Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4310-5_1
Donovan J Anderson, Marshall S Horwitz
Mutations are acquired frequently, such t`hat each cell's genome inscribes its history of cell divisions. Loss of heterozygosity (LOH) accumulates throughout the genome, offering large encoding capacity for phylogenetic inference of cell lineage.In this chapter, we demonstrate a method, using single-cell RNA sequencing, for reconstructing cell lineages from inferred LOH events in a Bayesian manner, annotating the lineage with cell phenotypes, and marking developmental time points based on X-chromosome inactivation. This type of retrospective analysis could be incorporated into scRNA-seq pipelines and was initially developed to investigate Emx1+ cortical projection neuron and glia lineages from C57Bl/6J (B6) and CAST/EiJ (CA) interstrain F1 mice, describing progenitor cells giving rise to multiple cortical cell types through stereotyped expansion and distinct waves of neurogenesis.
{"title":"Bayesian Phylogenetic Lineage Reconstruction with Loss of Heterozygosity Mutations Derived from Single-Cell RNA Sequencing.","authors":"Donovan J Anderson, Marshall S Horwitz","doi":"10.1007/978-1-0716-4310-5_1","DOIUrl":"https://doi.org/10.1007/978-1-0716-4310-5_1","url":null,"abstract":"<p><p>Mutations are acquired frequently, such t`hat each cell's genome inscribes its history of cell divisions. Loss of heterozygosity (LOH) accumulates throughout the genome, offering large encoding capacity for phylogenetic inference of cell lineage.In this chapter, we demonstrate a method, using single-cell RNA sequencing, for reconstructing cell lineages from inferred LOH events in a Bayesian manner, annotating the lineage with cell phenotypes, and marking developmental time points based on X-chromosome inactivation. This type of retrospective analysis could be incorporated into scRNA-seq pipelines and was initially developed to investigate Emx1+ cortical projection neuron and glia lineages from C57Bl/6J (B6) and CAST/EiJ (CA) interstrain F1 mice, describing progenitor cells giving rise to multiple cortical cell types through stereotyped expansion and distinct waves of neurogenesis.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2886 ","pages":"1-22"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915328","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4310-5_19
A S Eisele, D M Suter
Gene expression memory-based lineage inference (GEMLI) is a computational tool allowing to predict cell lineages solely from single-cell RNA-sequencing (scRNA-seq) datasets and is publicly available as an R package on GitHub. GEMLI is based on the occurrence of gene expression memory, i.e., the gene-specific maintenance of expression levels through cell divisions. This represents a shift away from experimental lineage tracing techniques based on genetic marks or physical cell lineage separation and greatly eases and expands lineage annotation. GEMLI allows to study cell lineages during differentiation in development, homeostasis, and regeneration, as well as disease onset and progression in various physiological and pathological contexts. This makes it possible to dissect cell type-specific gene expression memory, to discriminate symmetric and asymmetric cell fate decisions, and to reconstruct individual multicellular structures from pooled scRNA-seq datasets. GEMLI is particularly promising for its ability to identify small lineages in human samples, a context in which no other lineage tracing methods are applicable. In this chapter, we provide a detailed protocol of the GEMLI R package usage on gene expression matrices derived from standard scRNA-seq on various platforms. We cover the use of the main function to predict cell lineages and how to adjust its parameters to different tasks. We also show how lineage information is extracted, visualized, and fine-tuned. Finally, we describe the use of the package's functions for the detailed analysis of the predicted cell lineages. This includes the analysis of gene expression memory, cell type composition of individual large lineages, and identification of lineages at the transition point between two cell types.
{"title":"GEMLI: Gene Expression Memory-Based Lineage Inference from Single-Cell RNA-Sequencing Datasets.","authors":"A S Eisele, D M Suter","doi":"10.1007/978-1-0716-4310-5_19","DOIUrl":"https://doi.org/10.1007/978-1-0716-4310-5_19","url":null,"abstract":"<p><p>Gene expression memory-based lineage inference (GEMLI) is a computational tool allowing to predict cell lineages solely from single-cell RNA-sequencing (scRNA-seq) datasets and is publicly available as an R package on GitHub. GEMLI is based on the occurrence of gene expression memory, i.e., the gene-specific maintenance of expression levels through cell divisions. This represents a shift away from experimental lineage tracing techniques based on genetic marks or physical cell lineage separation and greatly eases and expands lineage annotation. GEMLI allows to study cell lineages during differentiation in development, homeostasis, and regeneration, as well as disease onset and progression in various physiological and pathological contexts. This makes it possible to dissect cell type-specific gene expression memory, to discriminate symmetric and asymmetric cell fate decisions, and to reconstruct individual multicellular structures from pooled scRNA-seq datasets. GEMLI is particularly promising for its ability to identify small lineages in human samples, a context in which no other lineage tracing methods are applicable. In this chapter, we provide a detailed protocol of the GEMLI R package usage on gene expression matrices derived from standard scRNA-seq on various platforms. We cover the use of the main function to predict cell lineages and how to adjust its parameters to different tasks. We also show how lineage information is extracted, visualized, and fine-tuned. Finally, we describe the use of the package's functions for the detailed analysis of the predicted cell lineages. This includes the analysis of gene expression memory, cell type composition of individual large lineages, and identification of lineages at the transition point between two cell types.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2886 ","pages":"375-400"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915347","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4310-5_5
Eugenio Gentile, Ashley Maynard, Zhisong He, Barbara Treutlein
Induced pluripotent stem cell (iPSC)-derived organoids provide models to study human organ development. Single-cell transcriptomics enables highly resolved descriptions of cell states within these systems; however, approaches are needed to directly determine the lineage relationship between cells. Here we provide a detailed protocol (Fig. 1) for the application of iTracer (He Z, Maynard A, Jain A, et al., Nat Methods 19:90-99, 2022), a recently published lineage recorder that combines reporter barcodes with inducible CRISPR-Cas9 scarring and is compatible with single-cell and spatial transcriptomics. iTracer is used to explore clonality and lineage dynamics during brain organoid development. More broadly, iTracer can be adapted to any iPSC-derived culture system to dissect lineage dynamics during normal or perturbed development.
诱导多能干细胞(iPSC)衍生的类器官为研究人体器官发育提供了模型。单细胞转录组学能够高度解析这些系统内的细胞状态;然而,需要直接确定细胞之间的谱系关系的方法。在这里,我们为iTracer的应用提供了详细的方案(图1)(He Z, Maynard a, Jain a, et ., Nat Methods 19:90- 99,2022), iTracer是一种最近发表的谱系记录器,将报告基因条形码与可诱导的CRISPR-Cas9瘢痕结合起来,与单细胞和空间转录组学兼容。iTracer用于探索脑类器官发育过程中的克隆和谱系动力学。更广泛地说,iTracer可以适用于任何ipsc衍生的培养系统,以解剖正常或受干扰发育期间的谱系动态。
{"title":"Lineage Recording in Human Brain Organoids with iTracer.","authors":"Eugenio Gentile, Ashley Maynard, Zhisong He, Barbara Treutlein","doi":"10.1007/978-1-0716-4310-5_5","DOIUrl":"https://doi.org/10.1007/978-1-0716-4310-5_5","url":null,"abstract":"<p><p>Induced pluripotent stem cell (iPSC)-derived organoids provide models to study human organ development. Single-cell transcriptomics enables highly resolved descriptions of cell states within these systems; however, approaches are needed to directly determine the lineage relationship between cells. Here we provide a detailed protocol (Fig. 1) for the application of iTracer (He Z, Maynard A, Jain A, et al., Nat Methods 19:90-99, 2022), a recently published lineage recorder that combines reporter barcodes with inducible CRISPR-Cas9 scarring and is compatible with single-cell and spatial transcriptomics. iTracer is used to explore clonality and lineage dynamics during brain organoid development. More broadly, iTracer can be adapted to any iPSC-derived culture system to dissect lineage dynamics during normal or perturbed development.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2886 ","pages":"85-101"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915379","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4322-8_12
Mika Saotome, Jill Goodman, Motoki Takaku
Hox genes play a pivotal role during development. Their expression is tightly controlled in a spatiotemporal manner, ensuring that specific body structures develop at the correct locations and times during development. Various genomics approaches have been used to capture temporal and dynamic regulation of Hox gene expression at the nucleosome/chromatin level. This chapter focuses on the utilization of capture MNase-seq and Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq), two advanced techniques that enable the exploration of chromatin accessibility and nucleosome positioning within these critical genomic regions.
{"title":"Capturing Chromatin Organization by MNase-seq and ATAC-seq.","authors":"Mika Saotome, Jill Goodman, Motoki Takaku","doi":"10.1007/978-1-0716-4322-8_12","DOIUrl":"https://doi.org/10.1007/978-1-0716-4322-8_12","url":null,"abstract":"<p><p>Hox genes play a pivotal role during development. Their expression is tightly controlled in a spatiotemporal manner, ensuring that specific body structures develop at the correct locations and times during development. Various genomics approaches have been used to capture temporal and dynamic regulation of Hox gene expression at the nucleosome/chromatin level. This chapter focuses on the utilization of capture MNase-seq and Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq), two advanced techniques that enable the exploration of chromatin accessibility and nucleosome positioning within these critical genomic regions.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2889 ","pages":"167-180"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142915523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4252-8_8
Ana Montero-Pedrazuela, Silvia Constanza Contreras-Jurado
Somatic cells can be transformed into induced pluripotent stem cells (iPSCs) using a technique called reprogramming. This process involves introducing Yamanaka factors (Oct4, Sox2, Klf4, and c-Myc) to the cells through retroviral supernatants. This chapter outlines a protocol for reprogramming mouse embryonic fibroblasts (MEFs) using the hormone triiodo-L-thyronine (T3) to enhance the generation of iPSCs. It also describes how to analyze these iPSCs by colony staining for alkaline phosphatase activity, a standard marker for identifying pluripotent embryonic stem cells. To further study iPSCs, individual colonies must be selected and expanded, and pluripotency is examined by analyzing gene expression profiles using quantitative real-time PCR to measure the endogenous expression of pluripotency genes. Integrating T3 into reprogramming methods may significantly improve the production of functional iPSCs. This advancement could open new avenues for research in cell plasticity, disease modeling, and regenerative therapies.
体细胞可以通过一种叫做重编程的技术转化为诱导多能干细胞(iPSC)。这一过程包括通过逆转录病毒上清液向细胞引入山中因子(Oct4、Sox2、Klf4和c-Myc)。本章概述了使用三碘-L-甲硫氨酸(T3)激素对小鼠胚胎成纤维细胞(MEFs)进行重编程的方案,以提高 iPSCs 的生成。报告还介绍了如何通过对碱性磷酸酶活性进行菌落染色来分析这些iPSC,碱性磷酸酶是识别多能胚胎干细胞的标准标记。要进一步研究 iPSC,必须对单个菌落进行筛选和扩增,并通过使用定量实时 PCR 分析基因表达谱来测量多能性基因的内源性表达,从而检验多能性。将 T3 整合到重编程方法中可能会大大改善功能性 iPSCs 的生产。这一进展将为细胞可塑性、疾病建模和再生疗法的研究开辟新的途径。
{"title":"Mouse Embryonic Fibroblasts Reprogramming to Induced Pluripotent Stem Cells by T3.","authors":"Ana Montero-Pedrazuela, Silvia Constanza Contreras-Jurado","doi":"10.1007/978-1-0716-4252-8_8","DOIUrl":"10.1007/978-1-0716-4252-8_8","url":null,"abstract":"<p><p>Somatic cells can be transformed into induced pluripotent stem cells (iPSCs) using a technique called reprogramming. This process involves introducing Yamanaka factors (Oct4, Sox2, Klf4, and c-Myc) to the cells through retroviral supernatants. This chapter outlines a protocol for reprogramming mouse embryonic fibroblasts (MEFs) using the hormone triiodo-L-thyronine (T3) to enhance the generation of iPSCs. It also describes how to analyze these iPSCs by colony staining for alkaline phosphatase activity, a standard marker for identifying pluripotent embryonic stem cells. To further study iPSCs, individual colonies must be selected and expanded, and pluripotency is examined by analyzing gene expression profiles using quantitative real-time PCR to measure the endogenous expression of pluripotency genes. Integrating T3 into reprogramming methods may significantly improve the production of functional iPSCs. This advancement could open new avenues for research in cell plasticity, disease modeling, and regenerative therapies.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2876 ","pages":"117-130"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142695079","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4252-8_14
Norberto Herrera Merino, María Victoria Redondo Vega
The thyroid gland, located at the base of the neck, regulates metabolism and hormone balance through hormones like T4 and T3, which are essential for growth, neurological development, and energy production. Thyroid diseases affect 10% of the global population, making accurate and up-to-date information on surgical interventions and advancements crucial for improving clinical outcomes. Thyroid gland surgery is a dynamic field that has experienced remarkable advances in diagnosis, surgical techniques, and postoperative management. These include new advances in surgical techniques that improve precision, reduce surgical trauma, and speed up patient recovery, identification of biomarkers, and understanding of the molecular characteristics of tumors that allow for more targeted therapeutic strategies, and incorporation of advanced technologies that improve diagnostic accuracy and efficacy. This review aims to guide healthcare professionals and lay the groundwork for future research and innovative treatments in thyroid surgery.
甲状腺位于颈部底部,通过 T4 和 T3 等激素调节新陈代谢和激素平衡,这些激素对生长、神经系统发育和能量生产至关重要。甲状腺疾病影响着全球10%的人口,因此准确、最新的外科干预和进展信息对于提高临床治疗效果至关重要。甲状腺手术是一个充满活力的领域,在诊断、手术技术和术后管理方面都取得了显著进步。这些进步包括:提高手术精确度、减少手术创伤、加快患者康复的外科技术新进展;生物标志物的确定;对肿瘤分子特征的了解,从而可以采取更有针对性的治疗策略;以及先进技术的应用,从而提高诊断的准确性和有效性。本综述旨在为医护人员提供指导,并为甲状腺外科的未来研究和创新治疗奠定基础。
{"title":"Review of Surgical Interventions in the Thyroid Gland: Recent Advances and Current Considerations.","authors":"Norberto Herrera Merino, María Victoria Redondo Vega","doi":"10.1007/978-1-0716-4252-8_14","DOIUrl":"10.1007/978-1-0716-4252-8_14","url":null,"abstract":"<p><p>The thyroid gland, located at the base of the neck, regulates metabolism and hormone balance through hormones like T4 and T3, which are essential for growth, neurological development, and energy production. Thyroid diseases affect 10% of the global population, making accurate and up-to-date information on surgical interventions and advancements crucial for improving clinical outcomes. Thyroid gland surgery is a dynamic field that has experienced remarkable advances in diagnosis, surgical techniques, and postoperative management. These include new advances in surgical techniques that improve precision, reduce surgical trauma, and speed up patient recovery, identification of biomarkers, and understanding of the molecular characteristics of tumors that allow for more targeted therapeutic strategies, and incorporation of advanced technologies that improve diagnostic accuracy and efficacy. This review aims to guide healthcare professionals and lay the groundwork for future research and innovative treatments in thyroid surgery.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2876 ","pages":"201-220"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142695340","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4252-8_1
Silvia Constanza Contreras-Jurado
The hypothalamus secretes the thyroid-releasing hormone (TRH) that induces the pituitary gland to release the thyroid-stimulating hormone (TSH) which stimulates thyroid follicular cells to release the thyroid hormones (THs), thyroxine (T4), and triiodothyronine (T3). The process of synthesizing T3 and T4 hormones involves various enzymatic steps, starting with the iodination of L-tyrosine residues present in the protein thyroglobulin. Thyroid hormones are released into the bloodstream, where they bind to thyroid hormone distributor proteins (THDPs) which transport them in the circulation. The conversion of T4 to T3 (the more biologically active hormone) in target tissues is facilitated by selenoprotein enzymes known as deiodinases. THs can bind to different molecules located on the plasma membrane, such as integrin αvβ3, through which they exercise regulatory non-genomic control. Nevertheless, most of thyroid hormone's actions are mediated intracellularly by binding to thyroid hormone receptors (TRs). Thyroid hormone receptors act as ligand-dependent transcription factors, Thyroid hormone receptors activate thyroid hormone response elements on gene promoters through canonical signaling. Thyroid hormones mediate several critical physiological processes including organ development, cell differentiation, metabolism, and cell growth and maintenance.
下丘脑分泌甲状腺释放激素(TRH),诱导垂体释放促甲状腺激素(TSH),后者刺激甲状腺滤泡细胞释放甲状腺激素(THs)、甲状腺素(T4)和三碘甲状腺原氨酸(T3)。合成 T3 和 T4 激素的过程涉及多个酶促步骤,首先是将存在于蛋白质甲状腺球蛋白中的 L-酪氨酸残基碘化。甲状腺激素释放到血液中,与甲状腺激素分配蛋白(THDPs)结合,后者在血液循环中运输甲状腺激素。T4在靶组织中转化为T3(生物活性更强的激素)的过程由被称为脱碘酶的硒蛋白酶促进。甲状腺激素可与位于质膜上的不同分子结合,如整合素αvβ3,通过这些分子进行非基因组控制。不过,甲状腺激素的大部分作用是通过与甲状腺激素受体(TRs)结合在细胞内介导的。甲状腺激素受体作为配体依赖性转录因子,通过典型信号激活基因启动子上的甲状腺激素反应元件。甲状腺激素介导了多个关键的生理过程,包括器官发育、细胞分化、新陈代谢以及细胞生长和维持。
{"title":"Thyroid Hormones and Co-workers: An Overview.","authors":"Silvia Constanza Contreras-Jurado","doi":"10.1007/978-1-0716-4252-8_1","DOIUrl":"10.1007/978-1-0716-4252-8_1","url":null,"abstract":"<p><p>The hypothalamus secretes the thyroid-releasing hormone (TRH) that induces the pituitary gland to release the thyroid-stimulating hormone (TSH) which stimulates thyroid follicular cells to release the thyroid hormones (THs), thyroxine (T4), and triiodothyronine (T3). The process of synthesizing T3 and T4 hormones involves various enzymatic steps, starting with the iodination of L-tyrosine residues present in the protein thyroglobulin. Thyroid hormones are released into the bloodstream, where they bind to thyroid hormone distributor proteins (THDPs) which transport them in the circulation. The conversion of T4 to T3 (the more biologically active hormone) in target tissues is facilitated by selenoprotein enzymes known as deiodinases. THs can bind to different molecules located on the plasma membrane, such as integrin αvβ3, through which they exercise regulatory non-genomic control. Nevertheless, most of thyroid hormone's actions are mediated intracellularly by binding to thyroid hormone receptors (TRs). Thyroid hormone receptors act as ligand-dependent transcription factors, Thyroid hormone receptors activate thyroid hormone response elements on gene promoters through canonical signaling. Thyroid hormones mediate several critical physiological processes including organ development, cell differentiation, metabolism, and cell growth and maintenance.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2876 ","pages":"3-16"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142695380","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4264-1_8
Magdalena Lebiedzinska-Arciszewska, Jan Suski, Massimo Bonora, Barbara Pakula, Paolo Pinton, Jerzy Duszynski, Patrycja Jakubek-Olszewska, Mariusz R Wieckowski
Mitochondria are considered one of the main sites of reactive oxygen species (ROS) production in the eukaryotic cells. For this reason, mitochondrial dysfunction associated with increased ROS production underlies various pathological conditions as well as promotes aging. Chronically increased rates of ROS production contribute to oxidative damage to macromolecules, i.e., DNA, proteins, and lipids. Accumulation of unrepaired oxidative damage may result in progressive cell dysfunction, which can finally trigger cell death. The main by-product of mitochondrial oxidative phosphorylation is superoxide, which is generated by the leak of electrons from the mitochondrial respiratory chain complexes leading to one-electron reduction of oxygen. Mitochondrial superoxide dismutase (MnSOD, SOD2) as well as cytosolic superoxide dismutase (Cu/ZnSOD, SOD1), whose smaller pool is localized in the mitochondrial intermembrane space, converts superoxide to H2O2, which can be then degraded by the catalase to harmless H2O.In this chapter, we focus on the relationship between one of the bioenergetic parameters, which is mitochondrial membrane potential, and the rate of ROS formation. We present a set of various methods enabling the characterization of these parameters applicable to isolated mitochondria or intact cells. We also present examples of experimental data demonstrating that the magnitude and direction (increase or decrease) of a change in mitochondrial ROS production depend on the mitochondrial metabolic state.
{"title":"The Relation Between Mitochondrial Membrane Potential and Reactive Oxygen Species Formation.","authors":"Magdalena Lebiedzinska-Arciszewska, Jan Suski, Massimo Bonora, Barbara Pakula, Paolo Pinton, Jerzy Duszynski, Patrycja Jakubek-Olszewska, Mariusz R Wieckowski","doi":"10.1007/978-1-0716-4264-1_8","DOIUrl":"10.1007/978-1-0716-4264-1_8","url":null,"abstract":"<p><p>Mitochondria are considered one of the main sites of reactive oxygen species (ROS) production in the eukaryotic cells. For this reason, mitochondrial dysfunction associated with increased ROS production underlies various pathological conditions as well as promotes aging. Chronically increased rates of ROS production contribute to oxidative damage to macromolecules, i.e., DNA, proteins, and lipids. Accumulation of unrepaired oxidative damage may result in progressive cell dysfunction, which can finally trigger cell death. The main by-product of mitochondrial oxidative phosphorylation is superoxide, which is generated by the leak of electrons from the mitochondrial respiratory chain complexes leading to one-electron reduction of oxygen. Mitochondrial superoxide dismutase (MnSOD, SOD2) as well as cytosolic superoxide dismutase (Cu/ZnSOD, SOD1), whose smaller pool is localized in the mitochondrial intermembrane space, converts superoxide to H<sub>2</sub>O<sub>2</sub>, which can be then degraded by the catalase to harmless H<sub>2</sub>O.In this chapter, we focus on the relationship between one of the bioenergetic parameters, which is mitochondrial membrane potential, and the rate of ROS formation. We present a set of various methods enabling the characterization of these parameters applicable to isolated mitochondria or intact cells. We also present examples of experimental data demonstrating that the magnitude and direction (increase or decrease) of a change in mitochondrial ROS production depend on the mitochondrial metabolic state.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2878 ","pages":"133-162"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142638482","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4264-1_13
Eliane Caseiro Soares de Menezes, Afshan Navid Malik
Mitochondrial DNA copy number (mtDNA-CN) in human body fluids is widely used as a biomarker of mitochondrial dysfunction in common metabolic diseases. Here we describe protocols to measure cellular and/or cell free (cf)-mtDNA-CN in human peripheral blood and urine. Cellular mtDNA is located inside the mitochondria where it encodes key subunits of the respiratory complexes in mitochondria and is usually normalized with reference to the nuclear genome as the mitochondrial genome to nuclear genome ratio (Mt/N) in either whole blood, peripheral blood mononuclear cells (PBMCs), or whole urine. Cf -mtDNA is usually found outside of the mitochondria, often released following mitochondrial damage, can trigger inflammatory pathways, and is usually measured as mtDNA-CN per volume of the starting material. Here we describe how to (1) separate whole blood into PBMCs, plasma, and serum fractions and whole urine into urinary supernatant and pellet, (2) prepare DNA from each of these fractions, (3) prepare reference standards for absolute quantification, (4) carry out qPCR for either relative or absolute quantification from test samples, (5) analyze qPCR data, and (6) calculate the sample size to adequately power studies. The protocol presented here is suitable for high throughput use and can be modified to quantify mtDNA from other body fluids, human cells, and tissues.
{"title":"Absolute Quantification of Cellular and Cell-Free Mitochondrial DNA Copy Number from Human Blood and Urinary Samples Using Real Time Quantitative PCR.","authors":"Eliane Caseiro Soares de Menezes, Afshan Navid Malik","doi":"10.1007/978-1-0716-4264-1_13","DOIUrl":"10.1007/978-1-0716-4264-1_13","url":null,"abstract":"<p><p>Mitochondrial DNA copy number (mtDNA-CN) in human body fluids is widely used as a biomarker of mitochondrial dysfunction in common metabolic diseases. Here we describe protocols to measure cellular and/or cell free (cf)-mtDNA-CN in human peripheral blood and urine. Cellular mtDNA is located inside the mitochondria where it encodes key subunits of the respiratory complexes in mitochondria and is usually normalized with reference to the nuclear genome as the mitochondrial genome to nuclear genome ratio (Mt/N) in either whole blood, peripheral blood mononuclear cells (PBMCs), or whole urine. Cf -mtDNA is usually found outside of the mitochondria, often released following mitochondrial damage, can trigger inflammatory pathways, and is usually measured as mtDNA-CN per volume of the starting material. Here we describe how to (1) separate whole blood into PBMCs, plasma, and serum fractions and whole urine into urinary supernatant and pellet, (2) prepare DNA from each of these fractions, (3) prepare reference standards for absolute quantification, (4) carry out qPCR for either relative or absolute quantification from test samples, (5) analyze qPCR data, and (6) calculate the sample size to adequately power studies. The protocol presented here is suitable for high throughput use and can be modified to quantify mtDNA from other body fluids, human cells, and tissues.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2878 ","pages":"233-257"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142639376","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-01DOI: 10.1007/978-1-0716-4264-1_9
Sónia A Pinho, Georgina L Gardner, Ricardo Alva, Jeff A Stuart, Teresa Cunha-Oliveira
In vitro and ex vivo studies are crucial for mitochondrial research, offering valuable insights into cellular mechanisms and aiding in diagnostic and therapeutic strategies. Accurate in vitro models rely on adequate cell culture conditions, such as the composition of culture media and oxygenation levels. These conditions can influence energy metabolism and mitochondrial activities, thus impacting studies involving mitochondrial components, such as the effectiveness of anticancer drugs. This chapter focuses on practical guidance for creating setups that replicate in vivo microenvironments, capturing the original metabolic context of cells. We explore protocols to better mimic the physiological cell environment, promote cellular reconfiguration, and prime cells according to the modeled context. The first part is dedicated to the use of human dermal fibroblasts, which are a promising model for pre-clinical mitochondrial research due to their adaptability and relevance to human mitochondrial physiology. We present an optimized protocol for gradually adjusting extracellular glucose levels, which demonstrated significant mitochondrial, metabolic, and redox remodeling in normal adult dermal fibroblasts. The second part is dedicated to replication of tumor microenvironments, which are relevant for studies targeting cellular energy metabolism to inhibit tumor growth. Currently available physiological media can mimic blood plasma metabolome but not the specific tumor microenvironment. To address this, we describe optimized media formulation and oxygenation protocols, which can simulate the tumor microenvironment in cell culture experiments. Replicating in vivo microenvironments in in vitro and ex vivo studies can enhance our understanding of cellular processes, facilitate drug development, and advance personalized therapeutics in mitochondrial medicine.
{"title":"Creating Physiological Cell Environments In Vitro: Adjusting Cell Culture Media Composition and Oxygen Levels to Investigate Mitochondrial Function and Cancer Metabolism.","authors":"Sónia A Pinho, Georgina L Gardner, Ricardo Alva, Jeff A Stuart, Teresa Cunha-Oliveira","doi":"10.1007/978-1-0716-4264-1_9","DOIUrl":"10.1007/978-1-0716-4264-1_9","url":null,"abstract":"<p><p>In vitro and ex vivo studies are crucial for mitochondrial research, offering valuable insights into cellular mechanisms and aiding in diagnostic and therapeutic strategies. Accurate in vitro models rely on adequate cell culture conditions, such as the composition of culture media and oxygenation levels. These conditions can influence energy metabolism and mitochondrial activities, thus impacting studies involving mitochondrial components, such as the effectiveness of anticancer drugs. This chapter focuses on practical guidance for creating setups that replicate in vivo microenvironments, capturing the original metabolic context of cells. We explore protocols to better mimic the physiological cell environment, promote cellular reconfiguration, and prime cells according to the modeled context. The first part is dedicated to the use of human dermal fibroblasts, which are a promising model for pre-clinical mitochondrial research due to their adaptability and relevance to human mitochondrial physiology. We present an optimized protocol for gradually adjusting extracellular glucose levels, which demonstrated significant mitochondrial, metabolic, and redox remodeling in normal adult dermal fibroblasts. The second part is dedicated to replication of tumor microenvironments, which are relevant for studies targeting cellular energy metabolism to inhibit tumor growth. Currently available physiological media can mimic blood plasma metabolome but not the specific tumor microenvironment. To address this, we describe optimized media formulation and oxygenation protocols, which can simulate the tumor microenvironment in cell culture experiments. Replicating in vivo microenvironments in in vitro and ex vivo studies can enhance our understanding of cellular processes, facilitate drug development, and advance personalized therapeutics in mitochondrial medicine.</p>","PeriodicalId":18490,"journal":{"name":"Methods in molecular biology","volume":"2878 ","pages":"163-199"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142639379","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}