1
|
Ilatovskaya DV, Behr A, Staruschenko A, Hall G, Palygin O. Mechanistic Insights Into Redox Damage of the Podocyte in Hypertension. Hypertension 2025; 82:14-25. [PMID: 39534957 PMCID: PMC11655258 DOI: 10.1161/hypertensionaha.124.22068] [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] [Indexed: 11/16/2024]
Abstract
Podocytes are specialized cells within the glomerular filtration barrier, which are crucial for maintaining glomerular structural integrity and convective ultrafiltration. Podocytes exhibit a unique arborized morphology with foot processes interfacing by slit diaphragms, ladder-like, multimolecular sieves, which provide size and charge selectivity for ultrafiltration and transmembrane signaling. Podocyte dysfunction, resulting from oxidative stress, dysregulated prosurvival signaling, or structural damage, can drive the development of proteinuria and glomerulosclerosis in hypertensive nephropathy. Functionally, podocyte injury leads to actin cytoskeleton rearrangements, foot process effacement, dysregulated slit diaphragm protein expression, and impaired ultrafiltration. Notably, the renin-angiotensin system plays a pivotal role in podocyte function, with beneficial AT2R (angiotensin receptor 2)-mediated nitric oxide (NO) signaling to counteract AT1R (angiotensin receptor 1)-driven calcium (Ca2+) influx and oxidative stress. Disruption of this balance contributes significantly to podocyte dysfunction and drives albuminuria, a marker of kidney damage and overall disease progression. Oxidative stress can also lead to sustained ion channel-mediated Ca2+ influx and precipitate cytoskeletal disorganization. The complex interplay between GPCR (G-protein coupled receptor) signaling, ion channel activation, and redox injury pathways underscores the need for additional research aimed at identifying targeted therapies to protect podocytes and preserve glomerular function. Earlier detection of albuminuria and podocyte injury through routine noninvasive diagnostics will also be critical in populations at the highest risk for the development of hypertensive kidney disease. In this review, we highlight the established mechanisms of oxidative stress-mediated podocyte damage in proteinuric kidney diseases, with an emphasis on a hypertensive renal injury. We will also consider emerging therapies that have the potential to selectively protect podocytes from redox-related injury.
Collapse
Affiliation(s)
- Daria V. Ilatovskaya
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA
| | - Amanda Behr
- Department of Medical Illustration, College of Allied Health Sciences, Augusta University, Augusta, GA
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL
- James A. Haley Veterans’ Hospital, Tampa, FL
| | - Gentzon Hall
- Division of Nephrology, Department of Internal Medicine, Duke University School of Medicine, Durham, NC
- Department of Medicine, Division of Nephrology, Duke Molecular Physiology Institute, Duke University, Durham, NC
| | - Oleg Palygin
- Department of Medicine, Division of Nephrology, Medical University of South Carolina, Charleston, SC
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
2
|
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: 1] [Impact Index Per Article: 0.5] [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
|
3
|
Zhao Q, Shao T, Zhu Y, Zong G, Zhang J, Tang S, Lin Y, Ma H, Jiang Z, Xu Y, Wu X, Zhang T. An MRTF-A-ZEB1-IRF9 axis contributes to fibroblast-myofibroblast transition and renal fibrosis. Exp Mol Med 2023:10.1038/s12276-023-00990-6. [PMID: 37121967 DOI: 10.1038/s12276-023-00990-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/23/2023] [Accepted: 02/14/2023] [Indexed: 05/02/2023] Open
Abstract
Myofibroblasts, characterized by the expression of the matricellular protein periostin (Postn), mediate the profibrogenic response during tissue repair and remodeling. Previous studies have demonstrated that systemic deficiency in myocardin-related transcription factor A (MRTF-A) attenuates renal fibrosis in mice. In the present study, we investigated the myofibroblast-specific role of MRTF-A in renal fibrosis and the underlying mechanism. We report that myofibroblast-specific deletion of MRTF-A, achieved through crossbreeding Mrtfa-flox mice with Postn-CreERT2 mice, led to amelioration of renal fibrosis. RNA-seq identified zinc finger E-Box binding homeobox 1 (Zeb1) as a downstream target of MRTF-A in renal fibroblasts. MRTF-A interacts with TEA domain transcription factor 1 (TEAD1) to bind to the Zeb1 promoter and activate Zeb1 transcription. Zeb1 knockdown retarded the fibroblast-myofibroblast transition (FMyT) in vitro and dampened renal fibrosis in mice. Transcriptomic assays showed that Zeb1 might contribute to FMyT by repressing the transcription of interferon regulatory factor 9 (IRF9). IRF9 knockdown overcame the effect of Zeb1 depletion and promoted FMyT, whereas IRF9 overexpression antagonized TGF-β-induced FMyT. In conclusion, our data unveil a novel MRTF-A-Zeb1-IRF9 axis that can potentially contribute to fibroblast-myofibroblast transition and renal fibrosis. Screening for small-molecule compounds that target this axis may yield therapeutic options for the mollification of renal fibrosis.
Collapse
Affiliation(s)
- Qianwen Zhao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Tinghui Shao
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yuwen Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Gengjie Zong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Junjie Zhang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Shifan Tang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Yanshan Lin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Hongzhen Ma
- Department of Geriatric Nephrology, First Affiliated Hospital to Nanjing Medical University, Nanjing, China
| | - Zhifan Jiang
- Department of Geriatric Nephrology, First Affiliated Hospital to Nanjing Medical University, Nanjing, China
| | - Yong Xu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Xiaoyan Wu
- School of Sports and Health, Nanjing Sport Institute, Nanjing, China.
| | - Tao Zhang
- Department of Geriatric Nephrology, First Affiliated Hospital to Nanjing Medical University, Nanjing, China.
| |
Collapse
|
4
|
An YA, Xiong W, Chen S, Bu D, Rutkowski JM, Berger JP, Kusminski CM, Zhang N, An Z, Scherer PE. Endotrophin neutralization through targeted antibody treatment protects from renal fibrosis in a podocyte ablation model. Mol Metab 2023; 69:101680. [PMID: 36696925 PMCID: PMC9918787 DOI: 10.1016/j.molmet.2023.101680] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/26/2022] [Accepted: 01/18/2023] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE Renal fibrosis is a hallmark for chronic kidney disease (CKD), and often leads to end stage renal disease (ESRD). However, limited interventions are available clinically to ameliorate or reverse renal fibrosis. METHODS Herein, we evaluated whether blockade of endotrophin through neutralizing antibodies protects from renal fibrosis in the podocyte insult model (the "POD-ATTAC" mouse). We determined the therapeutic effects of endotrophin targeted antibody through assessing renal function, renal inflammation and fibrosis at histological and transcriptional levels, and podocyte regeneration. RESULTS We demonstrated that neutralizing endotrophin antibody treatment significantly ameliorates renal fibrosis at the transcriptional, morphological, and functional levels. In the antibody treatment group, expression of pro-inflammatory and pro-fibrotic genes was significantly reduced, normal renal structures were restored, collagen deposition was decreased, and proteinuria and renal function were improved. We further performed a lineage tracing study confirming that podocytes regenerate as de novo podocytes upon injury and loss, and blockade of endotrophin efficiently enhances podocyte-specific marker expressions. CONCLUSION Combined, we provide pre-clinical evidence supporting neutralizing endotrophin as a promising therapy for intervening with renal fibrosis in CKD, and potentially in other chronic fibro-inflammatory diseases.
Collapse
Affiliation(s)
- Yu A An
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wei Xiong
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dawei Bu
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph M Rutkowski
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Division of Lymphatic Biology, Department of Medical Physiology, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Joel P Berger
- JP Berger Consulting, 580 Washington Street, #15C, Boston, MA, USA
| | - Christine M Kusminski
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Cell Biology, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
5
|
Xia Q, Tang Y, Li W, Liang T, Zhou Y, Liu J, Liu F. Surface-Engineered Monocyte Immunotherapy Combined Graphene Quantum Dots Effective Against Solid Tumor Targets. Int J Nanomedicine 2023; 18:2127-2140. [PMID: 37122502 PMCID: PMC10145394 DOI: 10.2147/ijn.s404486] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/16/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction The immunosuppressive tumor microenvironment (TME) of solid tumors inhibits most drug delivery system-based nanomaterials from achieving deep penetration in tumor tissue and interferes with T cell activity in terms of differentiation and exhaustion, which is becoming a critical therapy hurdle for solid tumors. Therefore, developing a therapeutic strategy with abilities of rapid establishment of tumor-targeted cells, elimination of immune obstacles, and enhanced active immunization is very important, while is still a big challenge. Methods A new strategy was explored to enhance immune therapy via the conjugation of microRNA155 (miR) to the surface of therapeutic monocyte with graphene quantum dots (GQDs). Results TME was reversed using surface-engineered monocyte immunotherapy via reprogramming pro-tumoral M2 TAMs into antitumor M1, and thus tumor elimination was dramatically enhanced. Conclusion Such a surface-engineered monocyte immunotherapy has been demonstrated to be well tolerated to intravenous administration and bio-compatible, showing the potential to be extended for the solid tumor treatment.
Collapse
Affiliation(s)
- Qing Xia
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Yue Tang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Wang Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Tingting Liang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China
- Correspondence: Feila Liu, School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, People’s Republic of China, Tel +86-15123002638, Fax +86 2362563190, Email
| |
Collapse
|
6
|
Gerges D, Hevesi Z, Schmidt SH, Kapps S, Pajenda S, Geist B, Schmidt A, Wagner L, Winnicki W. Tubular epithelial progenitors are excreted in urine during recovery from severe acute kidney injury and are able to expand and differentiate in vitro. PeerJ 2022; 10:e14110. [PMID: 36285332 PMCID: PMC9588302 DOI: 10.7717/peerj.14110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/02/2022] [Indexed: 01/21/2023] Open
Abstract
Background Acute kidney injury (AKI) is a serious condition associated with chronic kidney disease, dialysis requirement and a high risk of death. However, there are specialized repair mechanisms for the nephron, and migrated committed progenitor cells are the key players. Previous work has described a positive association between renal recovery and the excretion of tubular progenitor cells in the urine of kidney transplant recipients. The aim of this work was to describe such structures in non-transplanted AKI patients and to focus on their differentiation. Methods Morning urine was obtained from four patients with AKI stage 3 and need for RRT on a consecutive basis. Urine sediment gene expression was performed to assess which part of the tubular or glomerular segment was affected by injury, along with measurement of neprilysin. Urine output and sediment morphology were monitored, viable hyperplastic tubular epithelial clusters were isolated and characterized by antibody or cultured in vitro. These cells were monitored by phase contrast microscopy, gene, and protein expression over 9 days by qPCR and confocal immunofluorescence. Furthermore, UMOD secretion into the supernatant was quantitatively measured. Results Urinary neprilysin decreased rapidly with increasing urinary volume in ischemic, toxic, nephritic, and infection-associated AKI, whereas the decrease in sCr required at least 2 weeks. While urine output increased, dead cells were present in the sediment along with debris followed by hyperplastic agglomerates. Monitoring of urine sediment for tubular cell-specific gene transcript levels NPHS2 (podocyte), AQP1 and AQP6 (proximal tubule), and SLC12A1 (distal tubule) by qPCR revealed different components depending on the cause of AKI. Confocal immunofluorescence staining confirmed the presence of intact nephron-specific epithelial cells, some of which appeared in clusters expressing AQP1 and PAX8 and were 53% positive for the stem cell marker PROM1. Isolated tubule epithelial progenitor cells were grown in vitro, expanded, and reached confluence within 5-7 days, while the expression of AQP1 and UMOD increased, whereas PROM1 and Ki67 decreased. This was accompanied by a change in cell morphology from a disproportionately high nuclear/cytoplasmic ratio at day 2-7 with mitotic figures. In contrast, an apoptotic morphology of approximately 30% was found at day 9 with the appearance of multinucleated cells that were associable with different regions of the nephron tubule by marker proteins. At the same time, UMOD was detected in the culture supernatant. Conclusion During renal recovery, a high replicatory potential of tubular epithelial progenitor cells is found in urine. In vitro expansion and gene expression show differentiation into tubular cells with marker proteins specific for different nephron regions.
Collapse
Affiliation(s)
- Daniela Gerges
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Zsofia Hevesi
- Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Sophie H. Schmidt
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Sebastian Kapps
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Sahra Pajenda
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Barbara Geist
- Department of Biochemical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University Vienna, Vienna, Austria
| | - Alice Schmidt
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Ludwig Wagner
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| | - Wolfgang Winnicki
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna, Vienna, Austria
| |
Collapse
|
7
|
Melica ME, Antonelli G, Semeraro R, Angelotti ML, Lugli G, Landini S, Ravaglia F, La Regina G, Conte C, De Chiara L, Peired AJ, Mazzinghi B, Donati M, Molli A, Steiger S, Magi A, Bartalucci N, Raglianti V, Guzzi F, Maggi L, Annunziato F, Burger A, Lazzeri E, Anders HJ, Lasagni L, Romagnani P. Differentiation of crescent-forming kidney progenitor cells into podocytes attenuates severe glomerulonephritis in mice. Sci Transl Med 2022; 14:eabg3277. [PMID: 35947676 PMCID: PMC7614034 DOI: 10.1126/scitranslmed.abg3277] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Crescentic glomerulonephritis is characterized by vascular necrosis and parietal epithelial cell hyperplasia in the space surrounding the glomerulus, resulting in the formation of crescents. Little is known about the molecular mechanisms driving this process. Inducing crescentic glomerulonephritis in two Pax2Cre reporter mouse models revealed that crescents derive from clonal expansion of single immature parietal epithelial cells. Preemptive and delayed histone deacetylase inhibition with panobinostat, a drug used to treat hematopoietic stem cell disorders, attenuated crescentic glomerulonephritis with recovery of kidney function in the two mouse models. Three-dimensional confocal microscopy and stimulated emission depletion superresolution imaging of mouse glomeruli showed that, in addition to exerting an anti-inflammatory and immunosuppressive effect, panobinostat induced differentiation of an immature hyperplastic parietal epithelial cell subset into podocytes, thereby restoring the glomerular filtration barrier. Single-cell RNA sequencing of human renal progenitor cells in vitro identified an immature stratifin-positive cell subset and revealed that expansion of this stratifin-expressing progenitor cell subset was associated with a poor outcome in human crescentic glomerulonephritis. Treatment of human parietal epithelial cells in vitro with panobinostat attenuated stratifin expression in renal progenitor cells, reduced their proliferation, and promoted their differentiation into podocytes. These results offer mechanistic insights into the formation of glomerular crescents and demonstrate that selective targeting of renal progenitor cells can attenuate crescent formation and the deterioration of kidney function in crescentic glomerulonephritis in mice.
Collapse
Affiliation(s)
- Maria Elena Melica
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Giulia Antonelli
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Roberto Semeraro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Lucia Angelotti
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gianmarco Lugli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Samuela Landini
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Fiammetta Ravaglia
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Gilda La Regina
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Carolina Conte
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Letizia De Chiara
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Anna Julie Peired
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Benedetta Mazzinghi
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Marta Donati
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Alice Molli
- Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Stefanie Steiger
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Alberto Magi
- Department of Information Engineering, University of Florence, Florence, Italy
| | - Niccolò Bartalucci
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, AOUC, University of Florence, Florence 50139, Italy
| | - Valentina Raglianti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Francesco Guzzi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elena Lazzeri
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich 80336, Germany
| | - Laura Lasagni
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Corresponding authors. and
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), University of Florence, Florence 50139, Italy,Department of Experimental and Clinical Biomedical Sciences “Mario Serio,” University of Florence, Florence 50139, Italy,Nephrology and Dialysis Unit, Meyer Children’s Hospital, Florence 50139, Italy,Corresponding authors. and
| |
Collapse
|
8
|
Jackson AR, Narla ST, Bates CM, Becknell B. Urothelial progenitors in development and repair. Pediatr Nephrol 2022; 37:1721-1731. [PMID: 34471946 PMCID: PMC8942604 DOI: 10.1007/s00467-021-05239-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 10/20/2022]
Abstract
Urothelium is a specialized multilayer epithelium that lines the urinary tract from the proximal urethra to the kidney. In addition to proliferation and differentiation during development, urothelial injury postnatally triggers a robust regenerative capacity to restore the protective barrier between the urine and tissue. Mounting evidence supports the existence of dedicated progenitor cell populations that give rise to urothelium during development and in response to injury. Understanding the cellular and molecular basis for urothelial patterning and repair will inform tissue regeneration therapies designed to ameliorate a number of structural and functional defects of the urinary tract. Here, we review the current understanding of urothelial progenitors and the signaling pathways that govern urothelial development and repair. While most published studies have focused on bladder urothelium, we also discuss literature on upper tract urothelial progenitors. Furthermore, we discuss evidence supporting existence of context-specific progenitors. This knowledge is fundamental to the development of strategies to regenerate or engineer damaged or diseased urothelium.
Collapse
Affiliation(s)
- Ashley R Jackson
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute At Nationwide Children's Hospital, 700 Children's Drive, W308, Columbus, 43205, OH, USA
- Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA
| | - Sridhar T Narla
- Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, Rangos Research Building, 4401 Penn Avenue, Pittsburgh, 15224, PA, USA
| | - Carlton M Bates
- Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, Rangos Research Building, 4401 Penn Avenue, Pittsburgh, 15224, PA, USA.
- Division of Nephrology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| | - Brian Becknell
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute At Nationwide Children's Hospital, 700 Children's Drive, W308, Columbus, 43205, OH, USA.
- Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, USA.
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA.
| |
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
|
Little MH, Humphreys BD. Regrow or Repair: An Update on Potential Regenerative Therapies for the Kidney. J Am Soc Nephrol 2022; 33:15-32. [PMID: 34789545 PMCID: PMC8763179 DOI: 10.1681/asn.2021081073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Fifteen years ago, this journal published a review outlining future options for regenerating the kidney. At that time, stem cell populations were being identified in multiple tissues, the concept of stem cell recruitment to a site of injury was of great interest, and the possibility of postnatal renal stem cells was growing in momentum. Since that time, we have seen the advent of human induced pluripotent stem cells, substantial advances in our capacity to both sequence and edit the genome, global and spatial transcriptional analysis down to the single-cell level, and a pandemic that has challenged our delivery of health care to all. This article will look back over this period of time to see how our view of kidney development, disease, repair, and regeneration has changed and envision a future for kidney regeneration and repair over the next 15 years.
Collapse
Affiliation(s)
- Melissa H. Little
- Murdoch Children’s Research Institute, Parkville, Melbourne, Victoria, Australia,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Melbourne, Victoria, Australia,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Melbourne, Victoria, Australia
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis School of Medicine, Missouri,Department of Developmental Biology, Washington University in St. Louis School of Medicine, Missouri
| |
Collapse
|
11
|
Tubular Cell Cycle Response upon AKI: Revising Old and New Paradigms to Identify Novel Targets for CKD Prevention. Int J Mol Sci 2021; 22:ijms222011093. [PMID: 34681750 PMCID: PMC8537394 DOI: 10.3390/ijms222011093] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 02/07/2023] Open
Abstract
Acute kidney injury (AKI) is characterized by a rapid deterioration of kidney function, representing a global healthcare concern. In addition, AKI survivors frequently develop chronic kidney disease (CKD), contributing to a substantial proportion of disease burden globally. Yet, over the past 30 years, the burden of CKD has not declined to the same extent as many other important non-communicable diseases, implying a substantial deficit in the understanding of the disease progression. The assumption that the kidney response to AKI is based on a high proliferative potential of proximal tubular cells (PTC) caused a critical confounding factor, which has led to a limited development of strategies to prevent AKI and halt progression toward CKD. In this review, we discuss the latest findings on multiple mechanisms of response related to cell cycle behavior of PTC upon AKI, with a specific focus on their biological relevance. Collectively, we aim to (1) provide a new perspective on interpreting cell cycle progression of PTC in response to damage and (2) discuss how this knowledge can be used to choose the right therapeutic window of treatment for preserving kidney function while avoiding CKD progression.
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
|
Harari-Steinberg O, Omer D, Gnatek Y, Pleniceanu O, Goldberg S, Cohen-Zontag O, Pri-Chen S, Kanter I, Ben Haim N, Becker E, Ankawa R, Fuchs Y, Kalisky T, Dotan Z, Dekel B. Ex Vivo Expanded 3D Human Kidney Spheres Engraft Long Term and Repair Chronic Renal Injury in Mice. Cell Rep 2021; 30:852-869.e4. [PMID: 31968258 DOI: 10.1016/j.celrep.2019.12.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 10/04/2019] [Accepted: 12/12/2019] [Indexed: 12/26/2022] Open
Abstract
End-stage renal disease is a worldwide epidemic requiring renal replacement therapy. Harvesting tissue from failing kidneys and autotransplantation of tissue progenitors could theoretically delay the need for dialysis. Here we use healthy and end-stage human adult kidneys to robustly expand proliferative kidney epithelial cells and establish 3D kidney epithelial cultures termed "nephrospheres." Formation of nephrospheres reestablishes renal identity and function in primary cultures. Transplantation into NOD/SCID mice shows that nephrospheres restore self-organogenetic properties lost in monolayer cultures, allowing long-term engraftment as tubular structures, potentially adding nephron segments and demonstrating self-organization as critical to survival. Furthermore, long-term tubular engraftment of nephrospheres is functionally beneficial in murine models of chronic kidney disease. Remarkably, nephrospheres inhibit pro-fibrotic collagen production in cultured fibroblasts via paracrine modulation, while transplanted nephrospheres induce transcriptional signatures of proliferation and release from quiescence, suggesting re-activation of endogenous repair. These data support the use of human nephrospheres for renal cell therapy.
Collapse
Affiliation(s)
- Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sanja Goldberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Osnat Cohen-Zontag
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sara Pri-Chen
- The Maurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center, Ramat-Gan, Israel
| | - Itamar Kanter
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Nissim Ben Haim
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Eli Becker
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Roi Ankawa
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Tomer Kalisky
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan, Israel
| | - Zohar Dotan
- Department of Urology, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Sara Children's Hospital, Sheba Medical Center, Tel Hashomer, Ramat-Gan, Israel; Pediatric Research Center for Genetics, Development and Environment, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Division of Pediatric Nephrology, Safra Children's Hospital, Sheba Medical Center, Ramat-Gan, Israel.
| |
Collapse
|
14
|
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
|
15
|
Lee SA, Li KN, Tumbar T. Stem cell-intrinsic mechanisms regulating adult hair follicle homeostasis. Exp Dermatol 2020; 30:430-447. [PMID: 33278851 DOI: 10.1111/exd.14251] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Adult hair follicle stem cells (HFSCs) undergo dynamic and periodic molecular changes in their cellular states throughout the hair homeostatic cycle. These states are tightly regulated by cell-intrinsic mechanisms and by extrinsic signals from the microenvironment. HFSCs are essential not only for fuelling hair growth, but also for skin wound healing. Increasing evidence suggests an important role of HFSCs in organizing multiple skin components around the hair follicle, thus functioning as an organizing centre during adult skin homeostasis. Here, we focus on recent findings on cell-intrinsic mechanisms of HFSC homeostasis, which include transcription factors, histone modifications, DNA regulatory elements, non-coding RNAs, cell metabolism, cell polarity and post-transcriptional mRNA processing. Several transcription factors are now known to participate in well-known signalling pathways that control hair follicle homeostasis, as well as in super-enhancer activities to modulate HFSC and progenitor lineage progression. Interestingly, HFSCs have been shown to secrete molecules that are important in guiding the organization of several skin components around the hair follicle, including nerves, arrector pili muscle and vasculature. Finally, we discuss recent technological advances in the field such as single-cell RNA sequencing and live imaging, which revealed HFSC and progenitor heterogeneity and brought new light to understanding crosstalking between HFSCs and the microenvironment. The field is well on its way to generate a comprehensive map of molecular interactions that should serve as a solid theoretical platform for application in hair and skin disease and ageing.
Collapse
Affiliation(s)
- Seon A Lee
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Kefei Nina Li
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Tudorita Tumbar
- Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
16
|
Zhang K, Chen S, Sun H, Wang L, Li H, Zhao J, Zhang C, Li N, Guo Z, Han Z, Han ZC, Zheng G, Chen X, Li Z. In vivo two-photon microscopy reveals the contribution of Sox9 + cell to kidney regeneration in a mouse model with extracellular vesicle treatment. J Biol Chem 2020; 295:12203-12213. [PMID: 32641493 PMCID: PMC7443503 DOI: 10.1074/jbc.ra120.012732] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 07/02/2020] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have been shown to stimulate regeneration in the treatment of kidney injury. Renal regeneration is also thought to be stimulated by the activation of Sox9+ cells. However, whether and how the activation mechanisms underlying EV treatment and Sox9+ cell-dependent regeneration intersect is unclear. We reasoned that a high-resolution imaging platform in living animals could help to untangle this system. To test this idea, we first applied EVs derived from human placenta-derived MSCs (hP-MSCs) to a Sox9-CreERT2; R26mTmG transgenic mouse model of acute kidney injury (AKI). Then, we developed an abdominal imaging window in the mouse and tracked the Sox9+ cells in the inducible Sox9-Cre transgenic mice via in vivo lineage tracing with two-photon intravital microscopy. Our results demonstrated that EVs can travel to the injured kidneys post intravenous injection as visualized by Gaussia luciferase imaging and markedly increase the activation of Sox9+ cells. Moreover, the two-photon living imaging of lineage-labeled Sox9+ cells showed that the EVs promoted the expansion of Sox9+ cells in kidneys post AKI. Histological staining results confirmed that the descendants of Sox9+ cells contributed to nephric tubule regeneration which significantly ameliorated the renal function after AKI. In summary, intravital lineage tracing with two-photon microscopy through an embedded abdominal imaging window provides a practical strategy to investigate the beneficial functions and to clarify the mechanisms of regenerative therapies in AKI.
Collapse
Affiliation(s)
- Kaiyue Zhang
- Nankai University School of Medicine, Tianjin, China; The Key Laboratory of Bioactive Materials, Ministry of Education, the College of Life Sciences, Nankai University, Tianjin, China
| | - Shang Chen
- Nankai University School of Medicine, Tianjin, China; The Key Laboratory of Bioactive Materials, Ministry of Education, the College of Life Sciences, Nankai University, Tianjin, China
| | - Huimin Sun
- Nankai University School of Medicine, Tianjin, China
| | - Lina Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huifang Li
- Nankai University School of Medicine, Tianjin, China
| | - Jinglei Zhao
- Nankai University School of Medicine, Tianjin, China
| | - Chuyue Zhang
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Nana Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Zhibo Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, Jiangxi, China; Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China
| | - Zhong-Chao Han
- Jiangxi Engineering Research Center for Stem Cell, Shangrao, Jiangxi, China; Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China; Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health & Biotech Co., Beijing, China
| | - Guoguang Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiangmei Chen
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin, China; The Key Laboratory of Bioactive Materials, Ministry of Education, the College of Life Sciences, Nankai University, Tianjin, China; State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China; Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
17
|
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: 17] [Impact Index Per Article: 3.4] [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
|
18
|
Anguiano L, Kain R, Anders HJ. The glomerular crescent: triggers, evolution, resolution, and implications for therapy. Curr Opin Nephrol Hypertens 2020; 29:302-309. [PMID: 32132388 PMCID: PMC7170443 DOI: 10.1097/mnh.0000000000000596] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Crescents are classical histopathological lesions found in severe forms of rapidly progressive glomerulonephritis, also referred to as crescentic glomerulonephritis (CGN). Crescent formation is a consequence of diverse upstream pathomechanisms and unraveling these mechanisms is of great interest for improving the management of patients affected by CGN. Thus, in this review, we provide an update on the latest insight into the understanding on how crescents develop and how they resolve. RECENT FINDINGS Cellular crescents develop from activated parietal epithelial cells (PECs) residing along Bowman's capsule and their formation has as a consequence the decline in glomerular filtration rate (GFR). Cellular crescents can be reversible, but when multilevel growth of PECs associate with an epithelial--mesenchymal transition-like change in cell phenotype, fibrous crescents form, and crescents become irreversible also in terms of GFR recovery. Different molecular pathways trigger the activation of PECs and are a prime therapeutics target in CGN. First, crescent formation requires also vascular injury causing ruptures in the glomerular basement membrane that trigger plasmatic coagulation within Bowman's space. This vascular necrosis can be triggered by different upstream mechanisms, such as small vessel vasculitides, immune complex glomerulonephritis, anti-GBM disease, and C3 glomerulonephritis, that all share complement activation but involve diverse upstream immune mechanisms outside the kidney accessible for therapeutic intervention. SUMMARY Knowing the upstream mechanisms that triggered crescent formation provides a tool for the development of therapeutic interventions for CGN.
Collapse
Affiliation(s)
- Lidia Anguiano
- Renal Division, Department of Medicine IV, University Hospital of the Ludwig Maximilian University, Munich, Germany
| | - Renate Kain
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Hans-Joachim Anders
- Renal Division, Department of Medicine IV, University Hospital of the Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
19
|
Andrianova NV, Buyan MI, Zorova LD, Pevzner IB, Popkov VA, Babenko VA, Silachev DN, Plotnikov EY, Zorov DB. Kidney Cells Regeneration: Dedifferentiation of Tubular Epithelium, Resident Stem Cells and Possible Niches for Renal Progenitors. Int J Mol Sci 2019; 20:ijms20246326. [PMID: 31847447 PMCID: PMC6941132 DOI: 10.3390/ijms20246326] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/10/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022] Open
Abstract
A kidney is an organ with relatively low basal cellular regenerative potential. However, renal cells have a pronounced ability to proliferate after injury, which undermines that the kidney cells are able to regenerate under induced conditions. The majority of studies explain yielded regeneration either by the dedifferentiation of the mature tubular epithelium or by the presence of a resident pool of progenitor cells in the kidney tissue. Whether cells responsible for the regeneration of the kidney initially have progenitor properties or if they obtain a “progenitor phenotype” during dedifferentiation after an injury, still stays the open question. The major stumbling block in resolving the issue is the lack of specific methods for distinguishing between dedifferentiated cells and resident progenitor cells. Transgenic animals, single-cell transcriptomics, and other recent approaches could be powerful tools to solve this problem. This review examines the main mechanisms of kidney regeneration: dedifferentiation of epithelial cells and activation of progenitor cells with special attention to potential niches of kidney progenitor cells. We attempted to give a detailed description of the most controversial topics in this field and ways to resolve these issues.
Collapse
Affiliation(s)
- Nadezda V. Andrianova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Marina I. Buyan
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Ljubava D. Zorova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Irina B. Pevzner
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Vasily A. Popkov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Valentina A. Babenko
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Denis N. Silachev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Egor Y. Plotnikov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
- Sechenov First Moscow State Medical University, Institute of Molecular Medicine, 119991 Moscow, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P.)
| | - Dmitry B. Zorov
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
- Correspondence: (E.Y.P.); (D.B.Z.); Tel.: +7-495-939-5944 (E.Y.P.)
| |
Collapse
|
20
|
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
|
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
|
Ye Y, Song H, Zhang J, Shi S. Understanding the Biology and Pathogenesis of the Kidney by Single-Cell Transcriptomic Analysis. KIDNEY DISEASES 2018; 4:214-225. [PMID: 30574498 DOI: 10.1159/000492470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/26/2018] [Indexed: 12/20/2022]
Abstract
Background Single-cell RNA-seq (scRNA-seq) has recently emerged as a revolutionary and powerful tool for biomedical research. However, there have been relatively few studies using scRNA-seq in the field of kidney study. Summary scRNA-seq achieves gene expression profiling at single-cell resolution in contrast with the conventional methods of gene expression profiling, which are based on cell population and give averaged values of gene expression of the cells. Single-cell transcriptomic analysis is crucial because individual cells of the same type are highly heterogeneous in gene expression, which reflects the existence of subpopulations, different cellular states, or molecular dynamics, of the cells, and should be resolved for further insights. In addition, gene expression analysis of tissues or organs that usually comprise multiple cell types or subtypes results in data that are not fully applicable to any given cell type. scRNA-seq is capable of identifying all cell types and subtypes in a tissue, including those that are new or present in small quantity. With these unique capabilities, scRNA-seq has been used to dissect molecular processes in cell differentiation and to trace cell lineages in development. It is also used to analyze the cells in a lesion of disease to identify the cell types and molecular dynamics implicated in the injury. With continuous technical improvement, scRNA-seq has become extremely high throughput and cost effective, making it accessible to all laboratories. In the present review article, we provide an overall review of scRNA-seq concerning its history, improvements, and applications. In addition, we describe the available studies in which scRNA-seq was employed in the field of kidney research. Lastly, we discuss other potential uses of scRNA-seq for kidney research. Key Message This review article provides general information on scRNA-seq and its various uses. Particularly, we summarize the studies in the field of kidney diseases in which scRNA-seq was used and discuss potential additional uses of scRNA-seq for kidney research.
Collapse
Affiliation(s)
- Yuting Ye
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Hui Song
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jiong Zhang
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Shaolin Shi
- National Clinical Research Center for Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
23
|
Regenerative medicine in kidney disease: where we stand and where to go. Pediatr Nephrol 2018; 33:1457-1465. [PMID: 28735502 DOI: 10.1007/s00467-017-3754-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 05/23/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
The kidney is a complex organ with more than 20 types of specialized cells that play an important role in maintaining the body's homeostasis. The epithelial tubular cell is formed during embryonic development and has little proliferative capacity under physiological conditions, but after acute injury the kidney does have regenerative capacity. However, after repetitive or severe lesions, it may undergo a maladaptation process that predisposes it to chronic kidney injury. Regenerative medicine includes various repair and regeneration techniques, and these have gained increasing attention in the scientific literature. In the future, not only will these techniques contribute to the repair and regeneration of the human kidney, but probably also to the construction of an entire organ. New mechanisms studied for kidney regeneration and repair include circulating stem cells as mesenchymal stromal/stem cells and their paracrine mechanisms of action; renal progenitor stem cells; the leading role of tubular epithelial cells in the tubular repair process; the study of zebrafish larvae to understand the process of nephron development, kidney scaffold and its repopulation; and, finally, the development of organoids. This review elucidates where we are in terms of current scientific knowledge regarding these mechanisms and the promises of future scientific perspectives.
Collapse
|
24
|
Abstract
PURPOSE OF REVIEW Podocytes are critical components of the nephron filtration barrier and are depleted in many kidney injuries and disease states. Terminally differentiated adult podocytes are highly specialized, postmitotic cells, raising the question of whether the body has any ability to regenerate lost podocytes. This timely question has recently been illuminated by a series of innovative studies. Here, we review recent progress on this topic of significant interest and debate. RECENT FINDINGS The innovation of genetic labeling techniques enables fate tracing of individual podocytes, providing the strongest evidence yet that podocytes can be replaced by nearby progenitor cells. In particular, two progenitor pools have recently been identified in multiple studies: parietal epithelial cells and cells of renin lineage. These studies furthermore suggest that podocyte regeneration can be enhanced using ex-vivo or pharmacological interventions. SUMMARY Recent studies indicate that the podocyte compartment is more dynamic than previously believed. Bidirectional exchange with neighboring cellular compartments provides a mechanism for podocyte replacement. Based on these findings, we propose a set of criteria for evaluating podocyte regeneration and suggest that restoration of podocyte number to a subsclerotic threshold be targeted as a potentially achievable clinical goal.
Collapse
|
25
|
Lazzeri E, Angelotti ML, Peired A, Conte C, Marschner JA, Maggi L, Mazzinghi B, Lombardi D, Melica ME, Nardi S, Ronconi E, Sisti A, Antonelli G, Becherucci F, De Chiara L, Guevara RR, Burger A, Schaefer B, Annunziato F, Anders HJ, Lasagni L, Romagnani P. Endocycle-related tubular cell hypertrophy and progenitor proliferation recover renal function after acute kidney injury. Nat Commun 2018; 9:1344. [PMID: 29632300 PMCID: PMC5890293 DOI: 10.1038/s41467-018-03753-4] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/08/2018] [Indexed: 12/29/2022] Open
Abstract
Acute kidney injury (AKI) is considered largely reversible based on the capacity of surviving tubular cells to dedifferentiate and replace lost cells via cell division. Here we show by tracking individual tubular cells in conditional Pax8/Confetti mice that kidney function is recovered after AKI despite substantial tubular cell loss. Cell cycle and ploidy analysis upon AKI in conditional Pax8/FUCCI2aR mice and human biopsies identify endocycle-mediated hypertrophy of tubular cells. By contrast, a small subset of Pax2+ tubular progenitors enriches via higher stress resistance and clonal expansion and regenerates necrotic tubule segments, a process that can be enhanced by suitable drugs. Thus, renal functional recovery upon AKI involves remnant tubular cell hypertrophy via endocycle and limited progenitor-driven regeneration that can be pharmacologically enhanced.
Collapse
Affiliation(s)
- Elena Lazzeri
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Maria Lucia Angelotti
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Anna Peired
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Carolina Conte
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Julian A Marschner
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Laura Maggi
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | | | - Duccio Lombardi
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Maria Elena Melica
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| | - Sara Nardi
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Elisa Ronconi
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Alessandro Sisti
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Nephrology Unit and Meyer Children's University Hospital, Florence, Italy
| | - Giulia Antonelli
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | | | - Letizia De Chiara
- Nephrology Unit and Meyer Children's University Hospital, Florence, Italy
| | - Ricardo Romero Guevara
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Alexa Burger
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Beat Schaefer
- Department of Oncology and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Francesco Annunziato
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Laura Lasagni
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy
| | - Paola Romagnani
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy.
- Excellence Centre for Research, Transfer and High Education for the development of DE NOVO Therapies (DENOTHE), Florence, Italy.
- Nephrology Unit and Meyer Children's University Hospital, Florence, Italy.
| |
Collapse
|
26
|
Detection of renin lineage cell transdifferentiation to podocytes in the kidney glomerulus with dual lineage tracing. Kidney Int 2018; 93:1240-1246. [PMID: 29580637 DOI: 10.1016/j.kint.2018.01.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 12/21/2017] [Accepted: 01/04/2018] [Indexed: 12/14/2022]
Abstract
Understanding of cellular transdifferentiation is limited by the technical inability to track multiple lineages in vivo. To overcome this we developed a new tool to simultaneously fate map two distinct cell types in the kidney, and genetically test whether cells of renin lineage (CoRL) can transdifferentiate to a podocyte fate. Ren1cCreER/tdTomato/Nphs1-FLPo/FRT-EGFP mice (CoRL-PODO mice) were generated by crossing Ren1c-CreER/tdTomato CoRL reporter mice with Nphs1-FLPo/FRT-EGFP podocyte reporter mice. Following tamoxifen administration in these animals, CoRL were labeled with red fluorescence (tdTomato) and co-localized with renin. Podocytes were labeled green (enhanced green fluorescent protein) and co-localized with nephrin. Following podocyte loss by nephrotoxic antibody and subsequent enalapril-enhanced partial replacement, tdTomato-EGFP-labeled CoRL were detected as yellow-colored cells in a subset of glomerular tufts, without the use of antibodies. Co-localization with podocin indicated that these cells are podocytes, derived from CoRL origin. Thus, our novel study shows that two distinct cell types can be simultaneously labeled in the mouse kidney and provide strong genetic evidence in vivo that lost podocytes can be replaced in part by CoRL.
Collapse
|
27
|
A Population Phylogenetic View of Mitochondrial Heteroplasmy. Genetics 2018; 208:1261-1274. [PMID: 29343499 DOI: 10.1534/genetics.118.300711] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/12/2018] [Indexed: 11/18/2022] Open
Abstract
The mitochondrion has recently emerged as an active player in myriad cellular processes. Additionally, it was recently shown that >200 diseases are known to be linked to variants in mitochondrial DNA or in nuclear genes interacting with mitochondria. This has reinvigorated interest in its biology and population genetics. Mitochondrial heteroplasmy, or genotypic variation of mitochondria within an individual, is now understood to be common in humans and important in human health. However, it is still not possible to make quantitative predictions about the inheritance of heteroplasmy and its proliferation within the body, partly due to the lack of an appropriate model. Here, we present a population-genetic framework for modeling mitochondrial heteroplasmy as a process that occurs on an ontogenetic phylogeny, with genetic drift and mutation changing heteroplasmy frequencies during the various developmental processes represented in the phylogeny. Using this framework, we develop a Bayesian inference method for inferring rates of mitochondrial genetic drift and mutation at different stages of human life. Applying the method to previously published heteroplasmy frequency data, we demonstrate a severe effective germline bottleneck comprised of the cumulative genetic drift occurring between the divergence of germline and somatic cells in the mother, and the separation of germ layers in the offspring. Additionally, we find that the two somatic tissues we analyze here undergo tissue-specific bottlenecks during embryogenesis, less severe than the effective germline bottleneck, and that these somatic tissues experience little additional genetic drift during adulthood. We conclude with a discussion of possible extensions of the ontogenetic phylogeny framework and its possible applications to other ontogenetic processes in addition to mitochondrial heteroplasmy.
Collapse
|
28
|
Poulsen SB, Kristensen TB, Brooks HL, Kohan DE, Rieg T, Fenton RA. Role of adenylyl cyclase 6 in the development of lithium-induced nephrogenic diabetes insipidus. JCI Insight 2017; 2:e91042. [PMID: 28405619 DOI: 10.1172/jci.insight.91042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Psychiatric patients treated with lithium (Li+) may develop nephrogenic diabetes insipidus (NDI). Although the etiology of Li+-induced NDI (Li-NDI) is poorly understood, it occurs partially due to reduced aquaporin-2 (AQP2) expression in the kidney collecting ducts. A mechanism postulated for this is that Li+ inhibits adenylyl cyclase (AC) activity, leading to decreased cAMP, reduced AQP2 abundance, and less membrane targeting. We hypothesized that Li-NDI would not develop in mice lacking AC6. Whole-body AC6 knockout (AC6-/-) mice and potentially novel connecting tubule/principal cell-specific AC6 knockout (AC6loxloxCre) mice had approximately 50% lower urine osmolality and doubled water intake under baseline conditions compared with controls. Dietary Li+ administration increased water intake and reduced urine osmolality in control, AC6-/-, and AC6loxloxCre mice. Consistent with AC6-/- mice, medullary AQP2 and pS256-AQP2 abundances were lower in AC6loxloxCre mice compared with controls under standard conditions, and levels were further reduced after Li+ administration. AC6loxloxCre and control mice had a similar increase in the numbers of proliferating cell nuclear antigen-positive cells in response to Li+. However, AC6loxloxCre mice had a higher number of H+-ATPase B1 subunit-positive cells under standard conditions and after Li+ administration. Collectively, AC6 has a minor role in Li-NDI development but may be important for determining the intercalated cell-to-principal cell ratio.
Collapse
Affiliation(s)
- Søren Brandt Poulsen
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark.,VA San Diego Healthcare System, San Diego, California, USA
| | | | - Heddwen L Brooks
- Department of Physiology, College of Medicine, University of Arizona, Tucson, Arizona, USA
| | - Donald E Kohan
- Division of Nephrology, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Timo Rieg
- VA San Diego Healthcare System, San Diego, California, USA.,Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Robert A Fenton
- InterPrET Center, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
29
|
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
|
30
|
Abstract
Terminally differentiated cells can be reprogrammed to pluripotency or directly to another differentiated cell type in vitro, a capacity termed cellular plasticity. Plasticity is not limited to in vitro manipulations but rather represents an important aspect of the regenerative response to injury in organs. Differentiated adult cells retain the capacity to dedifferentiate, adopting a progenitor-like phenotype after injury or, alternatively, to transdifferentiate, directly converting to a different mature cell type. Emerging concepts on cellular plasticity have relevance to our understanding of repair after kidney injury, including epithelial regeneration. Here we discuss work published in the past 5 years on the cellular hierarchies and mechanisms underlying kidney injury and repair, with a particular focus on potential roles for cellular plasticity in this response.
Collapse
Affiliation(s)
- Monica Chang-Panesso
- Division of Nephrology, Department of Medicine, Washington University in Saint Louis School of Medicine, 660 S. Euclid Avenue, CB 8126, Saint Louis, Missouri 63110, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in Saint Louis School of Medicine, 660 S. Euclid Avenue, CB 8126, Saint Louis, Missouri 63110, USA
| |
Collapse
|
31
|
Swonger JM, Liu JS, Ivey MJ, Tallquist MD. Genetic tools for identifying and manipulating fibroblasts in the mouse. Differentiation 2016; 92:66-83. [PMID: 27342817 PMCID: PMC5079827 DOI: 10.1016/j.diff.2016.05.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 01/18/2023]
Abstract
The use of mouse genetic tools to track and manipulate fibroblasts has provided invaluable in vivo information regarding the activities of these cells. Recently, many new mouse strains have been described for the specific purpose of studying fibroblast behavior. Colorimetric reporter mice and lines expressing Cre are available for the study of fibroblasts in the organs prone to fibrosis, including heart, kidney, liver, lung, and skeletal muscle. In this review we summarize the current state of the models that have been used to define tissue resident fibroblast populations. While these complex genetic reagents provide unique insights into the process of fibrosis, they also require a thorough understanding of the caveats and limitations. Here, we discuss the specificity and efficiency of the available genetic models and briefly describe how they have been used to document the mechanisms of fibrosis.
Collapse
Affiliation(s)
- Jessica M Swonger
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Jocelyn S Liu
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Malina J Ivey
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Michelle D Tallquist
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| |
Collapse
|
32
|
Yang L, Kirby JE, Sunwoo H, Lee JT. Female mice lacking Xist RNA show partial dosage compensation and survive to term. Genes Dev 2016; 30:1747-60. [PMID: 27542829 PMCID: PMC5002979 DOI: 10.1101/gad.281162.116] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 07/22/2016] [Indexed: 11/25/2022]
Abstract
X-chromosome inactivation (XCI) compensates for differences in X-chromosome number between male and female mammals. XCI is orchestrated by Xist RNA, whose expression in early development leads to transcriptional silencing of one X chromosome in the female. Knockout studies have established a requirement for Xist with inviability of female embryos that inherit an Xist deletion from the father. Here, we report that female mice lacking Xist RNA can, surprisingly, develop and survive to term. Xist-null females are born at lower frequency and are smaller at birth, but organogenesis is mostly normal. Transcriptomic analysis indicates significant overexpression of hundreds of X-linked genes across multiple tissues. Therefore, Xist-null mice can develop to term in spite of a deficiency of dosage compensation. However, the degree of X-autosomal dosage imbalance was less than anticipated (1.14-fold to 1.36-fold). Thus, partial dosage compensation can be achieved without Xist, supporting the idea of inherent genome balance. Nevertheless, to date, none of the mutant mice has survived beyond weaning stage. Sudden death is associated with failure of postnatal organ maturation. Our data suggest Xist-independent mechanisms of dosage compensation and demonstrate that small deviations from X-autosomal balance can have profound effects on overall fitness.
Collapse
Affiliation(s)
- Lin Yang
- Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; Department of Genetics, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - James E Kirby
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Hongjae Sunwoo
- Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; Department of Genetics, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Jeannie T Lee
- Howard Hughes Medical Institute, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA; Department of Genetics, Harvard Medical School, Boston, Massachusetts 02114, USA
| |
Collapse
|
33
|
Chera S, Herrera PL. Regeneration of pancreatic insulin-producing cells by in situ adaptive cell conversion. Curr Opin Genet Dev 2016; 40:1-10. [PMID: 27266969 DOI: 10.1016/j.gde.2016.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/20/2016] [Accepted: 05/19/2016] [Indexed: 12/14/2022]
Abstract
The impaired ability to produce or respond to insulin, a hormone synthetized by the pancreatic β-cells, leads to diabetes. There is an excruciating need of finding new approaches to protect or restore these cells once they are lost. Replacement and ex vivo directed reprogramming methods have an undeniable therapeutic potential, yet they exhibit crucial flaws. The in vivo conversion of adult cells to functional insulin-producing cells is a promising alternative for regenerative treatments in diabetes. The stunning natural transdifferentiation potential of the adult endocrine pancreas was recently uncovered. Modulating molecular targets involved in β-cell fate maintenance or in general differentiation mechanisms can further potentiate this intrinsic cell plasticity, which leads to insulin production reconstitution.
Collapse
Affiliation(s)
- Simona Chera
- Department of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, Jonas Lies vei 65, 5021 Bergen, Norway
| | - Pedro L Herrera
- Department of Genetic Medicine & Development, Faculty of Medicine, Institute of Genetics and Genomics in Geneva (iGE3), and Centre facultaire du diabète, University of Geneva, 1 rue Michel-Servet, 1211 Geneva-4, Switzerland.
| |
Collapse
|
34
|
Meinke G, Bohm A, Hauber J, Pisabarro MT, Buchholz F. Cre Recombinase and Other Tyrosine Recombinases. Chem Rev 2016; 116:12785-12820. [PMID: 27163859 DOI: 10.1021/acs.chemrev.6b00077] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tyrosine-type site-specific recombinases (T-SSRs) have opened new avenues for the predictable modification of genomes as they enable precise genome editing in heterologous hosts. These enzymes are ubiquitous in eubacteria, prevalent in archaea and temperate phages, present in certain yeast strains, but barely found in higher eukaryotes. As tools they find increasing use for the generation and systematic modification of genomes in a plethora of organisms. If applied in host organisms, they enable precise DNA cleavage and ligation without the gain or loss of nucleotides. Criteria directing the choice of the most appropriate T-SSR system for genetic engineering include that, whenever possible, the recombinase should act independent of cofactors and that the target sequences should be long enough to be unique in a given genome. This review is focused on recent advancements in our mechanistic understanding of simple T-SSRs and their application in developmental and synthetic biology, as well as in biomedical research.
Collapse
Affiliation(s)
- Gretchen Meinke
- Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine , Boston, Massachusetts 02111, United States
| | - Andrew Bohm
- Department of Developmental, Molecular & Chemical Biology, Tufts University School of Medicine , Boston, Massachusetts 02111, United States
| | - Joachim Hauber
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology , 20251 Hamburg, Germany
| | | | - Frank Buchholz
- Medical Systems Biology, UCC, Medical Faculty Carl Gustav Carus TU Dresden , 01307 Dresden, Germany
| |
Collapse
|
35
|
A Subpopulation of Label-Retaining Cells of the Kidney Papilla Regenerates Injured Kidney Medullary Tubules. Stem Cell Reports 2016; 6:757-771. [PMID: 27117784 PMCID: PMC4939828 DOI: 10.1016/j.stemcr.2016.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 12/17/2022] Open
Abstract
To determine whether adult kidney papillary label-retaining cells (pLRCs) are specialized precursors, we analyzed their transcription profile. Among genes overexpressed in pLRCs, we selected candidate genes to perform qPCR and immunodetection of their encoded proteins. We found that Zfyve27, which encodes protrudin, identified a subpopulation of pLRCs. With Zfyve27-CreERT2 transgenic and reporter mice we generated bitransgenic animals and performed cell-lineage analysis. Post tamoxifen, Zfyve27-CreERT2 marked cells preferentially located in the upper part of the papilla. These cells were low cycling and did not generate progeny even after long-term observation, thus they did not appear to contribute to kidney homeostasis. However, after kidney injury, but only if severe, they activated a program of proliferation, migration, and morphogenesis generating multiple and long tubular segments. Remarkably these regenerated tubules were located preferentially in the kidney medulla, indicating that repair of injury in the kidney is regionally specified. These results suggest that different parts of the kidney have different progenitor cell pools.
Collapse
|
36
|
Kaverina NV, Eng DG, Schneider RRS, Pippin JW, Shankland SJ. Partial podocyte replenishment in experimental FSGS derives from nonpodocyte sources. Am J Physiol Renal Physiol 2016; 310:F1397-413. [PMID: 27076646 DOI: 10.1152/ajprenal.00369.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 04/12/2016] [Indexed: 12/29/2022] Open
Abstract
The current studies used genetic fate mapping to prove that adult podocytes can be partially replenished following depletion. Inducible NPHS2-rtTA/tetO-Cre/RS-ZsGreen-R reporter mice were generated to permanently label podocytes with the ZsGreen reporter. Experimental focal segmental glomerulosclerosis (FSGS) was induced with a cytotoxic podocyte antibody. On FSGS day 7, immunostaining for the podocyte markers p57, synaptopodin, and podocin were markedly decreased by 44%, and this was accompanied by a decrease in ZsGreen fluorescence. The nuclear stain DAPI was absent in segments of reduced ZsGreen and podocyte marker staining, which is consistent with podocyte depletion. Staining for p57, synaptopodin, podocin, and DAPI increased at FSGS day 28 and was augmented by the ACE inhibitor enalapril, which is consistent with a partial replenishment of podocytes. In contrast, ZsGreen fluorescence did not return and remained significantly low at day 28, indicating replenishment was from a nonpodocyte origin. Despite administration of bromodeoxyuridine (BrdU) thrice weekly throughout the course of disease, BrdU staining was not detected in podocytes, which is consistent with an absence of proliferation. Although ZsGreen reporting was reduced in the tuft at FSGS day 28, labeled podocytes were detected along the Bowman's capsule in a subset of glomeruli, which is consistent with migration from the tuft. Moreover, more than half of the migrated podocytes coexpressed the parietal epithelial cell (PEC) proteins claudin-1, SSeCKS, and PAX8. These results show that although podocytes can be partially replenished following abrupt depletion, a process augmented by ACE inhibition, the source or sources are nonpodocyte in origin and are independent of proliferation. Furthermore, a subset of podocytes migrate to the Bowman's capsule and begin to coexpress PEC markers.
Collapse
Affiliation(s)
| | - Diana G Eng
- Division of Nephrology, University of Washington, Seattle, Washington
| | | | - Jeffrey W Pippin
- Division of Nephrology, University of Washington, Seattle, Washington
| | | |
Collapse
|
37
|
Drummond BE, Wingert RA. Insights into kidney stem cell development and regeneration using zebrafish. World J Stem Cells 2016; 8:22-31. [PMID: 26981168 PMCID: PMC4766248 DOI: 10.4252/wjsc.v8.i2.22] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 11/28/2015] [Accepted: 01/11/2016] [Indexed: 02/06/2023] Open
Abstract
Kidney disease is an escalating global health problem, for which the formulation of therapeutic approaches using stem cells has received increasing research attention. The complexity of kidney anatomy and function, which includes the diversity of renal cell types, poses formidable challenges in the identification of methods to generate replacement structures. Recent work using the zebrafish has revealed their high capacity to regenerate the integral working units of the kidney, known as nephrons, following acute injury. Here, we discuss these findings and explore the ways that zebrafish can be further utilized to gain a deeper molecular appreciation of renal stem cell biology, which may uncover important clues for regenerative medicine.
Collapse
|
38
|
Hansson J, Ericsson AE, Axelson H, Johansson ME. Species diversity regarding the presence of proximal tubular progenitor cells of the kidney. Eur J Histochem 2016; 60:2567. [PMID: 26972712 PMCID: PMC4800248 DOI: 10.4081/ejh.2016.2567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/02/2015] [Accepted: 12/20/2015] [Indexed: 11/23/2022] Open
Abstract
The cellular source for tubular regeneration following kidney injury is a matter of dispute, with reports suggesting a stem or progenitor cells as the regeneration source while linage tracing studies in mice seemingly favor the classical theory, where regeneration is performed by randomly surviving cells. We, and others have previously described a scattered cell population localized to the tubules of human kidney, which increases in number following injury. Here we have characterized the species distribution of these proximal tubular progenitor cells (PTPCs) in kidney tissue from chimpanzee, pig, rat and mouse using a set of human PTPC markers. We detected PTPCs in chimpanzee and pig kidneys, but not in mouse tissue. Also, subjecting mice to the unilateral urethral obstruction model, caused clear signs of tubular injury, but failed to induce the PTPC phenotype in renal tubules.
Collapse
|
39
|
Meyer-Schwesinger C. The Role of Renal Progenitors in Renal Regeneration. Nephron Clin Pract 2016; 132:101-9. [DOI: 10.1159/000442180] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2015] [Accepted: 10/25/2015] [Indexed: 11/19/2022] Open
|
40
|
Wang J, Lin G, Alwaal A, Zhang X, Wang G, Jia X, Banie L, Villalta J, Lin CS, Lue TF. Kinetics of Label Retaining Cells in the Developing Rat Kidneys. PLoS One 2015; 10:e0144734. [PMID: 26650841 PMCID: PMC4674088 DOI: 10.1371/journal.pone.0144734] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 11/23/2015] [Indexed: 12/23/2022] Open
Abstract
Background The kidney is a specialized low-regenerative organ with several different types of cellular lineages. The BrdU label-retaining cell (LRCs) approach has been used as part of a strategy to identify tissue-specific stem cells in the kidney; however, because the complementary base pairing in double-stranded DNA blocks the access of the anti-BrdU antibody to BrdU subunits, the stem cell marker expression in BrdU-labeled cells are often difficult to detect. In this study, we introduced a new cell labeling and detection method in which BrdU was replaced with 5-ethynyl-2-deoxyuridine (EdU) and examined the time-dependent dynamic changes of EdU-labeled cells and potential stem/progenitor markers in the development of kidney. Methods Newborn rats were intraperitoneally injected with EdU, and their kidneys were harvested respectively at different time points at 1 day, 3 days, 1 week, 2 weeks, and 6 weeks post-injection. The kidney tissues were processed for EdU and cellular markers by immunofluorescence staining. Results At the early stage, LRCs labeled by EdU were 2176.0 ± 355.6 cells at day one in each renal tissue section, but dropped to 168 ± 48.4 cells by week 6. As time increased, the numbers of LRCs were differentially expressed in the renal cortex and papilla. At the postnatal day one, nearly twice as many cells in the cortex were EdU-labeled as compared to the papilla (28.6 ± 3.6% vs. 15.6 ± 3.4%, P<0.05), while there were more LRCs within the renal papilla since the postnatal week one, and at the postnatal week 6, one third as many cells in the cortex were EdU-labeled as compared to the papilla (2.5 ± 0.1% vs. 7.7 ± 2.7%, P<0.05). The long-term LRCs at 6-week time point were associated exclusively with the glomeruli in the cortex and the renal tubules in the papilla. At 6 weeks, the EdU-labeled LRCs combined with expression of CD34, RECA-1, Nestin, and Synaptopodin were discretely but widely distributed within the glomeruli; Stro-1 around the glomeruli; and α-smooth muscle actin (SMA) in arteries. Conversely, co-expression of CD34, RECA-1, and Nestin with the long term EdU-labeled LRCs was significantly lower in renal tubules (P<0.01), while Stro-1 and Synaptopodin were not detected. Conclusion Our data found that at 6-week time point, EdU-labeled LRCs existing in the glomeruli expressed undifferentiated podocyte and endothelial markers at high rates, while those in the renal tubules expressed Nestin and vascular markers at low rates. To understand the characterization and localization of these EdU-LRCs, further studies will be needed to test cell lineage tracing, clonogenicity and differentiation potency, and the contributions to the regeneration of the kidney in response to renal injury/repair.
Collapse
Affiliation(s)
- Jianwen Wang
- Department of Urology, Beijing ChaoYang Hospital, Capital Medical University, 8 Gongtinanlu, Beijing, 100020, China
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
- * E-mail:
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Amjad Alwaal
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Xiaoyu Zhang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Xingyuan Jia
- Department of Urology, Beijing ChaoYang Hospital, Capital Medical University, 8 Gongtinanlu, Beijing, 100020, China
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Jacqueline Villalta
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Ching-Shwun Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| | - Tom F. Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California San Francisco, San Francisco, CA, 94143-0738, United States of America
| |
Collapse
|
41
|
Podocyte Regeneration Driven by Renal Progenitors Determines Glomerular Disease Remission and Can Be Pharmacologically Enhanced. Stem Cell Reports 2015; 5:248-63. [PMID: 26235895 PMCID: PMC4618832 DOI: 10.1016/j.stemcr.2015.07.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 07/06/2015] [Accepted: 07/06/2015] [Indexed: 12/25/2022] Open
Abstract
Podocyte loss is a general mechanism of glomerular dysfunction that initiates and drives the progression of chronic kidney disease, which affects 10% of the world population. Here, we evaluate whether the regenerative response to podocyte injury influences chronic kidney disease outcome. In models of focal segmental glomerulosclerosis performed in inducible transgenic mice where podocytes are tagged, remission or progression of disease was determined by the amount of regenerated podocytes. When the same model was established in inducible transgenic mice where renal progenitors are tagged, the disease remitted if renal progenitors successfully differentiated into podocytes, while it persisted if differentiation was ineffective, resulting in glomerulosclerosis. Treatment with BIO, a GSK3s inhibitor, significantly increased disease remission by enhancing renal progenitor sensitivity to the differentiation effect of endogenous retinoic acid. These results establish renal progenitors as critical determinants of glomerular disease outcome and a pharmacological enhancement of their differentiation as a possible therapeutic strategy.
Collapse
|
42
|
Wang F, Zhao LJ. Cell lineage tracing in study of epithelial-to-mesenchymal transition during hepatic fibrosis. Shijie Huaren Xiaohua Zazhi 2015; 23:3235-3240. [DOI: 10.11569/wcjd.v23.i20.3235] [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] [Indexed: 02/06/2023] Open
Abstract
Hepatic fibrosis is the common pathologic process of chronic liver injury. Early studies mostly used immunohistochemistry to assess the role of epithelial-to-mesenchymal transition (EMT) in human or animal liver repair, and several types of liver cells including hepatocytes, cholangiocytes, hepatic stellate cells (HSCs) and liver progenitor cells have been shown to undergo EMT during hepatic fibrosis. However, this technique has several flaws. In recent years, with the rapid development of genetic engineering, especially the application of the recombinant enzyme Cre/loxP system, cell lineage tracing is becoming a popular and powerful tool to overcome the limitations of immunostaining for identifying EMT during hepatic fibrosis. Since this technique genetically labels cells, the marker will be present in any progeny of the labeled cells. Many groups have generated different lineages of double transgenic (DTG) mice and utilized different models of hepatic injury to investigate whether EMT contributes to hepatic injury or not. The purpose of this article is to summarize evidence, which is obtained using lineage cell tracing, for and against the possibility that EMT is involved in hepatic fibrosis.
Collapse
|
43
|
Lombardi D, Becherucci F, Romagnani P. How much can the tubule regenerate and who does it? An open question. Nephrol Dial Transplant 2015; 31:1243-50. [PMID: 26175143 PMCID: PMC4967725 DOI: 10.1093/ndt/gfv262] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/22/2015] [Indexed: 01/09/2023] Open
Abstract
The tubular compartment of the kidney is the primary site of a wide range of insults that can result in acute kidney injury (AKI), a condition associated with high mortality and an increased risk to develop end-stage renal disease. Nevertheless, kidney function is often quickly recovered after tubular injury. How this happens has only partially been unveiled. Indeed, although it has clearly been demonstrated that regenerated epithelial cells arise from survived intratubular cells, the true entity, as well as the cellular source of this regenerative process, remains mostly unknown. Is whichever proximal tubular epithelial cell able to dedifferentiate and divide to replace neighboring lost tubular cells, thus suggesting an extreme regenerative ability of residual tubular epithelium, or is the regenerative potential of tubular epithelium limited, and mostly related to a preexisting population of intratubular scattered progenitor cells which are more resistant to death? Gaining insights on how this process takes place is essential for developing new therapeutic strategies to prevent AKI, as well as AKI-related chronic kidney disease. The aim of this review is to discuss why the answers to these questions are still open, and how further investigations are needed to understand which is the true regenerative potential of the tubule and who are the players that allow functional recovery after AKI.
Collapse
Affiliation(s)
- Duccio Lombardi
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy
| | - Francesca Becherucci
- Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy
| | - Paola Romagnani
- Excellence Centre for Research, Transfer and High Education for the Development of DE NOVO Therapies (DENOTHE), University of Florence, Florence, Italy Nephrology and Dialysis Unit, Meyer Children's University Hospital, Florence, Italy Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|