1
|
Reum Kwon B, Jo AR, Lee I, Lee G, Joo Park Y, Pyo Lee J, Park NY, Kho Y, Kim S, Ji K, Choi K. Thyroid, neurodevelopmental, and kidney toxicities of common organic UV filters in embryo-larval zebrafish (Danio rerio), and their potential links. ENVIRONMENT INTERNATIONAL 2024; 192:109030. [PMID: 39341038 DOI: 10.1016/j.envint.2024.109030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024]
Abstract
Organic UV filters (OUVFs) have been commonly used in sunscreen and many consumer products. Following dermal application, these compounds can enter circulation and may cause systemic effects in humans. In the present study, we chose four OUVFs frequently detected in the environment, i.e., avobenzone (AVB), benzophenone-3 (BP-3), octocrylene (OC), and octyl methoxycinnamate (OMC), and evaluated their thyroid, neurodevelopmental, and kidney toxicities. For this purpose, zebrafish embryos (<4 h post fertilization, hpf) were exposed to sublethal concentrations of AVB, BP-3, OC, or OMC until 120 hpf. Exposure to all OUVFs decreased thyroid hormone (TH) levels, probably by enhanced metabolism and excretion of THs (ugt1ab and/or sult1 st5) in the larval fish. Exposure to the OUVFs also induced hypoactivities and/or anxiety-like behaviors: Regulatory changes of mbp, gfap, c-fos, syn2a, sty1a, and stxbp1b genes, support the changes in normal neurobehavior of the larval fish. Moreover, the OUVFs exposure caused increased proteinuria in the fish, along with transcriptional changes of wt1, nephrin, podocin, and cdh17 genes, which could explain the observed reduction in kidney functions. Principal component analysis (PCA) implied the potential interplay of THs with neurogenesis, or podocyte differentiation of the larval fish. Toxicological consequences of altered TH homeostasis, neurobehavior, and kidney function at the early life stage warrant further investigations not only in humans but also in aquatic ecosystems.
Collapse
Affiliation(s)
- Ba Reum Kwon
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Ah-Reum Jo
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Inae Lee
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Gowoon Lee
- Department of Safety Engineering, Korea National University of Transportation, Chungju, Chungbuk 27469, Republic of Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 03080, Republic of Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Republic of Korea
| | - Na-Youn Park
- Department of Health, Environment & Safety, Eulji University, Seongnam, Gyeonggi 13135, Republic of Korea
| | - Younglim Kho
- Department of Health, Environment & Safety, Eulji University, Seongnam, Gyeonggi 13135, Republic of Korea
| | - Sungkyoon Kim
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyunghee Ji
- Department of Environmental Health, Yongin University, Yongin, Gyeonggi 17092, Republic of Korea; Department of Occupational and Environmental Health, Yongin University, Yongin, Gyeonggi 17092, Republic of Korea
| | - Kyungho Choi
- Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
2
|
Song L, Li Q, Xia L, Sahay AE, Qiu Q, Li Y, Li H, Sasaki K, Susztak K, Wu H, Wan L. Single-cell multiomics reveals ENL mutation perturbs kidney developmental trajectory by rewiring gene regulatory landscape. Nat Commun 2024; 15:5937. [PMID: 39009564 PMCID: PMC11250843 DOI: 10.1038/s41467-024-50171-w] [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: 10/31/2023] [Accepted: 07/02/2024] [Indexed: 07/17/2024] Open
Abstract
How disruptions to normal cell differentiation link to tumorigenesis remains incompletely understood. Wilms tumor, an embryonal tumor associated with disrupted organogenesis, often harbors mutations in epigenetic regulators, but their role in kidney development remains unexplored. Here, we show at single-cell resolution that a Wilms tumor-associated mutation in the histone acetylation reader ENL disrupts kidney differentiation in mice by rewiring the gene regulatory landscape. Mutant ENL promotes nephron progenitor commitment while restricting their differentiation by dysregulating transcription factors such as Hox clusters. It also induces abnormal progenitors that lose kidney-associated chromatin identity. Furthermore, mutant ENL alters the transcriptome and chromatin accessibility of stromal progenitors, resulting in hyperactivation of Wnt signaling. The impacts of mutant ENL on both nephron and stroma lineages lead to profound kidney developmental defects and postnatal mortality in mice. Notably, a small molecule inhibiting mutant ENL's histone acetylation binding activity largely reverses these defects. This study provides insights into how mutations in epigenetic regulators disrupt kidney development and suggests a potential therapeutic approach.
Collapse
Affiliation(s)
- Lele Song
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qinglan Li
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lingbo Xia
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of the School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Arushi Eesha Sahay
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qi Qiu
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yuanyuan Li
- MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Haitao Li
- MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Kotaro Sasaki
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA, 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Katalin Susztak
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hao Wu
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Liling Wan
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
3
|
Allen B, Savoy L, Ryabinin P, Bottomly D, Chen R, Goff B, Wang A, McWheeny SK, Zhang H. Upregulation of HOXA3 by isoform-specific Wilms tumour 1 drives chemotherapy resistance in acute myeloid leukaemia. Br J Haematol 2024; 205:207-219. [PMID: 38867543 PMCID: PMC11448753 DOI: 10.1111/bjh.19563] [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: 03/04/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024]
Abstract
Upregulation of the Wilms' tumour 1 (WT1) gene is common in acute myeloid leukaemia (AML) and is associated with poor prognosis. WT1 generates 12 primary transcripts through different translation initiation sites and alternative splicing. The short WT1 transcripts express abundantly in primary leukaemia samples. We observed that overexpression of short WT1 transcripts lacking exon 5 with and without the KTS motif (sWT1+/- and sWT1-/-) led to reduced cell growth. However, only sWT1+/- overexpression resulted in decreased CD71 expression, G1 arrest, and cytarabine resistance. Primary AML patient cells with low CD71 expression exhibit resistance to cytarabine, suggesting that CD71 may serve as a potential biomarker for chemotherapy. RNAseq differential expressed gene analysis identified two transcription factors, HOXA3 and GATA2, that are specifically upregulated in sWT1+/- cells, whereas CDKN1A is upregulated in sWT1-/- cells. Overexpression of either HOXA3 or GATA2 reproduced the effects of sWT1+/-, including decreased cell growth, G1 arrest, reduced CD71 expression and cytarabine resistance. HOXA3 expression correlates with chemotherapy response and overall survival in NPM1 mutation-negative leukaemia specimens. Overexpression of HOXA3 leads to drug resistance against a broad spectrum of chemotherapeutic agents. Our results suggest that WT1 regulates cell proliferation and drug sensitivity in an isoform-specific manner.
Collapse
MESH Headings
- Humans
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, CD/biosynthesis
- Cell Line, Tumor
- Cytarabine/pharmacology
- Cytarabine/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Nucleophosmin
- Protein Isoforms
- Receptors, Transferrin
- Up-Regulation
- WT1 Proteins/genetics
- WT1 Proteins/metabolism
- WT1 Proteins/biosynthesis
Collapse
Affiliation(s)
- Basil Allen
- Division of Oncological Sciences, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Lindsey Savoy
- Division of Oncological Sciences, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Peter Ryabinin
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Daniel Bottomly
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Reid Chen
- Division of Oncological Sciences, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Bonnie Goff
- Division of Oncological Sciences, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Anthony Wang
- Division of Oncological Sciences, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| | - Shannon K McWheeny
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Knight Cancer Institute, Portland, OR
| | - Haijiao Zhang
- Division of Oncological Sciences, Oregon Health & Science University, Knight Cancer Institute, Portland, OR
| |
Collapse
|
4
|
Song L, Li Q, Xia L, Sahay A, Qiu Q, Li Y, Li H, Sasaki K, Susztak K, Wu H, Wan L. Single-Cell multiomics reveals ENL mutation perturbs kidney developmental trajectory by rewiring gene regulatory landscape. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.591709. [PMID: 38766219 PMCID: PMC11100752 DOI: 10.1101/2024.05.09.591709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Cell differentiation during organogenesis relies on precise epigenetic and transcriptional control. Disruptions to this regulation can result in developmental abnormalities and malignancies, yet the underlying mechanisms are not well understood. Wilms tumors, a type of embryonal tumor closely linked to disrupted organogenesis, harbor mutations in epigenetic regulators in 30-50% of cases. However, the role of these regulators in kidney development and pathogenesis remains unexplored. By integrating mouse modeling, histological characterizations, and single-cell transcriptomics and chromatin accessibility profiling, we show that a Wilms tumor-associated mutation in the chromatin reader protein ENL disrupts kidney development trajectory by rewiring the gene regulatory landscape. Specifically, the mutant ENL promotes the commitment of nephron progenitors while simultaneously restricting their differentiation by dysregulating key transcription factor regulons, particularly the HOX clusters. It also induces the emergence of abnormal progenitor cells that lose their chromatin identity associated with kidney specification. Furthermore, the mutant ENL might modulate stroma-nephron interactions via paracrine Wnt signaling. These multifaceted effects caused by the mutation result in severe developmental defects in the kidney and early postnatal mortality in mice. Notably, transient inhibition of the histone acetylation binding activity of mutant ENL with a small molecule displaces transcriptional condensates formed by mutant ENL from target genes, abolishes its gene activation function, and restores developmental defects in mice. This work provides new insights into how mutations in epigenetic regulators can alter the gene regulatory landscape to disrupt kidney developmental programs at single-cell resolution in vivo . It also offers a proof-of-concept for the use of epigenetics-targeted agents to rectify developmental defects.
Collapse
|
5
|
Gyarmati G, Shroff UN, Riquier-Brison A, Desposito D, Ju W, Stocker SD, Izuhara A, Deepak S, Becerra Calderon A, Burford JL, Kadoya H, Moon JY, Chen Y, Rinschen MM, Ahmadi N, Lau L, Biemesderfer D, James AW, Minichiello L, Zlokovic BV, Gill IS, Kretzler M, Peti-Peterdi J. Neuronally differentiated macula densa cells regulate tissue remodeling and regeneration in the kidney. J Clin Invest 2024; 134:e174558. [PMID: 38598837 PMCID: PMC11142747 DOI: 10.1172/jci174558] [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: 08/14/2023] [Accepted: 04/09/2024] [Indexed: 04/12/2024] Open
Abstract
Tissue regeneration is limited in several organs, including the kidney, contributing to the high prevalence of kidney disease globally. However, evolutionary and physiological adaptive responses and the presence of renal progenitor cells suggest an existing remodeling capacity. This study uncovered endogenous tissue remodeling mechanisms in the kidney that were activated by the loss of body fluid and salt and regulated by a unique niche of a minority renal cell type called the macula densa (MD). Here, we identified neuronal differentiation features of MD cells that sense the local and systemic environment and secrete angiogenic, growth, and extracellular matrix remodeling factors, cytokines and chemokines, and control resident progenitor cells. Serial intravital imaging, MD nerve growth factor receptor and Wnt mouse models, and transcriptome analysis revealed cellular and molecular mechanisms of these MD functions. Human and therapeutic translation studies illustrated the clinical potential of MD factors, including CCN1, as a urinary biomarker and therapeutic target in chronic kidney disease. The concept that a neuronally differentiated key sensory and regulatory cell type responding to organ-specific physiological inputs controls local progenitors to remodel or repair tissues may be applicable to other organs and diverse tissue-regenerative therapeutic strategies.
Collapse
Affiliation(s)
- Georgina Gyarmati
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Urvi Nikhil Shroff
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Anne Riquier-Brison
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Dorinne Desposito
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Wenjun Ju
- Division of Nephrology, Department of Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Sean D. Stocker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Audrey Izuhara
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Sachin Deepak
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Alejandra Becerra Calderon
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - James L. Burford
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Hiroyuki Kadoya
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Ju-Young Moon
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Yibu Chen
- USC Libraries Bioinformatics Service, University of Southern California, Los Angeles, California, USA
| | - Markus M. Rinschen
- Center for Molecular Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nariman Ahmadi
- Institute of Urology, Catherine and Joseph Aresty Department of Urology, University of Southern California, Los Angeles, California, USA
| | - Lester Lau
- Department of Biochemistry and Molecular Genetics, College of Medicine, The University of Illinois at Chicago, Chicago, Illinois, USA
| | - Daniel Biemesderfer
- Section of Nephrology and Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| | - Inderbir S. Gill
- Institute of Urology, Catherine and Joseph Aresty Department of Urology, University of Southern California, Los Angeles, California, USA
| | - Matthias Kretzler
- Division of Nephrology, Department of Medicine, and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - János Peti-Peterdi
- Department of Physiology and Neuroscience and Department of Medicine, Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
6
|
Bronstein R, Pace J, Gowthaman Y, Salant DJ, Mallipattu SK. Podocyte-Parietal Epithelial Cell Interdependence in Glomerular Development and Disease. J Am Soc Nephrol 2023; 34:737-750. [PMID: 36800545 PMCID: PMC10125654 DOI: 10.1681/asn.0000000000000104] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 02/04/2023] [Indexed: 02/19/2023] Open
Abstract
Podocytes and parietal epithelial cells (PECs) are among the few principal cell types within the kidney glomerulus, the former serving as a crucial constituent of the kidney filtration barrier and the latter representing a supporting epithelial layer that adorns the inner wall of Bowman's capsule. Podocytes and PECs share a circumscript developmental lineage that only begins to diverge during the S-shaped body stage of nephron formation-occurring immediately before the emergence of the fully mature nephron. These two cell types, therefore, share a highly conserved gene expression program, evidenced by recently discovered intermediate cell types occupying a distinct spatiotemporal gene expression zone between podocytes and PECs. In addition to their homeostatic functions, podocytes and PECs also have roles in kidney pathogenesis. Rapid podocyte loss in diseases, such as rapidly progressive GN and collapsing and cellular subtypes of FSGS, is closely allied with PEC proliferation and migration toward the capillary tuft, resulting in the formation of crescents and pseudocrescents. PECs are thought to contribute to disease progression and severity, and the interdependence between these two cell types during development and in various manifestations of kidney pathology is the primary focus of this review.
Collapse
Affiliation(s)
- Robert Bronstein
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Jesse Pace
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - Yogesh Gowthaman
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
| | - David J. Salant
- Division of Nephrology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Sandeep K. Mallipattu
- Division of Nephrology, Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, New York
- Renal Section, Northport VA Medical Center, Northport, New York
| |
Collapse
|
7
|
Ramos-Gonzalez MR, Vazquez-Garza E, Garcia-Rivas G, Rodriguez-Aguayo C, Chavez-Reyes A. Therapeutic Effects of WT1 Silencing via Respiratory Administration of Neutral DOPC Liposomal-siRNA in a Lung Metastasis Melanoma Murine Model. Noncoding RNA 2023; 9:ncrna9020021. [PMID: 36960966 PMCID: PMC10037624 DOI: 10.3390/ncrna9020021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/25/2023] Open
Abstract
The lungs represent a frequent target for metastatic melanoma as they offer a high-oxygen environment for tumor development. The overexpression of the WT1 protein has been associated with the occurrence of melanoma. In this study, we evaluated the effects of silencing the WT1 protein by siRNA in both in vitro in the B16F10 melanoma cell line and in vivo in a murine model of lung metastatic melanoma. We did this by implementing a novel respiratory delivery strategy of a neutral DOPC liposomal-siRNA system (L-siRNA). In vitro studies showed an effective silencing of the WT1 protein in the siRNAs' WT1-treated cells when compared with controls, resulting in a loss of the cell's viability and proliferation by inducing G1 arrest, the inhibition of the migration and invasion capacities of the cells, as well as the induction of apoptosis. In vivo, the respiratory administration of L-WT1 siRNA showed an efficient biodistribution on the lungs. After two weeks of treatment, the silencing of the WT1 protein resulted in an important antitumor activity that reduced the tumor weight. In the survival study, L-WT1 treatment could significantly delay the death of the animals. This work demonstrates the efficacy of the L-siRNA respiratory administration as a novel therapy to reduce pulmonary tumors and to increase survivability by silencing specific cancer oncogenes as WT1.
Collapse
Affiliation(s)
- Martin R Ramos-Gonzalez
- Department of Genetic Therapy, Monterrey Unit, Center for Research and Advanced Studies of the National Polytechnic Institute, Monterrey 66600, Nuevo León, Mexico
| | - Eduardo Vazquez-Garza
- Cátedra de Cardiología Y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey 64849, Nuevo León, Mexico
| | - Gerardo Garcia-Rivas
- Cátedra de Cardiología Y Medicina Vascular, Escuela de Medicina, Tecnologico de Monterrey, Monterrey 64849, Nuevo León, Mexico
| | - Cristian Rodriguez-Aguayo
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Arturo Chavez-Reyes
- Department of Genetic Therapy, Monterrey Unit, Center for Research and Advanced Studies of the National Polytechnic Institute, Monterrey 66600, Nuevo León, Mexico
- Basic Sciences Unit, Medical School, Universidad Finis Terrae, Santiago de Chile 7501015, Chile
| |
Collapse
|
8
|
Cetina-Palma A, Namorado-Tónix C, Rodríguez-Muñoz R, Vergara P, Reyes-Sánchez JL, Segovia J. Characterization of the pattern of expression of Gas1 in the kidney during postnatal development in the rat. PLoS One 2023; 18:e0284816. [PMID: 37093844 PMCID: PMC10124827 DOI: 10.1371/journal.pone.0284816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/09/2023] [Indexed: 04/25/2023] Open
Abstract
Growth Arrest-Specific 1 (Gas1) is a pleiotropic protein with different functions, in the adult kidney Gas1 acts as an endogenous inhibitor of cell proliferation but it is also necessary for the maintenance and proliferation of Renal Progenitor Cells (RPC) during early development, thus it fulfills important functions in the adult kidney. However, it is not known whether or not Gas1 is expressed during postnatal development, a critical stage for renal maturation. For this reason, the main objective of this work was to characterize the expression pattern of Gas1 in the different regions of the kidney by immunofluorescence and Western blot analysis during the postnatal development of the rat. We found that Gas1 is present and has a differential expression pattern in the various regions of the nephron during postnatal development. We observed that the highest levels of expression of Gas1 occur in the adult, however, Gas1 is also expressed in RPC and interestingly, the expression of RPC markers such as the Neural cell adhesion molecule (NCAM) and Cluster of differentiation 24 (CD24) were found to have an inverse pattern of expression to Gas1 (decreases as the kidney matures) during postnatal renal maturation, this indicates a role for Gas1 in the regulation of renal cell proliferation at this stage of development.
Collapse
Affiliation(s)
- Andrea Cetina-Palma
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Carmen Namorado-Tónix
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Rafael Rodríguez-Muñoz
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Paula Vergara
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - José Luis Reyes-Sánchez
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - José Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| |
Collapse
|
9
|
WhichTF is functionally important in your open chromatin data? PLoS Comput Biol 2022; 18:e1010378. [PMID: 36040971 PMCID: PMC9426921 DOI: 10.1371/journal.pcbi.1010378] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/11/2022] [Indexed: 11/19/2022] Open
Abstract
We present WhichTF, a computational method to identify functionally important transcription factors (TFs) from chromatin accessibility measurements. To rank TFs, WhichTF applies an ontology-guided functional approach to compute novel enrichment by integrating accessibility measurements, high-confidence pre-computed conservation-aware TF binding sites, and putative gene-regulatory models. Comparison with prior sheer abundance-based methods reveals the unique ability of WhichTF to identify context-specific TFs with functional relevance, including NF-κB family members in lymphocytes and GATA factors in cardiac cells. To distinguish the transcriptional regulatory landscape in closely related samples, we apply differential analysis and demonstrate its utility in lymphocyte, mesoderm developmental, and disease cells. We find suggestive, under-characterized TFs, such as RUNX3 in mesoderm development and GLI1 in systemic lupus erythematosus. We also find TFs known for stress response, suggesting routine experimental caveats that warrant careful consideration. WhichTF yields biological insight into known and novel molecular mechanisms of TF-mediated transcriptional regulation in diverse contexts, including human and mouse cell types, cell fate trajectories, and disease-associated cells. Transcription factors (TFs), a class of DNA binding proteins, regulate tissue- and cell-type-specific expression of genes. Identifying the critical TFs in a given cellular context leads to investigating molecular regulatory mechanisms in development, differentiation, and disease. Because there are more than 1,500 human TFs, experimental measurements of genome-wide occupancy across all TFs have been challenging. While computational approaches play pivotal roles, most existing methods rely on statistical enrichment, focusing either on sequence motif similarity recognized by TFs or the similarity of the genomic region of interest with the previously characterized TF occupancy profile. Here we propose WhichTF as an alternative, incorporating curated biomedical knowledge from ontology and integrating it with the high-confidence prediction of conserved TF binding sites in user-provided genomic regions of interest. We develop a new WhichTF score to rank TFs and demonstrate its applicability across human and mouse cell types, cellular differentiation trajectories, and disease-associated cells.
Collapse
|
10
|
Raming R, Cordasic N, Kirchner P, Ekici AB, Fahlbusch FB, Woelfle J, Hilgers KF, Hartner A, Menendez-Castro C. Neonatal nephron loss during active nephrogenesis results in altered expression of renal developmental genes and markers of kidney injury. Physiol Genomics 2021; 53:509-517. [PMID: 34704838 DOI: 10.1152/physiolgenomics.00059.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Preterm neonates are at a high risk for nephron loss under adverse clinical conditions. Renal damage potentially collides with postnatal nephrogenesis. Recent animal studies suggest that nephron loss within this vulnerable phase leads to renal damage later in life. Nephrogenic pathways are commonly reactivated after kidney injury supporting renal regeneration. We hypothesized that nephron loss during nephrogenesis affects renal development, which, in turn, impairs tissue repair after secondary injury. Neonates prior to 36 wk of gestation show an active nephrogenesis. In rats, nephrogenesis is ongoing until day 10 after birth. Mimicking the situation of severe nephron loss during nephrogenesis, male pups were uninephrectomized at day 1 of life (UNXd1). A second group of males was uninephrectomized at postnatal day 14 (UNXd14), after terminated nephrogenesis. Age-matched controls were sham operated. Three days after uninephrectomy transcriptional changes in the right kidney were analyzed by RNA-sequencing, followed by functional pathway analysis. In UNXd1, 1,182 genes were differentially regulated, but only 143 genes showed a regulation both in UNXd1 and UNXd14. The functional groups "renal development" and "kidney injury" were among the most differentially regulated groups and revealed distinctive alterations. Reduced expression of candidate genes concerning renal development (Bmp7, Gdnf, Pdgf-B, Wt1) and injury (nephrin, podocin, Tgf-β1) were detected. The downregulation of Bmp7 and Gdnf persisted until day 28. In UNXd14, Six2 was upregulated and Pax2 was downregulated. We conclude that nephron loss during nephrogenesis affects renal development and induces a specific regulation of genes that might hinder tissue repair after secondary kidney injury.
Collapse
Affiliation(s)
- Roman Raming
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Nada Cordasic
- Department of Nephrology and Hypertension, University Hospital of Erlangen, Erlangen, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, University Hospital of Erlangen, Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, University Hospital of Erlangen, Erlangen, Germany
| | - Fabian B Fahlbusch
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Joachim Woelfle
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Karl F Hilgers
- Department of Nephrology and Hypertension, University Hospital of Erlangen, Erlangen, Germany
| | - Andrea Hartner
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Carlos Menendez-Castro
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
11
|
Chan K, Li X. Current Epigenetic Insights in Kidney Development. Genes (Basel) 2021; 12:genes12081281. [PMID: 34440455 PMCID: PMC8391601 DOI: 10.3390/genes12081281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/07/2021] [Accepted: 08/19/2021] [Indexed: 12/31/2022] Open
Abstract
The kidney is among the best characterized developing tissues, with the genes and signaling pathways that regulate embryonic and adult kidney patterning and development having been extensively identified. It is now widely understood that DNA methylation and histone modification patterns are imprinted during embryonic development and must be maintained in adult cells for appropriate gene transcription and phenotypic stability. A compelling question then is how these epigenetic mechanisms play a role in kidney development. In this review, we describe the major genes and pathways that have been linked to epigenetic mechanisms in kidney development. We also discuss recent applications of single-cell RNA sequencing (scRNA-seq) techniques in the study of kidney development. Additionally, we summarize the techniques of single-cell epigenomics, which can potentially be used to characterize epigenomes at single-cell resolution in embryonic and adult kidneys. The combination of scRNA-seq and single-cell epigenomics will help facilitate the further understanding of early cell lineage specification at the level of epigenetic modifications in embryonic and adult kidney development, which may also be used to investigate epigenetic mechanisms in kidney diseases.
Collapse
Affiliation(s)
- Katrina Chan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
| | - Xiaogang Li
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
- Correspondence: ; Tel.: +1-507-266-0110
| |
Collapse
|
12
|
van der Velde A, Fan K, Tsuji J, Moore JE, Purcaro MJ, Pratt HE, Weng Z. Annotation of chromatin states in 66 complete mouse epigenomes during development. Commun Biol 2021; 4:239. [PMID: 33619351 PMCID: PMC7900196 DOI: 10.1038/s42003-021-01756-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 01/26/2021] [Indexed: 01/31/2023] Open
Abstract
The morphologically and functionally distinct cell types of a multicellular organism are maintained by their unique epigenomes and gene expression programs. Phase III of the ENCODE Project profiled 66 mouse epigenomes across twelve tissues at daily intervals from embryonic day 11.5 to birth. Applying the ChromHMM algorithm to these epigenomes, we annotated eighteen chromatin states with characteristics of promoters, enhancers, transcribed regions, repressed regions, and quiescent regions. Our integrative analyses delineate the tissue specificity and developmental trajectory of the loci in these chromatin states. Approximately 0.3% of each epigenome is assigned to a bivalent chromatin state, which harbors both active marks and the repressive mark H3K27me3. Highly evolutionarily conserved, these loci are enriched in silencers bound by polycomb repressive complex proteins, and the transcription start sites of their silenced target genes. This collection of chromatin state assignments provides a useful resource for studying mammalian development.
Collapse
Affiliation(s)
- Arjan van der Velde
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
- Bioinformatics Program, Boston University, Boston, MA, 02215, USA
| | - Kaili Fan
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Junko Tsuji
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Jill E Moore
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Michael J Purcaro
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Henry E Pratt
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Zhiping Weng
- Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
13
|
He LX, Abdolmaleky HM, Yin S, Wang Y, Zhou JR. Dietary Fermented Soy Extract and Oligo-Lactic Acid Alleviate Chronic Kidney Disease in Mice via Inhibition of Inflammation and Modulation of Gut Microbiota. Nutrients 2020; 12:E2376. [PMID: 32784477 PMCID: PMC7468970 DOI: 10.3390/nu12082376] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/03/2020] [Accepted: 08/03/2020] [Indexed: 01/04/2023] Open
Abstract
Chronic kidney disease (CKD) is a global epidemic with an increasing prevalence worldwide. Effective preventive strategies are urgently needed. This study aimed to investigate the effect of nutraceutical components, a fermented soybean product (ImmuBalance, IMB) and an oligo-lactic acid product (LAP), on the prevention of adenine-induced CKD in mice. Female C57BL/6 mice were randomly assigned into following experimental groups: negative control; model control; and models treated with IMB at 250 or 1000 mg/kg body weight (BW), LAP at 1000 or 2000 mg/kg BW, and IMB/LAP combinations. The CKD model was established by intraperitoneal injection of adenine daily for 4 weeks, and treatments started 2 weeks before adenine injection and ended after 10 weeks. Compared with the model control, the treatments did not significantly alter the body weight or food intake. Both IMB and LAP, especially their combination, significantly inhibited tubular dilation, tubulointerstitial degeneration or atrophy, interstitial chronic inflammation and acute inflammation in the kidneys of CKD mice, and significantly decreased serum cystatin C levels. IMB or LAP significantly reversed CKD-associated increases of circulating and kidney levels of inflammatory cytokines, circulating levels of kidney injury biomarkers, and kidney levels of stem cell biomarkers, and significantly reversed CKD-associated reduction of cecum Clostridium leptum group. Our results suggest that dietary supplementation of IMB or LAP may significantly delay the development and/or progression of CKD.
Collapse
Affiliation(s)
- Li-Xia He
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (L.-X.H.); (H.M.A.); (S.Y.)
- Feihe Nutrition Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hamid M. Abdolmaleky
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (L.-X.H.); (H.M.A.); (S.Y.)
| | - Sheng Yin
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (L.-X.H.); (H.M.A.); (S.Y.)
| | - Yihong Wang
- Department of Pathology and Laboratory of Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA;
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; (L.-X.H.); (H.M.A.); (S.Y.)
| |
Collapse
|
14
|
Sanford E, Wong T, Ellsworth KA, Ingulli E, Kingsmore SF. Clinical utility of ultra-rapid whole-genome sequencing in an infant with atypical presentation of WT1-associated nephrotic syndrome type 4. Cold Spring Harb Mol Case Stud 2020; 6:mcs.a005470. [PMID: 32843431 PMCID: PMC7476414 DOI: 10.1101/mcs.a005470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/10/2020] [Indexed: 12/31/2022] Open
Abstract
Relatively little is known about phenotypic variability in nonsyndromic nephropathy associated with the gene encoding the WT1 transcription factor. We report a 12-mo-old female who presented with vomiting, diarrhea, and fatigue in the setting of renal failure and malignant hypertension. Trio ultra-rapid whole-genome sequencing identified a novel, likely pathogenic, de novo missense variant (c.485T > A, p.Val162Asp) in WT1 in 46 h, consistent with a diagnosis of nephrotic syndrome type 4 (NPHS4; OMIM 256370). This disorder typically presents with nephrotic syndrome (gross proteinuria, hypoalbuminemia, and edema). Rapid diagnosis had an immediate impact on her clinical management in the pediatric intensive care unit. Diagnostic renal biopsy was avoided, and placement of permanent dialysis access, a gastrostomy tube, and bilateral nephrectomy were accelerated. This report expands the presenting phenotype of nonsyndromic nephrotic syndrome and/or renal failure due to heterozygous variants in WT1 (NPHS4). It also highlights the relationship between time to genomic diagnosis and clinical utility in critically ill infants.
Collapse
Affiliation(s)
- Erica Sanford
- Rady Children's Institute of Genomic Medicine, Rady Children's Hospital–San Diego, San Diego, California 92123, USA;,Division of Pediatric Intensive Care Medicine, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | - Terence Wong
- Rady Children's Institute of Genomic Medicine, Rady Children's Hospital–San Diego, San Diego, California 92123, USA
| | - Katarzyna A. Ellsworth
- Rady Children's Institute of Genomic Medicine, Rady Children's Hospital–San Diego, San Diego, California 92123, USA
| | - Elizabeth Ingulli
- Division of Pediatric Nephrology, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, USA
| | - Stephen F. Kingsmore
- Rady Children's Institute of Genomic Medicine, Rady Children's Hospital–San Diego, San Diego, California 92123, USA
| |
Collapse
|
15
|
The FGF, TGFβ and WNT axis Modulate Self-renewal of Human SIX2 + Urine Derived Renal Progenitor Cells. Sci Rep 2020; 10:739. [PMID: 31959818 PMCID: PMC6970988 DOI: 10.1038/s41598-020-57723-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022] Open
Abstract
Human urine is a non-invasive source of renal stem cells with regeneration potential. Urine-derived renal progenitor cells were isolated from 10 individuals of both genders and distinct ages. These renal progenitors express pluripotency-associated proteins- TRA-1-60, TRA-1-81, SSEA4, C-KIT and CD133, as well as the renal stem cell markers -SIX2, CITED1, WT1, CD24 and CD106. The transcriptomes of all SIX2+ renal progenitors clustered together, and distinct from the human kidney biopsy-derived epithelial proximal cells (hREPCs). Stimulation of the urine-derived renal progenitor cells (UdRPCs) with the GSK3β-inhibitor (CHIR99021) induced differentiation. Transcriptome and KEGG pathway analysis revealed upregulation of WNT-associated genes- AXIN2, JUN and NKD1. Protein interaction network identified JUN- a downstream target of the WNT pathway in association with STAT3, ATF2 and MAPK1 as a putative negative regulator of self-renewal. Furthermore, like pluripotent stem cells, self-renewal is maintained by FGF2-driven TGFβ-SMAD2/3 pathway. The urine-derived renal progenitor cells and the data presented should lay the foundation for studying nephrogenesis in human.
Collapse
|
16
|
Oberhammer L, Mitterberger MJ, Lusuardi L, Kunit T, Drerup M, Colleselli D, Griessner H, Hager M. Sporadic renal hemangioblastoma: A case report of a rare benign renal tumor. Clin Case Rep 2019; 7:2321-2326. [PMID: 31893050 PMCID: PMC6935610 DOI: 10.1002/ccr3.2466] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/30/2019] [Accepted: 08/27/2019] [Indexed: 11/25/2022] Open
Abstract
In renal tumors, suspicious for renal cell carcinoma, where there is any doubt and discrepancy between morphology and immune profile, we recommend performing further immunohistochemical staining for pan-cytokeratin, S100, NSE, and inhibin-alpha. Thus, follow-up overtreatment can be avoided in cases of benign kidney tumors.
Collapse
Affiliation(s)
- Lukas Oberhammer
- Department of Urology and AndrologyUniklinikum SalzburgSalzburgAustria
| | | | - Lukas Lusuardi
- Department of Urology and AndrologyUniklinikum SalzburgSalzburgAustria
| | - Thomas Kunit
- Department of Urology and AndrologyUniklinikum SalzburgSalzburgAustria
| | - Martin Drerup
- Department of Urology and AndrologyUniklinikum SalzburgSalzburgAustria
| | | | - Hubert Griessner
- Department of Urology and AndrologyUniklinikum SalzburgSalzburgAustria
| | - Martina Hager
- Department of PathologyUniklinikum SalzburgSalzburgAustria
| |
Collapse
|
17
|
Kang HM, Lim JH, Noh KH, Park D, Cho HS, Susztak K, Jung CR. Effective reconstruction of functional organotypic kidney spheroid for in vitro nephrotoxicity studies. Sci Rep 2019; 9:17610. [PMID: 31772214 PMCID: PMC6879515 DOI: 10.1038/s41598-019-53855-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/01/2019] [Indexed: 01/05/2023] Open
Abstract
Stable and reproducible kidney cellular models could accelerate our understanding of diseases, help therapeutics development, and improve nephrotoxicity screenings. Generation of a reproducible in vitro kidney models has been challenging owing to the cellular heterogeneity and structural complexity of the kidney. We generated mixed immortalized cell lines that stably maintained their characteristic expression of renal epithelial progenitor markers for the different lineages of kidney cellular compartments via the BMP7 signaling pathway from a mouse and a human whole kidney. These cells were used to generate functional and matured kidney spheroids containing multiple renal lineages, such as the proximal tubule, loop of Henle, distal tubules, and podocytes, using extracellular matrix and physiological force, named spheroid-forming unit (SFU). They expressed all apical and basolateral transporters that are important for drug metabolism and displayed key functional aspects of the proximal tubule, including protein endocytosis and increased gamma-glutamyltransferase activity, and cyclic AMP responded to external cues, such as parathyroid hormone. Following exposure, cells fluxed and took up drugs via proximal tubule-specific apical or basolateral transporters, and displayed increased cell death and expression of renal injury marker. Here, we developed a new differentiation method to generate kidney spheroids that structurally recapitulate important features of the kidney effectively and reproducibly using mixed immortalized renal cells, and showed their application for renal toxicity studies.
Collapse
Affiliation(s)
- Hyun Mi Kang
- Laboratory of Disease Modeling and Therapeutics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jung Hwa Lim
- Laboratory of Disease Modeling and Therapeutics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Kyung Hee Noh
- Laboratory of Disease Modeling and Therapeutics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Dongmin Park
- Laboratory of Disease Modeling and Therapeutics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Hyun-Soo Cho
- Stem Cell Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Katalin Susztak
- Division of Nephrology, Department of Medicine, Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Cho-Rok Jung
- Laboratory of Disease Modeling and Therapeutics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea. .,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
18
|
Francipane MG, Han B, Oxburgh L, Sims-Lucas S, Li Z, Lagasse E. Kidney-in-a-lymph node: A novel organogenesis assay to model human renal development and test nephron progenitor cell fates. J Tissue Eng Regen Med 2019; 13:1724-1731. [PMID: 31267702 DOI: 10.1002/term.2924] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/18/2019] [Accepted: 06/30/2019] [Indexed: 12/12/2022]
Abstract
Stem cell-derived organoids are emerging as sophisticated models for studying development and disease and as potential sources for developing organ substitutes. Unfortunately, although organoids containing renal structures have been generated from mouse and human pluripotent stem cells, there are still critical unanswered questions that are difficult to attain via in vitro systems, including whether these nonvascularized organoids have a stable and physiologically relevant phenotype or whether a suitable transplantation site for long-term in vivo studies can be identified. Even orthotopic engraftment of organoid cultures in the adult does not provide an environment conducive to vascularization and functional differentiation. Previously, we showed that the lymph node offers an alternative transplantation site where mouse metanephroi can differentiate into mature renal structures with excretory, homeostatic, and endocrine functions. Here, we show that the lymph node lends itself well as a niche to also grow human primary kidney rudiments and can additionally be viewed as a platform to interrogate emerging renal organoid cultures. Our study has a wide-ranging impact for tissue engineering approaches to rebuild functional tissues in vivo including-but not limited to-the kidney.
Collapse
Affiliation(s)
- Maria Giovanna Francipane
- McGowan Institute for Regenerative Medicine and Pathology Department, University of Pittsburgh, Pittsburgh, Pennsylvania.,Ri.MED Foundation, Palermo, Italy
| | - Bing Han
- McGowan Institute for Regenerative Medicine and Pathology Department, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, Maine
| | - Sunder Sims-Lucas
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania
| | - Zhongwei Li
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, Los Angeles, California, USA
| | - Eric Lagasse
- McGowan Institute for Regenerative Medicine and Pathology Department, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
19
|
Gcm1 is involved in cell proliferation and fibrosis during kidney regeneration after ischemia-reperfusion injury. Sci Rep 2019; 9:7883. [PMID: 31133638 PMCID: PMC6536531 DOI: 10.1038/s41598-019-44161-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 05/10/2019] [Indexed: 12/27/2022] Open
Abstract
In acute kidney injury (AKI), the S3 segment of the proximal tubule is particularly damaged, as it is most vulnerable to ischemia. However, this region is also involved in renal tubular regeneration. To deeply understand the mechanism of the repair process after ischemic injury in AKI, we focused on glial cells missing 1 (Gcm1), which is one of the genes expressed in the S3 segment. Gcm1 is essential for the development of the placenta, and Gcm1 knockout (KO) is embryonically lethal. Thus, the function of Gcm1 in the kidney has not been analyzed yet. We analyzed the function of Gcm1 in the kidney by specifically knocking out Gcm1 in the kidney. We created an ischemia-reperfusion injury (IRI) model to observe the repair process after AKI. We found that Gcm1 expression was transiently increased during the recovery phase of IRI. In Gcm1 conditional KO mice, during the recovery phase of IRI, tubular cell proliferation reduced and transforming growth factor-β1 expression was downregulated resulting in a reduction in fibrosis. In vitro, Gcm1 overexpression promoted cell proliferation and upregulated TGF-β1 expression. These findings indicate that Gcm1 is involved in the mechanisms of fibrosis and cell proliferation after ischemic injury of the kidney.
Collapse
|
20
|
Qian T, Hernday SE, Bao X, Olson WR, Panzer SE, Shusta EV, Palecek SP. Directed Differentiation of Human Pluripotent Stem Cells to Podocytes under Defined Conditions. Sci Rep 2019; 9:2765. [PMID: 30808965 PMCID: PMC6391455 DOI: 10.1038/s41598-019-39504-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 12/10/2018] [Indexed: 01/20/2023] Open
Abstract
A major cause of chronic kidney disease (CKD) is glomerular disease, which can be attributed to a spectrum of podocyte disorders. Podocytes are non-proliferative, terminally differentiated cells. Thus, the limited supply of primary podocytes impedes CKD research. Differentiation of human pluripotent stem cells (hPSCs) into podocytes has the potential to produce podocytes for disease modeling, drug screening, and cell therapies. In the podocyte differentiation process described here, hPSCs are first induced to primitive streak-like cells by activating canonical Wnt signaling. Next, these cells progress to mesoderm precursors, proliferative nephron progenitors, and eventually become mature podocytes by culturing in a serum-free medium. Podocytes generated via this protocol adopt podocyte morphology, express canonical podocyte markers, and exhibit podocyte phenotypes, including albumin uptake and TGF-β1 triggered cell death. This study provides a simple, defined strategy to generate podocytes for in vitro modeling of podocyte development and disease or for cell therapies.
Collapse
Affiliation(s)
- Tongcheng Qian
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Shaenah E Hernday
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Xiaoping Bao
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - William R Olson
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA
| | - Sarah E Panzer
- School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53706, USA
| | - Eric V Shusta
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA.
| | - Sean P Palecek
- Department of Chemical & Biological Engineering, University of Wisconsin, Madison, WI, 53706, USA.
| |
Collapse
|
21
|
Sontake V, Kasam RK, Sinner D, Korfhagen TR, Reddy GB, White ES, Jegga AG, Madala SK. Wilms' tumor 1 drives fibroproliferation and myofibroblast transformation in severe fibrotic lung disease. JCI Insight 2018; 3:121252. [PMID: 30135315 DOI: 10.1172/jci.insight.121252] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 07/11/2018] [Indexed: 12/29/2022] Open
Abstract
Wilms' tumor 1 (WT1) is a critical transcriptional regulator of mesothelial cells during lung development but is downregulated in postnatal stages and adult lungs. We recently showed that WT1 is upregulated in both mesothelial cells and mesenchymal cells in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a fatal fibrotic lung disease. Although WT1-positive cell accumulation leading to severe fibrotic lung disease has been studied, the role of WT1 in fibroblast activation and pulmonary fibrosis remains elusive. Here, we show that WT1 functions as a positive regulator of fibroblast activation, including fibroproliferation, myofibroblast transformation, and extracellular matrix (ECM) production. Chromatin immunoprecipitation experiments indicate that WT1 binds directly to the promoter DNA sequence of α-smooth muscle actin (αSMA) to induce myofibroblast transformation. In support, the genetic lineage tracing identifies WT1 as a key driver of mesothelial-to-myofibroblast and fibroblast-to-myofibroblast transformation. Importantly, the partial loss of WT1 was sufficient to attenuate myofibroblast accumulation and pulmonary fibrosis in vivo. Further, our coculture studies show that WT1 upregulation leads to non-cell autonomous effects on neighboring cells. Thus, our data uncovered a pathogenic role of WT1 in IPF by promoting fibroblast activation in the peripheral areas of the lung and as a target for therapeutic intervention.
Collapse
Affiliation(s)
- Vishwaraj Sontake
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Rajesh K Kasam
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Biochemistry, National Institute of Nutrition, Hyderabad, Telangana, India.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Debora Sinner
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Thomas R Korfhagen
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Geereddy B Reddy
- Department of Biochemistry, National Institute of Nutrition, Hyderabad, Telangana, India
| | - Eric S White
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, Michigan, USA
| | - Anil G Jegga
- Division of Biomedical Informatics Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Satish K Madala
- Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
22
|
Held M, Santeramo I, Wilm B, Murray P, Lévy R. Ex vivo live cell tracking in kidney organoids using light sheet fluorescence microscopy. PLoS One 2018; 13:e0199918. [PMID: 30048451 PMCID: PMC6062017 DOI: 10.1371/journal.pone.0199918] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/16/2018] [Indexed: 12/28/2022] Open
Abstract
Screening cells for their differentiation potential requires a combination of tissue culture models and imaging methods that allow for long-term tracking of the location and function of cells. Embryonic kidney re-aggregation in vitro assays have been established which allow for the monitoring of organotypic cell behaviour in re-aggregated and chimeric renal organoids. However, evaluation of cell integration is hampered by the high photonic load of standard fluorescence microscopy which poses challenges for imaging three-dimensional systems in real-time over a time course. Therefore, we employed light sheet microscopy, a technique that vastly reduces photobleaching and phototoxic effects. We have also developed a new method for culturing the re-aggregates which involves immersed culture, generating organoids which more closely reflect development in vivo. To facilitate imaging from various angles, we embedded the organoids in a freely rotatable hydrogel cylinder. Endpoint fixing and staining were performed to provide additional biomolecular information. We succeeded in imaging labelled cells within re-aggregated kidney organoids over 15 hours and tracking their fate while simultaneously monitoring the development of organotypic morphological structures. Our results show that Wt1-expressing embryonic kidney cells obtained from transgenic mice could integrate into re-aggregated chimeric kidney organoids and contribute to developing nephrons. Furthermore, the nascent proximal tubules that formed in the re-aggregated tissues using the new culture method displayed secretory function, as evidenced by their ability to secrete an organic anion mimic into the tubular lumen.
Collapse
Affiliation(s)
- Marie Held
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Ilaria Santeramo
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Bettina Wilm
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Patricia Murray
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Raphaël Lévy
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
23
|
Liao YJ, Huang RS, Lai WJ, Liu F, Ma L, Xie YS, Salerno S, Li Y, Fu P. Effects of Cyclosporine A on the Development of Metanephros in the Pregnant BALB/c Mice. Chin Med J (Engl) 2018; 130:2156-2162. [PMID: 28875951 PMCID: PMC5598326 DOI: 10.4103/0366-6999.213971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background: Cyclosporine A (CsA) is a commonly used clinical immunosuppressant. However, CsA exposure in rabbits during the gestation period was shown to cause a postnatal decrease in the number of nephrons, with the effects remaining unknown. In this study, we aimed to explore the effects of CsA on metanephros development in the pregnant BALB/c mice. Methods: Pregnant mice were randomly divided into two groups, and CsA (10 mg·kg−1·d−1) was subcutaneously injected from gestation day 10.5 to day 16.5 in the CsA group, whereas a comparable volume of normal saline was given to the control group. All of the mice were sacrificed on gestation day 17.5 and serum CsA concentration was measured. The fetuses were removed and weighed, and their kidneys were prepared for histological assessment and polymerase chain reaction assay. In an in vitro experiment, embryo kidneys of fetal mice on gestation day 12.5 were used, and CsA (10 μmol/L) was added in the culture of the CsA group. The growth pattern of the ureteric bud and nephrons was assessed by lectin staining. Results: No significant differences in the weight of embryo (4.54 ± 1.22 vs. 3.26 ± 1.09 mg) were observed between the CsA and control groups, the thickness of the cortical (510.0 ± 30.3 vs. 350.0 ± 29.7 μm, P < 0.05) and nephrogenic zone (272.5 ± 17.2 vs. 173.3 ± 24.0 μm, P < 0.05), and the number of glomeruli (36.5 ± 0.7 vs. 27.5 ± 2.1, P < 0.05) were reduced in the CsA group when compared to the control group. The cell proliferation of Ki-67 positive index between control and CsA group (307.0 ± 20.0 vs. 219.0 ± 25.0, P < 0.05) in the nephrogenic zone was decreased with the increase of apoptotic cells (17.0 ± 2.0 vs. 159.0 ± 33.0, P < 0.05). The mRNA expression of WT-1, Pax2, and Pax8 was downregulated by CsA treatment. As for the in vitro CsA group, the branch number of the ureteric bud was decreased in the CsA-treated group with the nephrons missing in contrast to control after the incubation for 24 h and 72 h (all P < 0.0001). Conclusion: Treatment of CsA suppressed metanephros development in the pregnant mice; however, the potential action of mechanism needs to be further investigated.
Collapse
Affiliation(s)
- Yu-Jie Liao
- Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Rong-Shuang Huang
- Department of Internal Medicine, Division of Nephrology, West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Wei-Jing Lai
- Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Fang Liu
- Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Liang Ma
- Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yuan-Sheng Xie
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing 100853, China
| | - Stephen Salerno
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Yi Li
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Ping Fu
- Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041; Department of Internal Medicine, Division of Nephrology, West China School of Medicine, Sichuan University, Chengdu 610041, China
| |
Collapse
|
24
|
Chou CW, Lin J, Jiang YJ, Liu YW. Aberrant Global and Jagged-Mediated Notch Signaling Disrupts Segregation Between wt1-Expressing and Steroidogenic Tissues in Zebrafish. Endocrinology 2017; 158:4206-4217. [PMID: 29029162 DOI: 10.1210/en.2017-00548] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/26/2017] [Indexed: 11/19/2022]
Abstract
Although the zebrafish interrenal tissue has been used as a model for steroidogenesis and genesis of the adrenal gland, its specification and morphogenesis remains largely unclear. In the present study, we explored how the Wilms tumor 1 (WT1)-expressing cells are segregated from the SF-1-expressing steroidogenic cells in the zebrafish model. The interrenal tissue precursors expressing ff1b, the equivalent of mammalian SF-1, were derived from wt1-expressing pronephric primordia in the zebrafish embryo. Through histochemistry and in situ hybridization, we demonstrated that the size of functionally differentiated interrenal tissue was substantially increased on global inhibition of the Notch signaling pathway and was accompanied by a disrupted segregation between the wt1- and ff1b-expressing cells. As the Notch pathway was conditionally activated during interrenal specification, differentiation, but not ff1b expression, of interrenal tissue was drastically compromised. In embryos deficient for Notch ligands jagged 1b and 2b, transgenic reporter activity of wt1b promoter was detected within the steroidogenic interrenal tissue. In conclusion, our results indicate that Jagged-Notch signaling is required (1) for segregation between wt1-expressing cells and differentiated steroidogenic tissue; and (2) to modulate the extent of functional differentiation in the steroidogenic interrenal tissue.
Collapse
Affiliation(s)
- Chih-Wei Chou
- Department of Life Science, Tunghai University, Taiwan
| | - Jamie Lin
- Department of Life Science, Tunghai University, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Taiwan
| | - Yun-Jin Jiang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Taiwan
| | - Yi-Wen Liu
- Department of Life Science, Tunghai University, Taiwan
| |
Collapse
|
25
|
Kumar AS, Srilakshmi R, Karthickeyan S, Balakrishnan K, Padmaraj R, Senguttuvan P. Wilms' tumour 1 gene mutations in south Indian children with steroid-resistant nephrotic syndrome. Indian J Med Res 2017; 144:276-280. [PMID: 27934809 PMCID: PMC5206881 DOI: 10.4103/0971-5916.195044] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background & objectives: Clinically, nephrotic syndrome (NS) is a diverse group of symptoms; about 20 per cent of NS cases are resistant to steroid treatment, and within ten years they progress to end-stage renal disease. The present study was undertaken to identify the mutations of Wilms’ tumour 1 (WT1) gene in steroid-resistant NS (SRNS) children. Methods: A total of 173 children with SRNS and 100 children in the control group were enrolled in the study. DNA extraction was done, screened for WT1 (exons 8 and 9) gene amplified by polymerase chain reaction and direct sequencing. Karyotype analyses were done for WT1 mutation cases. Results: WT1 mutations were found in three of 173 SRNS cases (2 girls, 1 boy). All of them had intron 9 (IVS 9 + 4 C>T, 2; IVS + 5 G>A, 1) mutation. Of these three cases, one had familial and another two had sporadic history. Renal histology analysis showed two cases with focal segmental glomerulosclerosis (FSGS) and they had external female genitalia but 46, XY karyotype. Both of them had streak gonads. Of the three cases, one expired. Interpretation & conclusions: The findings of the present study indicate that all females with SRNS-FSGS should be screened for WT1 gene mutation to diagnose whether they have FS for possible gonadectomy.
Collapse
Affiliation(s)
- Aravind Selvin Kumar
- Department of Paediatric Nephrology, Institute of Child Health and Hospital for Children, Madras Medical College, Chennai, India
| | - R Srilakshmi
- Department of Medical Genetics, Tamil Nadu Dr. M.G.R. Medical University, Guindy, India
| | - Smk Karthickeyan
- Department of Animal Genetics and Breeding, Madras Veterinary College, Chennai, India
| | - K Balakrishnan
- Department of Immunology, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - R Padmaraj
- Department of Paediatric Nephrology, Institute of Child Health and Hospital for Children, Madras Medical College, Chennai, India
| | - Prabha Senguttuvan
- Department of Paediatric Nephrology, Institute of Child Health and Hospital for Children, Madras Medical College, Chennai, India
| |
Collapse
|
26
|
Mata-Miranda MM, Vazquez-Zapien GJ, Rojas-Lopez M, Sanchez-Monroy V, Perez-Ishiwara DG, Delgado-Macuil RJ. Morphological, molecular and FTIR spectroscopic analysis during the differentiation of kidney cells from pluripotent stem cells. Biol Res 2017; 50:14. [PMID: 28376862 PMCID: PMC5379680 DOI: 10.1186/s40659-017-0119-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/28/2017] [Indexed: 12/23/2022] Open
Abstract
Background Kidney diseases are a global health problem. Currently, over 2 million people require dialysis or transplant which are associated with high morbidity and mortality; therefore, new researches focused on regenerative medicine have been developed, including the use of stem cells. Results In this research, we generate differentiated kidney cells (DKCs) from mouse pluripotent stem cells (mPSCs) analyzing their morphological, genetic, phenotypic, and spectroscopic characteristics along differentiation, highlighting that there are no reports of the use of Fourier transform infrared (FTIR) spectroscopy to characterize the directed differentiation of mPSCs to DKCs. The genetic and protein experiments proved the obtention of DKCs that passed through the chronological stages of embryonic kidney development. Regarding vibrational spectroscopy analysis by FTIR, bands related with biomolecules were shown on mPSCs and DKCs spectra, observing distinct differences between cell lineages and maturation stages. The second derivative of DKCs spectra showed changes in the protein bands compared to mPSCs. Finally, the principal components analysis obtained from FTIR spectra allowed to characterize chemical and structurally mPSCs and their differentiation process to DKCs in a rapid and non-invasive way. Conclusion Our results indicated that we obtained DKCs from mPSCs, which passed through the chronological stages of embryonic kidney development. Moreover, FTIR spectroscopy resulted in a non-invasive, rapid and precise technic that together with principal component analysis allows to characterize chemical and structurally both kind of cells and also discriminate and determine different stages along the cell differentiation process.
Collapse
Affiliation(s)
- Monica Maribel Mata-Miranda
- Centro de Investigación en Biotecnología Aplicada, CIBA-Tlaxcala, Instituto Politécnico Nacional, 90700, Tepetitla, Tlaxcala, Mexico.,Laboratorio de Biología Celular y Tisular, Escuela Médico Militar, Centro Militar de Ciencias de la Salud, Secretaría de la Defensa Nacional, 11200, Mexico City, Mexico
| | - Gustavo Jesus Vazquez-Zapien
- Centro de Investigación en Biotecnología Aplicada, CIBA-Tlaxcala, Instituto Politécnico Nacional, 90700, Tepetitla, Tlaxcala, Mexico.,Laboratorio de Embriología, Escuela Médico Militar, Centro Militar de Ciencias de la Salud, Secretaría de la Defensa Nacional, 11200, Mexico City, Mexico
| | - Marlon Rojas-Lopez
- Centro de Investigación en Biotecnología Aplicada, CIBA-Tlaxcala, Instituto Politécnico Nacional, 90700, Tepetitla, Tlaxcala, Mexico
| | - Virginia Sanchez-Monroy
- Escuela Nacional de Medicina y Homeopatía, Instituto Politécnico Nacional, 07320, Mexico City, Mexico
| | | | - Raul Jacobo Delgado-Macuil
- Centro de Investigación en Biotecnología Aplicada, CIBA-Tlaxcala, Instituto Politécnico Nacional, 90700, Tepetitla, Tlaxcala, Mexico.
| |
Collapse
|
27
|
Chuah JKC, Zink D. Stem cell-derived kidney cells and organoids: Recent breakthroughs and emerging applications. Biotechnol Adv 2016; 35:150-167. [PMID: 28017905 DOI: 10.1016/j.biotechadv.2016.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/12/2016] [Accepted: 12/17/2016] [Indexed: 02/09/2023]
Abstract
The global rise in the numbers of kidney patients and the shortage in transplantable organs have led to an increasing interest in kidney-specific regenerative therapies, renal disease modelling and bioartificial kidneys. Sources for large quantities of high-quality renal cells and tissues would be required, also for applications in in vitro platforms for compound safety and efficacy screening. Stem cell-based approaches for the generation of renal-like cells and tissues would be most attractive, but such methods were not available until recently. This situation has drastically changed since 2013, and various protocols for the generation of renal-like cells and precursors from pluripotent stem cells (PSC) have been established. The most recent breakthroughs were related to the establishment of various protocols for the generation of PSC-derived kidney organoids. In combination with recent advances in genome editing, bioprinting and the establishment of predictive renal screening platforms this results in exciting new possibilities. This review will give a comprehensive overview over current PSC-based protocols for the generation of renal-like cells, precursors and organoids, and their current and potential applications in regenerative medicine, compound screening, disease modelling and bioartificial organs.
Collapse
Affiliation(s)
- Jacqueline Kai Chin Chuah
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| | - Daniele Zink
- Institute of Bioengineering and Nanotechnology, Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, The Nanos, Singapore 138669, Singapore.
| |
Collapse
|
28
|
Nishimura Y, Hsu HH, Wang PC. Detection of initial angiogenesis from dorsal aorta into metanephroi and elucidation of its role in kidney development. Regen Ther 2016; 4:27-35. [PMID: 31245485 PMCID: PMC6581801 DOI: 10.1016/j.reth.2016.01.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/13/2015] [Accepted: 01/06/2016] [Indexed: 12/13/2022] Open
Abstract
Reconstruction of blood vessels is considered the most difficult part for the complicated organs, therefore, blood vessel construction is regarded as a key point for kidney regeneration in vitro. Vasculogenesis and angiogenesis are the two mechanisms to form blood vessels in embryonic organs, and most studies resided in vaculogenesis. Angiogenesis resided mostly in adult diseases such as wound healing, growth of tumors, and psoriasis diseases. However, renal angiogenesis is simply attributed to the sprouting of pre-existing blood vessel from dorsal aorta into metanephroi, and its occurrence is considered to be at a late stage of metanephric development. Since no techniques are available for delicate detection, the initial angiogenesis from dorsal aorta into metanephroi as well as its role in kidney development still remained unclear. In this study, we developed a method to detect the initial angiogenesis of dorsal aorta into metanephroi, and firstly clarified that dorsal aorta angiogenesis occurred at an early stage of metanephric development. We also elucidated the role of dorsal aorta angiogenesis in promoting the early blood vessel formation, tubule formation and glomeruli maturation. It is suggested that blood flow and dynamic circulation of various factors at the early developing stage may be prerequisite to a successful construction of blood vessels in the complicated organs either in vitro or in vivo. These findings contribute to a better understanding of dorsal aorta angiogenesis during kidney development and shed light on its significant value for the application of tissue engineering to complicated organs.
Collapse
Affiliation(s)
| | | | - Pi-Chao Wang
- Division of Bioindustrial Sciences, Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba City, Ibaraki 305-8572, Japan
| |
Collapse
|
29
|
Shukuya K, Ogura S, Tokuhara Y, Okubo S, Yatomi Y, Tozuka M, Shimosawa T. Novel round cells in urine sediment and their clinical implications. Clin Chim Acta 2016; 457:142-9. [PMID: 27101813 DOI: 10.1016/j.cca.2016.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 04/14/2016] [Accepted: 04/16/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND Voided urine contains a variety of cells from the kidney and urinary tract and urinalysis provides us various information by investigating cellular components. We investigated urine sediment from renal impaired patients. RESULTS We found 'round cell' to be distinct from known cells in sediment and is close to proximal convoluted tubule-derived cells based on morphology and molecular marker expression (GGT1 but not podocalyxin). Also it was positive for undifferentiated cell markers, including PAX2, WT1, OSR1, and SIX2. They were observed in end-stage renal failure patients and the number of cells was correlated with the severity of chronic kidney disease. A prospective analysis revealed that patients who had more round cells were more likely to require hemodialysis within a year. CONCLUSION Round cells are a novel marker that can be used to predict the need for hemodialysis.
Collapse
Affiliation(s)
- Kenichi Shukuya
- Department of Clinical Laboratory, School of Medicine, The University of Tokyo, Tokyo, Japan; Analytical Laboratory Chemistry, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sayoko Ogura
- Department of Clinical Laboratory, School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Pathology and Microbiology, Division of Laboratory Medicine, Nihon University School of Medicine, Tokyo, Japan; Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Japan
| | - Yasunori Tokuhara
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, Kagawa, Japan
| | - Shigeo Okubo
- Department of Clinical Laboratory, School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory, School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Minoru Tozuka
- Analytical Laboratory Chemistry, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, School of Medicine, The University of Tokyo, Tokyo, Japan; Japan Science and Technology Agency, CREST JST, Tokyo, Japan.
| |
Collapse
|
30
|
Arora H, Boulberdaa M, Qureshi R, Bitirim V, Messadeq N, Dolle P, Nebigil CG. Prokineticin receptor 1 is required for mesenchymal-epithelial transition in kidney development. FASEB J 2016; 30:2733-40. [PMID: 27084889 DOI: 10.1096/fj.201600181r] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/05/2016] [Indexed: 12/23/2022]
Abstract
Identification of factors regulating renal development is important to understand the pathogenesis of congenital kidney diseases. Little is known about the molecular mechanism of renal development and functions triggered by the angiogenic hormone prokineticin-2 and its receptor, PKR1. Utilizing the Gata5 (G5)-Cre and Wilms tumor 1 (Wt1)(GFP)cre transgenic lines, we generated mutant mice with targeted PKR1 gene disruptions in nephron progenitors. These mutant mice exhibited partial embryonic and postnatal lethality. Kidney developmental defects in PKR(G5-/-) mice are manifested in the adult stage as renal atrophy with glomerular defects, nephropathy, and uremia. PKR1(Wt1-/-) embryos exhibit hypoplastic kidneys with premature glomeruli and necrotic nephrons as a result of impaired proliferation and increased apoptosis in Wt1(+) renal mesenchymal cells. PKR1 regulates renal mesenchymal-epithelial transition (MET) that is involved in formation of renal progenitors, regulating glomerulogenesis toward forming nephrons during kidney development. In the isolated embryonic Wt1(+) renal cells, overexpression or activation of PKR1 promotes MET defined by the transition from elongated cell to octagonal cell morphology, and alteration of the expression of MET markers via activating NFATc3 signaling. Together, these results establish PKR1 via NFATc3 as a crucial modifier of MET processing to the development of nephron. Our study should facilitate new therapeutic opportunities in human renal disorders.-Arora, H., Boulberdaa, M., Qureshi, R., Bitirim, V., Messadeq, N., Dolle, P., Nebigil, C. G. Prokineticin receptor 1 is required for mesenchymal-epithelial transition in kidney development.
Collapse
Affiliation(s)
- Himanshu Arora
- Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Unité Mixte de Recherche (UMR) 7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France; and
| | - Mounia Boulberdaa
- Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Unité Mixte de Recherche (UMR) 7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France; and
| | - Rehana Qureshi
- Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Unité Mixte de Recherche (UMR) 7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France; and
| | - Verda Bitirim
- Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Unité Mixte de Recherche (UMR) 7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France; and
| | - Nadia Messadeq
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, UMR 7104 and INSERM Unité 964, Université de Strasbourg, Illkirch-Strasbourg, France
| | - Pascal Dolle
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS, UMR 7104 and INSERM Unité 964, Université de Strasbourg, Illkirch-Strasbourg, France
| | - Canan G Nebigil
- Centre National de la Recherche Scientifique (CNRS), Université de Strasbourg, Unité Mixte de Recherche (UMR) 7242, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France; and
| |
Collapse
|
31
|
Abstract
Wilms' tumor-1 protein (WT1) is a transcription factor that can either activate or repress genes to regulate cell growth, apoptosis and differentiation. WT1 can act as either a tumor suppressor or an oncogene. The cellular functions of WT1 are predominantly regulated by its various interacting partners. Recently we have found that WT1 can regulate the fidelity of chromosome segregation through its interaction with the spindle assembly checkpoint protein, Mitotic arrest deficient-2 (MAD2). WT1 delays anaphase entry by inhibiting the ubiquitination activity of the Anaphase promoting complex/cyclosome (APC/C). Our findings have revealed an important role of WT1 in the regulation of mitotic checkpoint and genomic stability.
Collapse
Affiliation(s)
- Jayasha Shandilya
- a Department of Biological Sciences ; University at Buffalo ; Buffalo , NY USA
| | | |
Collapse
|
32
|
Abstract
Recent investigations have highlighted the importance of the non-coding genome in regions of hypoxia in tumours. Such regions are frequently found in solid tumours, and are associated with worse patient survival and therapy resistance. Hypoxia stabilises the transcription factors, hypoxia inducible factors (HIF1α and HIF2α) which coordinate transcriptomic changes that occur in hypoxia. The changes in gene expression induced by HIF1α and HIF2α contribute to many of the hallmarks of cancer phenotypes and enable tumour growth, survival and invasion in the hypoxic tumour microenvironment. Non-coding RNAs, in particular microRNAs (miRNAs), which regulate mRNA stability and translation, and long-non-coding RNAs (lncRNAs), which have diverse functions including chromatin modification and transcriptional regulation, are also important in enabling the key hypoxia regulated processes. They have roles in the regulation of metabolism, angiogenesis, autophagy, invasion and metastasis in the hypoxic microenvironment. Furthermore, HIF1α and HIF2α expression and stabilisation are also regulated by both miRNAs and lncRNAs. Here we review the recent developments in the expression, regulation and functions of miRNAs, lncRNAs and other non-coding RNA classes in tumour hypoxia.
Collapse
Affiliation(s)
- Hani Choudhry
- Department of Biochemistry, Faculty of Science, Center of Innovation in Personalized Medicine, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adrian L Harris
- Molecular Oncology Laboratories, Department of Oncology, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Alan McIntyre
- Cancer Biology, Division of Cancer and Stem Cells, QMC, University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
33
|
Mari C, Winyard P. Concise Review: Understanding the Renal Progenitor Cell Niche In Vivo to Recapitulate Nephrogenesis In Vitro. Stem Cells Transl Med 2015; 4:1463-71. [PMID: 26494782 DOI: 10.5966/sctm.2015-0104] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/31/2015] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Chronic kidney disease (CKD), defined as progressive kidney damage and a reduction of the glomerular filtration rate, can progress to end-stage renal failure (CKD5), in which kidney function is completely lost. CKD5 requires dialysis or kidney transplantation, which is limited by the shortage of donor organs. The incidence of CKD5 is increasing annually in the Western world, stimulating an urgent need for new therapies to repair injured kidneys. Many efforts are directed toward regenerative medicine, in particular using stem cells to replace nephrons lost during progression to CKD5. In the present review, we provide an overview of the native nephrogenic niche, describing the complex signals that allow survival and maintenance of undifferentiated renal stem/progenitor cells and the stimuli that promote differentiation. Recapitulating in vitro what normally happens in vivo will be beneficial to guide amplification and direct differentiation of stem cells toward functional renal cells for nephron regeneration. SIGNIFICANCE Kidneys perform a plethora of functions essential for life. When their main effector, the nephron, is irreversibly compromised, the only therapeutic choices available are artificial replacement (dialysis) or renal transplantation. Research focusing on alternative treatments includes the use of stem cells. These are immature cells with the potential to mature into renal cells, which could be used to regenerate the kidney. To achieve this aim, many problems must be overcome, such as where to take these cells from, how to obtain enough cells to deliver to patients, and, finally, how to mature stem cells into the cell types normally present in the kidney. In the present report, these questions are discussed. By knowing the factors directing the proliferation and differentiation of renal stem cells normally present in developing kidney, this knowledge can applied to other types of stem cells in the laboratory and use them in the clinic as therapy for the kidney.
Collapse
Affiliation(s)
- Chiara Mari
- Developmental Biology and Cancer, Institute of Child Health, University College London, London, United Kingdom
| | - Paul Winyard
- Developmental Biology and Cancer, Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
34
|
Cabral de Almeida Cardoso L, Rodriguez-Laguna L, del Carmen Crespo M, Vallespín E, Palomares-Bralo M, Martin-Arenas R, Rueda-Arenas I, Silvestre de Faria PA, GT-CSGP Working Group, García-Miguel P, Lapunzina P, Regla Vargas F, Seuanez HN, Martínez-Glez V. Array CGH Analysis of Paired Blood and Tumor Samples from Patients with Sporadic Wilms Tumor. PLoS One 2015; 10:e0136812. [PMID: 26317783 PMCID: PMC4552764 DOI: 10.1371/journal.pone.0136812] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/07/2015] [Indexed: 11/24/2022] Open
Abstract
Wilms tumor (WT), the most common cancer of the kidney in infants and children, has a complex etiology that is still poorly understood. Identification of genomic copy number variants (CNV) in tumor genomes provides a better understanding of cancer development which may be useful for diagnosis and therapeutic targets. In paired blood and tumor DNA samples from 14 patients with sporadic WT, analyzed by aCGH, 22% of chromosome abnormalities were novel. All constitutional alterations identified in blood were segmental (in 28.6% of patients) and were also present in the paired tumor samples. Two segmental gains (2p21 and 20q13.3) and one loss (19q13.31) present in blood had not been previously described in WT. We also describe, for the first time, a small, constitutive partial gain of 3p22.1 comprising 2 exons of CTNNB1, a gene associated to WT. Among somatic alterations, novel structural chromosomal abnormalities were found, like gain of 19p13.3 and 20p12.3, and losses of 2p16.1-p15, 4q32.5-q35.1, 4q35.2-q28.1 and 19p13.3. Candidate genes included in these regions might be constitutively (SIX3, SALL4) or somatically (NEK1, PIAS4, BMP2) operational in the development and progression of WT. To our knowledge this is the first report of CNV in paired blood and tumor samples in sporadic WT.
Collapse
Affiliation(s)
| | - Lara Rodriguez-Laguna
- Section of Functional and Structural Genomics, Institute of Medical and Molecular Genetics (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - María del Carmen Crespo
- Section of Functional and Structural Genomics, Institute of Medical and Molecular Genetics (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Elena Vallespín
- Section of Functional and Structural Genomics, Institute of Medical and Molecular Genetics (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - María Palomares-Bralo
- Section of Functional and Structural Genomics, Institute of Medical and Molecular Genetics (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| | - Rubén Martin-Arenas
- Section of Functional and Structural Genomics, Institute of Medical and Molecular Genetics (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Inmaculada Rueda-Arenas
- Section of Functional and Structural Genomics, Institute of Medical and Molecular Genetics (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | | | | | | | - Pablo Lapunzina
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- Section of Clinical Genetics, Institute of Medical and Molecular Genetics (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
| | - Fernando Regla Vargas
- Genetics Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Birth Defects Epidemiology Laboratory, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Hector N. Seuanez
- Genetics Division, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
- Genetics Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Víctor Martínez-Glez
- Section of Functional and Structural Genomics, Institute of Medical and Molecular Genetics (INGEMM)-IdiPAZ, Hospital Universitario La Paz, Madrid, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
- * E-mail:
| |
Collapse
|
35
|
Monteiro Carvalho Mori da Cunha MG, Zia S, Oliveira Arcolino F, Carlon MS, Beckmann DV, Pippi NL, Luhers Graça D, Levtchenko E, Deprest J, Toelen J. Amniotic Fluid Derived Stem Cells with a Renal Progenitor Phenotype Inhibit Interstitial Fibrosis in Renal Ischemia and Reperfusion Injury in Rats. PLoS One 2015; 10:e0136145. [PMID: 26295710 PMCID: PMC4546614 DOI: 10.1371/journal.pone.0136145] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/31/2015] [Indexed: 12/19/2022] Open
Abstract
Objectives Mesenchymal stem cells derived from human amniotic fluid (hAFSCs) are a promising source for cellular therapy, especially for renal disorders, as a subpopulation is derived from the fetal urinary tract. The purpose of this study was to evaluate if hAFSCs with a renal progenitor phenotype demonstrate a nephroprotective effect in acute ischemia reperfusion (I/R) model and prevent late stage fibrosis. Methods A total of 45 male 12-wk-old Wistar rats were divided into three equal groups;: rats subjected to I/R injury and treated with Chang Medium, rats subjected to I/R injury and treated with hAFSCs and sham-operated animals. In the first part of this study, hAFSCs that highly expressed CD24, CD117, SIX2 and PAX2 were isolated and characterized. In the second part, renal I/R injury was induced in male rats and cellular treatment was performed 6 hours later via arterial injection. Functional and histological analyses were performed 24 hours, 48 hours and 2 months after treatment using serum creatinine, urine protein to creatinine ratio, inflammatory and regeneration markers and histomorphometric analysis of the kidney. Statistical analysis was performed by analysis of variance followed by the Tukey’s test for multiple comparisons or by nonparametric Kruskal-Wallis followed by Dunn. Statistical significance level was defined as p <0.05. Results hAFSCs treatment resulted in significantly reduced serum creatinine level at 24 hours, less tubular necrosis, less hyaline cast formation, higher proliferation index, less inflammatory cell infiltration and less myofibroblasts at 48h. The treated group had less fibrosis and proteinuria at 2 months after injury. Conclusion hAFSCs contain a renal progenitor cell subpopulation that has a nephroprotective effect when delivered intra-arterially in rats with renal I/R injury, and reduces interstitial fibrosis on long term follow-up.
Collapse
Affiliation(s)
- Marina Gabriela Monteiro Carvalho Mori da Cunha
- Department of Development and Regeneration, Organ System Cluster, Fetal therapy group, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
- Experimental Veterinary Surgery Laboratory, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Silvia Zia
- Department of Development and Regeneration, Organ System Cluster, Fetal therapy group, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Fanny Oliveira Arcolino
- Department of Development and Regeneration, Organ System Cluster, Laboratory of Pediatric Nephrology, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Marianne Sylvia Carlon
- Department of Pharmaceutical and Pharmacological Sciences, Laboratory of Molecular Virology and Gene Therapy, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
| | - Diego Vilibaldo Beckmann
- Experimental Veterinary Surgery Laboratory, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Ney Luis Pippi
- Experimental Veterinary Surgery Laboratory, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Dominguita Luhers Graça
- Experimental Veterinary Surgery Laboratory, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Elena Levtchenko
- Department of Development and Regeneration, Organ System Cluster, Laboratory of Pediatric Nephrology, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
| | - Jan Deprest
- Department of Development and Regeneration, Organ System Cluster, Fetal therapy group, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynecology, University Hospitals Leuven, Leuven, Belgium
| | - Jaan Toelen
- Department of Development and Regeneration, Organ System Cluster, Fetal therapy group, Group Biomedical Sciences, KU Leuven, Leuven, Belgium
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- * E-mail:
| |
Collapse
|
36
|
Prediction of drug-induced nephrotoxicity and injury mechanisms with human induced pluripotent stem cell-derived cells and machine learning methods. Sci Rep 2015. [PMID: 26212763 PMCID: PMC4515747 DOI: 10.1038/srep12337] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The renal proximal tubule is a main target for drug-induced toxicity. The prediction of proximal tubular toxicity during drug development remains difficult. Any in vitro methods based on induced pluripotent stem cell-derived renal cells had not been developed, so far. Here, we developed a rapid 1-step protocol for the differentiation of human induced pluripotent stem cells (hiPSC) into proximal tubular-like cells. These proximal tubular-like cells had a purity of >90% after 8 days of differentiation and could be directly applied for compound screening. The nephrotoxicity prediction performance of the cells was determined by evaluating their responses to 30 compounds. The results were automatically determined using a machine learning algorithm called random forest. In this way, proximal tubular toxicity in humans could be predicted with 99.8% training accuracy and 87.0% test accuracy. Further, we studied the underlying mechanisms of injury and drug-induced cellular pathways in these hiPSC-derived renal cells, and the results were in agreement with human and animal data. Our methods will enable the development of personalized or disease-specific hiPSC-based renal in vitro models for compound screening and nephrotoxicity prediction.
Collapse
|
37
|
Ambu R, Vinci L, Gerosa C, Fanni D, Obinu E, Faa A, Fanos V. WT1 expression in the human fetus during development. Eur J Histochem 2015; 59:2499. [PMID: 26150159 PMCID: PMC4503972 DOI: 10.4081/ejh.2015.2499] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/10/2015] [Accepted: 04/14/2015] [Indexed: 12/12/2022] Open
Abstract
Wilms’ Tumor 1 (WT1) is a transcription factor involved in the development of the urogenital system. The purpose of this study was to analyze the immunoreactivity for WT1 protein in different tissues and organs in human fetuses in early phases of gestation. To this end, samples from multiple organs were obtained from 4 human fetuses, ranging from 7 up to 12 weeks of gestation. Each sample was formalin-fixed, paraffin embedded and immunostained for WT1. Our data show that WT1 is involved in development of multiple human organs in a more vast series of cells types than previously reported. Immunostaining for WT1 was characterized by a predominant cytoplasmic reactivity in the vast majority of cell types. Mesenchimal progenitors in the fetal lung, ductal plate progenitors in fetal liver, cap mesenchimal cells in the developing kidney, fetal zone cells in adrenal glands, atrial and ventricular cardiomyocytes in the fetal heart, radial glial cells in the fetal cerebral cortex and skeletal muscle cell precursors showed the highest levels of WT1 immunoreactivity. Future studies will be needed to detect differences in the expression of WT1 in various organs at different gestational ages, in order to better evaluate the role of WT1 in cell proliferation and differentiation during intrauterine human development.
Collapse
|
38
|
Lindström NO, Lawrence ML, Burn SF, Johansson JA, Bakker ERM, Ridgway RA, Chang CH, Karolak MJ, Oxburgh L, Headon DJ, Sansom OJ, Smits R, Davies JA, Hohenstein P. Integrated β-catenin, BMP, PTEN, and Notch signalling patterns the nephron. eLife 2015; 3:e04000. [PMID: 25647637 PMCID: PMC4337611 DOI: 10.7554/elife.04000] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 12/28/2014] [Indexed: 12/13/2022] Open
Abstract
The different segments of the nephron and glomerulus in the kidney balance the processes of water homeostasis, solute recovery, blood filtration, and metabolite excretion. When segment function is disrupted, a range of pathological features are presented. Little is known about nephron patterning during embryogenesis. In this study, we demonstrate that the early nephron is patterned by a gradient in β-catenin activity along the axis of the nephron tubule. By modifying β-catenin activity, we force cells within nephrons to differentiate according to the imposed β-catenin activity level, thereby causing spatial shifts in nephron segments. The β-catenin signalling gradient interacts with the BMP pathway which, through PTEN/PI3K/AKT signalling, antagonises β-catenin activity and promotes segment identities associated with low β-catenin activity. β-catenin activity and PI3K signalling also integrate with Notch signalling to control segmentation: modulating β-catenin activity or PI3K rescues segment identities normally lost by inhibition of Notch. Our data therefore identifies a molecular network for nephron patterning.
Collapse
Affiliation(s)
- Nils O Lindström
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| | - Melanie L Lawrence
- Centre for Integrated Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Sally F Burn
- Department of Genetics and Development, Columbia University, New York, United States
| | - Jeanette A Johansson
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
| | - Elvira RM Bakker
- Laboratory of Gastroenterology and Hepatology, Erasmus MC, University Medical Centre, Rotterdam, Netherlands
| | - Rachel A Ridgway
- Department of Invasion and Metastasis, Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - C-Hong Chang
- Centre for Integrated Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Michele J Karolak
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, United States
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, United States
| | - Denis J Headon
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
| | - Owen J Sansom
- Beatston Institute for Cancer Research, Glasgow, United Kingdom
| | - Ron Smits
- Laboratory of Gastroenterology and Hepatology, Erasmus MC, University Medical Centre, Rotterdam, Netherlands
| | - Jamie A Davies
- Centre for Integrated Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter Hohenstein
- Division of Developmental Biology, The Roslin Institute, University of Edinburgh, Easter Bush, United Kingdom
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| |
Collapse
|
39
|
Shandilya J, Toska E, Richard DJ, Medler KF, Roberts SGE. WT1 interacts with MAD2 and regulates mitotic checkpoint function. Nat Commun 2014; 5:4903. [PMID: 25232865 PMCID: PMC4170573 DOI: 10.1038/ncomms5903] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 08/02/2014] [Indexed: 01/08/2023] Open
Abstract
Tumour suppressors safeguard the fidelity of the mitotic checkpoint by transcriptional regulation of genes that encode components of the mitotic checkpoint complex (MCC). Here we report a new role for the tumour suppressor and transcription factor, WT1, in the mitotic checkpoint. We show that WT1 regulates the MCC by directly interacting with the spindle assembly checkpoint protein, MAD2. WT1 colocalizes with MAD2 during mitosis and preferentially binds to the functionally active, closed-conformer, C-MAD2. Furthermore, WT1 associates with the MCC containing MAD2, BUBR1 and CDC20, resulting in prolonged inhibition of the anaphase-promoting complex/cyclosome (APC/C) and delayed degradation of its substrates SECURIN and CYCLIN B1. Strikingly, RNA interference-mediated depletion of WT1 leads to enhanced turnover of SECURIN, decreased lag time to anaphase and defects in chromosome segregation. Our findings identify WT1 as a regulator of the mitotic checkpoint and chromosomal stability.
Collapse
Affiliation(s)
- Jayasha Shandilya
- Department of Biological Sciences, University at Buffalo, Cooke Hall, North Campus, Buffalo, New York 14260, USA
| | - Eneda Toska
- Department of Biological Sciences, University at Buffalo, Cooke Hall, North Campus, Buffalo, New York 14260, USA
| | - Derek J Richard
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, Queensland 4102, Australia
| | - Kathryn F Medler
- Department of Biological Sciences, University at Buffalo, Cooke Hall, North Campus, Buffalo, New York 14260, USA
| | - Stefan G E Roberts
- 1] Department of Biological Sciences, University at Buffalo, Cooke Hall, North Campus, Buffalo, New York 14260, USA [2] School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
40
|
Kopan R, Chen S, Little M. Nephron progenitor cells: shifting the balance of self-renewal and differentiation. Curr Top Dev Biol 2014; 107:293-331. [PMID: 24439811 DOI: 10.1016/b978-0-12-416022-4.00011-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Within the developing mammalian kidney, several populations of progenitors form the discrete cellular components of the final organ. Fate mapping experiments revealed the cap mesenchyme (CM) to be the progenitor population for all nephron epithelial cells, whereas the neighboring stromal mesenchyme gives rise to mesangial, pericytic, renin-producing and interstitial cells. The collecting ducts are derived from a population of progenitors at the ureteric bud (UB) tip and a proportion of the endothelium is also derived from a dedicated mesenchymal progenitor. The stroma, CM, and UB interact to create spatially defined niches at the periphery of the developing organ. While the UB tip population persist, the CM represents a transient progenitor population that is exhausted to set the final organ size. The timing of CM exhaustion, and hence the final organ structure, is sensitive to disruptions such as premature birth. Here we will discuss our current understanding of the molecular processes allowing these populations to balance cell survival, self-renewal, support of branching, and maintain capacity to commit to differentiation.
Collapse
Affiliation(s)
- Raphael Kopan
- Department of Developmental Biology, Washington University, St. Louis, Missouri, USA; Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA.
| | - Shuang Chen
- Department of Developmental Biology, Washington University, St. Louis, Missouri, USA
| | - Melissa Little
- Department of Developmental Biology, Washington University, St. Louis, Missouri, USA; Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia.
| |
Collapse
|
41
|
From ureteric bud to the first glomeruli: genes, mediators, kidney alterations. Int Urol Nephrol 2014; 47:109-16. [PMID: 25201458 DOI: 10.1007/s11255-014-0784-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 07/04/2014] [Indexed: 12/23/2022]
Abstract
The development of the mammalian kidney is a complex and in part unknown process which requires interactions between pluripotential/stem cells, undifferentiated mesenchymal cells, epithelial and mesenchymal components, eventually leading to the coordinate development of multiple different specialized epithelial, endothelial and stromal cell types within the kidney architectural complexity. We will describe the embryology and molecular nephrogenetic mechanisms, a fascinating traffic of cells and tissues which takes place in five stages: (1) ureteric bud (UB) development; (2) cap mesenchyme formation; (3) mesenchymal-epithelial transition (MET); (4) glomerulogenesis and tubulogenesis; (5) interstitial cell development. In particular, we will analyze the multiple cell types involved in these dramatic events as characters moving between different worlds, from the mesenchymal to the epithelial world and back, and will start to define the multiple factors that propel these cells during their travels throughout the developing kidney. Moreover, according with the hypothesis of renal perinatal programing, we will present the results reached in the fields of immunohistochemistry and molecular biology, by means of which we can explain how a loss or excess of molecular factors governing nephrogenesis may cause the onset of pathologies of different gravity, in some cases leading to a chronic kidney disease at different times from birth.
Collapse
|
42
|
Abstract
The WT1 (Wilms' tumour 1) gene encodes a zinc finger transcription factor and RNA-binding protein that direct the development of several organs and tissues. WT1 manifests both tumour suppressor and oncogenic activities, but the reasons behind these opposing functions are still not clear. As a transcriptional regulator, WT1 can either activate or repress numerous target genes resulting in disparate biological effects such as growth, differentiation and apoptosis. The complex nature of WT1 is exemplified by a plethora of isoforms, post-translational modifications and multiple binding partners. How WT1 achieves specificity to regulate a large number of target genes involved in diverse physiological processes is the focus of the present review. We discuss the wealth of the growing molecular information that defines our current understanding of the versatility and utility of WT1 as a master regulator of organ development, a tumour suppressor and an oncogene.
Collapse
|
43
|
Kang M, Han YM. Differentiation of human pluripotent stem cells into nephron progenitor cells in a serum and feeder free system. PLoS One 2014; 9:e94888. [PMID: 24728509 PMCID: PMC3984279 DOI: 10.1371/journal.pone.0094888] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 03/20/2014] [Indexed: 12/13/2022] Open
Abstract
Objectives Kidney disease is emerging as a critical medical problem worldwide. Because of limited treatment options for the damaged kidney, stem cell treatment is becoming an alternative therapeutic approach. Of many possible human stem cell sources, pluripotent stem cells are most attractive due to their self-renewal and pluripotent capacity. However, little is known about the derivation of renal lineage cells from human pluripotent stem cells (hPSCs). In this study, we developed a novel protocol for differentiation of nephron progenitor cells (NPCs) from hPSCs in a serum- and feeder-free system. Materials and Methods We designed step-wise protocols for differentiation of human pluripotent stem cells toward primitive streak, intermediate mesoderm and NPCs by recapitulating normal nephrogenesis. Expression of key marker genes was examined by RT-PCR, real time RT-PCR and immunocytochemistry. Each experiment was independently performed three times to confirm its reproducibility. Results After modification of culture period and concentration of exogenous factors, hPSCs can differentiate into NPCs that markedly express specific marker genes such as SIX2, GDNF, HOXD11, WT1 and CITED1 in addition to OSR1, PAX2, SALL1 and EYA1. Moreover, NPCs possess the potential of bidirectional differentiation into both renal tubular epithelial cells and glomerular podocytes in defined culture conditions. In particular, approximately 70% of SYN-positive cells were obtained from hPSC-derived NPCs after podocytes induction. NPCs can also form in vitro tubule-like structures in three dimensional culture systems. Conclusions Our novel protocol for hPSCs differentiation into NPCs can be useful for producing alternative sources of cell replacement therapy and disease modeling for human kidney diseases.
Collapse
Affiliation(s)
- Minyong Kang
- Graduate Schools of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
| | - Yong-Mahn Han
- Graduate Schools of Medical Science and Engineering, KAIST, Daejeon, Republic of Korea
- Department of Biological Sciences, KAIST, Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
44
|
Kim SI, Lee SY, Wang Z, Ding Y, Haque N, Zhang J, Zhou J, Choi ME. TGF-β-activated kinase 1 is crucial in podocyte differentiation and glomerular capillary formation. J Am Soc Nephrol 2014; 25:1966-78. [PMID: 24652804 DOI: 10.1681/asn.2013030252] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
TGF-β-activated kinase 1 (TAK1) is a key intermediate in signal transduction induced by TGF-β or inflammatory cytokines, such as TNF-α and IL-1, which are potent inducers of podocyte injury responses that lead to proteinuria and glomerulosclerosis. Nevertheless, little is known about the physiologic and pathologic roles of TAK1 in podocytes. To examine the in vivo role of TAK1, we generated podocyte-specific Tak1 knockout mice (Nphs2-Cre(+):Tak1(fx/fx); Tak1(∆/∆)). Targeted deletion of Tak1 in podocytes resulted in perinatal lethality, with approximately 50% of animals dying soon after birth and 90% of animals dying within 1 week of birth. Tak1(∆/∆) mice developed proteinuria from P1 and exhibited delayed glomerulogenesis and reduced expression of Wilms' tumor suppressor 1 and nephrin in podocytes. Compared with Tak1(fx/fx) mice, Tak1(∆/∆) mice exhibited impaired formation of podocyte foot processes that caused disruption of the podocyte architecture with prominent foot process effacement. Intriguingly, Tak1(∆/∆) mice displayed increased expression of vascular endothelial growth factor within the glomerulus and abnormally enlarged glomerular capillaries. Furthermore, 4- and 7-week-old Tak1(∆/∆) mice with proteinuria had increased collagen deposition in the mesangium and the adjacent tubulointerstitial area. Thus, loss of Tak1 in podocytes is associated with the development of proteinuria and glomerulosclerosis. Taken together, our data show that TAK1 regulates the expression of Wilms' tumor suppressor 1, nephrin, and vascular endothelial growth factor and that TAK1 signaling has a crucial role in podocyte differentiation and attainment of normal glomerular microvasculature during kidney development and glomerular filtration barrier homeostasis.
Collapse
Affiliation(s)
- Sung Il Kim
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Division of Nephrology and Hypertension, Weill Cornell Medical College, New York, New York;
| | - So-Young Lee
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Internal Medicine, Bundang CHA Medical Center, CHA University School of Medicine, Seongnam, South Korea; and
| | - Zhibo Wang
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yan Ding
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Division of Nephrology and Hypertension, Weill Cornell Medical College, New York, New York
| | - Nadeem Haque
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jiwang Zhang
- Oncology Institute, Cardinal Bernardin Cancer Center, Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Jing Zhou
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mary E Choi
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Division of Nephrology and Hypertension, Weill Cornell Medical College, New York, New York;
| |
Collapse
|
45
|
Aydin M, Hakan N, Zenciroglu A, Aydog O, Okumus N. Different clinical presentations of WT1 gene mutations. Eur J Pediatr 2013; 172:1705-6. [PMID: 23835858 DOI: 10.1007/s00431-013-2085-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 06/22/2013] [Indexed: 11/28/2022]
Affiliation(s)
- Mustafa Aydin
- Division of Neonatology, Department of Pediatrics, Elazig Training and Research Hospital, Elazig, Turkey
| | | | | | | | | |
Collapse
|
46
|
Wt-1 Expression Linked to Nitric Oxide Availability during Neonatal Obstructive Nephropathy. Adv Urol 2013; 2013:401750. [PMID: 24288526 PMCID: PMC3833023 DOI: 10.1155/2013/401750] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 09/27/2013] [Accepted: 09/30/2013] [Indexed: 12/22/2022] Open
Abstract
The wt-1 gene encodes a zinc finger DNA-binding protein that acts as a transcriptional activator or repressor depending on the cellular or chromosomal context. The wt-1 regulates the expression of a large number of genes that have a critical role in kidney development. Congenital obstructive nephropathy disrupts normal renal development and causes chronic progressive interstitial fibrosis, which contributes to renal growth arrest, ultimately leading to chronic renal failure. Wt-1 is downregulated during congenital obstructive nephropathy, leading to apoptosis. Of great interest, nitric oxide bioavailability associated with heat shock protein 70 (Hsp70) interaction may modulate wt-1 mRNA expression, preventing obstruction-induced cell death during neonatal unilateral ureteral obstruction. Moreover, recent genetic researches have allowed characterization of many of the complex interactions among the individual components cited, but the realization of new biochemical, molecular, and functional experiments as proposed in our and other research labs allows us to establish a deeper level of commitment among proteins involved and the potential pathogenic consequences of their imbalance.
Collapse
|
47
|
Ma L, Qu L. The Function of MicroRNAs in Renal Development and Pathophysiology. J Genet Genomics 2013; 40:143-52. [DOI: 10.1016/j.jgg.2013.03.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 02/28/2013] [Accepted: 03/01/2013] [Indexed: 01/01/2023]
|
48
|
Chai OH, Song CH, Park SK, Kim W, Cho ES. Molecular regulation of kidney development. Anat Cell Biol 2013; 46:19-31. [PMID: 23560233 PMCID: PMC3615609 DOI: 10.5115/acb.2013.46.1.19] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 01/25/2013] [Accepted: 02/04/2013] [Indexed: 12/21/2022] Open
Abstract
Genetically engineered mice have provided much information about gene function in the field of developmental biology. Recently, conditional gene targeting using the Cre/loxP system has been developed to control the cell type and timing of the target gene expression. The increase in number of kidney-specific Cre mice allows for the analysis of phenotypes that cannot be addressed by conventional gene targeting. The mammalian kidney is a vital organ that plays a critical homeostatic role in the regulation of body fluid composition and excretion of waste products. The interactions between epithelial and mesenchymal cells are very critical events in the field of developmental biology, especially renal development. Kidney development is a complex process, requiring inductive interactions between epithelial and mesenchymal cells that eventually lead to the growth and differentiation of multiple highly specialized stromal, vascular, and epithelial cell types. Through the use of genetically engineered mouse models, the molecular bases for many of the events in the developing kidney have been identified. Defective morphogenesis may result in clinical phenotypes that range from complete renal agenesis to diseases such as hypertension that exist in the setting of grossly normal kidneys. In this review, we focus on the growth and transcription factors that define kidney progenitor cell populations, initiate ureteric bud branching, induce nephron formation within the metanephric mesenchyme, and differentiate stromal and vascular progenitors in the metanephric mesenchyme.
Collapse
Affiliation(s)
- Ok-Hee Chai
- Department of Anatomy, Institute for Medical Sciences, Chonbuk National University Medical School, Jeonju, Korea
| | | | | | | | | |
Collapse
|
49
|
Narayanan K, Schumacher KM, Tasnim F, Kandasamy K, Schumacher A, Ni M, Gao S, Gopalan B, Zink D, Ying JY. Human embryonic stem cells differentiate into functional renal proximal tubular-like cells. Kidney Int 2013; 83:593-603. [PMID: 23389418 DOI: 10.1038/ki.2012.442] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Renal cells are used in basic research, disease models, tissue engineering, drug screening, and in vitro toxicology. In order to provide a reliable source of human renal cells, we developed a protocol for the differentiation of human embryonic stem cells into renal epithelial cells. The differentiated stem cells expressed markers characteristic of renal proximal tubular cells and their precursors, whereas markers of other renal cell types were not expressed or expressed at low levels. Marker expression patterns of these differentiated stem cells and in vitro cultivated primary human renal proximal tubular cells were comparable. The differentiated stem cells showed morphological and functional characteristics of renal proximal tubular cells, and generated tubular structures in vitro and in vivo. In addition, the differentiated stem cells contributed in organ cultures for the formation of simple epithelia in the kidney cortex. Bioreactor experiments showed that these cells retained their functional characteristics under conditions as applied in bioartificial kidneys. Thus, our results show that human embryonic stem cells can differentiate into renal proximal tubular-like cells. Our approach would provide a source for human renal proximal tubular cells that are not affected by problems associated with immortalized cell lines or primary cells.
Collapse
|
50
|
Menendez-Castro C, Hilgers KF, Amann K, Daniel C, Cordasic N, Wachtveitl R, Fahlbusch F, Plank C, Dötsch J, Rascher W, Hartner A. Intrauterine growth restriction leads to a dysregulation of Wilms' tumour supressor gene 1 (WT1) and to early podocyte alterations. Nephrol Dial Transplant 2012; 28:1407-17. [PMID: 23229934 DOI: 10.1093/ndt/gfs517] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Intrauterine growth restriction (IUGR) leads to low nephron number and higher incidence of renal disease. We hypothesized that IUGR induces early podocyte alterations based on a dysregulation of Wilms' tumour suppressor gene 1 (WT1), a key player of nephrogenesis and mediator of podocyte integrity. METHODS IUGR was induced in rats by maternal protein restriction during pregnancy. Kidneys were harvested from male offspring at Days 1 and 70 of life. qRT-PCR, immunohistochemistry and electron microscopy were performed in renal tissue. Albuminuria was assessed by enzyme-linked immunosorbent assay. RESULTS At Day 70 of life, higher albuminuria and overt alterations of podocyte ultrastructure were detected in IUGR animals in spite of normal blood pressure. Moreover, we found increased glomerular immunoreactivity and expression of desmin, while synaptopodin and nephrin were decreased. Glomerular immunoreactivity and expression of WT1 were increased in IUGR animals at this time point with an altered expressional ratio of WT1 +KTS and -KTS isoforms. These changes of WT1 expression were already present at the time of birth. CONCLUSIONS IUGR results in early podocyte damage possibly due to a dysregulation of WT1. We suggest that an imbalance of WT1 isoforms to the disadvantage of -KTS affects nephrogenesis in IUGR rats and that persistent dysregulation of WT1 results in a reduced ability to maintain podocyte integrity, rendering IUGR rats more susceptible for renal disease.
Collapse
Affiliation(s)
- Carlos Menendez-Castro
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nuremberg, Erlangen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|