1
|
Yoon B, Kim H, Jung SW, Park J. Single-cell lineage tracing approaches to track kidney cell development and maintenance. Kidney Int 2024; 105:1186-1199. [PMID: 38554991 DOI: 10.1016/j.kint.2024.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 12/06/2023] [Accepted: 01/09/2024] [Indexed: 04/02/2024]
Abstract
The kidney is a complex organ consisting of various cell types. Previous studies have aimed to elucidate the cellular relationships among these cell types in developing and mature kidneys using Cre-loxP-based lineage tracing. However, this methodology falls short of fully capturing the heterogeneous nature of the kidney, making it less than ideal for comprehensively tracing cellular progression during kidney development and maintenance. Recent technological advancements in single-cell genomics have revolutionized lineage tracing methods. Single-cell lineage tracing enables the simultaneous tracing of multiple cell types within complex tissues and their transcriptomic profiles, thereby allowing the reconstruction of their lineage tree with cell state information. Although single-cell lineage tracing has been successfully applied to investigate cellular hierarchies in various organs and tissues, its application in kidney research is currently lacking. This review comprehensively consolidates the single-cell lineage tracing methods, divided into 4 categories (clustered regularly interspaced short palindromic repeat [CRISPR]/CRISPR-associated protein 9 [Cas9]-based, transposon-based, Polylox-based, and native barcoding methods), and outlines their technical advantages and disadvantages. Furthermore, we propose potential future research topics in kidney research that could benefit from single-cell lineage tracing and suggest suitable technical strategies to apply to these topics.
Collapse
Affiliation(s)
- Baul Yoon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Hayoung Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea
| | - Su Woong Jung
- Division of Nephrology, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul, Republic of Korea; Division of Nephrology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Seoul, Republic of Korea.
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, Republic of Korea.
| |
Collapse
|
2
|
Fu Y, Xiang Y, Wei Q, Ilatovskaya D, Dong Z. Rodent models of AKI and AKI-CKD transition: an update in 2024. Am J Physiol Renal Physiol 2024; 326:F563-F583. [PMID: 38299215 PMCID: PMC11208034 DOI: 10.1152/ajprenal.00402.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024] Open
Abstract
Despite known drawbacks, rodent models are essential tools in the research of renal development, physiology, and pathogenesis. In the past decade, rodent models have been developed and used to mimic different etiologies of acute kidney injury (AKI), AKI to chronic kidney disease (CKD) transition or progression, and AKI with comorbidities. These models have been applied for both mechanistic research and preclinical drug development. However, current rodent models have their limitations, especially since they often do not fully recapitulate the pathophysiology of AKI in human patients, and thus need further refinement. Here, we discuss the present status of these rodent models, including the pathophysiologic compatibility, clinical translational significance, key factors affecting model consistency, and their main limitations. Future efforts should focus on establishing robust models that simulate the major clinical and molecular phenotypes of human AKI and its progression.
Collapse
Affiliation(s)
- Ying Fu
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
| | - Yu Xiang
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
| | - Daria Ilatovskaya
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, United States
| | - Zheng Dong
- Department of Nephrology, Institute of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, People's Republic of China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
- Research Department, Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, United States
| |
Collapse
|
3
|
Long HY, Qian ZP, Lan Q, Xu YJ, Da JJ, Yu FX, Zha Y. Human pluripotent stem cell-derived kidney organoids: Current progress and challenges. World J Stem Cells 2024; 16:114-125. [PMID: 38455108 PMCID: PMC10915962 DOI: 10.4252/wjsc.v16.i2.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/18/2023] [Accepted: 01/29/2024] [Indexed: 02/26/2024] Open
Abstract
Human pluripotent stem cell (hPSC)-derived kidney organoids share similarities with the fetal kidney. However, the current hPSC-derived kidney organoids have some limitations, including the inability to perform nephrogenesis and lack of a corticomedullary definition, uniform vascular system, and coordinated exit pathway for urinary filtrate. Therefore, further studies are required to produce hPSC-derived kidney organoids that accurately mimic human kidneys to facilitate research on kidney development, regeneration, disease modeling, and drug screening. In this review, we discussed recent advances in the generation of hPSC-derived kidney organoids, how these organoids contribute to the understanding of human kidney development and research in disease modeling. Additionally, the limitations, future research focus, and applications of hPSC-derived kidney organoids were highlighted.
Collapse
Affiliation(s)
- Hong-Yan Long
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Zu-Ping Qian
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Qin Lan
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
| | - Yong-Jie Xu
- Department of Laboratory Medicine, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Jing-Jing Da
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Fu-Xun Yu
- Key Laboratory of Diagnosis and Treatment of Pulmonary Immune Diseases, National Health Commission, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China
| | - Yan Zha
- Graduate School, Zunyi Medical University, Zunyi 563000, Guizhou Province, China
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang 550002, Guizhou Province, China.
| |
Collapse
|
4
|
Li Z, Yang W, Wu P, Shan Y, Zhang X, Chen F, Yang J, Yang JR. Reconstructing cell lineage trees with genomic barcoding: approaches and applications. J Genet Genomics 2024; 51:35-47. [PMID: 37269980 DOI: 10.1016/j.jgg.2023.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/05/2023]
Abstract
In multicellular organisms, developmental history of cell divisions and functional annotation of terminal cells can be organized into a cell lineage tree (CLT). The reconstruction of the CLT has long been a major goal in developmental biology and other related fields. Recent technological advancements, especially those in editable genomic barcodes and single-cell high-throughput sequencing, have sparked a new wave of experimental methods for reconstructing CLTs. Here we review the existing experimental approaches to the reconstruction of CLT, which are broadly categorized as either image-based or DNA barcode-based methods. In addition, we present a summary of the related literature based on the biological insight provided by the obtained CLTs. Moreover, we discuss the challenges that will arise as more and better CLT data become available in the near future. Genomic barcoding-based CLT reconstructions and analyses, due to their wide applicability and high scalability, offer the potential for novel biological discoveries, especially those related to general and systemic properties of the developmental process.
Collapse
Affiliation(s)
- Zizhang Li
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Wenjing Yang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Peng Wu
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Yuyan Shan
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Xiaoyu Zhang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Feng Chen
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Junnan Yang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jian-Rong Yang
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
5
|
Logotheti S, Papadaki E, Zolota V, Logothetis C, Vrahatis AG, Soundararajan R, Tzelepi V. Lineage Plasticity and Stemness Phenotypes in Prostate Cancer: Harnessing the Power of Integrated "Omics" Approaches to Explore Measurable Metrics. Cancers (Basel) 2023; 15:4357. [PMID: 37686633 PMCID: PMC10486655 DOI: 10.3390/cancers15174357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Prostate cancer (PCa), the most frequent and second most lethal cancer type in men in developed countries, is a highly heterogeneous disease. PCa heterogeneity, therapy resistance, stemness, and lethal progression have been attributed to lineage plasticity, which refers to the ability of neoplastic cells to undergo phenotypic changes under microenvironmental pressures by switching between developmental cell states. What remains to be elucidated is how to identify measurements of lineage plasticity, how to implement them to inform preclinical and clinical research, and, further, how to classify patients and inform therapeutic strategies in the clinic. Recent research has highlighted the crucial role of next-generation sequencing technologies in identifying potential biomarkers associated with lineage plasticity. Here, we review the genomic, transcriptomic, and epigenetic events that have been described in PCa and highlight those with significance for lineage plasticity. We further focus on their relevance in PCa research and their benefits in PCa patient classification. Finally, we explore ways in which bioinformatic analyses can be used to determine lineage plasticity based on large omics analyses and algorithms that can shed light on upstream and downstream events. Most importantly, an integrated multiomics approach may soon allow for the identification of a lineage plasticity signature, which would revolutionize the molecular classification of PCa patients.
Collapse
Affiliation(s)
- Souzana Logotheti
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| | - Eugenia Papadaki
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
- Department of Informatics, Ionian University, 49100 Corfu, Greece;
| | - Vasiliki Zolota
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | | | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vasiliki Tzelepi
- Department of Pathology, University of Patras, 26504 Patras, Greece; (S.L.); (E.P.); (V.Z.)
| |
Collapse
|
6
|
Gross A, Buschang PH, Shakya A, Jing Y. Short-term effects of mechanical loading on the transdifferentiation of condylar chondrocytes. Am J Orthod Dentofacial Orthop 2023; 164:201-214. [PMID: 36922241 PMCID: PMC10659147 DOI: 10.1016/j.ajodo.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 03/14/2023]
Abstract
INTRODUCTION Transdifferentiation of chondrocytes into bone cells explains most of the prenatal and early postnatal condylar growth, but its role during later postnatal growth and the mechanisms regulating transdifferentiation remain unknown. This study aimed to quantify the effects of mechanical loading on chondrocyte-derived osteogenesis during late postnatal condylar growth using a short-term mandibular laterotrusion model. METHODS Thirty 4-week-old Aggrecan-CreERT2, R26RtdTomato, and 2.3Col1a1-GFP compound mice received tamoxifen injections and were divided into control and experimental groups. Appliances were bonded to shift the mandibles of the experimental mice for 5 days, causing protrusion and retrusion of the right and left condyles, respectively. Radiographic, microcomputed tomographic, and histomorphometric analyses were performed. RESULTS The experimental and control groups showed substantial transdifferentiation of chondrocytes into bone cells. The experimental mice developed asymmetric mandibles, with the protrusive side significantly longer than the retrusive side. The protrusive condyles showed significantly increased chondrogenesis and greater numbers of chondrocyte-derived osteogenic cells, especially in the posterior third. The opposite effects were seen on the retrusive side. CONCLUSIONS Transdifferentiation of chondrocytes into bone cells occurs during late postnatal condylar growth. Laterotrusion regulates condylar chondrogenesis and chondrocyte transdifferentiation, which alters the amount and direction of condylar growth. Our study demonstrated that chondrocytes are key players in condylar bone formation and should be the focus of studies to control and further understand condylar growth.
Collapse
Affiliation(s)
- Amanda Gross
- Department of Orthodontics, Texas A&M University School of Dentistry, Dallas, Tex
| | - Peter H Buschang
- Department of Orthodontics, Texas A&M University School of Dentistry, Dallas, Tex
| | - Ajay Shakya
- Department of Biomedical Sciences, Texas A&M University School of Dentistry, Dallas, Tex
| | - Yan Jing
- Department of Orthodontics, Texas A&M University School of Dentistry, Dallas, Tex.
| |
Collapse
|
7
|
Kumar S, Fan X, Rasouly HM, Sharma R, Salant DJ, Lu W. ZEB2 controls kidney stromal progenitor differentiation and inhibits abnormal myofibroblast expansion and kidney fibrosis. JCI Insight 2023; 8:e158418. [PMID: 36445780 PMCID: PMC9870089 DOI: 10.1172/jci.insight.158418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022] Open
Abstract
FOXD1+ cell-derived stromal cells give rise to pericytes and fibroblasts that support the kidney vasculature and interstitium but are also major precursors of myofibroblasts. ZEB2 is a SMAD-interacting transcription factor that is expressed in developing kidney stromal progenitors. Here we show that Zeb2 is essential for normal FOXD1+ stromal progenitor development. Specific conditional knockout of mouse Zeb2 in FOXD1+ stromal progenitors (Zeb2 cKO) leads to abnormal interstitial stromal cell development, differentiation, and kidney fibrosis. Immunofluorescent staining analyses revealed abnormal expression of interstitial stromal cell markers MEIS1/2/3, CDKN1C, and CSPG4 (NG2) in newborn and 3-week-old Zeb2-cKO mouse kidneys. Zeb2-deficient FOXD1+ stromal progenitors also took on a myofibroblast fate that led to kidney fibrosis and kidney failure. Cell marker studies further confirmed that these myofibroblasts expressed pericyte and resident fibroblast markers, including PDGFRβ, CSPG4, desmin, GLI1, and NT5E. Notably, increased interstitial collagen deposition associated with loss of Zeb2 in FOXD1+ stromal progenitors was accompanied by increased expression of activated SMAD1/5/8, SMAD2/3, SMAD4, and AXIN2. Thus, our study identifies a key role of ZEB2 in maintaining the cell fate of FOXD1+ stromal progenitors during kidney development, whereas loss of ZEB2 leads to differentiation of FOXD1+ stromal progenitors into myofibroblasts and kidney fibrosis.
Collapse
|
8
|
Safi W, Marco A, Moya D, Prado P, Garreta E, Montserrat N. Assessing kidney development and disease using kidney organoids and CRISPR engineering. Front Cell Dev Biol 2022; 10:948395. [PMID: 36120564 PMCID: PMC9479189 DOI: 10.3389/fcell.2022.948395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/06/2022] [Indexed: 11/26/2022] Open
Abstract
The differentiation of human pluripotent stem cells (hPSCs) towards organoids is one of the biggest scientific advances in regenerative medicine. Kidney organoids have not only laid the groundwork for various organ-like tissue systems but also provided insights into kidney embryonic development. Thus, several protocols for the differentiation of renal progenitors or mature cell types have been established. Insights into the interplay of developmental pathways in nephrogenesis and determination of different cell fates have enabled the in vitro recapitulation of nephrogenesis. Here we first provide an overview of kidney morphogenesis and patterning in the mouse model in order to dissect signalling pathways that are key to define culture conditions sustaining renal differentiation from hPSCs. Secondly, we also highlight how genome editing approaches have provided insights on the specific role of different genes and molecular pathways during renal differentiation from hPSCs. Based on this knowledge we further review how CRISPR/Cas9 technology has enabled the recapitulation and correction of cellular phenotypes associated with human renal disease. Last, we also revise how the field has positively benefited from emerging technologies as single cell RNA sequencing and discuss current limitations on kidney organoid technology that will take advantage from bioengineering solutions to help standardizing the use of this model systems to study kidney development and disease.
Collapse
Affiliation(s)
- Wajima Safi
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
- *Correspondence: Wajima Safi, ; Elena Garreta, ; Nuria Montserrat,
| | - Andrés Marco
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
| | | | - Patricia Prado
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Elena Garreta
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
- *Correspondence: Wajima Safi, ; Elena Garreta, ; Nuria Montserrat,
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration. Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- *Correspondence: Wajima Safi, ; Elena Garreta, ; Nuria Montserrat,
| |
Collapse
|
9
|
Zhang W, Gao C, Tsilosani A, Samarakoon R, Plews R, Higgins P. Potential renal stem/progenitor cells identified by in vivo lineage tracing. Am J Physiol Renal Physiol 2022; 322:F379-F391. [PMID: 35100814 PMCID: PMC8934668 DOI: 10.1152/ajprenal.00326.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mammalian kidneys consist of more than 30 different types of cells. A challenging task is to identify and characterize the stem/progenitor subpopulations that establish the lineage relationships among these cellular elements during nephrogenesis in the embryonic and neonate kidneys and during tissue homeostasis and/or injury repair in the mature kidney. Moreover, the potential clinical utility of stem/progenitor cells holds promise for development of new regenerative medicine approaches for the treatment of renal diseases. Stem cells are defined by unlimited self-renewal capacity and pluripotentiality. Progenitor cells have pluripotentiality, but no or limited self-renewal potential. Cre-LoxP-based in vivo genetic lineage tracing is a powerful tool to identify the stem/progenitor cells in their native environment. Hypothetically, this technique enables investigators to accurately track the progeny of a single cell, or a group of cells. The Cre/loxP system has been widely employed to uncover the function of genes in various mammalian tissues and to identify stem/progenitor cells through in vivo lineage tracing analyses. In this review, we summarize the recent advances in the development and characterization of various Cre drivers, and their use in identifying potential renal stem/progenitor cells in both developing and mature mouse kidneys.
Collapse
Affiliation(s)
- Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Chao Gao
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Akaki Tsilosani
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| | - Robert Plews
- Department of General Surgery, Albany Medical College, Albany, NY, United States
| | - Paul Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, United States
| |
Collapse
|
10
|
Direct evidence of proximal tubular proliferation in early diabetic nephropathy. Sci Rep 2022; 12:778. [PMID: 35039597 PMCID: PMC8763925 DOI: 10.1038/s41598-022-04880-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/28/2021] [Indexed: 12/26/2022] Open
Abstract
Kidney hypertrophy is a common clinical feature in patients with diabetes and is associated with poor renal outcomes. Initial cell proliferation followed by cellular hypertrophy are considered the responsible mechanisms for diabetic kidney hypertrophy. However, whether similar responses against hyperglycemia continue in the chronic phase in diabetes is unclear. We performed lineage tracing analysis of proximal tubular epithelia using novel type 2 diabetic mice with a tamoxifen-inducible proximal tubule-specific fluorescent reporter. Clonal analysis of proximal tubular epithelia demonstrated that the labeled epithelia proliferated in type 2 diabetic mice. Based on the histological analysis and protein/DNA ratio of sorted labeled tubular epithelia, there was no evidence of cellular hypertrophy in type 2 diabetic mice. Lineage tracing and histological analyses of streptozocin-induced type 1 diabetes also revealed that cellular proliferation occurs in the chronic phase of type 1 diabetes induction. According to our study, epithelial proliferation accompanied by SGLT2 upregulation, rather than cellular hypertrophy, predominantly occurs in the hypertrophic kidney in both type 1 and type 2 diabetes. An increased number of SGLT2+ tubular epithelia may be an adaptive response against hyperglycemia, and linked to the hyper-reabsorption of sodium and glucose observed in type 2 diabetes patients.
Collapse
|
11
|
Muto Y, Humphreys BD. Recent advances in lineage tracing for the kidney. Kidney Int 2021; 100:1179-1184. [PMID: 34217781 PMCID: PMC8608712 DOI: 10.1016/j.kint.2021.05.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 11/19/2022]
Abstract
Lineage tracing was originally developed by developmental biologists to identify all progeny of a single cell during morphogenesis. More recently this approach has been applied to other fields, including organ homeostasis and recovery from injury. Modern lineage tracing techniques typically rely on reporter gene expression induced by cell-specific DNA recombination. There have been important scientific advances in the last 10 years that have impacted lineage tracing approaches, including intersectional genetics, optical clearing techniques, and the use of sequencing-based genomic lineage tracing. The latter combines CRISPR-Cas9-based genetic scarring with single-cell RNA-sequencing that, in theory, could allow comprehensive reconstruction of a lineage tree for an entire organism. This review summarizes recent advances in lineage tracing technologies and outlines potential applications for kidney research.
Collapse
Affiliation(s)
- Yoshiharu Muto
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA; Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
12
|
Abstract
AbstractAcute kidney injury (AKI) is a common clinical symptom, which is mainly manifested by elevated serum creatinine and blood urea nitrogen levels. When AKI is not repaired in time, the patient is prone to develop chronic kidney disease (CKD). The kidney is composed of more than 30 different cells, and its structure is complex. It is extremely challenging to understand the lineage relationships and cell fate of these cells in the process of kidney injury and regeneration. Since the 20th century, lineage tracing technology has provided an important mean for studying organ development, tissue damage repair, and the differentiation and fate of single cells. However, traditional lineage tracing methods rely on sacrificing animals to make tissue slices and then take snapshots with conventional imaging tools to obtain interesting information. This method cannot achieve dynamic and continuous monitoring of cell actions on living animals. As a kind of intravital microscopy (IVM), two-photon microscopy (TPM) has successfully solved the above problems. Because TPM has the ability to penetrate deep tissues and can achieve imaging at the single cell level, lineage tracing technology with TPM is gradually becoming popular. In this review, we provided the key technical elements of lineage tracing, and how to use intravital imaging technology to visualize and quantify the fate of renal cells.
Collapse
|
13
|
Cell Lineage Tracing: Colocalization of Cell Lineage Markers with a Fluorescent Reporter. Methods Mol Biol 2021; 2230:325-335. [PMID: 33197022 DOI: 10.1007/978-1-0716-1028-2_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cell lineage tracing, an old technique which originated in the nineteenth century, regains popularity and relevance due to introduction of a more sensitive tomato fluorescent protein under the control of a ubiquitous promoter (Rosa 26 gene). In addition, various tissue specific CreERT2 mouse lines are widely available, making cell lineage tracing studies more specific and powerful. In this protocol, we provide a practical guide for researchers to map progeny of specific cells such as chondrocytes during development using a fluorescent reporter (tomato, red) and multiple chondrocyte Cre lines. Further, we provide valuable examples in which these tracing lines, combined with a bone reporter mouse line (2.3 Col 1a1-GFP) or costained with different immunofluorescent proteins, revealed how a chondrocyte transdifferentiates into a bone cell in vivo.
Collapse
|
14
|
Moeller MJ, Tharaux PL. Cellular regeneration of podocytes from parietal cells: the debate is still open. Kidney Int 2020; 96:542-544. [PMID: 31445579 DOI: 10.1016/j.kint.2019.04.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 04/23/2019] [Indexed: 12/16/2022]
Abstract
The study by Kaverina et al. in this issue addresses an important question: can podocytes be replenished by parietal epithelial cells (PECs)? The authors use a complex transgenic mouse model in which podocytes are labeled with GFP and PECs are simultaneously labeled with tdTomato. When Kaverina and colleagues induce focal segmental glomerulosclerosis (FSGS), they find that individual PECs are doubly labeled, coexpress podocyte markers, and form structures similar to foot processes, suggesting that these PECs may have transdifferentiated into podocytes.
Collapse
Affiliation(s)
- Marcus J Moeller
- Division of Nephrology and Clinical Immunology, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.
| | - Pierre-Louis Tharaux
- Université; de Paris, Paris Cardiovascular Centre PARCC, Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| |
Collapse
|
15
|
Jing Y, Wang Z, Li H, Ma C, Feng J. Chondrogenesis Defines Future Skeletal Patterns Via Cell Transdifferentiation from Chondrocytes to Bone Cells. Curr Osteoporos Rep 2020; 18:199-209. [PMID: 32219639 PMCID: PMC7717675 DOI: 10.1007/s11914-020-00586-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to obtain a better understanding of how chondrogenesis defines skeletal development via cell transdifferentiation from chondrocytes to bone cells. RECENT FINDINGS A breakthrough in cell lineage tracing allows bone biologists to trace the cell fate and demonstrate that hypertrophic chondrocytes can directly transdifferentiate into bone cells during endochondral bone formation. However, there is a knowledge gap for the biological significance of this lineage extension and the mechanisms controlling this process. This review first introduces the history of the debate on the cell fate of chondrocytes in endochondral bone formation; then summarizes key findings obtained in recent years, which strongly support a new theory: the direct cell transdifferentiation from chondrocytes to bone cells precisely connects chondrogenesis (for providing a template of the future skeleton, classified as phase I) and osteogenesis (for finishing skeletal construction, or phase II) in a continuous lineage-linked process of endochondral bone formation and limb elongation; and finally outlines nutrition factors and molecules that regulate the cell transdifferentiation process during the relay from chondrogenesis to osteogenesis.
Collapse
Affiliation(s)
- Yan Jing
- Department of Orthodontics, Texas A&M University College of Dentistry, 3302 Gaston ave, Dallas, TX, 75246, USA.
| | - Zheng Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Hui Li
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
- State Key Laboratory of Oral Diseases, Department of Traumatic and Plastic Surgery, , West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chi Ma
- Department of Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Jian Feng
- Department of Orthodontics, Texas A&M University College of Dentistry, 3302 Gaston ave, Dallas, TX, 75246, USA.
| |
Collapse
|
16
|
Wu H, Humphreys BD. Single Cell Sequencing and Kidney Organoids Generated from Pluripotent Stem Cells. Clin J Am Soc Nephrol 2020; 15:550-556. [PMID: 31992574 PMCID: PMC7133134 DOI: 10.2215/cjn.07470619] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Methods to differentiate human pluripotent stem cells into kidney organoids were first introduced about 5 years ago, and since that time, the field has grown substantially. Protocols are producing increasingly complex three-dimensional structures, have been used to model human kidney disease, and have been adapted for high-throughput screening. Over this same time frame, technologies for massively parallel, single-cell RNA sequencing (scRNA-seq) have matured. Now, both of these powerful approaches are being combined to better understand how kidney organoids can be applied to the understanding of kidney development and disease. There are several reasons why this is a synergistic combination. Kidney organoids are complicated and contain many different cell types of variable maturity. scRNA-seq is an unbiased technology that can comprehensively categorize cell types, making it ideally suited to catalog all cell types present in organoids. These same characteristics also make scRNA-seq a powerful approach for quantitative comparisons between protocols, batches, and pluripotent cell lines as it becomes clear that reproducibility and quality can vary across all three variables. Lineage trajectories can be reconstructed using scRNA-seq data, enabling the rational adjustment of differentiation strategies to promote maturation of desired kidney cell types or inhibit differentiation of undesired off-target cell types. Here, we review the ways that scRNA-seq has been successfully applied in the organoid field and predict future applications for this powerful technique. We also review other developing single-cell technologies and discuss how they may be combined, using "multiomic" approaches, to improve our understanding of kidney organoid differentiation and usefulness in modeling development, disease, and toxicity testing.
Collapse
Affiliation(s)
- Haojia Wu
- Division of Nephrology, Department of Medicine; and
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine; and
- Department of Developmental Biology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| |
Collapse
|
17
|
Frahsek M, Schulte K, Chia-Gil A, Djudjaj S, Schueler H, Leuchtle K, Smeets B, Dijkman H, Floege J, Moeller MJ. Cre recombinase toxicity in podocytes: a novel genetic model for FSGS in adolescent mice. Am J Physiol Renal Physiol 2019; 317:F1375-F1382. [PMID: 31588799 DOI: 10.1152/ajprenal.00573.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Here, we show that inducible overexpression of Cre recombinase in glomerular podocytes but not in parietal epithelial cells may trigger focal segmental glomerulosclerosis (FSGS) in juvenile transgenic homocygous Pod-rtTA/LC1 mice. Administration of doxycycline shortly after birth, but not at any other time point later in life, resulted in podocyte injury and development of classical FSGS lesions in these mice. Sclerotic lesions were formed as soon as 3 wk of age, and FSGS progressed with low variability until 13 wk of age. In addition, our experiments identified Cre toxicity as a potentially relevant limitation for studies in podocytes of transgenic animals. In summary, our study establishes a novel genetic model for FSGS in mice, which exhibits low variability and manifests already at a young age.
Collapse
Affiliation(s)
- Madeleine Frahsek
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Kevin Schulte
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany.,Department of Nephrology and Hypertension, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Arnaldo Chia-Gil
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Sonja Djudjaj
- Institute of Pathology, RWTH University of Aachen, Aachen, Germany
| | - Herdit Schueler
- Institute of Human Genetics, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Katja Leuchtle
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Bart Smeets
- Department of Pathology, Radboud University, Nijmegen, The Netherlands
| | - Henry Dijkman
- Department of Pathology, Radboud University, Nijmegen, The Netherlands
| | - Jürgen Floege
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany
| | - Marcus J Moeller
- Nephrology and Clinical Immunology, University Hospital of RWTH Aachen University, Aachen, Germany.,Heisenberg Chair for Preventive and Translational Nephrology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
18
|
Drake KA, Fessler AR, Carroll TJ. Methods for renal lineage tracing: In vivo and beyond. Methods Cell Biol 2019; 154:121-143. [PMID: 31493814 DOI: 10.1016/bs.mcb.2019.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Lineage tracing has resulted in fundamental discoveries in kidney development and disease and remains a powerful technique to study mechanisms of organogenesis, homeostasis, and repair/regeneration. Following decades of research on the cellular and molecular regulation of renal organogenesis, the kidney has become one of the most well-characterized organs, resulting in exciting advancements in pluripotent stem cell differentiation, tissue bioengineering, and the potential for developing novel regenerative therapies for kidney disease. Lineage tracing, or the labeling of progeny cells arising from a single cell or group of cells, allows for spatial and temporal analyses of dynamic in vivo and in vitro processes. As lineage tracing techniques expand across disciplines of developmental biology, stem cell biology, and regenerative medicine, careful experimental design and interpretation, along with an understanding of the basic principles and technical limitations, are essential for utilizing genetically complex lineage tracing models to further understand kidney development and disease.
Collapse
Affiliation(s)
- Keri A Drake
- Division of Pediatric Nephrology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Alicia R Fessler
- Department of Internal Medicine (Nephrology), University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Thomas J Carroll
- Department of Internal Medicine (Nephrology), University of Texas Southwestern Medical Center, Dallas, TX, United States; Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| |
Collapse
|
19
|
Vanslambrouck JM, Wilson SB, Tan KS, Soo JYC, Scurr M, Spijker HS, Starks LT, Neilson A, Cui X, Jain S, Little MH, Howden SE. A Toolbox to Characterize Human Induced Pluripotent Stem Cell-Derived Kidney Cell Types and Organoids. J Am Soc Nephrol 2019; 30:1811-1823. [PMID: 31492807 DOI: 10.1681/asn.2019030303] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The generation of reporter lines for cell identity, lineage, and physiologic state has provided a powerful tool in advancing the dissection of mouse kidney morphogenesis at a molecular level. Although use of this approach is not an option for studying human development in vivo, its application in human induced pluripotent stem cells (iPSCs) is now feasible. METHODS We used CRISPR/Cas9 gene editing to generate ten fluorescence reporter iPSC lines designed to identify nephron progenitors, podocytes, proximal and distal nephron, and ureteric epithelium. Directed differentiation to kidney organoids was performed according to published protocols. Using immunofluorescence and live confocal microscopy, flow cytometry, and cell sorting techniques, we investigated organoid patterning and reporter expression characteristics. RESULTS Each iPSC reporter line formed well patterned kidney organoids. All reporter lines showed congruence of endogenous gene and protein expression, enabling isolation and characterization of kidney cell types of interest. We also demonstrated successful application of reporter lines for time-lapse imaging and mouse transplantation experiments. CONCLUSIONS We generated, validated, and applied a suite of fluorescence iPSC reporter lines for the study of morphogenesis within human kidney organoids. This fluorescent iPSC reporter toolbox enables the visualization and isolation of key populations in forming kidney organoids, facilitating a range of applications, including cellular isolation, time-lapse imaging, protocol optimization, and lineage-tracing approaches. These tools offer promise for enhancing our understanding of this model system and its correspondence with human kidney morphogenesis.
Collapse
Affiliation(s)
| | - Sean B Wilson
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Ker Sin Tan
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Joanne Y-C Soo
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Michelle Scurr
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - H Siebe Spijker
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lakshi T Starks
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Amber Neilson
- Department of Genetics, Genome Engineering and iPSC Center and
| | - Xiaoxia Cui
- Department of Genetics, Genome Engineering and iPSC Center and
| | - Sanjay Jain
- Department of Medicine, Kidney Translational Research Center, Washington University School of Medicine, St. Louis, Missouri; and
| | - Melissa Helen Little
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia; .,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics and.,Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Sara E Howden
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Faculty of Medicine, Dentistry and Health Sciences, Department of Paediatrics and
| |
Collapse
|
20
|
Puelles VG, Fleck D, Ortz L, Papadouri S, Strieder T, Böhner AM, van der Wolde JW, Vogt M, Saritas T, Kuppe C, Fuss A, Menzel S, Klinkhammer BM, Müller-Newen G, Heymann F, Decker L, Braun F, Kretz O, Huber TB, Susaki EA, Ueda HR, Boor P, Floege J, Kramann R, Kurts C, Bertram JF, Spehr M, Nikolic-Paterson DJ, Moeller MJ. Novel 3D analysis using optical tissue clearing documents the evolution of murine rapidly progressive glomerulonephritis. Kidney Int 2019; 96:505-516. [DOI: 10.1016/j.kint.2019.02.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 01/23/2019] [Accepted: 02/28/2019] [Indexed: 12/17/2022]
|
21
|
Application of Cell Lineage Tracing Combined with Immunofluorescence in the Study of Dentinogenesis. Methods Mol Biol 2019; 1922:39-48. [PMID: 30838563 DOI: 10.1007/978-1-4939-9012-2_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
Abstract
The cell lineage tracing system has been used predominantly in developmental biology studies. The Cre recombinase allows for the activation of the reporter in a specific cell line and all progeny. In this protocol, we will introduce how the cell lineage tracing technique can be performed in the investigation of dentinogenesis by using Gli1-CreERT2; R26RTomato compound mice. Moreover, we combined cell lineage tracing in conjunction with immunofluorescence-to further define cell fate by analyzing the expression of specific cell markers for odontoblasts. This combination not only broadens the application of cell lineage tracing but also simplifies the generation of compound mice. More importantly, the number, location, and differentiation status of parent cell progeny can be displayed simultaneously, providing more information than cell lineage tracing or immunofluorescence alone. In conclusion, the co-application of cell lineage tracing technique and immunofluorescence is a powerful tool for investigating cell biology in the field of dentinogenesis and tooth development.
Collapse
|
22
|
Howden SE, Vanslambrouck JM, Wilson SB, Tan KS, Little MH. Reporter-based fate mapping in human kidney organoids confirms nephron lineage relationships and reveals synchronous nephron formation. EMBO Rep 2019; 20:embr.201847483. [PMID: 30858339 DOI: 10.15252/embr.201847483] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 01/15/2023] Open
Abstract
Nephron formation continues throughout kidney morphogenesis in both mice and humans. Lineage tracing studies in mice identified a self-renewing Six2-expressing nephron progenitor population able to give rise to the full complement of nephrons throughout kidney morphogenesis. To investigate the origin of nephrons within human pluripotent stem cell-derived kidney organoids, we performed a similar fate-mapping analysis of the SIX2-expressing lineage in induced pluripotent stem cell (iPSC)-derived kidney organoids to explore the feasibility of investigating lineage relationships in differentiating iPSCs in vitro Using CRISPR/Cas9 gene-edited lineage reporter lines, we show that SIX2-expressing cells give rise to nephron epithelial cell types but not to presumptive ureteric epithelium. The use of an inducible (CreERT2) line revealed a declining capacity for SIX2+ cells to contribute to nephron formation over time, but retention of nephron-forming capacity if provided an exogenous WNT signal. Hence, while human iPSC-derived kidney tissue appears to maintain lineage relationships previously identified in developing mouse kidney, unlike the developing kidney in vivo, kidney organoids lack a nephron progenitor niche capable of both self-renewal and ongoing nephrogenesis.
Collapse
Affiliation(s)
- Sara E Howden
- Murdoch Children's Research Institute, Parkville, Vic., Australia .,Department of Paediatrics, The University of Melbourne, Melbourne, Vic., Australia
| | - Jessica M Vanslambrouck
- Murdoch Children's Research Institute, Parkville, Vic., Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Vic., Australia
| | - Sean B Wilson
- Murdoch Children's Research Institute, Parkville, Vic., Australia
| | - Ker Sin Tan
- Murdoch Children's Research Institute, Parkville, Vic., Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Vic., Australia .,Department of Paediatrics, The University of Melbourne, Melbourne, Vic., Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, Vic., Australia
| |
Collapse
|
23
|
Chen L, Higgins PJ, Zhang W. Development and Diseases of the Collecting Duct System. Results Probl Cell Differ 2017; 60:165-203. [PMID: 28409346 DOI: 10.1007/978-3-319-51436-9_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The collecting duct of the mammalian kidney is important for the regulation of extracellular volume, osmolarity, and pH. There are two major structurally and functionally distinct cell types: principal cells and intercalated cells. The former regulates Na+ and water homeostasis, while the latter participates in acid-base homeostasis. In vivo lineage tracing using Cre recombinase or its derivatives such as CreGFP and CreERT2 is a powerful new technique to identify stem/progenitor cells in their native environment and to decipher the origins of the tissue that they give rise to. Recent studies using this technique in mice have revealed multiple renal progenitor cell populations that differentiate into various nephron segments and collecting duct. In particular, emerging evidence suggests that like principal cells, most of intercalated cells originate from the progenitor cells expressing water channel Aquaporin 2. Mutations or malfunctions of the channels, pumps, and transporters expressed in the collecting duct system cause various human diseases. For example, gain-of-function mutations in ENaC cause Liddle's syndrome, while loss-of-function mutations in ENaC lead to Pseudohypoaldosteronism type 1. Mutations in either AE1 or V-ATPase B1 result in distal renal tubular acidosis. Patients with disrupted AQP2 or AVPR2 develop nephrogenic diabetes insipidus. A better understanding of the function and development of the collecting duct system may facilitate the discovery of new therapeutic strategies for treating kidney disease.
Collapse
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, NHLBI, Bethesda, MD, 20892-1603, USA
| | - Paul J Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
24
|
Jing Y, Hinton RJ, Chan KS, Feng JQ. Co-localization of Cell Lineage Markers and the Tomato Signal. J Vis Exp 2016. [PMID: 28060349 DOI: 10.3791/54982] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The cell lineage tracing system has been used predominantly in developmental biology studies. The use of Cre recombinase allows for the activation of the reporter in a specific cell line and all progeny. Here, we used the cell lineage tracing technique to demonstrate that chondrocytes directly transform into osteoblasts and osteocytes during long bone and mandibular condyle development using two kinds of Cre, Col10a1-Cre and Aggrecan-CreERT2 (Agg-CreERT2), crossed with Rosa26tdTomato. Both Col10 and aggrecan are well-recognized markers for chondrocytes. On this basis, we developed a new method-cell lineage tracing in conjunction with fluorescent immunohistochemistry-to define cell fate by analyzing the expression of specific cell markers. Runx2 (a marker for early-stage osteogenic cells) and Dentin matrix protein1 (DMP1; a marker for late-stage osteogenic cells) were used to identify chondrocyte-derived bone cells and their differentiation status. This combination not only broadens the application of cell lineage tracing, but also simplifies the generation of compound mice. More importantly, the number, location, and differentiation statuses of parent cell progeny are displayed simultaneously, providing more information than cell lineage tracing alone. In conclusion, the co-application of cell lineage tracing techniques and immunofluorescence is a powerful tool for investigating cell biology in vivo.
Collapse
Affiliation(s)
- Yan Jing
- Department of Biomedical Sciences, Texas A&M University College of Dentistry;
| | - Robert J Hinton
- Department of Biomedical Sciences, Texas A&M University College of Dentistry
| | - Kevin S Chan
- Department of Biomedical Sciences, Texas A&M University College of Dentistry
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry;
| |
Collapse
|
25
|
Eymael J, Smeets B. Origin and fate of the regenerating cells of the kidney. Eur J Pharmacol 2016; 790:62-73. [DOI: 10.1016/j.ejphar.2016.07.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/08/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022]
|
26
|
Le Clef N, Verhulst A, D’Haese PC, Vervaet BA. Unilateral Renal Ischemia-Reperfusion as a Robust Model for Acute to Chronic Kidney Injury in Mice. PLoS One 2016; 11:e0152153. [PMID: 27007127 PMCID: PMC4805266 DOI: 10.1371/journal.pone.0152153] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/09/2016] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury (AKI) is an underestimated, yet important risk factor for development of chronic kidney disease (CKD). Even after initial total recovery of renal function, some patients develop progressive and persistent deterioration of renal function and these patients are more likely to progress to end-stage renal disease (ESRD). Animal models are indispensable for unravelling the mechanisms underlying this progression towards CKD and ESRD and for the development of new therapeutic strategies in its prevention or treatment. Ischemia (i.e. hypoperfusion after surgery, bleeding, dehydration, shock, or sepsis) is a major aetiology in human AKI, yet unilateral ischemia-reperfusion is a rarely used animal model for research on CKD and fibrosis. Here, we demonstrate in C57Bl/6J mice, by both histology and gene expression, that unilateral ischemia-reperfusion without contralateral nephrectomy is a very robust model to study the progression from acute renal injury to long-term tubulo-interstitial fibrosis, i.e. the histopathological hallmark of CKD. Furthermore, we report that the extent of renal fibrosis, in terms of Col I, TGFβ, CCN2 and CCN3 expression and collagen I immunostaining, increases with increasing body temperature during ischemia and ischemia-time. Thus, varying these two main determinants of ischemic injury allows tuning the extent of the long-term fibrotic outcome in this model. Finally, in order to cover the whole practical finesse of ischemia-reperfusion and allow model and data transfer, we provide a referenced overview on crucial technical issues (incl. anaesthesia, analgesia, and pre- and post-operative care) with the specific aim of putting starters in the right direction of implementing ischemia in their research and stimulate them, as well as the community, to have a critical view on ischemic literature data.
Collapse
Affiliation(s)
- Nathalie Le Clef
- Departement Biomedical Sciences, Laboratory of Pathophysiology, University of Antwerp, Wilrijk (Antwerp), Belgium
| | - Anja Verhulst
- Departement Biomedical Sciences, Laboratory of Pathophysiology, University of Antwerp, Wilrijk (Antwerp), Belgium
| | - Patrick C. D’Haese
- Departement Biomedical Sciences, Laboratory of Pathophysiology, University of Antwerp, Wilrijk (Antwerp), Belgium
- * E-mail:
| | - Benjamin A. Vervaet
- Departement Biomedical Sciences, Laboratory of Pathophysiology, University of Antwerp, Wilrijk (Antwerp), Belgium
| |
Collapse
|
27
|
Liu Y, Dong Z, Liu H, Zhu J, Liu F, Chen G. Transition of mesothelial cell to fibroblast in peritoneal dialysis: EMT, stem cell or bystander? Perit Dial Int 2015; 35:14-25. [PMID: 25700459 DOI: 10.3747/pdi.2014.00188] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Long-term peritoneal dialysis (PD) can lead to fibrotic changes in the peritoneum, characterized by loss of mesothelial cells (MCs) and thickening of the submesothelial area with an accumulation of collagen and myofibroblasts. The origin of myofibroblasts is a central question in peritoneal fibrosis that remains unanswered at present. Numerous clinical and experimental studies have suggested that MCs, through epithelial-mesenchymal transition (EMT), contribute to the pool of peritoneal myofibroblasts. However, recent work has placed significant doubts on the paradigm of EMT in organ fibrogenesis (in the kidney particularly), highlighting the need to reconsider the role of EMT in the generation of myofibroblasts in peritoneal fibrosis. In particular, selective cell isolation and lineage-tracing experiments have suggested the existence of progenitor cells in the peritoneum, which are able to switch to fibroblast-like cells when stimulated by the local environment. These findings highlight the plastic nature of MCs and its contribution to peritoneal fibrogenesis. In this review, we summarize the key findings and caveats of EMT in organ fibrogenesis, with a focus on PD-related peritoneal fibrosis, and discuss the potential of peritoneal MCs as a source of myofibroblasts.
Collapse
Affiliation(s)
- Yu Liu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Zheng Dong
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Hong Liu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Jiefu Zhu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Fuyou Liu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| | - Guochun Chen
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, PR China; Department of Cellular Biology and Anatomy, Georgia Regents University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| |
Collapse
|
28
|
Transgenic Strategies to Study Podocyte Loss and Regeneration. Stem Cells Int 2015; 2015:678347. [PMID: 26089920 PMCID: PMC4451768 DOI: 10.1155/2015/678347] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 01/22/2023] Open
Abstract
Podocyte death and regeneration are major topics in kidney research but remain controversial. Data obtained in humans demonstrate the existence of cells sited along Bowman's capsule that behave as podocyte progenitors in vitro and in in vivo mouse models of podocyte injury xenotrasplanted with this human-derived population. However, this podocyte reservoir still remains elusive in murine models, where it could be more easily studied. Transgenic models can be a powerful tool to identify this population and to better understand its dynamics and hierarchies in both physiological and pathological conditions. Indeed, exploiting transgenic approaches allows detecting, at the single cell level, movements, cell death, and replacement. Moreover, through lineage tracing it is now possible to identify specific population increase and to point out clonal expansions during or after the regenerative processes. However, applying transgenic strategies to study glomerular regeneration requires the search of markers to unequivocally identify this progenitor population. Achieving this aim would lead to a deep comprehension of the biological processes that underlie glomerular regeneration and clarify how different cell pools interface during this phase. Here we discuss strategies that have been used and new approaches in transgenic models finalized to study podocyte loss and subsequent replacement.
Collapse
|
29
|
Abstract
Lineage tracing is a powerful tool to track cells in vivo and provides enhanced spatial, temporal, and kinetic resolution of the mechanisms that underlie tissue renewal and repair. The data obtained from novel mouse models engineered for lineage tracing has started to transform our understanding of the changes in cell fate that underlie renal pathophysiology, the role of stem and/or progenitor cells in kidney development, and the mechanisms of kidney regeneration. The complexity of the genetic systems that are engineered for lineage tracing requires careful analysis and interpretation. In this Review we emphasize that close attention in lineage tracing studies must be paid to the specificity of the promoter, the use of drug-controlled activation of Cre recombinase as a genetic switch, and the type of reporter that should be engineered into lineage tracing genetic constructs. We evaluate the optimal experimental conditions required to achieve the pre-specified aims of the study and discuss the novel genetic techniques that are becoming available to study putative renal progenitor cells and the mechanisms of kidney regeneration.
Collapse
|
30
|
Humphreys BD. Cutting to the chase: taking the pulse of label-retaining cells in kidney. Am J Physiol Renal Physiol 2014; 308:F29-30. [PMID: 25298529 DOI: 10.1152/ajprenal.00538.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Benjamin D Humphreys
- Brigham and Women's Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; and Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|