1
|
Parvez RK, Csipán RL, Liu J, Gevorgyan A, Rutledge EA, Guo J, Kim DK, McMahon AP. Developmental and Cell Fate Analyses Support a Postnatal Origin for the Cortical Collecting System in the Mouse Kidney. J Am Soc Nephrol 2025; 36:812-824. [PMID: 39665296 DOI: 10.1681/asn.0000000579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
Key Points
An adult-like corticomedullary organization underlying kidney function is established 10 days after birth in the mouse kidney.Genetic lineage tracing demonstrates the cortical collecting duct network is generated from progenitors after birth.Mature cell types of the nephron progenitor–derived connecting tubule and ureteric progenitor–derived collecting epithelium are established by P15.
Background
Structure and function in the mammalian kidney are organized along a radial axis highlighted by the corticomedullary organization and regional patterning of the collecting system. The arborized collecting epithelium arises through controlled growth, branching, and commitment of Wnt11+ ureteric progenitor cells within cortically localized branch tips until postnatal day 3.
Methods
We applied in situ hybridization and immunofluorescence to key markers of collecting duct cell types to examine their distribution in the embryonic and postnatal mouse kidneys. To address the contribution of ureteric progenitor cells at a given time to cell diversity and spatial organization in the adult mouse kidney, we performed genetic lineage tracing of Wnt11
+
cells in the embryonic and early postnatal mouse kidney.
Results
Cell fate analyses showed much of the cortical collecting duct network was established postnatally. Furthermore, epithelial reorganization, regional differentiation, and functional maturation of key cell types to an adult-like collecting epithelium was not complete until around 2 weeks after birth in both ureteric progenitor cell–derived collecting system and structurally homologous nephron progenitor cell–derived connecting tubule.
Conclusions
These studies underline the importance of the relatively understudied early postnatal period to the development of a functional mammalian kidney.
Collapse
Affiliation(s)
- Riana K Parvez
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Réka L Csipán
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Jing Liu
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ara Gevorgyan
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Elisabeth A Rutledge
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Amgen, Inc., Thousand Oaks, California
| | - Jinjin Guo
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Doh Kyung Kim
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
| | - Andrew P McMahon
- Department of Stem Cell and Regenerative Medicine, University of Southern California, Los Angeles, California
- Division of Biology and Biological Engineering, California Institute for Technology, Pasadena, California
| |
Collapse
|
2
|
Peloggia J, Lush ME, Tsai YY, Wood C, Piotrowski T. Environmental and molecular control of tissue-specific ionocyte differentiation in zebrafish. Development 2024; 151:dev202809. [PMID: 39324331 PMCID: PMC11528218 DOI: 10.1242/dev.202809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024]
Abstract
Organisms cope with environmental fluctuations and maintain fitness in part via reversible phenotypic changes (acclimation). Aquatic animals are subject to dramatic seasonal fluctuations in water salinity, which affect osmolarity of their cells and consequently cellular function. Mechanosensory lateral line hair cells detect water motion for swimming behavior and are especially susceptible to salinity changes due to their direct contact with the environment. To maintain hair cell function when salinity decreases, neuromast (Nm)-associated ionocytes differentiate and invade lateral line neuromasts. The signals that trigger the adaptive differentiation of Nm ionocytes are unknown. We demonstrate that new Nm ionocytes are rapidly specified and selectively triggered to proliferate by low Ca2+ and Na+/Cl- levels. We further show that Nm ionocyte recruitment and induction is affected by hair cell activity. Once specified, Nm ionocyte differentiation and survival are associated with sequential activation of different Notch pathway components, a process different from other tissue-specific ionocytes. In summary, we show how environmental changes activate a signaling cascade that leads to physiological adaptation. This may prove essential for survival not only in seasonal changing environments but also in changing climates.
Collapse
Affiliation(s)
- Julia Peloggia
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Mark E. Lush
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Ya-Yin Tsai
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Christopher Wood
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | | |
Collapse
|
3
|
Wu ST, Feng Y, Song R, Qi Y, Li L, Lu D, Wang Y, Wu W, Morgan A, Wang X, Xia Y, Liu R, Alexander SI, Wong J, Zhang Y, Zheng X. Foxp1 Is Required for Renal Intercalated Cell Differentiation and Acid-Base Regulation. J Am Soc Nephrol 2024; 35:533-548. [PMID: 38332484 PMCID: PMC11149051 DOI: 10.1681/asn.0000000000000319] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 02/05/2024] [Indexed: 02/10/2024] Open
Abstract
Key Points Foxp1 is a key transcriptional factor for the differentiation of intercalated cells in collecting ducts. Dmrt2 and Hmx2 act downstream of Foxp1 to control the differentiation of type A and type B intercalated cells, respectively. Foxp1 and Dmrt2 are essential for body acid–base balance regulation. Background Kidney collecting ducts comprise principal cells and intercalated cells, with intercalated cells playing a crucial role in kidney acid–base regulation through H+ and HCO3− secretion. Despite its significance, the molecular mechanisms controlling intercalated cell development remain incompletely understood. Methods To investigate the specific role of Foxp1 in kidney tubular system, we specifically deleted Foxp1 expression in kidney distal nephrons and collecting ducts. We examined the effects of Foxp1 on intercalated cell differentiation and urine acidification. RNA sequencing and Chip-seq were used to identify Foxp1 target genes. To dissect the genetic network that regulates intercalated cell differentiation, Dmrt2 -deficient mice were generated to determine the role of Dmrt2 in intercalated cell differentiation. Foxp1 -deficient mice were crossed with Notch2 -deficient mice to dissect the relation between Foxp1 and Notch signaling. Results Foxp1 was selectively expressed in intercalated cells in collecting ducts. The absence of Foxp1 in kidney tubules led to the abolishment of intercalated cell differentiation in the collecting ducts, resulting in distal renal tubular acidosis. Foxp1 regulates the expression of Dmrt2 and Hmx2 , two genes encoding transcription factors specifically expressed in type A and type B intercalated cell cells, respectively. Further genetic analysis revealed that Dmrt2 was essential for type A intercalated cell differentiation, and Foxp1 was necessary downstream of Notch for the regulation of intercalated cell differentiation. Conclusions Foxp1 is required for the renal intercalated cell differentiation and participated in acid–base regulation. Foxp1 regulated downstream transcriptional factors, Dmrt2 and Hmx2, which were involved in the specification of distinct subsets of intercalated cells.
Collapse
Affiliation(s)
- Shi-Ting Wu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Yu Feng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Renhua Song
- Epigenetics and RNA Biology Program Centenary Institute and the Faulty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Yanmiao Qi
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Lin Li
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Dongbo Lu
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Yixuan Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Wenrun Wu
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Angela Morgan
- Murdoch Children's Research Institute, The Royal Children's Hospital and Department of Audiology and Speech Pathology and Department of Pediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Xiaohong Wang
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Renjing Liu
- Vascular Epigenetics Laboratory, Victor Chang Cardiac Research Institute and School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Stephen I. Alexander
- Department of Pediatric Nephrology, The Children's Hospital at Westmead and Centre for Kidney Research, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Justin Wong
- Epigenetics and RNA Biology Program Centenary Institute and the Faulty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Yuzhen Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of China, Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiangjian Zheng
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, and Center for Cardiovascular Diseases, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
deRiso J, Mukherjee M, Janga M, Simmons A, Kareta M, Tao J, Chandrasekar I, Surendran K. Kidney collecting duct cell type composition is regulated by Notch signaling via modulation of mTORC1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.587573. [PMID: 38645025 PMCID: PMC11030444 DOI: 10.1101/2024.04.09.587573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The plasticity and diversity of cell types with specialized functions likely defines the capacity of multicellular organisms to adapt to physiologic stressors. The kidney collecting ducts contribute to water, electrolyte, and pH homeostasis and are composed of mature intermingled epithelial cell types that are susceptible to transdifferentiate. The conversion of kidney collecting duct principal cells to intercalated cells is actively inhibited by Notch signaling to ensure urine concentrating capability. Here we identify Hes1, a target of Notch signaling, allows for maintenance of functionally distinct epithelial cell types within the same microenvironment by regulating mechanistic target of rapamycin complex 1 (mTORC1) activity. Hes1 directly represses the expression of insulin receptor substrate 1 ( Irs1 ), an upstream component of mTOR pathway and suppresses mTORC1 activity in principal cells. Genetic inactivation of tuberous sclerosis complex 2 ( Tsc2 ) to increase mTORC1 activity in mature principal cells is sufficient to promote acquisition of intercalated cell properties, while inhibition of mTORC1 in adult kidney epithelia suppresses intercalated cell properties. Considering that mTORC1 integrates environmental cues, the linkage of functionally distinct epithelial cell types to mTORC1 activity levels likely allows for cell plasticity to be regulated by physiologic and metabolic signals and the ability to sense/transduce these signals.
Collapse
|
5
|
Han S, Xu Y, Chen D, Yang F, Wang M, Zhou Q, Wang G, Li L, Xu C, Wang W, Cai S, Xing N. Notch activation defines immune-suppressive subsets of ccRCCs with unfavorable benefits from immunotherapy over VEGFR/mTOR inhibitors. iScience 2024; 27:108290. [PMID: 38179060 PMCID: PMC10765066 DOI: 10.1016/j.isci.2023.108290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 01/06/2024] Open
Abstract
The evolutionarily conserved Notch pathway, involved in cancer stem cell capacity and cancer immunity, may predict the benefit from immune checkpoint inhibitors (ICIs) in clear cell renal cell carcinoma (ccRCC). In the TCGA dataset, mRNA expression of Notch pathway genes identified three clusters with different prognoses and molecular characteristics. Based on the differentially expressed Notch pathway genes between clusters, we constructed the Notch-score, correlated with Notch activation, angiogenesis, PI3K-AKT-mTOR activity, and sensitivities to VEGFR/mTOR inhibitors. A high Notch-score was linked with more "resting"/"anti-inflammatory" rather than "activated"/"pro-inflammatory" tumor-infiltrating immune cells, inactivated immune pathways, and scarce any benefits from ICI-based therapies over VEGFR/mTOR inhibitors in the JAVELIN Renal 101 (avelumab plus axitinib vs. sunitinib) and the CheckMate-009/010/025 trials (nivolumab vs. everolimus). For the Notch-activated ccRCCs, ICIs provide limited advantages and might not be strongly recommended, by which the cost-effectiveness of treatments in ccRCCs may be potentially improved.
Collapse
Affiliation(s)
- Sujun Han
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Xu
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Dong Chen
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feiya Yang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingshuai Wang
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiaoxia Zhou
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | | | - Leo Li
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Chunwei Xu
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wenxian Wang
- Department of Clinical Trial, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Shangli Cai
- Burning Rock Biotech, Guangzhou, Guangdong, China
| | - Nianzeng Xing
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Urology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
6
|
Peloggia J, Lush ME, Tsai YY, Wood C, Piotrowski T. Environmental and molecular control of tissue-specific ionocyte differentiation in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575421. [PMID: 38260427 PMCID: PMC10802608 DOI: 10.1101/2024.01.12.575421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Organisms adjust their physiology to cope with environmental fluctuations and maintain fitness. These adaptations occur via genetic changes over multiple generations or through acclimation, a set of reversible phenotypic changes that confer resilience to the individual. Aquatic organisms are subject to dramatic seasonal fluctuations in water salinity, which can affect the function of lateral line mechanosensory hair cells. To maintain hair cell function when salinity decreases, ion-regulating cells, Neuromast-associated ionocytes (Nm ionocytes), increase in number and invade lateral line neuromasts. How environmental changes trigger this adaptive differentiation of Nm ionocytes and how these cells are specified is still unknown. Here, we identify Nm ionocyte progenitors as foxi3a/foxi3b-expressing skin cells and show that their differentiation is associated with sequential activation of different Notch pathway components, which control ionocyte survival. We demonstrate that new Nm ionocytes are rapidly specified by absolute salinity levels, independently of stress response pathways. We further show that Nm ionocyte differentiation is selectively triggered by depletion of specific ions, such as Ca2+ and Na+/Cl-, but not by low K+ levels, and is independent of media osmolarity. Finally, we demonstrate that hair cell activity plays a role in Nm ionocyte recruitment and that systemic factors are not necessary for Nm ionocyte induction. In summary, we have identified how environmental changes activate a signaling cascade that triggers basal skin cell progenitors to differentiate into Nm ionocytes and invade lateral line organs. This adaptive behavior is an example of physiological plasticity that may prove essential for survival in changing climates.
Collapse
Affiliation(s)
- Julia Peloggia
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Mark E. Lush
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Ya-Yin Tsai
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Christopher Wood
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Tatjana Piotrowski
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Lead Contact
| |
Collapse
|
7
|
Kang MJ, Ioannou S, Lougheide Q, Dittmar M, Hsu Y, Pastor-Soler NM. The study of intercalated cells using ex vivo techniques: primary cell culture, cell lines, kidney slices, and organoids. Am J Physiol Cell Physiol 2024; 326:C229-C251. [PMID: 37899748 DOI: 10.1152/ajpcell.00479.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 10/23/2023] [Accepted: 10/24/2023] [Indexed: 10/31/2023]
Abstract
This review summarizes methods to study kidney intercalated cell (IC) function ex vivo. While important for acid-base homeostasis, IC dysfunction is often not recognized clinically until it becomes severe. The advantage of using ex vivo techniques is that they allow for the differential evaluation of IC function in controlled environments. Although in vitro kidney tubular perfusion is a classical ex vivo technique to study IC, here we concentrate on primary cell cultures, immortalized cell lines, and ex vivo kidney slices. Ex vivo techniques are useful in evaluating IC signaling pathways that allow rapid responses to extracellular changes in pH, CO2, and bicarbonate (HCO3-). However, these methods for IC work can also be challenging, as cell lines that recapitulate IC do not proliferate easily in culture. Moreover, a "pure" IC population in culture does not necessarily replicate its collecting duct (CD) environment, where ICs are surrounded by the more abundant principal cells (PCs). It is reassuring that many findings obtained in ex vivo IC systems signaling have been largely confirmed in vivo. Some of these newly identified signaling pathways reveal that ICs are important for regulating NaCl reabsorption, thus suggesting new frontiers to target antihypertensive treatments. Moreover, recent single-cell characterization studies of kidney epithelial cells revealed a dual developmental origin of IC, as well as the presence of novel CD cell types with certain IC characteristics. These exciting findings present new opportunities for the study of IC ex vivo and will likely rediscover the importance of available tools in this field.NEW & NOTEWORTHY The study of kidney intercalated cells has been limited by current cell culture and kidney tissue isolation techniques. This review is to be used as a reference to select ex vivo techniques to study intercalated cells. We focused on the use of cell lines and kidney slices as potential useful models to study membrane transport proteins. We also review how novel collecting duct organoids may help better elucidate the role of these intriguing cells.
Collapse
Affiliation(s)
- Min Ju Kang
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Silvia Ioannou
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Quinn Lougheide
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Michael Dittmar
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Young Hsu
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| | - Nuria M Pastor-Soler
- Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of USC, Los Angeles, California, United States
| |
Collapse
|
8
|
Sørtvedt X, Nielsen R, Praetorius J, Christensen BM. Absence of E-Cadherin and β-Catenin in the Basal Plasma Membrane of Collecting Duct Cells During NDI Development and Recovery. J Histochem Cytochem 2023; 71:357-375. [PMID: 37439659 PMCID: PMC10363910 DOI: 10.1369/00221554231185809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/12/2023] [Indexed: 07/14/2023] Open
Abstract
Lithium (Li) induces severe polyuria and polydipsia in up to 40% of patients undergoing Li treatment. In rats, Li treatment induces a reversible cellular remodeling of the collecting duct (CD), decreasing the fraction of principal-to-intercalated cells. To investigate the potential role of adherens junction proteins, we performed immunohistochemistry on kidney cross-sections from rats treated with Li as well as rats undergoing recovery on a normal diet following 4 weeks of Li-treatment. We performed immunoelectron microscopy on cryosections to determine the ultrastructural localizations. Immunohistochemistry showed that E-cadherin and β-catenin were present in both the lateral and basal plasma membrane domains of CD cells. Immunoelectron microscopy confirmed that β-catenin was localized both to the lateral and the basal plasma membrane. The basal localization of both proteins was absent from a fraction of mainly principal cells after 10 and 15 days of Li-treatment. After 4 weeks of Li-treatment few to no cells were absent of E-cadherin and β-catenin at the basal plasma membrane. After 12 and 19 days of recovery some cells exhibited an absence of basal localization of both proteins. Thus, the observed localizational changes of E-cadherin and β-catenin appear before the cellular remodeling during both development and recovery from Li-NDI.
Collapse
Affiliation(s)
- Xabier Sørtvedt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | |
Collapse
|
9
|
Kui M, Pluznick JL, Zaidman NA. The transcription factor Foxi1 promotes expression of V-ATPase and Gpr116 in M-1 cells. Am J Physiol Renal Physiol 2023; 324:F267-F273. [PMID: 36603001 PMCID: PMC9942906 DOI: 10.1152/ajprenal.00272.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 01/06/2023] Open
Abstract
The diverse functions of each nephron segment rely on the coordinated action of specialized cell populations that are uniquely defined by their transcriptional profile. In the collecting duct, there are two critical and distinct cell populations: principal cells and intercalated cells. Principal cells play key roles in the regulation of water, Na+, and K+, whereas intercalated cells are best known for their role in acid-base homeostasis. Currently, there are no in vitro systems that recapitulate the heterogeneity of the collecting ducts, which limits high-throughput and replicate investigations of genetic and physiological phenomena. Here, we demonstrated that the transcription factor Foxi1 is sufficient to alter the transcriptional identity of M-1 cells, a murine cortical collecting duct cell line. Specifically, overexpression of Foxi1 induces the expression of intercalated cell transcripts including Gpr116, Atp6v1b1, Atp6v1g3, Atp6v0d2, Slc4a9, and Slc26a4. These data indicate that overexpression of Foxi1 differentiates M-1 cells toward a non-A, non-B type intercalated cell phenotype and may provide a novel in vitro tool to study transcriptional regulation and physiological function of the renal collecting duct.NEW & NOTEWORTHY Transfection of M-1 cells with the transcription factor Foxi1 generates cells that express V-ATPase and Gpr116 as well as other genes associated with renal intercalated cells. This straightforward and novel in vitro system could be used to study processes including transcriptional regulation and cell specification and differentiation in renal intercalated cells.
Collapse
Affiliation(s)
- Mackenzie Kui
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Jennifer L Pluznick
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Nathan A Zaidman
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, New Mexico, United States
| |
Collapse
|
10
|
Gao C, Zhang L, Chen E, Zhang W. Aqp2 + Progenitor Cells Maintain and Repair Distal Renal Segments. J Am Soc Nephrol 2022; 33:1357-1376. [PMID: 35318267 PMCID: PMC9257824 DOI: 10.1681/asn.2021081105] [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] [Received: 08/18/2021] [Accepted: 03/08/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Adult progenitor cells presumably demonstrate clonogenicity, self-renewal, and multipotentiality, and can regenerate cells under various conditions. Definitive evidence demonstrating the existence of such progenitor cells in adult mammalian kidneys is lacking. METHOD We performed in vivo lineage tracing and thymidine analogue labeling using adult tamoxifen-inducible (Aqp2ECE/+ RFP/+, Aqp2ECE/+ Brainbow/+, and Aqp2ECE/+ Brainbow/Brainbow) and WT mice. The tamoxifen-inducible mice were analyzed between 1 and 300 days postinduction. Alternatively, WT and tamoxifen-induced mice were subjected to unilateral ureteral obstruction and thymidine analogue labeling and analyzed 2-14 days post-surgery. Multiple cell-specific markers were used for high-resolution immunofluorescence confocal microscopy to identify the cell types derived from Aqp2+ cells. RESULTS Like their embryonic counterparts, adult cells expressing Aqp2 and V-ATPase subunits B1 and B2 (Aqp2+ B1B2+) are the potential Aqp2+ progenitor cells (APs). Adult APs rarely divide to generate daughter cells, either maintaining the property of the AP (self-renewal) or differentiating into DCT2/CNT/CD cells (multipotentiality), forming single cell-derived, multiple-cell clones (clonogenicity) during tissue maintenance. APs selectively and continuously regenerate DCT2/CNT/CD cells in response to injury resulting from ureteral ligation. AP proliferation demonstrated direct correlation with Notch activation and was inversely correlated with development of kidney fibrosis. Derivation of both intercalated and DCT2 cells was found to be cell division-dependent and -independent, most likely through AP differentiation which requires cell division and through direct conversion of APs and/or regular principal cells without cell division, respectively. CONCLUSION Our study demonstrates that Aqp2+ B1B2+ cells behave as adult APs to maintain and repair DCT2/CNT1/CNT2/CD segments.
Collapse
Affiliation(s)
- Chao Gao
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Long Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Enuo Chen
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York
| |
Collapse
|
11
|
Kumar P, Zadjali F, Yao Y, Johnson D, Siroky B, Astrinidis A, Vogel P, Gross KW, Bissler JJ. Tsc2 mutation induces renal tubular cell nonautonomous disease. Genes Dis 2022; 9:187-200. [PMID: 35005118 PMCID: PMC8720703 DOI: 10.1016/j.gendis.2021.03.010] [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: 11/11/2020] [Revised: 03/25/2021] [Accepted: 03/31/2021] [Indexed: 01/22/2023] Open
Abstract
TSC renal cystic disease is poorly understood and has no approved treatment. In a new principal cell-targeted murine model of Tsc cystic disease, the renal cystic epithelium is mostly composed of type A intercalated cells with an intact Tsc2 gene confirmed by sequencing, although these cells exhibit a Tsc-mutant disease phenotype. We used a newly derived targeted murine model in lineage tracing and extracellular vesicle (EV) characterization experiments and a cell culture model in EV characterization and cellular induction experiments to understand TSC cystogenesis. Using lineage tracing experiments, we found principal cells undergo clonal expansion but contribute very few cells to the cyst. We determined that cystic kidneys contain more interstitial EVs than noncystic kidneys, excrete fewer EVs in urine, and contain EVs in cyst fluid. Moreover, the loss of Tsc2 gene in EV-producing cells greatly changes the effect of EVs on renal tubular epithelium, such that the epithelium develops increased secretory and proliferative pathway activity. We demonstate that the mTORC1 pathway activity is independent form the EV production, and that the EV effects for a single cell line can vary significantly. TSC cystogenesis involves significant contribution from genetically intact cells conscripted to the mutant phenotype by mutant cell derived EVs.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - Fahad Zadjali
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Ying Yao
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - Daniel Johnson
- Molecular Bioinformatics Center, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Brian Siroky
- Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - Aristotelis Astrinidis
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| | - Peter Vogel
- Department of Veterinary Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - John J Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38103, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Pediatric Medicine Department, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
12
|
Kumar P, Zadjali F, Yao Y, Bissler JJ. Renal cystic disease in tuberous sclerosis complex. Exp Biol Med (Maywood) 2021; 246:2111-2117. [PMID: 34488473 DOI: 10.1177/15353702211038378] [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: 11/16/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is associated with TSC1 or TSC2 gene mutations resulting in hyperactivation of the mTORC1 pathway. This mTORC1 activation is associated with abnormal tissue development and proliferation such that in the kidney there are both solid tumors and cystic lesions. This review summarizes recent advances in tuberous sclerosis complex nephrology and focuses on the genetics and cell biology of tuberous sclerosis complex renal disease, highlighting a role of extracellular vesicles and the innate immune system in disease pathogenesis.
Collapse
Affiliation(s)
- Prashant Kumar
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - Fahad Zadjali
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Department of Clinical Biochemistry, College of Medicine & Health Sciences, Sultan Qaboos University, Muscat, PC 123, Oman
| | - Ying Yao
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA
| | - John J Bissler
- Department of Pediatrics, University of Tennessee Health Science Center and Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Children's Foundation Research Institute (CFRI), Le Bonheur Children's Hospital, Memphis, TN 38105, USA.,Pediatric Medicine Department, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
13
|
Klemens CA, Staruschenko A, Palygin O. The Mechanisms of Cellular Plasticity in Collecting Duct Cells: Intermediate Cell Type and Notch-mediated Transdifferentiation. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab032. [PMID: 34223174 PMCID: PMC8244659 DOI: 10.1093/function/zqab032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 01/06/2023]
Affiliation(s)
- Christine A Klemens
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Alexander Staruschenko
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA,Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53295, USA
| | | |
Collapse
|
14
|
Kidney intercalated cells and the transcription factor FOXi1 drive cystogenesis in tuberous sclerosis complex. Proc Natl Acad Sci U S A 2021; 118:2020190118. [PMID: 33536341 PMCID: PMC8017711 DOI: 10.1073/pnas.2020190118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by mutations in TSC1 or TSC2 gene and affects multiple organs, including the kidney, where it presents with angiomyolipomata and cysts that can result in kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Current studies demonstrate that kidney cyst epithelia in TSC mouse models and in humans with TSC are composed of hyperproliferating intercalated cells, along with activation of H+-ATPase and carbonic anhydrase 2. Interfering with intercalated cell proliferation completely inhibited and inactivating carbonic anhydrase 2 significantly protected against cyst formation in TSC. Targeting the acid base and/or electrolyte transporters of intercalated cells may provide a therapeutic approach for the treatment of kidney cysts in TSC. Tuberous sclerosis complex (TSC) is caused by mutations in either TSC1 or TSC2 genes and affects multiple organs, including kidney, lung, and brain. In the kidney, TSC presents with the enlargement of benign tumors (angiomyolipomata) and cysts, which eventually leads to kidney failure. The factors promoting cyst formation and tumor growth in TSC are incompletely understood. Here, we report that mice with principal cell-specific inactivation of Tsc1 develop numerous cortical cysts, which are overwhelmingly composed of hyperproliferating A-intercalated (A-IC) cells. RNA sequencing and confirmatory expression studies demonstrated robust expression of Forkhead Transcription Factor 1 (Foxi1) and its downstream targets, apical H+-ATPase and cytoplasmic carbonic anhydrase 2 (CAII), in cyst epithelia in Tsc1 knockout (KO) mice but not in Pkd1 mutant mice. In addition, the electrogenic 2Cl−/H+ exchanger (CLC-5) is significantly up-regulated and shows remarkable colocalization with H+-ATPase on the apical membrane of cyst epithelia in Tsc1 KO mice. Deletion of Foxi1, which is vital to intercalated cells viability and H+-ATPase expression, completely abrogated the cyst burden in Tsc1 KO mice, as indicated by MRI images and histological analysis in kidneys of Foxi1/Tsc1 double-knockout (dKO) mice. Deletion of CAII, which is critical to H+-ATPase activation, caused significant reduction in cyst burden and increased life expectancy in CAII/Tsc1 dKO mice vs. Tsc1 KO mice. We propose that intercalated cells and their acid/base/electrolyte transport machinery (H+-ATPase/CAII/CLC-5) are critical to cystogenesis, and their inhibition or inactivation is associated with significant protection against cyst generation and/or enlargement in TSC.
Collapse
|
15
|
Generation of patterned kidney organoids that recapitulate the adult kidney collecting duct system from expandable ureteric bud progenitors. Nat Commun 2021; 12:3641. [PMID: 34131121 PMCID: PMC8206157 DOI: 10.1038/s41467-021-23911-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 05/21/2021] [Indexed: 01/19/2023] Open
Abstract
Current kidney organoids model development and diseases of the nephron but not the contiguous epithelial network of the kidney’s collecting duct (CD) system. Here, we report the generation of an expandable, 3D branching ureteric bud (UB) organoid culture model that can be derived from primary UB progenitors from mouse and human fetal kidneys, or generated de novo from human pluripotent stem cells. In chemically-defined culture conditions, UB organoids generate CD organoids, with differentiated principal and intercalated cells adopting spatial assemblies reflective of the adult kidney’s collecting system. Aggregating 3D-cultured nephron progenitor cells with UB organoids in vitro results in a reiterative process of branching morphogenesis and nephron induction, similar to kidney development. Applying an efficient gene editing strategy to remove RET activity, we demonstrate genetically modified UB organoids can model congenital anomalies of kidney and urinary tract. Taken together, these platforms will facilitate an enhanced understanding of development, regeneration and diseases of the mammalian collecting duct system. Here, the authors model the collecting duct system in kidneys by taking ureteric bud (UB) progenitor cells from both mouse and human primary tissues, as well as from hESC and hiPSC to generate organoids, which can model congenital anomalies of the kidney and urinary tract.
Collapse
|
16
|
Petrillo F, Iervolino A, Angrisano T, Jelen S, Costanzo V, D’Acierno M, Cheng L, Wu Q, Guerriero I, Mazzarella MC, De Falco A, D’Angelo F, Ceccarelli M, Caraglia M, Capasso G, Fenton RA, Trepiccione F. Dysregulation of Principal Cell miRNAs Facilitates Epigenetic Regulation of AQP2 and Results in Nephrogenic Diabetes Insipidus. J Am Soc Nephrol 2021; 32:1339-1354. [PMID: 33727367 PMCID: PMC8259636 DOI: 10.1681/asn.2020010031] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 02/02/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs), formed by cleavage of pre-microRNA by the endoribonuclease Dicer, are critical modulators of cell function by post-transcriptionally regulating gene expression. METHODS Selective ablation of Dicer in AQP2-expressing cells (DicerAQP2Cre+ mice) was used to investigate the role of miRNAs in the kidney collecting duct of mice. RESULTS The mice had severe polyuria and nephrogenic diabetes insipidus, potentially due to greatly reduced AQP2 and AQP4 levels. Although epithelial sodium channel levels were decreased in cortex and increased in inner medulla, amiloride-sensitive sodium reabsorption was equivalent in DicerAQP2Cre+ mice and controls. Small-RNA sequencing and proteomic analysis revealed 31 and 178 significantly regulated miRNAs and proteins, respectively. Integrated bioinformatic analysis of the miRNAome and proteome suggested alterations in the epigenetic machinery and various transcription factors regulating AQP2 expression in DicerAQP2Cre+ mice. The expression profile and function of three miRNAs (miR-7688-5p, miR-8114, and miR-409-3p) whose predicted targets were involved in epigenetic control (Phf2, Kdm5c, and Kdm4a) or transcriptional regulation (GATA3, GATA2, and ELF3) of AQP2 were validated. Luciferase assays could not demonstrate direct interaction of AQP2 or the three potential transcription factors with miR-7688-5p, miR-8114, and miR-409-3p. However, transfection of respective miRNA mimics reduced AQP2 expression. Chromatin immunoprecipitation assays demonstrated decreased Phf2 and significantly increased Kdm5c interactions at the Aqp2 gene promoter in DicerAQP2Cre+ mice, resulting in decreased RNA Pol II association. CONCLUSIONS Novel evidence indicates miRNA-mediated epigenetic regulation of AQP2 expression.
Collapse
Affiliation(s)
- Federica Petrillo
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anna Iervolino
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | - Tiziana Angrisano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Sabina Jelen
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | - Vincenzo Costanzo
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | | | - Lei Cheng
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Qi Wu
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ilaria Guerriero
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | | | - Alfonso De Falco
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | - Fulvio D’Angelo
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy
| | - Michele Ceccarelli
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy,Department of Electrical Engineering and Information Technology (DIETI) University of Naples “Federico II”, Naples, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Giovambattista Capasso
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy,Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Francesco Trepiccione
- Biogem, Institute of Genetic Research “Gaetano Salvatore”, Ariano Irpino, Italy,Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
17
|
Assmus A, Mullins L, Ward M, Dobie R, Hunter R, Henderson NC, Mullins JJ. Loss of Adam10 Disrupts Ion Transport in Immortalized Kidney Collecting Duct Cells. FUNCTION 2021; 2:zqab024. [PMID: 34131651 PMCID: PMC8187228 DOI: 10.1093/function/zqab024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 01/06/2023] Open
Abstract
The kidney cortical collecting duct (CCD) comprises principal cells (PCs), intercalated cells (IC), and the recently discovered intermediate cell type. Kidney pathology in a mouse model of the syndrome of apparent aldosterone excess revealed plasticity of the CCD, with altered PC:intermediate cell:IC ratio. The self-immortalized mouse CCD cell line, mCCDcl1, shows functional characteristics of PCs, but displays a range of cell types, including intermediate cells, making it ideal to study plasticity. We knocked out Adam10, a key component of the Notch pathway, in mCCDcl1 cells, using CRISPR-Cas9 technology, and isolated independent clones, which exhibited severely affected sodium transport capacity and loss of aldosterone response. Single-cell RNA sequencing revealed significantly reduced expression of major PC-specific markers, such as Scnn1g (γ-ENaC) and Hsd11b2 (11βHSD2), but no significant changes in transcription of components of the Notch pathway were observed. Immunostaining in the knockout clone confirmed the decrease in expression of γ-ENaC and importantly, showed an altered, diffuse distribution of PC and IC markers, suggesting altered trafficking in the Adam10 knockout clone as an explanation for the loss of polarization.
Collapse
Affiliation(s)
- Adrienne Assmus
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Linda Mullins
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Mairi Ward
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Ross Dobie
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Robert Hunter
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Neil C Henderson
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - John J Mullins
- University of Edinburgh/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
18
|
Chen L, Chou CL, Knepper MA. Targeted Single-Cell RNA-seq Identifies Minority Cell Types of Kidney Distal Nephron. J Am Soc Nephrol 2021; 32:886-896. [PMID: 33769948 PMCID: PMC8017539 DOI: 10.1681/asn.2020101407] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/03/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Proximal tubule cells dominate the kidney parenchyma numerically, although less abundant cell types of the distal nephron have disproportionate roles in water and electrolyte balance. METHODS Coupling of a FACS-based enrichment protocol with single-cell RNA-seq profiled the transcriptomes of 9099 cells from the thick ascending limb (CTAL)/distal convoluted tubule (DCT) region of the mouse nephron. RESULTS Unsupervised clustering revealed Slc12a3 +/Pvalb + and Slc12a3 +/Pvalb - cells, identified as DCT1 and DCT2 cells, respectively. DCT1 cells appear to be heterogeneous, with orthogonally variable expression of Slc8a1, Calb1, and Ckb. An additional DCT1 subcluster showed marked enrichment of cell cycle-/cell proliferation-associated mRNAs (e.g., Mki67, Stmn1, and Top2a), which fit with the known plasticity of DCT cells. No DCT2-specific transcripts were found. DCT2 cells contrast with DCT1 cells by expression of epithelial sodium channel β- and γ-subunits and much stronger expression of transcripts associated with calcium transport (Trpv5, Calb1, S100g, and Slc8a1). Additionally, scRNA-seq identified three distinct CTAL (Slc12a1 +) cell subtypes. One of these expressed Nos1 and Avpr1a, consistent with macula densa cells. The other two CTAL clusters were distinguished by Cldn10 and Ptger3 in one and Cldn16 and Foxq1 in the other. These two CTAL cell types were also distinguished by expression of alternative Iroquois homeobox transcription factors, with Irx1 and Irx2 in the Cldn10 + CTAL cells and Irx3 in the Cldn16 + CTAL cells. CONCLUSIONS Single-cell transcriptomics revealed unexpected diversity among the cells of the distal nephron in mouse. Web-based data resources are provided for the single-cell data.
Collapse
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
19
|
Li H, Hohenstein P, Kuure S. Embryonic Kidney Development, Stem Cells and the Origin of Wilms Tumor. Genes (Basel) 2021; 12:genes12020318. [PMID: 33672414 PMCID: PMC7926385 DOI: 10.3390/genes12020318] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 12/23/2022] Open
Abstract
The adult mammalian kidney is a poorly regenerating organ that lacks the stem cells that could replenish functional homeostasis similarly to, e.g., skin or the hematopoietic system. Unlike a mature kidney, the embryonic kidney hosts at least three types of lineage-specific stem cells that give rise to (a) a ureter and collecting duct system, (b) nephrons, and (c) mesangial cells together with connective tissue of the stroma. Extensive interest has been raised towards these embryonic progenitor cells, which are normally lost before birth in humans but remain part of the undifferentiated nephrogenic rests in the pediatric renal cancer Wilms tumor. Here, we discuss the current understanding of kidney-specific embryonic progenitor regulation in the innate environment of the developing kidney and the types of disruptions in their balanced regulation that lead to the formation of Wilms tumor.
Collapse
Affiliation(s)
- Hao Li
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
| | - Peter Hohenstein
- Department of Human Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Satu Kuure
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, FIN-00014 Helsinki, Finland;
- GM-Unit, Laboratory Animal Center, Helsinki Institute of Life Science, University of Helsinki, FIN-00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-2941-59395
| |
Collapse
|
20
|
Mukherjee M, DeRiso J, Janga M, Fogarty E, Surendran K. Foxi1 inactivation rescues loss of principal cell fate selection in Hes1-deficient kidneys but does not ensure maintenance of principal cell gene expression. Dev Biol 2020; 466:1-11. [PMID: 32800756 DOI: 10.1016/j.ydbio.2020.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/23/2020] [Accepted: 08/06/2020] [Indexed: 01/12/2023]
Abstract
The distal nephron and collecting duct segments of the mammalian kidney consist of intercalated cell types intermingled among principal cell types. Notch signaling ensures that a sufficient number of cells select a principal instead of an intercalated cell fate. However, the precise mechanisms by which Notch signaling patterns the distal nephron and collecting duct cell fates is unknown. Here we observed that Hes1, a direct target of Notch signaling pathway, is required within the mouse developing collecting ducts for repression of Foxi1 expression, an essential intercalated cell specific transcription factor. Interestingly, inactivation of Foxi1 in Hes1-deficient collecting ducts rescues the deficiency in principal cell fate selection, overall urine concentrating deficiency, and reduces the occurrence of hydronephrosis. However, Foxi1 inactivation does not rescue the reduction in expression of all principal cell genes in the Hes1-deficient kidney collecting duct cells that select the principal cell fate. Additionally, suppression of Notch/Hes1 signaling in mature principal cells reduces principal cell gene expression without activating Foxi1. We conclude that Hes1 is a Notch signaling target that is essential for normal patterning of the collecting ducts with intermingled cell types by repressing Foxi1, and for maintenance of principal cell gene expression independent of repressing Foxi1.
Collapse
Affiliation(s)
- Malini Mukherjee
- Sanford Research, 2301 East 60th Street North, Sioux Falls, SD, 57104, USA
| | - Jennifer DeRiso
- Sanford Research, 2301 East 60th Street North, Sioux Falls, SD, 57104, USA
| | - Madhusudhana Janga
- Sanford Research, 2301 East 60th Street North, Sioux Falls, SD, 57104, USA
| | - Eric Fogarty
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, 57069, SD, USA
| | - Kameswaran Surendran
- Sanford Research, 2301 East 60th Street North, Sioux Falls, SD, 57104, USA; Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD, 57104, USA.
| |
Collapse
|
21
|
Dratwa-Chałupnik A, Wojdyła K, Ożgo M, Lepczyński A, Michałek K, Herosimczyk A, Rogowska-Wrzesińska A. Urinary Proteome of Newborn Calves-New Potential in Non-Invasive Neonatal Diagnostic. Animals (Basel) 2020; 10:E1257. [PMID: 32722141 PMCID: PMC7459841 DOI: 10.3390/ani10081257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/18/2020] [Accepted: 07/22/2020] [Indexed: 11/16/2022] Open
Abstract
Urine is a biological diagnostic material suitable not only for the analysis of kidney and urinary tract functions but also the function of other tissues and organs. The urine proteome of adult mammals differs from the urine proteome of neonatal ones. The establishment of urinary protein maps of healthy newborn calves is important for diagnosing and monitoring the progression of various diseases. The experiment was carried out on a Polish-Friesian var. of Black-and-White male calves in the sixth day of postnatal life. The two proteomics approaches used for separation and identification of urinary proteins were: 2-DE with MALDI-TOF-TOF-MS/MS and 1-DE with LC-MS/MS. This resulted in the identification of 692 urinary proteins. The majority of them were classified as extracellular proteins (40.32%), as well as proteins involved in regulation of major cellular processes (31.07%). We have observed the presence of unique proteins associated with embryonic (ameloblastin, alpha-fetoprotein, Delta-like protein, embryo-specific fibronectin 1 transcript variant, Indian hedgehog homolog) and kidney development (angiotensin-converting enzyme, angiotensinogen, aquaporin-1, calbindin, glypican 3, nidogen 1, pro-cathepsin H). Additionally, proteins involved in the renal regulation of water and electrolyte balance (angiotensinogen, angiotensin-converting enzyme, aquaporin-1, ezrin, uromodulin) were detected. Presented in the current study 1-D and 2-D urinary proteomic maps are the basis for the identification and detection of prognostic biomarkers important for defining a calf's health status.
Collapse
Affiliation(s)
- Alicja Dratwa-Chałupnik
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland; (M.O.); (A.L.); (K.M.); (A.H.)
| | - Katarzyna Wojdyła
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, M-DK-5230 Odense, Denmark; (K.W.); (A.R.-W.)
| | - Małgorzata Ożgo
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland; (M.O.); (A.L.); (K.M.); (A.H.)
| | - Adam Lepczyński
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland; (M.O.); (A.L.); (K.M.); (A.H.)
| | - Katarzyna Michałek
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland; (M.O.); (A.L.); (K.M.); (A.H.)
| | - Agnieszka Herosimczyk
- Department of Physiology, Cytobiology and Proteomics, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology Szczecin, Klemensa Janickiego 29, 71-270 Szczecin, Poland; (M.O.); (A.L.); (K.M.); (A.H.)
| | - Adelina Rogowska-Wrzesińska
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, M-DK-5230 Odense, Denmark; (K.W.); (A.R.-W.)
| |
Collapse
|
22
|
Wall SM, Verlander JW, Romero CA. The Renal Physiology of Pendrin-Positive Intercalated Cells. Physiol Rev 2020; 100:1119-1147. [PMID: 32347156 PMCID: PMC7474261 DOI: 10.1152/physrev.00011.2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Intercalated cells (ICs) are found in the connecting tubule and the collecting duct. Of the three IC subtypes identified, type B intercalated cells are one of the best characterized and known to mediate Cl- absorption and HCO3- secretion, largely through the anion exchanger pendrin. This exchanger is thought to act in tandem with the Na+-dependent Cl-/HCO3- exchanger, NDCBE, to mediate net NaCl absorption. Pendrin is stimulated by angiotensin II and aldosterone administration via the angiotensin type 1a and the mineralocorticoid receptors, respectively. It is also stimulated in models of metabolic alkalosis, such as with NaHCO3 administration. In some rodent models, pendrin-mediated HCO3- secretion modulates acid-base balance. However, of probably more physiological or clinical significance is the role of these pendrin-positive ICs in blood pressure regulation, which occurs, at least in part, through pendrin-mediated renal Cl- absorption, as well as their effect on the epithelial Na+ channel, ENaC. Aldosterone stimulates ENaC directly through principal cell mineralocorticoid hormone receptor (ligand) binding and also indirectly through its effect on pendrin expression and function. In so doing, pendrin contributes to the aldosterone pressor response. Pendrin may also modulate blood pressure in part through its action in the adrenal medulla, where it modulates the release of catecholamines, or through an indirect effect on vascular contractile force. In addition to its role in Na+ and Cl- balance, pendrin affects the balance of other ions, such as K+ and I-. This review describes how aldosterone and angiotensin II-induced signaling regulate pendrin and the contribution of pendrin-positive ICs in the kidney to distal nephron function and blood pressure.
Collapse
Affiliation(s)
- Susan M Wall
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| | - Jill W Verlander
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| | - Cesar A Romero
- Departments of Medicine and Physiology, Emory University School of Medicine, Atlanta, Georgia; and Department of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
23
|
Genini A, Mohebbi N, Daryadel A, Bettoni C, Wagner CA. Adaptive response of the murine collecting duct to alkali loading. Pflugers Arch 2020; 472:1079-1092. [DOI: 10.1007/s00424-020-02423-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/31/2020] [Accepted: 06/19/2020] [Indexed: 01/14/2023]
|
24
|
Ransick A, Lindström NO, Liu J, Zhu Q, Guo JJ, Alvarado GF, Kim AD, Black HG, Kim J, McMahon AP. Single-Cell Profiling Reveals Sex, Lineage, and Regional Diversity in the Mouse Kidney. Dev Cell 2020; 51:399-413.e7. [PMID: 31689386 DOI: 10.1016/j.devcel.2019.10.005] [Citation(s) in RCA: 291] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/26/2019] [Accepted: 10/04/2019] [Indexed: 01/13/2023]
Abstract
Chronic kidney disease affects 10% of the population with notable differences in ethnic and sex-related susceptibility to kidney injury and disease. Kidney dysfunction leads to significant morbidity and mortality and chronic disease in other organ systems. A mouse-organ-centered understanding underlies rapid progress in human disease modeling and cellular approaches to repair damaged systems. To enhance an understanding of the mammalian kidney, we combined anatomy-guided single-cell RNA sequencing of the adult male and female mouse kidney with in situ expression studies and cell lineage tracing. These studies reveal cell diversity and marked sex differences, distinct organization and cell composition of nephrons dependent on the time of nephron specification, and lineage convergence, in which contiguous functionally related cell types are specified from nephron and collecting system progenitor populations. A searchable database, Kidney Cell Explorer (https://cello.shinyapps.io/kidneycellexplorer/), enables gene-cell relationships to be viewed in the anatomical framework of the kidney.
Collapse
Affiliation(s)
- Andrew Ransick
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Qin Zhu
- Graduate Program in Genomics and Computational Biology, Biomedical Graduate Studies, University of Pennsylvania, 160 BRB II/III - 421 Curie Blvd, Philadelphia, PA 19104-6064, USA
| | - Jin-Jin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Gregory F Alvarado
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Hannah G Black
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Junhyong Kim
- Department of Biology, University of Pennsylvania, 415 S. University Ave, Philadelphia, PA 19104, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
25
|
Iervolino A, Prosperi F, De La Motte LR, Petrillo F, Spagnuolo M, D'Acierno M, Siccardi S, Perna AF, Christensen BM, Frische S, Capasso G, Trepiccione F. Potassium depletion induces cellular conversion in the outer medullary collecting duct altering Notch signaling pathway. Sci Rep 2020; 10:5708. [PMID: 32235870 PMCID: PMC7109050 DOI: 10.1038/s41598-020-61882-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 02/24/2020] [Indexed: 11/22/2022] Open
Abstract
Potassium depletion affects AQP2 expression and the cellular composition of the kidney collecting duct. This, in turn, contributes to the development of a secondary form of nephrogenic diabetes insipidus and hypokalemic nephropathy. Here we show that after 14 days of potassium depletion, the cellular fraction of A-type intercalated cells increases while the fraction of principal cells decreases along the outer medullary collecting duct in rats. The intercalated cells acquired a novel distribution pattern forming rows of cells attached to each other. These morphological changes occur progressively and reverse after 7 days of recovery on normal rat chow diet. The cellular remodeling mainly occurred in the inner stripe of outer medulla similar to the previously seen effect of lithium on the collecting duct cellular profile. The cellular remodeling is associated with the appearance of cells double labelled with both specific markers of principal and type-A intercalated cells. The appearance of this cell type was associated with the downregulation of the Notch signaling via the Hes1 pathways. These results show that the epithelium of the collecting duct has a high degree of plasticity and that Notch signaling likely plays a key role during hypokalemia.
Collapse
Affiliation(s)
- Anna Iervolino
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy
| | - Federica Prosperi
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy
| | - Luigi R De La Motte
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy
| | - Federica Petrillo
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy
| | - Manuela Spagnuolo
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy
| | - Mariavittoria D'Acierno
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy.,Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Sabrina Siccardi
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy.,Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Alessandra F Perna
- Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | | | | | - Giovambattista Capasso
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy.,Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Francesco Trepiccione
- Biogem S.c.a.r.l., Istituto di Ricerche Genetiche "Gaetano Salvatore", Ariano Irpino, Italy. .,Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy.
| |
Collapse
|
26
|
Khoshdel Rad N, Aghdami N, Moghadasali R. Cellular and Molecular Mechanisms of Kidney Development: From the Embryo to the Kidney Organoid. Front Cell Dev Biol 2020; 8:183. [PMID: 32266264 PMCID: PMC7105577 DOI: 10.3389/fcell.2020.00183] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/04/2020] [Indexed: 12/27/2022] Open
Abstract
Development of the metanephric kidney is strongly dependent on complex signaling pathways and cell-cell communication between at least four major progenitor cell populations (ureteric bud, nephron, stromal, and endothelial progenitors) in the nephrogenic zone. In recent years, the improvement of human-PSC-derived kidney organoids has opened new avenues of research on kidney development, physiology, and diseases. Moreover, the kidney organoids provide a three-dimensional (3D) in vitro model for the study of cell-cell and cell-matrix interactions in the developing kidney. In vitro re-creation of a higher-order and vascularized kidney with all of its complexity is a challenging issue; however, some progress has been made in the past decade. This review focuses on major signaling pathways and transcription factors that have been identified which coordinate cell fate determination required for kidney development. We discuss how an extensive knowledge of these complex biological mechanisms translated into the dish, thus allowed the establishment of 3D human-PSC-derived kidney organoids.
Collapse
Affiliation(s)
- Niloofar Khoshdel Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reza Moghadasali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
27
|
Zhang MZ. Notch signaling is essential in collecting duct epithelial cell fate determination during development and maintenance of cell type homeostasis in adult. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:S376. [PMID: 32016094 PMCID: PMC6976490 DOI: 10.21037/atm.2019.12.121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/20/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
- The Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
28
|
Mukherjee M, Fogarty E, Janga M, Surendran K. Notch Signaling in Kidney Development, Maintenance, and Disease. Biomolecules 2019; 9:E692. [PMID: 31690016 PMCID: PMC6920979 DOI: 10.3390/biom9110692] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/31/2019] [Accepted: 11/02/2019] [Indexed: 02/06/2023] Open
Abstract
Kidney development involves formation of nephrons intricately aligned with the vasculature and connected to a branched network of collecting ducts. Notch signaling plays multiple roles during kidney development involving the formation of nephrons composed of diverse epithelial cell types arranged into tubular segments, all the while maintaining a nephron progenitor niche. Here, we review the roles of Notch signaling identified from rodent kidney development and injury studies, while discussing human kidney diseases associated with aberrant Notch signaling. We also review Notch signaling requirement in maintenance of mature kidney epithelial cell states and speculate that Notch activity regulation mediates certain renal physiologic adaptations.
Collapse
Affiliation(s)
- Malini Mukherjee
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA.
| | - Eric Fogarty
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA.
| | - Madhusudhana Janga
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA.
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60th Street North, Sioux Falls, SD 57104, USA.
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57105, USA.
| |
Collapse
|
29
|
Abstract
The epithelium of the kidney collecting duct (CD) is composed mainly of two different types of cells with distinct and complementary functions. CD principal cells traditionally have been considered to have a major role in Na+ and water regulation, while intercalated cells (ICs) were thought to largely modulate acid-base homeostasis. In recent years, our understanding of IC function has improved significantly owing to new research findings. Thus, we now have a new model for CD transport that integrates mechanisms of salt and water reabsorption, K+ homeostasis, and acid-base status between principal cells and ICs. There are three main types of ICs (type A, type B, and non-A, non-B), which first appear in the late distal convoluted tubule or in the connecting segment in a species-dependent manner. ICs can be detected in CD from cortex to the initial part of the inner medulla, although some transport proteins that are key components of ICs also are present in medullary CD, cells considered inner medullary. Of the three types of ICs, each has a distinct morphology and expresses different complements of membrane transport proteins that translate into very different functions in homeostasis and contributions to CD luminal pro-urine composition. This review includes recent discoveries in IC intracellular and paracrine signaling that contributes to acid-base regulation as well as Na+, Cl-, K+, and Ca2+ homeostasis. Thus, these new findings highlight the potential role of ICs as targets for potential hypertension treatments.
Collapse
Affiliation(s)
- Renee Rao
- University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA
| | - Vivek Bhalla
- Division of Nephrology, Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Núria M Pastor-Soler
- University of Southern California/University Kidney Research Organization, Kidney Research Center, Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine of University of Southern California, Los Angeles, CA.
| |
Collapse
|
30
|
Chen L, Clark JZ, Nelson JW, Kaissling B, Ellison DH, Knepper MA. Renal-Tubule Epithelial Cell Nomenclature for Single-Cell RNA-Sequencing Studies. J Am Soc Nephrol 2019; 30:1358-1364. [PMID: 31253652 DOI: 10.1681/asn.2019040415] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Jevin Z Clark
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Jonathan W Nelson
- Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, Oregon; and
| | | | - David H Ellison
- Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, Oregon; and
| | - Mark A Knepper
- Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland;
| |
Collapse
|
31
|
Yoshioka W, Tohyama C. Mechanisms of Developmental Toxicity of Dioxins and Related Compounds. Int J Mol Sci 2019; 20:E617. [PMID: 30708991 PMCID: PMC6387164 DOI: 10.3390/ijms20030617] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Dioxins and related compounds induce morphological abnormalities in developing animals in an aryl hydrocarbon receptor (AhR)-dependent manner. Here we review the studies in which 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is used as a prototypical compound to elucidate the pathogenesis of morphological abnormalities. TCDD-induced cleft palate in fetal mice involves a delay in palatogenesis and dissociation of fused palate shelves. TCDD-induced hydronephrosis, once considered to be caused by the anatomical obstruction of the ureter, is now separated into TCDD-induced obstructive and non-obstructive hydronephrosis, which develops during fetal and neonatal periods, respectively. In the latter, a prostaglandin E₂ synthesis pathway and urine concentration system are involved. TCDD-induced abnormal development of prostate involves agenesis of the ventral lobe. A suggested mechanism is that AhR activation in the urogenital sinus mesenchyme by TCDD modulates the wingless-type MMTV integration site family (WNT)/β-catenin signaling cascade to interfere with budding from urogenital sinus epithelium. TCDD exposure to zebrafish embryos induces loss of epicardium progenitor cells and heart malformation. AHR2-dependent downregulation of Sox9b expression in cardiomyocytes is a suggested underlying mechanism. TCDD-induced craniofacial malformation in zebrafish is considered to result from the AHR2-dependent reduction in SRY-box 9b (SOX9b), probably partly via the noncoding RNA slincR, resulting in the underdevelopment of chondrocytes and cartilage.
Collapse
Affiliation(s)
- Wataru Yoshioka
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan.
| |
Collapse
|
32
|
Zhang Y, Riquier-Brison A, Liu T, Huang Y, Carlson NG, Peti-Peterdi J, Kishore BK. Genetic Deletion of P2Y 2 Receptor Offers Long-Term (5 Months) Protection Against Lithium-Induced Polyuria, Natriuresis, Kaliuresis, and Collecting Duct Remodeling and Cell Proliferation. Front Physiol 2018; 9:1765. [PMID: 30618788 PMCID: PMC6304354 DOI: 10.3389/fphys.2018.01765] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 11/22/2018] [Indexed: 11/15/2022] Open
Abstract
Chronic lithium administration for the treatment of bipolar disorder leads to nephrogenic diabetes insipidus (NDI), characterized by polyuria, natriuresis, kaliuresis, and collecting duct remodeling and cell proliferation among other features. Previously, using a 2-week lithium-induced NDI model, we reported that P2Y2 receptor (R) knockout mice are significantly resistant to polyuria, natriuresis, kaliuresis, and decrease in AQP2 protein abundance in the kidney relative to wild type mice. Here we show this protection is long-lasting, and is also associated with significant amelioration of lithium-induced collecting duct remodeling and cell proliferation. Age-matched wild type and knockout mice were fed regular (n = 5/genotype) or lithium-added (40 mmol/kg chow; n = 10/genotype) diet for 5 months and euthanized. Water intake, urine output and osmolality were monitored once in every month. Salt blocks were provided to mice on lithium-diet to prevent sodium loss. At the end of 5 months mice were euthanized and serum and kidney samples were analyzed. There was a steady increase in lithium-induced polyuria, natriuresis and kaliuresis in wild type mice over the 5-month period. Increases in these urinary parameters were very low in lithium-fed knockout mice, resulting in significantly widening differences between the wild type and knockout mice. Terminal AQP2 and NKCC2 protein abundances in the kidney were significantly higher in lithium-fed knockout vs. wild type mice. There were no significant differences in terminal serum lithium or sodium levels between the wild type and knockout mice. Confocal immunofluorescence microscopy revealed that lithium-induced marked remodeling of collecting duct with significantly increased proportion of [H+]-ATPase-positive intercalated cells and decreased proportion of AQP2-positive principal cells in the wild type, but not in knockout mice. Lithium-induced collecting duct cell proliferation (indicated by Ki67 labeling), was significantly lower in knockout vs. wild type mice. This is the first piece of evidence that purinergic signaling is potentially involved in lithium-induced collecting duct remodeling and cell proliferation. Our results demonstrate that genetic deletion of P2Y2-R protects against the key structural and functional alterations in Li-induced NDI, and underscore the potential utility of targeting this receptor for the treatment of NDI in bipolar patients on chronic lithium therapy.
Collapse
Affiliation(s)
- Yue Zhang
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, United States
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Anne Riquier-Brison
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, United States
| | - Tao Liu
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, United States
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
| | - Yufeng Huang
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
| | - Noel G. Carlson
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, United States
- Department of Neurobiology and Anatomy, University of Utah Health, Salt Lake City, UT, United States
- Center on Aging, University of Utah Health, Salt Lake City, UT, United States
| | - János Peti-Peterdi
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA, United States
| | - Bellamkonda K. Kishore
- Nephrology Research, Department of Veterans Affairs Salt Lake City Health Care System, Salt Lake City, UT, United States
- Department of Internal Medicine, University of Utah Health, Salt Lake City, UT, United States
- Center on Aging, University of Utah Health, Salt Lake City, UT, United States
- Department of Nutrition and Integrative Physiology, University of Utah Health, Salt Lake City, UT, United States
| |
Collapse
|
33
|
Mukherjee M, deRiso J, Otterpohl K, Ratnayake I, Kota D, Ahrenkiel P, Chandrasekar I, Surendran K. Endogenous Notch Signaling in Adult Kidneys Maintains Segment-Specific Epithelial Cell Types of the Distal Tubules and Collecting Ducts to Ensure Water Homeostasis. J Am Soc Nephrol 2018; 30:110-126. [PMID: 30514723 DOI: 10.1681/asn.2018040440] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 11/07/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Notch signaling is required during kidney development for nephron formation and principal cell fate selection within the collecting ducts. Whether Notch signaling is required in the adult kidney to maintain epithelial diversity, or whether its loss can trigger principal cell transdifferentiation (which could explain acquired diabetes insipidus in patients receiving lithium) is unclear. METHODS To investigate whether loss of Notch signaling can trigger principal cells to lose their identity, we genetically inactivated Notch1 and Notch2, inactivated the Notch signaling target Hes1, or induced expression of a Notch signaling inhibitor in all of the nephron segments and collecting ducts in mice after kidney development. We examined renal function and cell type composition of control littermates and mice with conditional Notch signaling inactivation in adult renal epithelia. In addition, we traced the fate of genetically labeled adult kidney collecting duct principal cells after Hes1 inactivation or lithium treatment. RESULTS Notch signaling was required for maintenance of Aqp2-expressing cells in distal nephron and collecting duct segments in adult kidneys. Fate tracing revealed mature principal cells in the inner stripe of the outer medulla converted to intercalated cells after genetic inactivation of Hes1 and, to a lesser extent, lithium treatment. Hes1 ensured repression of Foxi1 to prevent the intercalated cell program from turning on in mature Aqp2+ cell types. CONCLUSIONS Notch signaling via Hes1 regulates maintenance of mature renal epithelial cell states. Loss of Notch signaling or use of lithium can trigger transdifferentiation of mature principal cells to intercalated cells in adult kidneys.
Collapse
Affiliation(s)
| | | | - Karla Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota
| | - Ishara Ratnayake
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; and
| | - Divya Kota
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; and
| | - Phil Ahrenkiel
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; and
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group and .,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota
| |
Collapse
|
34
|
Hardt S, Valek L, Zeng-Brouwers J, Wilken-Schmitz A, Schaefer L, Tegeder I. Progranulin Deficient Mice Develop Nephrogenic Diabetes Insipidus. Aging Dis 2018; 9:817-830. [PMID: 30271659 PMCID: PMC6147595 DOI: 10.14336/ad.2017.1127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/27/2017] [Indexed: 12/23/2022] Open
Abstract
Loss-of-function mutations of progranulin are associated with frontotemporal dementia in humans, and its deficiency in mice is a model for this disease but with normal life expectancy and mild cognitive decline on aging. The present study shows that aging progranulin deficient mice develop progressive polydipsia and polyuria under standard housing conditions starting at middle age (6-9 months). They showed high water licking behavior and doubling of the normal daily drinking volume, associated with increased daily urine output and a decrease of urine osmolality, all maintained during water restriction. Creatinine clearance, urine urea, urine albumin and glucose were normal. Hence, there were no signs of osmotic diuresis or overt renal disease, other than a concentrating defect. In line, the kidney morphology and histology revealed a 50% increase of the kidney weight, kidney enlargement, mild infiltrations of the medulla with pro-inflammatory cells, widening of tubules but no overt signs of a glomerular or tubular pathology. Plasma vasopressin levels were on average about 3-fold higher than normal levels, suggesting that the water loss resulted from unresponsiveness of the collecting tubules towards vasopressin, and indeed aquaporin-2 immunofluorescence in collecting tubules was diminished, whereas renal and hypothalamic vasopressin were increased, the latter in spite of substantial astrogliosis in the hypothalamus. The data suggest that progranulin deficiency causes nephrogenic diabetes insipidus in mice during aging. Possibly, polydipsia in affected patients - eventually interpreted as psychogenic polydipsia - may point to a similar concentrating defect.
Collapse
Affiliation(s)
- Stefanie Hardt
- 1Clinical Pharmacology, Goethe-University Hospital Frankfurt am Main, Germany
| | - Lucie Valek
- 1Clinical Pharmacology, Goethe-University Hospital Frankfurt am Main, Germany
| | - Jinyang Zeng-Brouwers
- 2General Pharmacology and Toxicology, Goethe-University Hospital Frankfurt am Main, Germany
| | | | - Liliana Schaefer
- 2General Pharmacology and Toxicology, Goethe-University Hospital Frankfurt am Main, Germany
| | - Irmgard Tegeder
- 1Clinical Pharmacology, Goethe-University Hospital Frankfurt am Main, Germany
| |
Collapse
|
35
|
Lindström NO, De Sena Brandine G, Ransick A, McMahon AP. Single-Cell RNA Sequencing of the Adult Mouse Kidney: From Molecular Cataloging of Cell Types to Disease-Associated Predictions. Am J Kidney Dis 2018; 73:140-142. [PMID: 30241960 DOI: 10.1053/j.ajkd.2018.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 07/22/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Guilherme De Sena Brandine
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern California, Los Angeles, CA
| | - Andrew Ransick
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA.
| |
Collapse
|
36
|
Walton SL, Singh RR, Little MH, Bowles J, Li J, Moritz KM. Prolonged prenatal hypoxia selectively disrupts collecting duct patterning and postnatal function in male mouse offspring. J Physiol 2018; 596:5873-5889. [PMID: 29676801 DOI: 10.1113/jp275918] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS In the present study, we investigated whether hypoxia during late pregnancy impairs kidney development in mouse offspring, and also whether this has long-lasting consequences affecting kidney function in adulthood. Hypoxia disrupted growth of the kidney, particularly the collecting duct network, in juvenile male offspring. By mid-late adulthood, these mice developed early signs of kidney disease, notably a compromised response to water deprivation. Female offspring showed no obvious signs of impaired kidney development and did not develop kidney disease, suggesting an underlying protection mechanism from the hypoxia insult. These results help us better understand the long-lasting impact of gestational hypoxia on kidney development and the increased risk of chronic kidney disease. ABSTRACT Prenatal hypoxia is a common perturbation to arise during pregnancy, and can lead to adverse health outcomes in later life. The long-lasting impact of prenatal hypoxia on postnatal kidney development and maturation of the renal tubules, particularly the collecting duct system, is relatively unknown. In the present study, we used a model of moderate chronic maternal hypoxia throughout late gestation (12% O2 exposure from embryonic day 14.5 until birth). Histological analyses revealed marked changes in the tubular architecture of male hypoxia-exposed neonates as early as postnatal day 7, with disrupted medullary development and altered expression of Ctnnb1 and Crabp2 (encoding a retinoic acid binding protein). Kidneys of the RARElacZ line offspring exposed to hypoxia showed reduced β-galactosidase activity, indicating reduced retinoic acid-directed transcriptional activation. Wild-type male mice exposed to hypoxia had an early decline in urine concentrating capacity, evident at 4 months of age. At 12 months of age, hypoxia-exposed male mice displayed a compromised response to a water deprivation challenge, which was was correlated with an altered cellular composition of the collecting duct and diminished expression of aquaporin 2. There were no differences in the tubular structures or urine concentrating capacity between the control and hypoxia-exposed female offspring at any age. The findings of the present study suggest that prenatal hypoxia selectively disrupts collecting duct patterning through altered Wnt/β-catenin and retinoic acid signalling and this results in impaired function in male mouse offspring in later life.
Collapse
Affiliation(s)
- Sarah L Walton
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Reetu R Singh
- Department of Physiology, Monash University, Melbourne, VIC, Australia
| | - Melissa H Little
- Murdoch Childrens Research Institute, Parkville, VIC, Australia.,Department of Pediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Josephine Bowles
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Joan Li
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | - Karen M Moritz
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.,Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
37
|
Choi A, Nam SA, Kim WY, Park SH, Kim H, Yang CW, Kim J, Kim YK. Notch signaling in the collecting duct regulates renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction in mice. Korean J Intern Med 2018; 33:774-782. [PMID: 28602064 PMCID: PMC6030409 DOI: 10.3904/kjim.2016.230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/24/2016] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND/AIMS Mind bomb-1 (Mib1) encodes an E3 ubiquitin ligase, which is required for the initiation of Notch signaling. Recently, it was demonstrated that the renal collecting duct plays an important role in renal fibrosis. Here, we investigated the role of Notch signaling in renal fibrosis using conditional knockout mice with the specific ablation of Mib1 in renal collecting duct principal cells. METHODS Mib1-floxed mice (Mib1f/f) were crossed with aquaporin 2 (AQP2)-Cre mice in order to generate principal cell-specific Mib1 knockout mice (Mib1f/f :AQP2-Cre+). Unilateral ureteral obstruction (UUO) was performed, and mice were sacrificed 7 days after UUO. RESULTS After performing the UUO, renal tubulointerstitial fibrosis and the expression of transforming growth factor β were markedly enhanced in the obstructed kidneys of Mib1f/f mice compared with the sham-operated kidney of Mib1f/f mice. These changes were shown to be even more pronounced in the obstructed kidneys of Mib1f/f :AQP2-Cre+ mice than in those of the Mib1f/f mice . Furthermore, the number of TUNNEL-positive cells in renal collecting duct was higher in the obstructed kidneys of Mib1f/f :AQP2-Cre+ mice than in the kidneys of Mib1f/f mice. CONCLUSIONS Notch signaling in the renal collecting duct plays an important role in the regulation of renal tubulointerstitial fibrosis and apoptosis after UUO.
Collapse
Affiliation(s)
- Arum Choi
- Department of Anatomy and Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sun Ah Nam
- Department of Anatomy and Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Wan-Young Kim
- Department of Anatomy and Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang Hee Park
- Institute of Clinical Medicine Research of Bucheon St. Mary’s Hospital, Bucheon, Korea
| | - Hyang Kim
- Division of Nephrology, Department of Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Chul Woo Yang
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jin Kim
- Department of Anatomy and Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Correspondence to Jin Kim, M.D. Department of Anatomy and Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-7258 Fax: +82-2-2536-3110 E-mail:
| | - Yong Kyun Kim
- Department of Anatomy and Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
38
|
Affiliation(s)
- Benjamin D Humphreys
- Division of Nephrology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
39
|
Park J, Shrestha R, Qiu C, Kondo A, Huang S, Werth M, Li M, Barasch J, Suszták K. Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease. Science 2018; 360:758-763. [PMID: 29622724 DOI: 10.1126/science.aar2131] [Citation(s) in RCA: 744] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/27/2018] [Indexed: 12/12/2022]
Abstract
Our understanding of kidney disease pathogenesis is limited by an incomplete molecular characterization of the cell types responsible for the organ's multiple homeostatic functions. To help fill this knowledge gap, we characterized 57,979 cells from healthy mouse kidneys by using unbiased single-cell RNA sequencing. On the basis of gene expression patterns, we infer that inherited kidney diseases that arise from distinct genetic mutations but share the same phenotypic manifestation originate from the same differentiated cell type. We also found that the collecting duct in kidneys of adult mice generates a spectrum of cell types through a newly identified transitional cell. Computational cell trajectory analysis and in vivo lineage tracing revealed that intercalated cells and principal cells undergo transitions mediated by the Notch signaling pathway. In mouse and human kidney disease, these transitions were shifted toward a principal cell fate and were associated with metabolic acidosis.
Collapse
Affiliation(s)
- Jihwan Park
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rojesh Shrestha
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chengxiang Qiu
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ayano Kondo
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shizheng Huang
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max Werth
- Renal Division, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan Barasch
- Renal Division, Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Katalin Suszták
- Renal Electrolyte and Hypertension Division, Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
40
|
Chen L, Higgins PJ, Zhang W. Development and Diseases of the Collecting Duct System. Results Probl Cell Differ 2017; 60:165-203. [PMID: 28409346 DOI: 10.1007/978-3-319-51436-9_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The collecting duct of the mammalian kidney is important for the regulation of extracellular volume, osmolarity, and pH. There are two major structurally and functionally distinct cell types: principal cells and intercalated cells. The former regulates Na+ and water homeostasis, while the latter participates in acid-base homeostasis. In vivo lineage tracing using Cre recombinase or its derivatives such as CreGFP and CreERT2 is a powerful new technique to identify stem/progenitor cells in their native environment and to decipher the origins of the tissue that they give rise to. Recent studies using this technique in mice have revealed multiple renal progenitor cell populations that differentiate into various nephron segments and collecting duct. In particular, emerging evidence suggests that like principal cells, most of intercalated cells originate from the progenitor cells expressing water channel Aquaporin 2. Mutations or malfunctions of the channels, pumps, and transporters expressed in the collecting duct system cause various human diseases. For example, gain-of-function mutations in ENaC cause Liddle's syndrome, while loss-of-function mutations in ENaC lead to Pseudohypoaldosteronism type 1. Mutations in either AE1 or V-ATPase B1 result in distal renal tubular acidosis. Patients with disrupted AQP2 or AVPR2 develop nephrogenic diabetes insipidus. A better understanding of the function and development of the collecting duct system may facilitate the discovery of new therapeutic strategies for treating kidney disease.
Collapse
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, NHLBI, Bethesda, MD, 20892-1603, USA
| | - Paul J Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
41
|
Werth M, Schmidt-Ott KM, Leete T, Qiu A, Hinze C, Viltard M, Paragas N, Shawber CJ, Yu W, Lee P, Chen X, Sarkar A, Mu W, Rittenberg A, Lin CS, Kitajewski J, Al-Awqati Q, Barasch J. Transcription factor TFCP2L1 patterns cells in the mouse kidney collecting ducts. eLife 2017; 6. [PMID: 28577314 PMCID: PMC5484618 DOI: 10.7554/elife.24265] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/03/2017] [Indexed: 12/19/2022] Open
Abstract
Although most nephron segments contain one type of epithelial cell, the collecting ducts consists of at least two: intercalated (IC) and principal (PC) cells, which regulate acid-base and salt-water homeostasis, respectively. In adult kidneys, these cells are organized in rosettes suggesting functional interactions. Genetic studies in mouse revealed that transcription factor Tfcp2l1 coordinates IC and PC development. Tfcp2l1 induces the expression of IC specific genes, including specific H+-ATPase subunits and Jag1. Jag1 in turn, initiates Notch signaling in PCs but inhibits Notch signaling in ICs. Tfcp2l1 inactivation deletes ICs, whereas Jag1 inactivation results in the forfeiture of discrete IC and PC identities. Thus, Tfcp2l1 is a critical regulator of IC-PC patterning, acting cell-autonomously in ICs, and non-cell-autonomously in PCs. As a result, Tfcp2l1 regulates the diversification of cell types which is the central characteristic of 'salt and pepper' epithelia and distinguishes the collecting duct from all other nephron segments. DOI:http://dx.doi.org/10.7554/eLife.24265.001
Collapse
Affiliation(s)
- Max Werth
- Columbia University, New York, United States
| | - Kai M Schmidt-Ott
- Columbia University, New York, United States.,Max Delbruck Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology and Intensive Care Medicine, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | | | - Andong Qiu
- Columbia University, New York, United States.,Tongji University, Shanghai, China
| | | | - Melanie Viltard
- Columbia University, New York, United States.,Institute for European Expertise in Physiology, Paris, France
| | - Neal Paragas
- Columbia University, New York, United States.,University of Washington, Seattle, United States
| | | | - Wenqiang Yu
- Columbia University, New York, United States.,Fudan University, Shanghai, China
| | - Peter Lee
- Columbia University, New York, United States
| | - Xia Chen
- Columbia University, New York, United States
| | - Abby Sarkar
- Columbia University, New York, United States
| | - Weiyi Mu
- Columbia University, New York, United States
| | | | | | - Jan Kitajewski
- Columbia University, New York, United States.,University of Illinois at Chicago, Chicago, United States
| | | | | |
Collapse
|
42
|
El-Dahr SS, Li Y, Liu J, Gutierrez E, Hering-Smith KS, Signoretti S, Pignon JC, Sinha S, Saifudeen Z. p63+ ureteric bud tip cells are progenitors of intercalated cells. JCI Insight 2017; 2:89996. [PMID: 28469077 DOI: 10.1172/jci.insight.89996] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 03/23/2017] [Indexed: 01/13/2023] Open
Abstract
During renal branching morphogenesis, ureteric bud tip cells (UBTC) serve as the progenitor epithelium for all cell types of the collecting duct. While the transcriptional circuitry of ureteric bud (UB) branching has been intensively studied, the transcriptional control of UBTC differentiation has been difficult to ascertain. This is partly due to limited knowledge of UBTC-specific transcription factors that mark the progenitor state. Here, we identify the transcription factor p63 (also known as TP63), a master regulator of basal stem cells in stratified epithelia, as a specific marker of mouse and human UBTC. Nuclear p63 marks Ret+ UBTC transiently and is silenced by the end of nephrogenesis. Lineage tracing revealed that a subset of UBTC expressing the ΔNp63 isoform (N-terminus truncated p63) is dedicated to generating cortical intercalated cells. Germline targeting of ΔNp63 in mice caused a marked reduction in intercalated cells near the time of birth, indicating that p63 not only marks UBTC, but also is essential for their differentiation. We conclude that the choice of UBTC progenitors to differentiate is determined earlier than previously recognized and that UBTC progenitors are prepatterned and fate restricted. These findings prompt the rethinking of current paradigms of collecting duct differentiation and may have implications for regenerative renal medicine.
Collapse
Affiliation(s)
| | | | | | | | | | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Harvard School of Medicine, Boston, Massachusetts, USA
| | - Jean-Christophe Pignon
- Department of Pathology, Brigham and Women's Hospital, Harvard School of Medicine, Boston, Massachusetts, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine, University of Buffalo, New York, USA
| | | |
Collapse
|
43
|
Yosypiv IV. Prorenin receptor in kidney development. Pediatr Nephrol 2017; 32:383-392. [PMID: 27160552 DOI: 10.1007/s00467-016-3365-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 02/26/2016] [Accepted: 02/29/2016] [Indexed: 01/11/2023]
Abstract
Prorenin receptor (PRR), a receptor for renin and prorenin and an accessory subunit of the vacuolar proton pump H+-ATPase, is expressed in the developing kidney. Global loss of PRR is lethal in mice, and PRR mutations are associated with a high blood pressure, left ventricular hypertrophy and X-linked mental retardation in humans. With the advent of modern gene targeting techniques, including conditional knockout approaches, several recent studies have demonstrated critical roles for the PRR in several lineages of the developing kidney. PRR signaling has been shown to be essential for branching morphogenesis of the ureteric bud (UB), nephron progenitor survival and nephrogenesis. PRR regulates these developmental events through interactions with other transcription and growth factors. Several targeted PRR knockout animal models have structural defects mimicking congenital anomalies of the kidney and urinary tract observed in humans. The aim of this review, is to highlight new insights into the cellular and molecular mechanisms by which PRR may regulate UB branching, terminal differentiation and function of UB-derived collecting ducts, nephron progenitor maintenance, progression of nephrogenesis and normal structural kidney development and function.
Collapse
Affiliation(s)
- Ihor V Yosypiv
- Section of Pediatric Nephrology, Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University Health Sciences Center, 1430 Tulane Avenue, SL-37, New Orleans, LA, 70112, USA.
| |
Collapse
|
44
|
Grassmeyer J, Mukherjee M, deRiso J, Hettinger C, Bailey M, Sinha S, Visvader JE, Zhao H, Fogarty E, Surendran K. Elf5 is a principal cell lineage specific transcription factor in the kidney that contributes to Aqp2 and Avpr2 gene expression. Dev Biol 2017; 424:77-89. [PMID: 28215940 DOI: 10.1016/j.ydbio.2017.02.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 11/25/2022]
Abstract
The mammalian kidney collecting ducts are critical for water, electrolyte and acid-base homeostasis and develop as a branched network of tubular structures composed of principal cells intermingled with intercalated cells. The intermingled nature of the different collecting duct cell types has made it challenging to identify unique and critical factors that mark and/or regulate the development of the different collecting duct cell lineages. Here we report that the canonical Notch signaling pathway components, RBPJ and Presinilin1 and 2, are involved in patterning the mouse collecting duct cell fates by maintaining a balance between principal cell and intercalated cell fates. The relatively reduced number of principal cells in Notch-signaling-deficient kidneys offered a unique genetic leverage to identify critical principal cell-enriched factors by transcriptional profiling. Elf5, which codes for an ETS transcription factor, is one such gene that is down-regulated in kidneys with Notch-signaling-deficient collecting ducts. Additionally, Elf5 is among the earliest genes up regulated by ectopic expression of activated Notch1 in the developing collecting ducts. In the kidney, Elf5 is first expressed early within developing collecting ducts and remains on in mature principal cells. Lineage tracing of Elf5-expressing cells revealed that they are committed to the principal cell lineage by as early as E16.5. Over-expression of ETS Class IIa transcription factors, including Elf5, Elf3 and Ehf, increase the transcriptional activity of the proximal promoters of Aqp2 and Avpr2 in cultured ureteric duct cell lines. Conditional inactivation of Elf5 in the developing collecting ducts results in a small but significant reduction in the expression levels of Aqp2 and Avpr2 genes. We have identified Elf5 as an early maker of the principal cell lineage that contributes to the expression of principal cell specific genes.
Collapse
Affiliation(s)
- Justin Grassmeyer
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60(th) Street North, Sioux Falls, SD 57104, USA
| | - Malini Mukherjee
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60(th) Street North, Sioux Falls, SD 57104, USA
| | - Jennifer deRiso
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60(th) Street North, Sioux Falls, SD 57104, USA
| | - Casey Hettinger
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60(th) Street North, Sioux Falls, SD 57104, USA
| | | | - Satrajit Sinha
- Department of Biochemistry, State University of New York at Buffalo, Center for Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA
| | - Jane E Visvader
- Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Haotian Zhao
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60(th) Street North, Sioux Falls, SD 57104, USA; Department of Pediatrics, Sanford School of Medicine, Sioux Falls, SD 57104, USA
| | - Eric Fogarty
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60(th) Street North, Sioux Falls, SD 57104, USA; Basic Biomedical Sciences graduate program, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069 USA
| | - Kameswaran Surendran
- Sanford Children's Health Research Center, Sanford Research, 2301 East 60(th) Street North, Sioux Falls, SD 57104, USA; Department of Pediatrics, Sanford School of Medicine, Sioux Falls, SD 57104, USA.
| |
Collapse
|
45
|
Rahman SS, Boesen EI. Outside the mainstream: novel collecting duct proteins regulating water balance. Am J Physiol Renal Physiol 2016; 311:F1341-F1345. [PMID: 27784697 DOI: 10.1152/ajprenal.00488.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/06/2016] [Accepted: 10/18/2016] [Indexed: 12/26/2022] Open
Abstract
Body water balance is critical to survival and, therefore, very tightly regulated by the hypothalamus and kidney. A key mechanism involved in this process, the arginine vasopressin-mediated phosphorylation and apical membrane insertion of aquaporin 2 in the collecting duct, has been extensively studied; however, with the increased availability of conditional knockout animals, several novel collecting duct proteins have recently been implicated in water homeostasis. In this Mini-Review, we briefly discuss these novel proteins and their roles in the regulation of water homeostasis.
Collapse
Affiliation(s)
- Shamma S Rahman
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Erika I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
46
|
Kim JH, Han GC, Seo JY, Park I, Park W, Jeong HW, Lee SH, Bae SH, Seong J, Yum MK, Hann SH, Kwon YG, Seo D, Choi MH, Kong YY. Sex hormones establish a reserve pool of adult muscle stem cells. Nat Cell Biol 2016; 18:930-40. [PMID: 27548913 DOI: 10.1038/ncb3401] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/19/2016] [Indexed: 01/02/2023]
Abstract
Quiescent satellite cells, known as adult muscle stem cells, possess a remarkable ability to regenerate skeletal muscle following injury throughout life. Although they mainly originate from multipotent stem/progenitor cells of the somite, the mechanism underlying the establishment of quiescent satellite cell populations is unknown. Here, we show that sex hormones induce Mind bomb 1 (Mib1) expression in myofibres at puberty, which activates Notch signalling in cycling juvenile satellite cells and causes them to be converted into adult quiescent satellite cells. Myofibres lacking Mib1 fail to send Notch signals to juvenile satellite cells, leading to impaired cell cycle exit and depletion. Our findings reveal that the hypothalamic-pituitary-gonadal axis drives Mib1 expression in the myofibre niche. Moreover, the same axis regulates the re-establishment of quiescent satellite cell populations following injury. Our data show that sex hormones establish adult quiescent satellite cell populations by regulating the myofibre niche at puberty and re-establish them during regeneration.
Collapse
Affiliation(s)
- Ji-Hoon Kim
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Gi-Chan Han
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Ji-Yun Seo
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Inkuk Park
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Wookjin Park
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Hyun-Woo Jeong
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Su Hyeon Lee
- Materials and Life Science Research Division, Korea Institute of Science and Technology, Seoul 136-791, South Korea
| | - Sung-Hwan Bae
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Jinwoo Seong
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Min-Kyu Yum
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Sang-Hyeon Hann
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-752, South Korea
| | - Daekwan Seo
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea.,Center for RNA Research Institute for Basic Science, Seoul 151-742, South Korea
| | - Man Ho Choi
- Materials and Life Science Research Division, Korea Institute of Science and Technology, Seoul 136-791, South Korea
| | - Young-Yun Kong
- School of Biological Sciences, Seoul National University, Seoul 151-747, South Korea
| |
Collapse
|
47
|
Akchurin O, Du Z, Ramkellawan N, Dalal V, Han SH, Pullman J, Müsch A, Susztak K, Reidy KJ. Partitioning-Defective 1a/b Depletion Impairs Glomerular and Proximal Tubule Development. J Am Soc Nephrol 2016; 27:3725-3737. [PMID: 27185860 DOI: 10.1681/asn.2014111124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/30/2016] [Indexed: 12/21/2022] Open
Abstract
The kidney is a highly polarized epithelial organ that develops from undifferentiated mesenchyme, although the mechanisms that regulate the development of renal epithelial polarity are incompletely understood. Partitioning-defective 1 (Par1) proteins have been implicated in cell polarity and epithelial morphogenesis; however, the role of these proteins in the developing kidney has not been established. Therefore, we studied the contribution of Par1a/b to renal epithelial development. We examined the renal phenotype of newborn compound mutant mice carrying only one allele of Par1a or Par1b. Loss of three out of four Par1a/b alleles resulted in severe renal hypoplasia, associated with impaired ureteric bud branching. Compared with kidneys of newborn control littermates, kidneys of newborn mutant mice exhibited dilated proximal tubules and immature glomeruli, and the renal proximal tubular epithelia lacked proper localization of adhesion complexes. Furthermore, Par1a/b mutants expressed low levels of renal Notch ligand Jag1, activated Notch2, and Notch effecter Hes1. Together, these data demonstrate that Par1a/b has a key role in glomerular and proximal tubule development, likely via modulation of Notch signaling.
Collapse
Affiliation(s)
- Oleh Akchurin
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Zhongfang Du
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Nadira Ramkellawan
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Vidhi Dalal
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York
| | - Seung Hyeok Han
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James Pullman
- Department of Pathology, Montefiore Medical Center, Bronx, New York; and
| | - Anne Müsch
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kimberly J Reidy
- Division of Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, New York; .,Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
48
|
Abstract
The basic unit of kidney function is the nephron. In the mouse, around 14,000 nephrons form in a 10-day period extending into early neonatal life, while the human fetus forms the adult complement of nephrons in a 32-week period completed prior to birth. This review discusses our current understanding of mammalian nephrogenesis: the contributing cell types and the regulatory processes at play. A conceptual developmental framework has emerged for the mouse kidney. This framework is now guiding studies of human kidney development enabled in part by in vitro systems of pluripotent stem cell-seeded nephrogenesis. A near future goal will be to translate our developmental knowledge-base to the productive engineering of new kidney structures for regenerative medicine.
Collapse
Affiliation(s)
- Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
49
|
Aquaporin 2-labeled cells differentiate to intercalated cells in response to potassium depletion. Histochem Cell Biol 2015; 145:17-24. [PMID: 26496924 DOI: 10.1007/s00418-015-1372-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2015] [Indexed: 10/22/2022]
Abstract
The mammalian renal collecting duct consists of principal cells (PCs) and intercalated cells (ICs). Both PCs and ICs are involved in potassium (K(+)) homeostasis, PCs through their role in K(+) secretion and ICs through their ability to facilitate K(+) resorption. We previously hypothesized that PCs may differentiate into ICs upon K(+) depletion. However, no direct evidence has yet been obtained to conclusively demonstrate that PCs differentiate into ICs in response to K(+) depletion. Here, we present direct evidence for the differentiation of PCs into ICs by cell lineage tracing using aquaporin 2 (AQP2)-Cre mice and R26R-EYFP transgenic mice. In control mice, AQP2-EYFP(+) cells exhibited mainly a PC phenotype (AQP2-positive/H(+)-ATPase-negative). Interestingly, some AQP2-EYFP(+) cells exhibited an IC phenotype (H(+)-ATPase-positive/AQP2-negative); these cells accounted for 1.7 %. After K(+) depletion, the proportion of AQP2-EYFP(+) cells with an IC phenotype was increased to 4.1 %. Furthermore, some AQP2-EYFP(+) cells exhibited a "null cell" phenotype (AQP2-negative/H(+)-ATPase-negative) after K(+) depletion. Collectively, our data demonstrate that AQP2-labeled cells can differentiate into ICs, as well as null cells, in response to K(+) depletion. This finding indicates that some of AQP2-labeled cells possess properties of progenitor cells and that they can differentiate into ICs in the adult mouse kidney.
Collapse
|
50
|
Grimm PR, Lazo-Fernandez Y, Delpire E, Wall SM, Dorsey SG, Weinman EJ, Coleman R, Wade JB, Welling PA. Integrated compensatory network is activated in the absence of NCC phosphorylation. J Clin Invest 2015; 125:2136-50. [PMID: 25893600 DOI: 10.1172/jci78558] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 02/09/2015] [Indexed: 12/11/2022] Open
Abstract
Thiazide diuretics are used to treat hypertension; however, compensatory processes in the kidney can limit antihypertensive responses to this class of drugs. Here, we evaluated compensatory pathways in SPAK kinase-deficient mice, which are unable to activate the thiazide-sensitive sodium chloride cotransporter NCC (encoded by Slc12a3). Global transcriptional profiling, combined with biochemical, cell biological, and physiological phenotyping, identified the gene expression signature of the response and revealed how it establishes an adaptive physiology. Salt reabsorption pathways were created by the coordinate induction of a multigene transport system, involving solute carriers (encoded by Slc26a4, Slc4a8, and Slc4a9), carbonic anhydrase isoforms, and V-type H⁺-ATPase subunits in pendrin-positive intercalated cells (PP-ICs) and ENaC subunits in principal cells (PCs). A distal nephron remodeling process and induction of jagged 1/NOTCH signaling, which expands the cortical connecting tubule with PCs and replaces acid-secreting α-ICs with PP-ICs, were partly responsible for the compensation. Salt reabsorption was also activated by induction of an α-ketoglutarate (α-KG) paracrine signaling system. Coordinate regulation of a multigene α-KG synthesis and transport pathway resulted in α-KG secretion into pro-urine, as the α-KG-activated GPCR (Oxgr1) increased on the PP-IC apical surface, allowing paracrine delivery of α-KG to stimulate salt transport. Identification of the integrated compensatory NaCl reabsorption mechanisms provides insight into thiazide diuretic efficacy.
Collapse
|