipid metabolism and amino acid metabolism, and changes tumor metabolism and tumor microenvironment. This will help to find new targets and strategies for TNBC immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    HER2+乳腺肿瘤有丰富的免疫抑制细胞,包括M2型肿瘤相关巨噬细胞(TAMs)。尽管TAM由免疫刺激M1型和免疫抑制M2型组成,在免疫抑制性肿瘤中M1/M2-TAM比值降低,有助于他们的免疫疗法难治性。M1-与M2-TAM的形成取决于不同的精氨酸代谢,其中M1-TAM将精氨酸转化为一氧化氮(NO),M2-TAM将精氨酸转化为多胺(PA)。我们假设M1-与M2-TAMs中这种不同的精氨酸代谢归因于BH4(NO合酶辅因子)的不同可用性,并且其补充将M2-TAMs重新编程为M1-TAMs。最近,我们报道了墨蝶呤(SEP),内源性BH4前体,提高M1-TAM标志物在HER2+肿瘤中的表达。这里,我们显示SEP恢复M2样巨噬细胞中的BH4水平,然后将精氨酸代谢重定向到NO合成,并将M2型转化为M1型。重编程的巨噬细胞表现出完全成熟的抗原呈递能力和诱导效应T细胞以触发HER2+癌细胞的免疫原性细胞死亡。这项研究证实了SEP在HER2乳腺肿瘤微环境的代谢转变中作为一种新型的免疫治疗策略的实用性。
    HER2+ breast tumors have abundant immune-suppressive cells, including M2-type tumor-associated macrophages (TAMs). Although TAMs consist of the immune-stimulatory M1 type and immune-suppressive M2 type, the M1/M2-TAM ratio is reduced in immune-suppressive tumors, contributing to their immunotherapy refractoriness. M1- versus M2-TAM formation depends on differential arginine metabolism, where M1-TAMs convert arginine to nitric oxide (NO) and M2-TAMs convert arginine to polyamines (PAs). We hypothesize that such distinct arginine metabolism in M1- versus M2-TAMs is attributed to different availability of BH4 (NO synthase cofactor) and that its replenishment would reprogram M2-TAMs to M1-TAMs. Recently, we reported that sepiapterin (SEP), the endogenous BH4 precursor, elevates the expression of M1-TAM markers within HER2+ tumors. Here, we show that SEP restores BH4 levels in M2-like macrophages, which then redirects arginine metabolism to NO synthesis and converts M2 type to M1 type. The reprogrammed macrophages exhibit full-fledged capabilities of antigen presentation and induction of effector T cells to trigger immunogenic cell death of HER2+ cancer cells. This study substantiates the utility of SEP in the metabolic shift of the HER2+ breast tumor microenvironment as a novel immunotherapeutic strategy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    从成体细胞产生的诱导多能干细胞(iPSC)被诱导表达通过重编程技术使其具有多能性的基因。凭借其无限的增殖能力和多方面的分化潜力,并规避了胚胎干细胞(ESC)应用中遇到的伦理问题,iPSCs在细胞治疗领域有着广泛的应用,药物筛选,和疾病模型,并可能为未来再生医学治疗疾病开辟新的可能性。在这次审查中,我们从不同的重编程细胞技术开始获得iPSCs,包括生物技术,化学,和物理调制技术,展示各自的长处,和限制,以及最近的研究进展。其次,我们综述了基于iPSC重编程的再生疗法的最新研究进展.iPSCs目前被广泛用于研究毛囊缺损的各种临床疾病,心肌梗塞,神经系统疾病,肝脏疾病,和脊髓损伤。本文重点介绍了iPSCs的转化临床研究及其在医学领域的发展潜力。最后,我们总结了总体综述,并展望了iPSCs在细胞治疗以及组织再生工程领域的潜在前景和可能存在的问题。我们相信,正在推进的iPSC研究将有助于推动期待已久的细胞治疗突破。
    Induced pluripotent stem cells (iPSCs) that are generated from adult somatic cells are induced to express genes that make them pluripotent through reprogramming techniques. With their unlimited proliferative capacity and multifaceted differentiation potential and circumventing the ethical problems encountered in the application of embryonic stem cells (ESC), iPSCs have a broad application in the fields of cell therapy, drug screening, and disease models and may open up new possibilities for regenerative medicine to treat diseases in the future. In this review, we begin with different reprogramming cell technologies to obtain iPSCs, including biotechnological, chemical, and physical modulation techniques, and present their respective strengths, and limitations, as well as the recent progress of research. Secondly, we review recent research advances in iPSC reprogramming-based regenerative therapies. iPSCs are now widely used to study various clinical diseases of hair follicle defects, myocardial infarction, neurological disorders, liver diseases, and spinal cord injuries. This review focuses on the translational clinical research around iPSCs as well as their potential for growth in the medical field. Finally, we summarize the overall review and look at the potential future of iPSCs in the field of cell therapy as well as tissue regeneration engineering and possible problems. We believe that the advancing iPSC research will help drive long-awaited breakthroughs in cellular therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    OBJECTIVE: To establish a methodological system for reprogramming rat embryonic fibroblasts (REF) into chemically induced neurons (ciNCs) via small molecule compounds to provide safe and effective donor cells for treatment of neurodegenerative diseases.
    METHODS: Based on the method established by PEI Gang\'s research group to directly reprogram human fibroblasts into neurons, the induction medium and maturation medium was optimized by replacing the coating solution, mitigating oxidative stress injury, adding neurogenic protective factors, adjusting the concentration of trichothecenes, performing small-molecule removal experiments, and carrying out immunofluorescence and Western blotting on cells at different stages of induction to validate the effect of induction.
    RESULTS: When the original protocol was used for induction, the cell survival rate was (34.24±2.77)%. After replacing the coating solution gelatin with matrigel, the cell survival rate increased to (45.41±4.27)%; after adding melatonin, the cell survival rate increased to (67.95±5.61)% and (23.43±1.42)% were transformed into neural-like cells; after adding the small molecule P7C3-A20, the cell survival rate was further increased to (76.27±1.41)%, and (39.72±4.75)% of the cells were transformed into neural-like cells. When the concentration of trichothecene was increased to 30 μmol/L, the proportion of neural-like cells reached (55.79±1.90)%; after the removal of SP600125, (86.96±2.15)% of the cells survived, and the rate of neural-like cell production increased to (63.43±1.60)%. With the optimized protocol, REF could be successfully induced into ciNC through the neural precursor cell stage, in which the neural precursor cells were able to highly express the neural precursor cell markers SRY-related HMG-box gene 2 (Sox2) and paired box 6 (Pax6) as well as neuron-specific marker tubulin 1 (Tuj1), while the expression of fiber-associated protein vimentin was reduced. After two weeks of induction of neural precursor cells in a maturation medium, most cells displayed neuronal-like cell morphology. The induced ciNCs were able to highly express the mature neuronal surface markers Tuj1 and microtubule-associated protein 2 (MAP2), while the expression of vimentin was reduced.
    CONCLUSIONS: The small molecule combinations optimized in this study can reprogram REF to ciNCs under normoxic conditions.
    目的: 建立通过小分子化合物将大鼠胚胎成纤维细胞(REF)重编程为化学诱导神经元(ciNC)的方法体系,为细胞移植治疗神经退行性疾病提供安全有效的供体细胞。方法: 以裴钢研究团队建立的人成纤维细胞直接重编程为神经元的方法为基础,通过更换包被液、缓解氧化应激损伤、添加神经源性保护因子、调整毛喉素浓度和小分子去除实验对诱导培养基和成熟培养基进行优化,并通过免疫荧光法和蛋白质印迹法验证不同诱导阶段细胞的相关蛋白质。结果: 以原方案进行诱导时,细胞存活率仅(34.24±2.77)%。包被液更换为基质胶后,细胞存活率上升到(45.41±4.27)%;添加褪黑素后细胞存活率提高至(67.95±5.61)%,(23.43±1.42)%的细胞转变为神经样细胞;添加小分子P7C3-A20后细胞存活率进一步提升至(76.27±1.41)%,(39.72±4.75)%的细胞转变为神经样细胞;毛喉素浓度提升至30 μmol/L时,神经样细胞比例达到(55.79±1.90)%;去除SP600125后,(86.96±2.15)%细胞存活,神经样细胞生成率提高至(63.43±1.60)%。以优化后的方案诱导,可经过神经前体细胞将REF诱导为ciNC。其中,神经前体细胞能够高表达神经前体细胞标志物SRY-box转录因子2、配对盒6以及神经元特异性标志物Ⅲ类β-微管蛋白,而成纤维相关蛋白波形蛋白表达量减少。神经前体细胞在成熟培养基中诱导两周后,可见大部分细胞呈神经样细胞形态,诱导后的ciNC高表达Ⅲ类β-微管蛋白和微管相关蛋白2,而成纤维相关蛋白波形蛋白表达减少。结论: 优化后的小分子组合在常氧条件下能将REF重编程为ciNC。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    造血干细胞(HSC)在肺癌微环境中表现出显著的功能和代谢改变,通过增加向髓源性抑制细胞(MDSC)的分化来促进肿瘤进展和免疫逃避。我们的目的是分析肺癌中HSC从糖酵解到氧化磷酸化(OXPHOS)的代谢转变,并确定其对HSC功能的影响。使用小鼠Lewis肺癌肺癌模型,我们对长期和短期HSC进行了代谢分析,以及多能祖细胞,比较它们在正常和癌症条件下的代谢状态。我们使用2-[N-(7-Nitrobenz-2-氧杂-1,3-三唑-4-基)氨基]-2-脱氧葡萄糖(2-NBDG)测量葡萄糖摄取,并评估乳酸水平,乙酰辅酶A,和ATP。线粒体功能通过流式细胞术评估,以及葡萄糖代谢抑制剂2-DG对HSC分化和线粒体活性的影响。肺癌条件下的HSC显示葡萄糖摄取和乳酸产生增加,随着OXPHOS活性的增加,标志着新陈代谢的转变。2-DG治疗导致T-HSCs和MDSCs减少,红细胞计数增加,强调其影响HSC代谢和分化途径的潜力。这项研究为肺癌中HSC的代谢重编程提供了新的见解,强调从糖酵解到OXPHOS的关键转变及其对癌症相关代谢途径的治疗靶向的意义。
    Hematopoietic stem cells (HSCs) exhibit significant functional and metabolic alterations within the lung cancer microenvironment, contributing to tumor progression and immune evasion by increasing differentiation into myeloid-derived suppressor cells (MDSCs). Our aim is to analyze the metabolic transition of HSCs from glycolysis to oxidative phosphorylation (OXPHOS) in lung cancer and determine its effects on HSC functionality. Using a murine Lewis Lung Carcinoma lung cancer model, we conducted metabolic profiling of long-term and short-term HSCs, as well as multipotent progenitors, comparing their metabolic states in normal and cancer conditions. We measured glucose uptake using 2-[N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)Amino]-2-Deoxyglucose (2-NBDG) and assessed levels of lactate, acetyl-coenzyme A, and ATP. Mitochondrial functionality was evaluated through flow cytometry, alongside the impact of the glucose metabolism inhibitor 2-DG on HSC differentiation and mitochondrial activity. HSCs under lung cancer conditions showed increased glucose uptake and lactate production, with an associated rise in OXPHOS activity, marking a metabolic shift. Treatment with 2-DG led to decreased T-HSCs and MDSCs and an increased red blood cell count, highlighting its potential to influence metabolic and differentiation pathways in HSCs. This study provides novel insights into the metabolic reprogramming of HSCs in lung cancer, emphasizing the critical shift from glycolysis to OXPHOS and its implications for the therapeutic targeting of cancer-related metabolic pathways.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    细胞命运的趋势对大多数扰动是稳健的,然而,对某些扰动的敏感提出了一个有趣的问题,即底层分子网络中关键路径的存在,关键路径决定了不同的细胞命运。重编程和转分化清楚地显示了通过仅调节几个或甚至单个分子开关而改变细胞命运的实例。然而,还不知道如何识别这种开关,叫做主调节器,以及细胞命运如何由其调节决定。这里,我们介绍凯撒,一个计算框架,可以系统地识别主调节器并解开产生的规范化内核,互连反馈的关键子结构,对细胞命运决定至关重要。我们证明了CAESAR可以成功预测重编程因子去分化为小鼠胚胎干细胞和造血干细胞的转分化。同时通过规范内核揭示了潜在的基本机制。CAESAR提供了复杂分子网络如何决定细胞命运的系统级理解。
    The tendency for cell fate to be robust to most perturbations, yet sensitive to certain perturbations raises intriguing questions about the existence of a key path within the underlying molecular network that critically determines distinct cell fates. Reprogramming and trans-differentiation clearly show examples of cell fate change by regulating only a few or even a single molecular switch. However, it is still unknown how to identify such a switch, called a master regulator, and how cell fate is determined by its regulation. Here, we present CAESAR, a computational framework that can systematically identify master regulators and unravel the resulting canalizing kernel, a key substructure of interconnected feedbacks that is critical for cell fate determination. We demonstrate that CAESAR can successfully predict reprogramming factors for de-differentiation into mouse embryonic stem cells and trans-differentiation of hematopoietic stem cells, while unveiling the underlying essential mechanism through the canalizing kernel. CAESAR provides a system-level understanding of how complex molecular networks determine cell fates.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    巨噬细胞在先天免疫反应中起着至关重要的作用,表现出上下文相关的行为。在肿瘤微环境中,巨噬细胞以肿瘤相关或M2样巨噬细胞的形式存在,提出了重新编程的挑战。在这项研究中,我们开发了一种肽水凝胶,能够通过激活NF-κB信号通路将M0巨噬细胞极化为促炎M1巨噬细胞。重要的是,还发现该系统能够通过激活CD206受体将M2巨噬细胞重编程为促炎性M1样巨噬细胞。纳米纤维水凝胶自组装由包含先天防御调节肽和自组装促进基序的短肽,呈现密集排列的调节因子,这些调节因子与巨噬细胞膜受体多价结合,不仅使M0巨噬细胞极化,而且使M2巨噬细胞重新极化为M1样巨噬细胞。总的来说,这项工作为重新编程巨噬细胞提供了一个有前途的策略,通过重塑免疫抗性微环境来增强免疫治疗的潜力。
    Macrophages play crucial roles in the innate immune response, exhibiting context-dependent behaviors. Within the tumor microenvironment, macrophages exist as tumor-associated or M2-like macrophages, presenting reprogramming challenges. In this study, we develop a peptide hydrogel that is able to polarize M0 macrophages into pro-inflammatory M1 macrophages through the activation of NF-κB signaling pathways. Importantly, this system is also found to be capable of reprogramming M2 macrophages into pro-inflammatory M1-like macrophages by activating CD206 receptors. The nanofibrous hydrogel self-assembles from a short peptide that contains an innate defense regulator peptide and a self-assembly promoting motif, presenting densely arrayed regulators that multivalently engage with macrophage membrane receptors to not only polarize M0 macrophages but also repolarize M2 macrophages into M1-like macrophages. Overall, this work offers a promising strategy for reprogramming macrophages, holding the potential to enhance immunotherapy by remodeling immune-resistant microenvironments.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号