1
|
Janek A, Badeński A, Badeńska M, Szuster M, Szymańska-Kurek K, Trembecka-Dubel E, Szczepańska M. WT1 Gene Pathogenic Variants: Clinical Challenges and Treatment Strategies in Pediatric Nephrology-One Center Practice. Int J Mol Sci 2025; 26:3642. [PMID: 40332152 PMCID: PMC12026538 DOI: 10.3390/ijms26083642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/01/2025] [Accepted: 04/10/2025] [Indexed: 05/08/2025] Open
Abstract
Pathogenic variants in the Wilms' tumor suppressor gene 1 (WT1 gene) can lead to serious disorders within the kidney and urogenital system, including chronic kidney disease. There is still much uncertainty regarding the optimal management of diseases caused by WT1 dysfunction, posing a challenge for physicians caring for these patients. The aim of our study is to present experiences related to the course and treatment of patients with confirmed WT1 pathogenic variants. Data from seven patients (five girls, two boys), who were at the age of 4.8 ± 5.1 years (0.3-14 years) at their first admission and were treated between 1997-2022, were analyzed. The analysis included each patient's age at the day of diagnosis, anthropometric measurements, comorbidities, and laboratory and genetic test results, as well as their treatment, oncological procedures, and performed surgeries. Wilms' tumor was the first manifestation of the disease in three patients. Arterial hypertension was diagnosed in three patients, and anemia in four. Treatment of patients with nephrotic syndrome included glucocorticosteroid therapy (GCS), calcineurin inhibitors (CNIs), and mycophenolate mofetil (MMF). Nephrectomy was performed in five children, while kidney transplantation was carried out in two patients. An interdisciplinary approach to WT1 gene pathogenic variants, including early diagnosis, individualization, regular monitoring of treatment, and oncological vigilance, is crucial for improving prognosis and ensuring proper care for patients with nephrological manifestations of WT1 gene region disorders. Furthermore, for a comprehensive understanding of the scope of this disease and the development of effective therapy methods, continued research on the clinical manifestations of WT1 pathogenic variants is essential.
Collapse
Affiliation(s)
- Artur Janek
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (A.J.); (M.B.); (M.S.); (E.T.-D.); (M.S.)
| | - Andrzej Badeński
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (A.J.); (M.B.); (M.S.); (E.T.-D.); (M.S.)
| | - Marta Badeńska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (A.J.); (M.B.); (M.S.); (E.T.-D.); (M.S.)
| | - Martyna Szuster
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (A.J.); (M.B.); (M.S.); (E.T.-D.); (M.S.)
| | - Karolina Szymańska-Kurek
- Department of Pediatric Nephrology with Dialysis Division for Children, Independent Public Clinical Hospital No. 1, ul. 3 Maja 13/15, 41-800 Zabrze, Poland;
| | - Elżbieta Trembecka-Dubel
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (A.J.); (M.B.); (M.S.); (E.T.-D.); (M.S.)
| | - Maria Szczepańska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (A.J.); (M.B.); (M.S.); (E.T.-D.); (M.S.)
| |
Collapse
|
2
|
Fan X, Li J, Gao Y, Li L, Zhang H, Bi Z. The mechanism of enterogenous toxin methylmalonic acid aggravating calcium-phosphorus metabolic disorder in uremic rats by regulating the Wnt/β-catenin pathway. Mol Med 2025; 31:19. [PMID: 39844078 PMCID: PMC11756144 DOI: 10.1186/s10020-025-01067-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/03/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Uremia (UR) is caused by increased UR-related toxins in the bloodstream. We explored the mechanism of enterogenous toxin methylmalonic acid (MMA) in calcium-phosphorus metabolic disorder in UR rats via the Wnt/β-catenin pathway. METHODS The UR rat model was established by 5/6 nephrectomy. The fecal bacteria of UR rats were transplanted into Sham rats. Sham rats were injected with exogenous MMA or Salinomycin (SAL). Pathological changes in renal/colon tissues were analyzed. MMA concentration, levels of renal function indicators, serum inflammatory factors, Ca2+/P3+, and parathyroid hormone, intestinal flora structure, fecal metabolic profile, intestinal permeability, and glomerular filtration rate (GFR) were assessed. Additionally, rat glomerular podocytes were cultured, with cell viability and apoptosis measured. RESULTS Intestinal flora richness and diversity in UR rats were decreased, along with unbalanced flora structure. Among the screened 133 secondary differential metabolites, the MMA concentration rose, showing the most significant difference. UR rat fecal transplantation caused elevated MMA concentration in the serum and renal tissues of Sham rats. The intestinal flora metabolite MMA or exogenous MMA promoted intestinal barrier impairment, increased intestinal permeability, induced glomerular podocyte loss, and reduced GFR, causing calcium-phosphorus metabolic disorder. The intestinal flora metabolite MMA or exogenous MMA induced inflammatory responses and facilitated glomerular podocyte apoptosis by activating the Wnt/β-catenin pathway, which could be counteracted by repressing the Wnt/β-catenin pathway. CONCLUSIONS Enterogenous toxin MMA impelled intestinal barrier impairment in UR rats, enhanced intestinal permeability, and activated the Wnt/β-catenin pathway to induce glomerular podocyte loss and reduce GFR, thus aggravating calcium-phosphorus metabolic disorder.
Collapse
Affiliation(s)
- Xing Fan
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
| | - Jing Li
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
| | - Yan Gao
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China.
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China.
| | - Lin Li
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China.
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China.
| | - Haisong Zhang
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
| | - Zhaoyu Bi
- Department of Nephrology, The Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, No. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei Province, China
| |
Collapse
|
3
|
Nagano C, Nozu K. A review of the genetic background in complicated WT1-related disorders. Clin Exp Nephrol 2025; 29:1-9. [PMID: 39002031 PMCID: PMC11807054 DOI: 10.1007/s10157-024-02539-x] [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: 05/07/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
The Wilms tumor 1 (WT1) gene was first identified in 1990 as a strong candidate for conferring a predisposition to Wilms tumor. The WT1 protein has four zinc finger structures (DNA binding domain) at the C-terminus, which bind to transcriptional regulatory sequences on DNA, and acts as a transcription factor. WT1 is expressed during kidney development and regulates differentiation, and is also expressed in glomerular epithelial cells after birth to maintain the structure of podocytes. WT1-related disorders are a group of conditions associated with an aberrant or absent copy of the WT1 gene. This group of conditions encompasses a wide phenotypic spectrum that includes Denys-Drash syndrome (DDS), Frasier syndrome (FS), Wilms-aniridia-genitourinary-mental retardation syndrome, and isolated manifestations of nephropathy or Wilms tumor. The genotype-phenotype correlation is becoming clearer: patients with missense variants in DNA binding sites including C2H2 sites manifest DDS and develop early-onset and rapidly developing end-stage kidney disease. A deeper understanding of the genotype-phenotype correlation has also been obtained in DDS, but no such correlation has been observed in FS. The incidence of Wilms tumor is higher in patients with DDS and exon-truncating variants than in those with non-truncating variants. Here, we briefly describe the genetic background of this highly complicated WT1-related disorders.
Collapse
Affiliation(s)
- China Nagano
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan.
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| |
Collapse
|
4
|
Fleming AM, Gehle DB, Perrino MR, Graetz DE, Bissler JJ, McCarville B, Krasin MJ, Brennan RC, Zhang J, Yang W, Sapkota Y, Hudson MM, Davidoff AM, Green DM, Murphy AJ. Concomitant Wilms tumor and autosomal dominant polycystic kidney disease. Pediatr Blood Cancer 2024; 71:e31230. [PMID: 39085996 PMCID: PMC11369902 DOI: 10.1002/pbc.31230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/01/2024] [Accepted: 07/16/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Concomitant Wilms tumor (WT) and autosomal dominant polycystic kidney disease (ADPKD) is exceedingly rare, presenting a diagnostic and technical challenge to pediatric surgical oncologists. The simultaneous workup and management of these disease processes are incompletely described. PROCEDURE We performed a retrospective analysis of patients treated at our institution with concomitant diagnoses of WT and ADPKD. We also review the literature on the underlying biology and management principles of these conditions. RESULTS We present three diverse cases of concomitant unilateral WT and ADPKD who underwent nephrectomy. One patient had preoperative imaging consistent with ADPKD with confirmatory testing postoperatively, one was found to have contralateral renal cysts intraoperatively with confirmatory imaging post nephrectomy, and one was diagnosed in childhood post nephrectomy. All patients are alive at last follow-up, and the patient with longest follow-up has progressed to end-stage kidney failure requiring transplantation and dialysis in adulthood. All patients underwent germline testing and were found to have no cancer predisposition syndrome or pathogenic or likely pathogenic variants for WT. CONCLUSION Concomitant inheritance of ADPKD and development of WT are extremely rare, and manifestations of ADPKD may not present until late childhood or adulthood. ADPKD is not a known predisposing condition for WT. When ADPKD diagnosis is made by family history, imaging, and/or genetic testing before WT diagnosis and treatment, the need for extensive preoperative characterization of cystic kidney lesions in children and increased risk of post-nephrectomy kidney failure warrant further discussion of surgical approach and perioperative management strategies.
Collapse
Affiliation(s)
- Andrew M. Fleming
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Daniel B. Gehle
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Melissa R. Perrino
- Department of Oncology, Division of Cancer Predisposition, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Dylan E. Graetz
- Department of Oncology, Solid Tumor Division, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - John J. Bissler
- Department of Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, TN 38105
- Department of Pediatrics, Division of Pediatric Nephrology, University of Tennessee Health Science Center, TN 38163
| | - Beth McCarville
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Matthew J. Krasin
- Department of Radiation Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Rachel C. Brennan
- Department of Pediatric Hematology & Oncology, Logan Health, Kalispell, MT 59901
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Wentao Yang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105
- Department of Oncology, Division of Cancer Survivorship, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Andrew M. Davidoff
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Daniel M. Green
- Department of Oncology, Division of Cancer Survivorship, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Andrew J. Murphy
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN 38105
- Department of Surgery, Division of Pediatric Surgery, University of Tennessee Health Science Center, Memphis, TN 38163
| |
Collapse
|
5
|
Li X, Chen M, Cao J, Chen X, Song H, Shi S, He B, Zhang B, Zhang Z. Human umbilical cord mesenchymal stem cell-derived exosomes mitigate diabetic nephropathy via enhancing M2 macrophages polarization. Heliyon 2024; 10:e37002. [PMID: 39286156 PMCID: PMC11402917 DOI: 10.1016/j.heliyon.2024.e37002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Background and objectives Exosomes, which are small nanoscale vesicles capable of secretion, have garnered significant attention in recent years because of their therapeutic potential, particularly in the context of kidney diseases. Notably, human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Exos) are emerging as promising targeted therapies for renal conditions. The aim of this study was to investigate the therapeutic effects of hucMSC-Exos on diabetic kidney disease (DKD) both in vivo and in vitro. Additionally, this study seeks to elucidate cellular and molecular differentials, as well as the expression of relevant signaling pathways, through single-cell RNA sequencing. This endeavor was designed to enhance our understanding of the connection between hucMSC-Exos and the pathogenesis of DKD. Methods and results The study commenced with the extraction and characterization of hucMSC-Exos, including the determination of their concentrations. Animal experiments were conducted to evaluate the therapeutic potential of hucMSC-Exos in a DKD mouse model. Subsequently, single-cell sequencing was employed to investigate the molecular mechanisms underlying the efficacy of extracellular vesicles in ameliorating DKD. These findings were further substantiated by cell-based experiments. Importantly, the results indicate that hucMSC-Exos can impede the progression of DKD in mice, with macrophage activation playing a pivotal role in this process. Conclusions The in vivo experiments conclusively established hucMSC-Exos as a pivotal component in preserving renal function and retarding the progression of DKD. Our utilization of single-cell sequencing technology, in conjunction with in vivo and in vitro experiments, provides compelling evidence that M2 macrophages are instrumental in enhancing the amelioration of diabetic nephropathy.
Collapse
Affiliation(s)
- Xueting Li
- Department of Nephrology, Affiliated Hospital of Jining Medical University, Jining, Shandong, PR China
| | - Mingkai Chen
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Jinghe Cao
- Department of Reproductive Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Xinke Chen
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Hui Song
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Shuo Shi
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Baoyu He
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, PR China
| | - Ziteng Zhang
- Departments of Thoracic Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong, 272000, PR China
- Departments of Thoracic Surgery, Qinghai Red Cross Hospital, Xining, Qinghai, 81000, PR China
| |
Collapse
|
6
|
Lim KT, Loh AHP. Inter-Ethnic Variations in the Clinical, Pathological, and Molecular Characteristics of Wilms Tumor. Cancers (Basel) 2024; 16:3051. [PMID: 39272909 PMCID: PMC11393868 DOI: 10.3390/cancers16173051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Wilms tumor is the commonest primary renal malignancy in children and demonstrates substantial inter-ethnic variation in clinical, pathological, and molecular characteristics. Wilms tumor occurs at a lower incidence and at a younger age in Asians compared to Caucasians and Africans. Asians also present at an earlier stage of disease, with a higher incidence of favorable histology tumors and a lower incidence of perilobar nephrogenic rests compared to Caucasians, while African children present with more advanced disease. Studies have implicated population differences in the incidence of WT1 mutations, loss of imprinting of the IGF2 locus, and loss of heterozygosity of 1p/16q, or 1q gain as possible bases for epidemiological differences in the disease profile of Wilms tumors in various ethnic groups. Yet, evidence to support these associations is confounded by differences in treatment protocols and inequalities in the availability of treatment resources and remains limited by the quality of population-based data, especially in resource-limited settings.
Collapse
Affiliation(s)
- Kia Teng Lim
- Ministry of Health Holdings, Singapore 139691, Singapore
| | - Amos H P Loh
- VIVA-KKH Paediatric Brain and Solid Tumour Programme, Children's Blood and Cancer Centre, KK Women's and Children's Hospital, Singapore 229899, Singapore
- SingHealth-Duke NUS Global Health Institute, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Paediatric Surgery, KK Women's and Children's Hospital, Singapore 229899, Singapore
| |
Collapse
|
7
|
Wang G, Wu H, Zhai X, Zhang L, Zhang C, Cheng C, Xu X, Gao E, Xiong X, Zhang J, Liu Z. Kidney Organoid Modeling of WT1 Mutations Reveals Key Regulatory Paths Underlying Podocyte Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308556. [PMID: 38810140 PMCID: PMC11304319 DOI: 10.1002/advs.202308556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/16/2024] [Indexed: 05/31/2024]
Abstract
Wilms tumor-1(WT1) is a crucial transcription factor that regulates podocyte development. However, the epigenomic mechanism underlying the function of WT1 during podocyte development has yet to be fully elucidated. Here, single-cell chromatin accessibility and gene expression maps of foetal kidneys and kidney organoids are generated. Functional implications of WT1-targeted genes, which are crucial for the development of podocytes and the maintenance of their structure, including BMPER/PAX2/MAGI2 that regulates WNT signaling pathway, MYH9 that maintains actin filament organization and NPHS1 that modulates cell junction assembly are identified. To further illustrate the functional importance of WT1-mediated transcriptional regulation during podocyte development, cultured and implanted patient-derived kidney organoids derived from the Induced Pluripotent Stem Cell (iPSCs) of a patient with a heterozygous missense mutation in WT1 are generated. Results from single-cell RNA sequencing (scRNA-seq) and functional assays confirm that the WT1 mutation leads to delays in podocyte development and causes damage to cell structures, due to its failure to activate the targeting genes MAGI2, MYH9, and NPHS1. Notably, correcting the mutation in the patient iPSCs using CRISPR-Cas9 gene editing rescues the podocyte phenotype. Collectively, this work elucidates the WT1-related epigenomic landscape with respect to human podocyte development and identifies the disease-causing role of a WT1 mutation.
Collapse
Affiliation(s)
- Gang Wang
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Hangdi Wu
- Department of Basic Medical SciencesZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Xiuwen Zhai
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Li Zhang
- Department of Basic Medical SciencesZhejiang University School of MedicineHangzhouZhejiang310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
- Center for Stem Cell and Regenerative MedicineDepartment of Basic Medical Sciences & The First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Changming Zhang
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Chen Cheng
- Department of Basic Medical SciencesZhejiang University School of MedicineHangzhouZhejiang310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
- Center for Stem Cell and Regenerative MedicineDepartment of Basic Medical Sciences & The First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
| | - Xiaodong Xu
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Erzhi Gao
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Xushen Xiong
- Department of Basic Medical SciencesZhejiang University School of MedicineHangzhouZhejiang310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
- State Key Laboratory of Transvascular Implantation DevicesThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou311121China
| | - Jin Zhang
- Department of Basic Medical SciencesZhejiang University School of MedicineHangzhouZhejiang310058China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
- Center for Stem Cell and Regenerative MedicineDepartment of Basic Medical Sciences & The First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058China
- Hematology InstituteZhejiang UniversityHangzhouZhejiang310058China
| | - Zhihong Liu
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
- Liangzhu LaboratoryZhejiang UniversityHangzhou311121China
| |
Collapse
|
8
|
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
|
9
|
Cheng C, Wang G, Zhu Y, Wu H, Zhang L, Liu Z, Huang Y, Zhang J. Multiplexed bulk and single-cell RNA-seq hybrid enables cost-efficient disease modeling with chimeric organoids. Nat Commun 2024; 15:3946. [PMID: 38729950 PMCID: PMC11087505 DOI: 10.1038/s41467-024-48282-5] [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: 07/28/2022] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
Disease modeling with isogenic Induced Pluripotent Stem Cell (iPSC)-differentiated organoids serves as a powerful technique for studying disease mechanisms. Multiplexed coculture is crucial to mitigate batch effects when studying the genetic effects of disease-causing variants in differentiated iPSCs or organoids, and demultiplexing at the single-cell level can be conveniently achieved by assessing natural genetic barcodes. Here, to enable cost-efficient time-series experimental designs via multiplexed bulk and single-cell RNA-seq of hybrids, we introduce a computational method in our Vireo Suite, Vireo-bulk, to effectively deconvolve pooled bulk RNA-seq data by genotype reference, and thereby quantify donor abundance over the course of differentiation and identify differentially expressed genes among donors. Furthermore, with multiplexed scRNA-seq and bulk RNA-seq, we demonstrate the usefulness and necessity of a pooled design to reveal donor iPSC line heterogeneity during macrophage cell differentiation and to model rare WT1 mutation-driven kidney disease with chimeric organoids. Our work provides an experimental and analytic pipeline for dissecting disease mechanisms with chimeric organoids.
Collapse
Affiliation(s)
- Chen Cheng
- Center for Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong SAR, China
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow for Transplantation Center of the First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Gang Wang
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
- Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuqing Zhu
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow for Transplantation Center of the First Affiliated Hospital, Zhejiang University, Hangzhou, China
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, Anhui, China
| | - Hangdi Wu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
- Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Zhang
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow for Transplantation Center of the First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Zhihong Liu
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China.
- Department of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yuanhua Huang
- Center for Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong SAR, China.
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
- Department of Statistics and Actuarial Science, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| | - Jin Zhang
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow for Transplantation Center of the First Affiliated Hospital, Zhejiang University, Hangzhou, China.
- Center of Gene/Cell Engineering and Genome Medicine of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
10
|
Rajão Martins F, Gonçalves F, Guedes A, Sequeira G, Ribeiro C. Polyarthritis due to metastatic calcinosis in a patient with new WT1 gene mutation: resolution after renal transplantation. Scand J Rheumatol 2024; 53:220-222. [PMID: 38224230 DOI: 10.1080/03009742.2023.2299610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 01/16/2024]
Affiliation(s)
- F Rajão Martins
- Rheumatology Department, Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | - F Gonçalves
- Nephrology Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - A Guedes
- Nephrology Department, Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | - G Sequeira
- Rheumatology Department, Centro Hospitalar Universitário do Algarve, Faro, Portugal
| | - C Ribeiro
- Rheumatology Department, Centro Hospitalar Universitário do Algarve, Faro, Portugal
| |
Collapse
|
11
|
Goh J, Wei H, Lai AHM, Chang B, Khan S, Syn Y, Jamuar SS, Tan EC. Novel and recurrent variants in PAX6 in four patients with ocular phenotypes from Southeast Asia. Clin Dysmorphol 2024; 33:63-68. [PMID: 38441200 DOI: 10.1097/mcd.0000000000000487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Aniridia is an autosomal dominant condition characterized by the complete or partial absence of the iris, often with additional presentations such as foveal hypoplasia, nystagmus, cataract, glaucoma and other ocular abnormalities. Most cases are caused by heterozygous mutations in the paired box 6 gene (PAX6), which codes for a transcription factor that regulates eye development. Four patients from our hospital who presented with ocular phenotypes were recruited for research sequencing with informed consent. Sanger sequencing of PAX6 coding exons or exome sequencing was performed on genomic DNA from venous blood samples. Variants in PAX6 were identified in the four patients. Two variants are recurrent single-nucleotide substitutions - one is a substitution found in a patient with bilateral aniridia, whereas the other is a splice variant in a patient with nystagmus and neuroblastoma. The other two variants are novel and found in two patients with isolated aniridia. Both are small duplications that are predicted to lead to premature termination. For the recurrent variants, the comparison of phenotypes for patients with identical variants would shed light on the mechanisms of pathogenesis, and the discovery of two novel variants expands the spectrum of PAX6 mutations.
Collapse
Affiliation(s)
- Jeannette Goh
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital
- Paediatrics Academic Clinical Programme, SingHealth Duke-NUS Medical School
| | - Heming Wei
- Research Laboratory, KK Women's and Children's Hospital
| | - Angeline H M Lai
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital
- Paediatrics Academic Clinical Programme, SingHealth Duke-NUS Medical School
| | - Benjamin Chang
- Opthalmology Service, KK Women's and Children's Hospital
| | - Shazia Khan
- Opthalmology Service, KK Women's and Children's Hospital
| | - Yamon Syn
- Singapore National Eye Centre, Singapore
| | - Saumya S Jamuar
- Genetics Service, Department of Paediatrics, KK Women's and Children's Hospital
- Paediatrics Academic Clinical Programme, SingHealth Duke-NUS Medical School
| | - Ene-Choo Tan
- Paediatrics Academic Clinical Programme, SingHealth Duke-NUS Medical School
- Research Laboratory, KK Women's and Children's Hospital
| |
Collapse
|
12
|
Mura E, Parazzini C, Tonduti D. Rare forms of hypomyelination and delayed myelination. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:225-252. [PMID: 39322381 DOI: 10.1016/b978-0-323-99209-1.00002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Hypomyelination is defined by the evidence of an unchanged pattern of deficient myelination on two MRIs performed at least 6 months apart in a child older than 1 year. When the temporal criteria are not fulfilled, and the follow-up MRI shows a progression of the myelination even if still not adequate for age, hypomyelination is excluded and the pattern is instead consistent with delayed myelination. This can be mild and nonspecific in some cases, while in other cases there is a severe delay that in the first disease stages could be difficult to differentiate from hypomyelination. In hypomyelinating leukodystrophies, hypomyelination is due to a primary impairment of myelin deposition, such as in Pelizaeus Merzabcher disease. Conversely, myelin lack is secondary, often to primary neuronal disorders, in delayed myelination and some condition with hypomyelination. Overall, the group of inherited white matter disorders with abnormal myelination has expanded significantly during the past 20 years. Many of these disorders have only recently been described, for many of them only a few patients have been reported and this contributes to make challenging the diagnostic process and the interpretation of Next Generation Sequencing results. In this chapter, we review the clinical and radiologic features of rare and lesser known forms of hypomyelination and delayed myelination not mentioned in other chapters of this handbook.
Collapse
Affiliation(s)
- Eleonora Mura
- Unit of Pediatric Neurology, Department of Biomedical and Clinical Sciences, V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy; C.O.A.L.A (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy
| | - Cecilia Parazzini
- C.O.A.L.A (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy; Pediatric Radiology and Neuroradiology Department, V. Buzzi Children's Hospital, Milan, Italy
| | - Davide Tonduti
- Unit of Pediatric Neurology, Department of Biomedical and Clinical Sciences, V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy; C.O.A.L.A (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children's Hospital, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
13
|
Khandpur S, Srivastava M, Sharma R, Asif S, Bhadauria DS, Mishra P, Purty AJ, Tiwari S. Association of Wilms tumor-1 protein in urinary exosomes with kidney injury: a population-based cross-sectional study. Front Med (Lausanne) 2023; 10:1220309. [PMID: 37795410 PMCID: PMC10545876 DOI: 10.3389/fmed.2023.1220309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/25/2023] [Indexed: 10/06/2023] Open
Abstract
Objective Loss of Wilms tumor-1 (WT1) protein, a podocytopathy marker, through urine exosome (uE), could be an early indication of kidney injury. We examined WT1 in uE (uE-WT1), along with other urine markers of glomerular and kidney tubule injury, in individuals without chronic kidney disease (CKD). Methodology The cross-sectional study included individuals who reported having no evidence of chronic kidney disease (CKD). Albumin-to-creatinine ratio (ACR) and estimated glomerular filtration rate (eGFR) were used to assess kidney function. eGFR was calculated using the 2009 CKD-EPI (CKD-Epidemiological) equation. WT1 was analyzed in uE from humans and Wistar rats (before and after the 9th week of diabetes, n = 20). uE-WT1, urinary neutrophil gelatinase-associated lipocalin (NGAL), and kidney injury molecule-1 (KIM-1) were estimated using ELISA. The Kruskal-Wallis H test, Mann-Whitney U test, and stepwise multivariable linear regression were performed. Results Urine NGAL and ACR increase with uE-WT1 quartiles (n = 146/quarter). Similarly, uE-WT1, KIM-1, and NGAL were positively associated with ACR. Furthermore, KIM-1, NGAL, and uE-WT1 correlated with ACR. uE-WT1 outperformed KMI-1 and NGAL to explain ACR variability (25% vs. 6% or 9%, respectively). Kidney injury in streptozotocin-induced diabetic rats was associated with a significant rise in uE-WT1. Moreover, the findings were confirmed by the histopathology of kidney tissues from rats. Conclusion uE-WT1 was strongly associated with kidney function in rats. In individuals without CKD, uE-WT1 outperformed NGAL as a determinant of differences in ACR.
Collapse
Affiliation(s)
- Sukhanshi Khandpur
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Medha Srivastava
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Rajni Sharma
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Shafaque Asif
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Dharmendra S. Bhadauria
- Department of Nephrology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Prabhaker Mishra
- Department of Biostatistics and Health Informatics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Anil J. Purty
- Department of Community Medicine, Pondicherry Institute of Medical Sciences (A Unit of Madras Medical Mission), Puducherry, India
| | - Swasti Tiwari
- Department of Molecular Medicine and Biotechnology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
14
|
Xu J, Zhou X, Zhang T, Zhang B, Xu PX. Smarca4 deficiency induces Pttg1 oncogene upregulation and hyperproliferation of tubular and interstitial cells during kidney development. Front Cell Dev Biol 2023; 11:1233317. [PMID: 37727504 PMCID: PMC10506413 DOI: 10.3389/fcell.2023.1233317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
Kidney formation and nephrogenesis are controlled by precise spatiotemporal gene expression programs, which are coordinately regulated by cell-cycle, cell type-specific transcription factors and epigenetic/chromatin regulators. However, the roles of epigenetic/chromatin regulators in kidney development and disease remain poorly understood. In this study, we investigated the impact of deleting the chromatin remodeling factor Smarca4 (Brg1), a human Wilms tumor-associated gene, in Wnt4-expressing cells. Smarca4 deficiency led to severe tubular defects and a shortened medulla. Through unbiased single-cell RNA sequencing analyses, we identified multiple types of Wnt4 Cre-labeled interstitial cells, along with nephron-related cells. Smarca4 deficiency increased interstitial cells but markedly reduced tubular cells, resulting in cells with mixed identity and elevated expression of cell-cycle regulators and genes associated with extracellular matrix and epithelial-to-mesenchymal transition/fibrosis. We found that Smarca4 loss induced a significant upregulation of the oncogene Pttg1 and hyperproliferation of Wnt4 Cre-labeled cells. These changes in the cellular state could hinder the cellular transition into characteristic tubular structures, eventually leading to fibrosis. In conclusion, our findings shed light on novel cell types and genes associated with Wnt4 Cre-labeled cells and highlight the critical role of Smarca4 in regulating tubular cell differentiation and the expression of the cancer-causing gene Pttg1 in the kidney. These findings may provide valuable insights into potential therapeutic strategies for renal cell carcinoma resulting from SMARCA4 deficiency.
Collapse
Affiliation(s)
- Jinshu Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ting Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Pin-Xian Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
15
|
Chen H, Zhang M, Lin J, Lu J, Zhong F, Zhong F, Gao X, Liao X. Genotype-phenotype correlation of WT1 mutation-related nephropathy in Chinese children. Front Pediatr 2023; 11:1192021. [PMID: 37576146 PMCID: PMC10416235 DOI: 10.3389/fped.2023.1192021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 07/05/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction This study aimed to analyze the clinical characteristics of nephropathy associated with WT1 gene mutations in Chinese children and explore the relationship between genotype and clinical phenotype. Methods Cases diagnosed at the Guangzhou Women and Children's Medical Center, were combined with those retrieved from PubMed and China National Knowledge Infrastructure (CNKI) databases from January 2015 to June 2022 and integrated into a study cohort; grouped according to gene mutation sites, clinical phenotype, and renal pathological types. The clinical characteristics between groups were compared, and the relationship between genotype and age of onset, clinical phenotype, and pathological type were retrospectively analyzed. Results The center enrolled 15 confirmed children: seven cases of non-simple nephropathy, including Denys-Drash syndrome (DDS) and Frasier syndrome (FS); eight cases of isolated steroid-resistant nephrotic syndrome (ISRNS); and 13 cases (86.7%) that progressed to end-stage renal disease (ESRD). The initial hemoglobin and bicarbonate levels of patients with clinical non-simple nephropathy were significantly lower than those with simple nephropathy, whereas the serum creatinine levels were higher than those of patients with simple nephropathy. A total of 75 cases of nephropathy associated with WT1 mutations in the study cohort met the inclusion and exclusion criteria. The most common clinical manifestations of WT1 mutations in this cohort were DDS (29/75, 38.7%) and ISRNS (37/75, 49.3%). A renal biopsy was performed in 43 patients, and the common types of renal pathology were focal segmental glomerulosclerosis (23/43, 53.5%) and DMS (13/43, 30.2%). Within the cohort, there were 12 cases (16.0%) in the exon 8 mutation group, 32 (42.6%) in the exon 9 group, 19 (25.3%) in the intron 9 group, and 12 (16.0%) in other gene site mutation groups. Common sites of WT1 mutations in Chinese children were exons 9 and intron 9. Exon 8 mutations were uniquely correlated with the age of onset within three months [5/7; 71.4%; Adjusted standardized residual (AR) = 4.2]. The renal survival time in the exon 8 mutation group was the shortest (P = 0.003). Discussion The molecular and biological characteristics of WT1 mutation-related nephropathy determine the clinical type, pathological features, and renal survival time of the disease; and there was a strong correlation between the genotype and clinical phenotype.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xin Liao
- Department of Nephrology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
16
|
Tabei A, Sakairi T, Hamatani H, Ohishi Y, Watanabe M, Nakasatomi M, Ikeuchi H, Kaneko Y, Kopp JB, Hiromura K. The miR-143/145 cluster induced by TGF-β1 suppresses Wilms' tumor 1 expression in cultured human podocytes. Am J Physiol Renal Physiol 2023; 325:F121-F133. [PMID: 37167274 PMCID: PMC10511167 DOI: 10.1152/ajprenal.00313.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/01/2023] [Accepted: 05/01/2023] [Indexed: 05/13/2023] Open
Abstract
Transforming growth factor (TGF)-β1 contributes to podocyte injury in various glomerular diseases, including diabetic kidney disease, probably at least in part by attenuating the expression of Wilms' tumor 1 (WT1). However, the precise mechanisms remain to be defined. We performed miRNA microarray analysis in a human podocyte cell line cultured with TGF-β1 to examine the roles of miRNAs in podocyte damage. The microarray analysis identified miR-143-3p as the miRNA with the greatest increase following exposure to TGF-β1. Quantitative RT-PCR confirmed a significant increase in the miR-143-3p/145-5p cluster in TGF-β1-supplemented cultured podocytes and demonstrated upregulation of miR-143-3p in the glomeruli of mice with type 2 diabetes. Ectopic expression of miR-143-3p and miR-145-5p suppressed WT1 expression in cultured podocytes. Furthermore, inhibition of Smad or mammalian target of rapamycin signaling each partially reversed the TGF-β1-induced increase in miR-143-3p/145-5p and decrease in WT1. In conclusion, TGF-β1 induces expression of miR-143-3p/145-5p in part through Smad and mammalian target of rapamycin pathways, and miR-143-3p/145-5p reduces expression of WT1 in cultured human podocytes. miR-143-3p/145-5p may contribute to TGF-β1-induced podocyte injury.NEW & NOTEWORTHY This study by miRNA microarray analysis demonstrated that miR-143-3p expression was upregulated in cultured human podocytes following exposure to transforming growth factor (TGF)-β1. Furthermore, we report that the miR-143/145 cluster contributes to decreased expression of Wilms' tumor 1, which represents a possible mechanism for podocyte injury induced by TGF-β1. This study is important because it presents a novel mechanism for TGF-β-associated glomerular diseases, including diabetic kidney disease (DKD), and suggests potential therapeutic strategies targeting miR-143-3p/145-5p.
Collapse
Affiliation(s)
- Akifumi Tabei
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Toru Sakairi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hiroko Hamatani
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yuko Ohishi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Mitsuharu Watanabe
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Masao Nakasatomi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Hidekazu Ikeuchi
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Yoriaki Kaneko
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Keiju Hiromura
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Gunma, Japan
| |
Collapse
|
17
|
Hong X, Nie H, Deng J, Liang S, Chen L, Li J, Gong S, Wang G, Zuo W, Hou F, Zhang F. WT1 + glomerular parietal epithelial progenitors promote renal proximal tubule regeneration after severe acute kidney injury. Theranostics 2023; 13:1311-1324. [PMID: 36923529 PMCID: PMC10008742 DOI: 10.7150/thno.79326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/07/2023] [Indexed: 03/14/2023] Open
Abstract
Rationale: Mammalian renal proximal tubules can partially regenerate after acute kidney injury (AKI). However, cells participating in the renal proximal tubule regeneration remain to be elucidated. Wilms' tumor 1 (WT1) expresses in a subtype of glomeruli parietal epithelial cells (PECs) in adult kidneys, it remains unclear whether these WT1+ PECs play a role in renal regeneration/repair after AKI. Methods: Ischemia-reperfusion injury (IRI) mouse model was used to investigate the expression pattern of WT1 in the kidney after severe AKI. Conditional deletion of WT1 gene mice were generated using Pax8CreERT2 and WT1fl/fl mice to examine the function of WT1. Then, genetic cell lineage tracing and single-cell RNA sequencing were performed to illustrate that WT1+ PECs develop into WT1+ proximal tubular epithelial cells (PTECs). Furthermore, in vitro clonogenicity, direct differentiation analysis and in vivo transplantation were used to reveal the stem cell-like properties of these WT1+ PECs. Results: The expression of WT1 protein in PECs and PTECs was increased after severe AKI. Conditional deletion of WT1 gene in PTECs and PECs aggravated renal tubular injury after severe AKI. WT1+ PECs develop into WT1+ PTECs via the transient scattered tubular cell stage, and these WT1+ PECs possess specific stem cell-like properties. Conclusions: We discovered a group of WT1+ PECs that promote renal proximal tubule regeneration/repair after severe AKI, and the expression of WT1 in PECs and PTECs is essential for renal proximal tubule regeneration after severe kidney injury.
Collapse
Affiliation(s)
- Xizhen Hong
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China.,Division of Nephrology, Second Affiliated Hospital of Zhejiang University School of Medicine, No.88, Jiefang Road, Shangcheng District, Hangzhou, Zhejiang, 310009, China
| | - Hao Nie
- East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.,Kiangnan Stem Cell Institute, Zhejiang 311300, China
| | - Juan Deng
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China
| | - Shiting Liang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China
| | - Liting Chen
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China
| | - Jing Li
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China
| | - Siqiao Gong
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China
| | - Guobao Wang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China
| | - Wei Zuo
- East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.,Kiangnan Stem Cell Institute, Zhejiang 311300, China
| | - Fanfan Hou
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| | - Fujian Zhang
- Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China.,National Clinical Research Center of Kidney Diseases, Nanfang Hospital, Guangzhou, China.,Guangdong Provincial Clinical Research Center for Kidney Disease, Guangzhou, China.,Guangdong Provincial Key Laboratory of Renal Failure Research, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, 510005 Guangzhou, China
| |
Collapse
|
18
|
Tekguc M, Gaal RCVAN, Uzel SGM, Gupta N, Riella LV, Lewis JA, Morizane R. Kidney organoids: a pioneering model for kidney diseases. Transl Res 2022; 250:1-17. [PMID: 35750295 PMCID: PMC9691572 DOI: 10.1016/j.trsl.2022.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 11/18/2022]
Abstract
The kidney is a vital organ that regulates the bodily fluid and electrolyte homeostasis via tailored urinary excretion. Kidney injuries that cause severe or progressive chronic kidney disease have driven the growing population of patients with end-stage kidney disease, leading to substantial patient morbidity and mortality. This irreversible kidney damage has also created a huge socioeconomical burden on the healthcare system, highlighting the need for novel translational research models for progressive kidney diseases. Conventional research methods such as in vitro 2D cell culture or animal models do not fully recapitulate complex human kidney diseases. By contrast, directed differentiation of human induced pluripotent stem cells enables in vitro generation of patient-specific 3D kidney organoids, which can be used to model acute or chronic forms of hereditary, developmental, and metabolic kidney diseases. Furthermore, when combined with biofabrication techniques, organoids can be used as building blocks to construct vascularized kidney tissues mimicking their in vivo counterpart. By applying gene editing technology, organoid building blocks may be modified to minimize the process of immune rejection in kidney transplant recipients. In the foreseeable future, the universal kidney organoids derived from HLA-edited/deleted induced pluripotent stem cell (iPSC) lines may enable the supply of bioengineered organotypic kidney structures that are immune-compatible for the majority of the world population. Here, we summarize recent advances in kidney organoid research coupled with novel technologies such as organoids-on-chip and biofabrication of 3D kidney tissues providing convenient platforms for high-throughput drug screening, disease modelling, and therapeutic applications.
Collapse
Affiliation(s)
- Murat Tekguc
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Harvard Stem Cell Institute (HSCI), Cambridge, Massachusetts
| | - Ronald C VAN Gaal
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts; School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Sebastien G M Uzel
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts; School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Navin Gupta
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Harvard Stem Cell Institute (HSCI), Cambridge, Massachusetts
| | - Leonardo V Riella
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Jennifer A Lewis
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts; School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Ryuji Morizane
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Harvard Stem Cell Institute (HSCI), Cambridge, Massachusetts; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts.
| |
Collapse
|
19
|
Luan J, Kopp JB, Zhou H. N6-methyladenine RNA Methylation Epigenetic Modification and Kidney Diseases. Kidney Int Rep 2022; 8:36-50. [PMID: 36644366 PMCID: PMC9831943 DOI: 10.1016/j.ekir.2022.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/01/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022] Open
Abstract
RNA methylation modification is a rapidly developing field in epigenetics. N6-methyladensine (m6A) is the most common internal modification in eukaryotic mRNA. m6A group regulates RNA splicing, stability, translocation, and translation. Enzymes catalyzing this process were termed as writers, erasers, and readers. Recent studies have focused on exploring the role of RNA methylation in human diseases. RNA methylation modifications, particularly m6A, play important roles in the pathogenesis of kidney diseases. In this review, we provide a brief description of m6A and summarize the impact of m6A on acute and chronic kidney disease (CKD) and possible future study directions for this research.
Collapse
Affiliation(s)
- Junjun Luan
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jeffrey B. Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases/National Institutes of Health, Bethesda, Maryland, USA,Jeffrey B. Kopp, Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases/National Institutes of Health, 10 Center Drive, 3N116, Bethesda, Maryland 20892-1268, USA.
| | - Hua Zhou
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China,Correspondence: Hua Zhou, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning 110004, China.
| |
Collapse
|
20
|
Ahmed SF, Alimusina M, Batista RL, Domenice S, Lisboa Gomes N, McGowan R, Patjamontri S, Mendonca BB. The Use of Genetics for Reaching a Diagnosis in XY DSD. Sex Dev 2022; 16:207-224. [DOI: 10.1159/000524881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/03/2022] [Indexed: 11/19/2022] Open
Abstract
Reaching a firm diagnosis is vital for the long-term management of a patient with a difference or disorder of sex development (DSD). This is especially the case in XY DSD where the diagnostic yield is particularly low. Molecular genetic technology is playing an increasingly important role in the diagnostic process, and it is highly likely that it will be used more often at an earlier stage in the diagnostic process. In many cases of DSD, the clinical utility of molecular genetics is unequivocally clear, but in many other cases there is a need for careful exploration of the benefit of genetic diagnosis through long-term monitoring of these cases. Furthermore, the incorporation of molecular genetics into the diagnostic process requires a careful appreciation of the strengths and weaknesses of the evolving technology, and the interpretation of the results requires a clear understanding of the wide range of conditions that are associated with DSD.
Collapse
|
21
|
Zhai R, Liu Y, Tong J, Yu Y, Yang L, Gu Y, Niu J. Empagliflozin Ameliorates Preeclampsia and Reduces Postpartum Susceptibility to Adriamycin in a Mouse Model Induced by Angiotensin Receptor Agonistic Autoantibodies. Front Pharmacol 2022; 13:826792. [PMID: 35401209 PMCID: PMC8984158 DOI: 10.3389/fphar.2022.826792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/08/2022] [Indexed: 01/10/2023] Open
Abstract
Preeclampsia (PE) is the leading cause of maternal and perinatal morbidity and mortality and also is a risk factor for cardiovascular and kidney disease later in life. PE is associated with oversecretion of autoantibodies against angiotensin II type 1 receptor (AT1-AA) by the placenta into the maternal circulation. Here, we sought to determine the therapeutic value of the sodium-glucose co-transporter 2 (SGLT2) inhibitor empagliflozin (EMPA) in mice with AT1-AA-induced preeclampsia. Pregnant mice were injected with AT1-AA at gestation day (GD) 13 and treated daily with EMPA until GD 19, at which point some of the maternal mice were sacrificed and assessed. The other maternal mice were labored on time and challenged with adriamycin (ADR) at 12 weeks postpartum; their offspring were assessed for fetal outcomes. We showed that EMPA treatment significantly relieved high systolic blood pressure and proteinuria and ameliorated kidney injury in PE mice without affecting fetal outcomes. EMPA also ameliorated podocyte injury and oxidative stress, reduced the expression of SGLT2 and activated the AMPK/SIRT1 signaling pathway in vivo and in vitro. Remarkably, EMPA treatment during pregnancy reduced ADR-induced kidney and podocyte injury postpartum. These findings suggest that EMPA could be a potential pharmacological agent for PE.
Collapse
Affiliation(s)
- Ruonan Zhai
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yuan Liu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Jiahao Tong
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Ying Yu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Lin Yang
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| | - Yong Gu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China.,Department of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianying Niu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Zhao D, Zhu L, Zhang S, Guo Z, Wang L, Pan T, Sa R, Chen Z, Jiang J, Chen G. Case Report: Successful ABO-Incompatible Deceased Donor Kidney Transplantation in an Infant Without Pre-transplant Immunological Treatment. Front Med (Lausanne) 2022; 9:838738. [PMID: 35308516 PMCID: PMC8924516 DOI: 10.3389/fmed.2022.838738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/27/2022] [Indexed: 11/15/2022] Open
Abstract
ABO blood group antibodies have not been generated or are at low titer during early infancy. Therefore, in theory, ABO-incompatible kidney transplantation (ABOi KT) may be successfully achieved in small infants without any pre-transplant treatment. We report here the first ABO-incompatible deceased donor kidney transplantation (ABOi DDKT) in an infant. The recipient infant was ABO blood group O, and the donor group A. The recipient was diagnosed with a Wilms tumor gene 1 (WT1) mutation and had received peritoneal dialysis for 4 months prior to transplant. At 7 months and 27 days of age, the infant underwent bilateral native nephrectomy and single-kidney transplantation from a 3-year-old brain-dead donor. No pre- or post-transplantation antibody removal treatment was performed, since the recipient's anti-iso-hemagglutinin-A Ig-M/G antibody titers were both low (1:2) before transplantation and have remained at low levels or undetectable to date. At 11 months post-transplant, the recipient is at home, thriving, with normal development and graft function. This outcome suggests that ABOi DDKT without antibody removal preparatory treatment is feasible in small infants, providing a new option for kidney transplantation in this age range.
Collapse
Affiliation(s)
- Daqiang Zhao
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lan Zhu
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, China
| | - Shengyuan Zhang
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiliang Guo
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Wang
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, China
| | - Tianhui Pan
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rula Sa
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhishui Chen
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, China
| | - Jipin Jiang
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, China
| | - Gang Chen
- Institution of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Ministry of Public Health, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
23
|
Hopfenmüller VL, Perner B, Reuter H, Bates TJD, Große A, Englert C. The Wilms Tumor Gene wt1a Contributes to Blood-Cerebrospinal Fluid Barrier Function in Zebrafish. Front Cell Dev Biol 2022; 9:809962. [PMID: 35087838 PMCID: PMC8786916 DOI: 10.3389/fcell.2021.809962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022] Open
Abstract
The Wilms tumor suppressor gene Wt1 encodes a zinc finger transcription factor, which is highly conserved among vertebrates. It is a key regulator of urogenital development and homeostasis but also plays a role in other organs including the spleen and the heart. More recently additional functions for Wt1 in the mammalian central nervous system have been described. In contrast to mammals, bony fish possess two paralogous Wt1 genes, namely wt1a and wt1b. By performing detailed in situ hybridization analyses during zebrafish development, we discovered new expression domains for wt1a in the dorsal hindbrain, the caudal medulla and the spinal cord. Marker analysis identified wt1a expressing cells of the dorsal hindbrain as ependymal cells of the choroid plexus in the myelencephalic ventricle. The choroid plexus acts as a blood-cerebrospinal fluid barrier and thus is crucial for brain homeostasis. By employing wt1a mutant larvae and a dye accumulation assay with fluorescent tracers we demonstrate that Wt1a is required for proper choroid plexus formation and function. Thus, Wt1a contributes to the barrier properties of the choroid plexus in zebrafish, revealing an unexpected role for Wt1 in the zebrafish brain.
Collapse
Affiliation(s)
| | - Birgit Perner
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Hanna Reuter
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Thomas J D Bates
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Andreas Große
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany
| | - Christoph Englert
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Jena, Germany.,Institute of Biochemistry and Biophysics, Friedrich-Schiller-University Jena, Jena, Germany
| |
Collapse
|
24
|
HEK293-Conditioned Medium Altered the Expression of Renal Markers WT1, CD2AP, and CDH16 in the Human Adipose Mesenchymal Stem Cells. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2022. [DOI: 10.1007/s40883-021-00246-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Omidali M, Korani M. Study the effect of crocin on the expression of PAX2 and WT1 genes in renal tissues and serum levels of NGAL and cystatin C in cadmium-treated rats. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
He S, Yang L, Xiao Z, Tang K, Xu D. Identification of key carcinogenic genes in Wilms' tumor. Genes Genet Syst 2021; 96:141-149. [PMID: 34334530 DOI: 10.1266/ggs.21-00015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
This study aimed to probe carcinogenic genes and pathways associated with Wilms' tumor (WT) onset and malignancy progression. After screening, three datasets acquired from the Gene Expression Omnibus database were analyzed. Differentially expressed genes (DEGs) were identified and GO functional enrichment, KEGG pathway enrichment and protein-protein interaction (PPI) were analyzed. The DEGs with top fold change values or top protein interaction scores were used to analyze overall survival based on the TARGET WT dataset. Together, 866 up-regulated genes in GDS1791, 585 up-regulated genes in GDS2010, and 277 down-regulated genes in GDS4802 were found, from which 46 key DEGs were selected for further analysis. In the PPI network, hub positions included COL5A1, COL4A1, ARPP21, SPARCL1, CD86, LY96 and PPP1R12B. The top DEGs (ARPP21, SYNPO, PRRC2B, PPP1R12B, EFCAB2 and LY96) were selected for survival analysis, and they consistently showed a significantly positive correlation with poor survival. Together, five key carcinogenic genes (SYNPO, PRRC2B, PPP1R12B, EFCAB2 and LY96) were highly associated with WT onset and patient survival. These risk genes, interaction networks and enrichments should improve our understanding of the complex molecular mechanisms in WT development and help clinical applications.
Collapse
Affiliation(s)
- Shaohua He
- Department of Pediatric Surgery, Fujian Provincial Hospital
| | | | - Zhixiang Xiao
- Department of Pediatric Surgery, Fujian Provincial Hospital
| | - Kunbin Tang
- Department of Pediatric Surgery, Fujian Provincial Hospital
| | - Di Xu
- Department of Pediatric Surgery, Fujian Provincial Hospital
| |
Collapse
|
27
|
Yang J, Zhang D, Motojima M, Kume T, Hou Q, Pan Y, Duan A, Zhang M, Jiang S, Hou J, Shi J, Qin Z, Liu Z. Super-Enhancer-Associated Transcription Factors Maintain Transcriptional Regulation in Mature Podocytes. J Am Soc Nephrol 2021; 32:1323-1337. [PMID: 33771836 PMCID: PMC8259645 DOI: 10.1681/asn.2020081177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 01/30/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Transcriptional programs control cell fate, and identifying their components is critical for understanding diseases caused by cell lesion, such as podocytopathy. Although many transcription factors (TFs) are necessary for cell-state maintenance in glomeruli, their roles in transcriptional regulation are not well understood. METHODS The distribution of H3K27ac histones in human glomerulus cells was analyzed to identify superenhancer-associated TFs, and ChIP-seq and transcriptomics were performed to elucidate the regulatory roles of the TFs. Transgenic animal models of disease were further investigated to confirm the roles of specific TFs in podocyte maintenance. RESULTS Superenhancer distribution revealed a group of potential TFs in core regulatory circuits in human glomerulus cells, including FOXC1/2, WT1, and LMX1B. Integration of transcriptome and cistrome data of FOXC1/2 in mice resolved transcriptional regulation in podocyte maintenance. FOXC1/2 regulated differentiation-associated transcription in mature podocytes. In both humans and animal models, mature podocyte injury was accompanied by deregulation of FOXC1/2 expression, and FOXC1/2 overexpression could protect podocytes in zebrafish. CONCLUSIONS FOXC1/2 maintain podocyte differentiation through transcriptional stabilization. The genome-wide chromatin resources support further investigation of TFs' regulatory roles in glomeruli transcription programs.
Collapse
Affiliation(s)
- Jingping Yang
- Medical School of Nanjing University, Nanjing, China
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Difei Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Masaru Motojima
- Department of Clinical Pharmacology, Tokai University School of Medicine, Isehara, Japan
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Qing Hou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yu Pan
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Aiping Duan
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Song Jiang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jinhua Hou
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jingsong Shi
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhaohui Qin
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Zhihong Liu
- Medical School of Nanjing University, Nanjing, China
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
28
|
Sun X, Yi J, Yang J, Han Y, Qian X, Liu Y, Li J, Lu B, Zhang J, Pan X, Liu Y, Liang M, Chen E, Liu P, Lu Y. An integrated epigenomic-transcriptomic landscape of lung cancer reveals novel methylation driver genes of diagnostic and therapeutic relevance. Am J Cancer Res 2021; 11:5346-5364. [PMID: 33859751 PMCID: PMC8039961 DOI: 10.7150/thno.58385] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/21/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Aberrant DNA methylation occurs commonly during carcinogenesis and is of clinical value in human cancers. However, knowledge of the impact of DNA methylation changes on lung carcinogenesis and progression remains limited. Methods: Genome-wide DNA methylation profiles were surveyed in 18 pairs of tumors and adjacent normal tissues from non-small cell lung cancer (NSCLC) patients using Reduced Representation Bisulfite Sequencing (RRBS). An integrated epigenomic-transcriptomic landscape of lung cancer was depicted using the multi-omics data integration method. Results: We discovered a large number of hypermethylation events pre-marked by poised promoter in embryonic stem cells, being a hallmark of lung cancer. These hypermethylation events showed a high conservation across cancer types. Eight novel driver genes with aberrant methylation (e.g., PCDH17 and IRX1) were identified by integrated analysis of DNA methylome and transcriptome data. Methylation level of the eight genes measured by pyrosequencing can distinguish NSCLC patients from lung tissues with high sensitivity and specificity in an independent cohort. Their tumor-suppressive roles were further experimentally validated in lung cancer cells, which depend on promoter hypermethylation. Similarly, 13 methylation-driven ncRNAs (including 8 lncRNAs and 5 miRNAs) were identified, some of which were co-regulated with their host genes by the same promoter hypermethylation. Finally, by analyzing the transcription factor (TF) binding motifs, we uncovered sets of TFs driving the expression of epigenetically regulated genes and highlighted the epigenetic regulation of gene expression of TCF21 through DNA methylation of EGR1 binding motifs. Conclusions: We discovered several novel methylation driver genes of diagnostic and therapeutic relevance in lung cancer. Our findings revealed that DNA methylation in TF binding motifs regulates target gene expression by affecting the binding ability of TFs. Our study also provides a valuable epigenetic resource for identifying DNA methylation-based diagnostic biomarkers, developing cancer drugs for epigenetic therapy and studying cancer pathogenesis.
Collapse
|
29
|
Panfoli I, Granata S, Candiano G, Verlato A, Lombardi G, Bruschi M, Zaza G. Analysis of urinary exosomes applications for rare kidney disorders. Expert Rev Proteomics 2021; 17:735-749. [PMID: 33395324 DOI: 10.1080/14789450.2020.1866993] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Exosomes are nanovesicles that play important functions in a variety of physiological and pathological conditions. They are powerful cell-to-cell communication tool thanks to the protein, mRNA, miRNA, and lipid cargoes they carry. They are also emerging as valuable diagnostic and prognostic biomarker sources. Urinary exosomes carry information from all the cells of the urinary tract, downstream of the podocyte. Rare kidney diseases are a subset of an inherited diseases whose genetic diagnosis can be unclear, and presentation can vary due to genetic, epigenetic, and environmental factors. Areas covered: In this review, we focus on a group of rare and often neglected kidney diseases, for which we have sufficient available literature data on urinary exosomes. The analysis of their content can help to comprehend pathological mechanisms and to identify biomarkers for diagnosis, prognosis, and therapeutic targets. Expert opinion: The foreseeable large-scale application of system biology approach to the profiling of exosomal proteins as a source of renal disease biomarkers will be also useful to stratify patients with rare kidney diseases whose penetrance, phenotypic presentation, and age of onset vary sensibly. This can ameliorate the clinical management.
Collapse
Affiliation(s)
- Isabella Panfoli
- Department of Pharmacy-DIFAR, University of Genoa , Genoa, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Giovanni Candiano
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini , Genoa, Italy
| | - Alberto Verlato
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Gianmarco Lombardi
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini , Genoa, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University-Hospital of Verona , Verona, Italy
| |
Collapse
|
30
|
Chen Q, Chen J, Wang C, Chen X, Liu J, Zhou L, Liu Y. MicroRNA-466o-3p mediates β-catenin-induced podocyte injury by targeting Wilms tumor 1. FASEB J 2020; 34:14424-14439. [PMID: 32888352 DOI: 10.1096/fj.202000464r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/04/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022]
Abstract
Podocytes are highly specialized cells that play an essential role in maintaining the integrity and function of the glomerular filtration barrier. Wilms tumor 1 (WT1) and β-catenin are two master regulators that play opposing roles in podocyte biology and mutually antagonize each other. However, exactly how β-catenin inhibits WT1 remains incompletely understood. In this study, we demonstrated the role of miR-466o-3p in mediating β-catenin-triggered podocyte injury by targeting WT1. The expression of miR-466o-3p was upregulated in cultured podocytes after β-catenin activation and in glomerular podocytes in adriamycin (ADR) nephropathy, remnant kidney after 5/6 renal ablation, and diabetic kidney disease. Bioinformatics analysis and luciferase reporter assay confirmed that miR-466o-3p directly targeted WT1 mRNA. Furthermore, overexpression of miR-466o-3p downregulated WT1 protein and promoted podocyte injury in vitro. Conversely, inhibition of miR-466o-3p alleviated β-catenin-induced podocyte dysfunction. In mouse model of ADR nephropathy, overexpression of miR-466o-3p inhibited WT1, aggravated podocytes injury and deteriorated proteinuria. In contrast, inhibition of renal miR-466o-3p by antagomiR, either prior to or after ADR injection, substantially restored WT1, alleviated podocytes injury and reduced renal fibrosis. These studies reveal a critical role for miR-466o-3p, a novel microRNA that has not been characterized previously, in mediating β-catenin-triggered WT1 inhibition. Our findings also uncover a new pathogenic mechanism by which β-catenin promotes podocyte injury and proteinuria in glomerular diseases.
Collapse
Affiliation(s)
- Qiyan Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiongcheng Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunhong Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaowen Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiafeng Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
31
|
Weiss AC, Rivera-Reyes R, Englert C, Kispert A. Expansion of the renal capsular stroma, ureteric bud branching defects and cryptorchidism in mice with Wilms tumor 1 gene deletion in the stromal compartment of the developing kidney. J Pathol 2020; 252:290-303. [PMID: 32715478 DOI: 10.1002/path.5518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022]
Abstract
Development of the mammalian kidney is orchestrated by reciprocal interactions of stromal and nephrogenic mesenchymal cells with the ureteric bud epithelium. Previous work showed that the transcription factor Wilms tumor 1 (WT1) acts in the nephrogenic lineage to maintain precursor cells, to drive the epithelial transition of aggregating precursors into a renal vesicle and to specify and maintain the podocyte fate. However, WT1 is expressed not only in the nephrogenic lineage but also transiently in stromal progenitors in the renal cortex. Here we report that specific deletion of Wt1 in the stromal lineage using the Foxd1cre driver line results at birth in cryptorchidism and hypoplastic kidneys that harbour fewer and enlarged ureteric bud tips and display an expansion of capsular stroma into the cortical region. In vivo and ex vivo analysis at earlier stages revealed that stromal loss of Wt1 reduces stromal proliferation, and delays and alters branching morphogenesis, resulting in a variant architecture of the collecting duct tree with an increase of single at the expense of bifurcated ureteric bud tips. Molecular analysis identified a transient reduction of Aldh1a2 expression and of retinoic acid signalling activity in stromal progenitors, and of Ret in ureteric bud tips. Administration of retinoic acid partly rescued the branching defects of mutant kidneys in culture. We propose that WT1 maintains retinoic acid signalling in the cortical stroma, which, in turn, assures proper levels and dynamics of Ret expression in the ureteric bud tips, and thus normal ramification of the ureteric tree. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Anna-Carina Weiss
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | | | - Christoph Englert
- Molecular Genetics, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| |
Collapse
|
32
|
Downregulation of megalin, cubilin, ClC-5 and podocin in Fabry nephropathy: potential implications in the decreased effectiveness of enzyme replacement therapy. J Nephrol 2020; 34:1307-1314. [PMID: 32840752 DOI: 10.1007/s40620-020-00835-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/11/2020] [Indexed: 01/21/2023]
Abstract
Fabry disease is an X-linked disorder due to mutations in α-galactosidase A, resulting in the accumulation of enzyme substrates and cell malfunction. Kidney involvement is frequent, affecting all native kidney cell types. Podocyte damage results in proteinuria and chronic kidney disease. End-stage kidney disease is the rule in middle-aged males and some females with the classic phenotype. In podocytes and kidney proximal tubular cells, megalin is one of the molecules involved in enzyme replacement therapy (ERT) cellular absorption. After podocyte damage, podocin concentration is decreased and contributes to progressive proteinuria. We report in a male and a female patient the decreased expression of megalin, cubilin, ClC-5 and podocin compared to controls and chronic kidney disease (CKD) biopsies. Moreover, the decrease in ClC-5, a molecule engaged in endosomal-lysosomal acidification, could also affect ERT. These findings may partially explain some of the dysfunctions described in Fabry nephropathy and could highlight possible alterations in the pharmacokinetics of the delivered enzyme.
Collapse
|
33
|
Cell polarity and oncogenesis: common mutations contribute to altered cellular polarity and promote malignancy. THE NUCLEUS 2020. [DOI: 10.1007/s13237-020-00313-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
34
|
Ettou S, Jung YL, Miyoshi T, Jain D, Hiratsuka K, Schumacher V, Taglienti ME, Morizane R, Park PJ, Kreidberg JA. Epigenetic transcriptional reprogramming by WT1 mediates a repair response during podocyte injury. SCIENCE ADVANCES 2020; 6:eabb5460. [PMID: 32754639 PMCID: PMC7380960 DOI: 10.1126/sciadv.abb5460] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/10/2020] [Indexed: 06/11/2023]
Abstract
In the context of human disease, the mechanisms whereby transcription factors reprogram gene expression in reparative responses to injury are not well understood. We have studied the mechanisms of transcriptional reprogramming in disease using murine kidney podocytes as a model for tissue injury. Podocytes are a crucial component of glomeruli, the filtration units of each nephron. Podocyte injury is the initial event in many processes that lead to end-stage kidney disease. Wilms tumor-1 (WT1) is a master regulator of gene expression in podocytes, binding nearly all genes known to be crucial for maintenance of the glomerular filtration barrier. Using murine models and human kidney organoids, we investigated WT1-mediated transcriptional reprogramming during the course of podocyte injury. Reprogramming the transcriptome involved highly dynamic changes in the binding of WT1 to target genes during a reparative injury response, affecting chromatin state and expression levels of target genes.
Collapse
Affiliation(s)
- Sandrine Ettou
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Youngsook L. Jung
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Tomoya Miyoshi
- Nephrology Division, Massachusetts General Hospital, Boston, MA 02114, USA
- Renal Division, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Dhawal Jain
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Ken Hiratsuka
- Nephrology Division, Massachusetts General Hospital, Boston, MA 02114, USA
- Renal Division, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
| | - Valerie Schumacher
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Mary E. Taglienti
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ryuji Morizane
- Nephrology Division, Massachusetts General Hospital, Boston, MA 02114, USA
- Renal Division, Brigham and Women’s Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Peter J. Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Jordan A. Kreidberg
- Department of Urology, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
35
|
Münch J, Kirschner KM, Schlee H, Kraus C, Schönauer R, Jin W, Le Duc D, Scholz H, Halbritter J. Autosomal dominant polycystic kidney disease in absence of renal cyst formation illustrates genetic interaction between WT1 and PKD1. J Med Genet 2020; 58:jmedgenet-2019-106633. [PMID: 32381729 DOI: 10.1136/jmedgenet-2019-106633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 03/05/2020] [Accepted: 03/20/2020] [Indexed: 11/04/2022]
Abstract
PURPOSE Autosomal dominant polycystic kidney disease (ADPKD), caused by pathogenic variants of either PKD1 or PKD2, is characterised by wide interfamilial and intrafamilial phenotypic variability. This study aimed to determine the molecular basis of marked clinical variability in ADPKD family members and sought to analyse whether alterations of WT1 (Wilms tumour 1), encoding a regulator of gene expression, may have an impact on renal cyst formation. METHODS ADPKD family members underwent clinical and molecular evaluation. Functionally, Pkd1 mRNA and protein expression upon Wt1 knockdown was evaluated in mouse embryonic kidneys and mesonephric M15 cells. RESULTS By renal gene panel analysis, we identified two pathogenic variants in an individual with maternal history of ADPKD, however, without cystic kidneys but polycystic liver disease: a known PKD1 missense variant (c.8311G>A, p.Glu2771Lys) and a known de novo WT1 splice site variant (c.1432+4C>T). The latter was previously associated with imbalanced +/-KTS isoform ratio of WT1. In ex vivo organ cultures from mouse embryonic kidneys, Wt1 knockdown resulted in decreased Pkd1 expression on mRNA and protein level. CONCLUSION While the role of WT1 in glomerulopathies has been well established, this report by illustrating genetic interaction with PKD1 proposes WT1 as potential modifier in ADPKD.
Collapse
Affiliation(s)
- Johannes Münch
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Saxony, Germany
| | - Karin M Kirschner
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Berlin, Germany
| | - Hendrik Schlee
- Dialysis Weissenfels, Nephrology Burgenlandkreis, Weissenfels, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Ria Schönauer
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Saxony, Germany
| | - Wenjun Jin
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Saxony, Germany
| | - Diana Le Duc
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Saxony, Germany
| | - Holger Scholz
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Berlin, Germany
| | - Jan Halbritter
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Saxony, Germany
| |
Collapse
|
36
|
Eroglu FK, Orhan D, İnözü M, Duzova A, Gulhan B, Ozaltin F, Topaloglu R. CD80 expression and infiltrating regulatory T cells in idiopathic nephrotic syndrome of childhood. Pediatr Int 2019; 61:1250-1256. [PMID: 31513327 DOI: 10.1111/ped.14005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/05/2019] [Accepted: 09/06/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND CD80 (also known as B7-1) is a co-stimulatory molecule that is expressed in biopsies and also excreted in urine in patients with minimal change disease (MCD) and focal segmental glomerulosclerosis (FSGS). CD80 is inhibited by the cytotoxic T-lymphocyte-associated-antigen 4 (CTLA4), which is mainly expressed on regulatory T cells (Tregs). Ineffective circulating Treg response is involved in the pathogenesis of nephrotic syndrome. In this study, we evaluated CD80 expression and infiltrating Tregs in children with MCD and FSGS. METHODS Evaluation of CD80 expression and semi-quantitative evaluation of Tregs (FOXP3-positive CD4 T cells) were carried out in 31 kidney biopsies (12 MCD, 19 FSGS) with immunofluorescence and immunohistochemistry staining. RESULTS All MCD sections were stained negative; whereas six out of 19 FSGS sections (all from steroid-resistant (SR) patients), including one from a Wilms' tumor 1 (WT1) mutation-positive FSGS patient, stained positive for anti-CD80 goat antibody, and negative for anti-CD80 rabbit antibody. FSGS biopsy specimens had significantly higher FOXP3-positive cells/mm2 compared with MCD and control samples (P < 0.001). Biopsy samples from SR-FSGS patients (n = 12) with positive CD80 staining (n = 6) had significantly less Tregs (FOXP3-positive CD4 T cells) compared with CD80 (-) biopsies (n = 6; P = 0.004). CONCLUSION CD80 expression was not detected in the majority of the archival biopsy sections and the results were not consistent across the different antibodies. In the SR-FSGS sections, however, CD80-positive biopsies had decreased FOXP3-positive CD4 T cells, suggesting that a decreased anti-inflammatory milieu may be the cause of increased CD80 expression.
Collapse
Affiliation(s)
- Fehime Kara Eroglu
- Division of Pediatric Nephrology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Diclehan Orhan
- Department of Pathology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Mihriban İnözü
- Division of Pediatric Nephrology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ali Duzova
- Division of Pediatric Nephrology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Bora Gulhan
- Division of Pediatric Nephrology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Fatih Ozaltin
- Division of Pediatric Nephrology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Rezan Topaloglu
- Division of Pediatric Nephrology, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
37
|
Li TS, Chen L, Wang SC, Yang YZ, Xu HJ, Gu HM, Zhao XJ, Dong P, Pan Y, Shang ZQ, Zhang XQ, Kong LD. Magnesium isoglycyrrhizinate ameliorates fructose-induced podocyte apoptosis through downregulation of miR-193a to increase WT1. Biochem Pharmacol 2019; 166:139-152. [DOI: 10.1016/j.bcp.2019.05.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/08/2019] [Indexed: 02/07/2023]
|
38
|
Hamatani H, Sakairi T, Ikeuchi H, Kaneko Y, Maeshima A, Nojima Y, Hiromura K. TGF‐β1 alters DNA methylation levels in promoter and enhancer regions of the
WT1
gene in human podocytes. Nephrology (Carlton) 2019; 24:575-584. [DOI: 10.1111/nep.13411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Hiroko Hamatani
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Toru Sakairi
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Hidekazu Ikeuchi
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Yoriaki Kaneko
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Akito Maeshima
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| | - Yoshihisa Nojima
- Department of Rheumatology and NephrologyJapan Red Cross Maebashi Hospital Maebashi Japan
| | - Keiju Hiromura
- Department of Nephrology and RheumatologyGunma University Graduate School of Medicine Maebashi Japan
| |
Collapse
|
39
|
Abstract
Wilms tumour is the most common renal malignancy of childhood. The disease is curable in the majority of cases, albeit at considerable cost in terms of late treatment-related effects in some children. However, one in ten children with Wilms tumour will die of their disease despite modern treatment approaches. The genetic changes that underpin Wilms tumour have been defined by studies of familial cases and by unbiased DNA sequencing of tumour genomes. Together, these approaches have defined the landscape of cancer genes that are operative in Wilms tumour, many of which are intricately linked to the control of fetal nephrogenesis. Advances in our understanding of the germline and somatic genetic changes that underlie Wilms tumour may translate into better patient outcomes. Improvements in risk stratification have already been seen through the introduction of molecular biomarkers into clinical practice. A host of additional biomarkers are due to undergo clinical validation. Identifying actionable mutations has led to potential new targets, with some novel compounds undergoing testing in early phase trials. Avenues that warrant further exploration include targeting Wilms tumour cancer genes with a non-redundant role in nephrogenesis and targeting the fetal renal transcriptome.
Collapse
Affiliation(s)
- Taryn Dora Treger
- Wellcome Sanger Institute, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Tanzina Chowdhury
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, London, UK.
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| | - Sam Behjati
- Wellcome Sanger Institute, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| |
Collapse
|
40
|
Chiba Y, Inoue CN. Once-Daily Low-Dose Cyclosporine A Treatment with Angiotensin Blockade for Long-Term Remission of Nephropathy in Frasier Syndrome. TOHOKU J EXP MED 2019; 247:35-40. [PMID: 30651406 DOI: 10.1620/tjem.247.35] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cyclosporine A is known to be effective in some genetic podocyte injury. However, the efficacy of cyclosporine A depends on the degree of histopathological findings, and the relationship between long-term use and renal prognosis remains unknown. Frasier syndrome is a rare genetic disorder caused by intronic mutations in WT1, and is characterized by progressive glomerulopathy, a 46,XY disorder of sex development, and an increased risk of gonadoblastoma. We report here a 16-year-old phenotypically female patient with Frasier syndrome. A renal biopsy at the age of seven years showed segmentally effaced podocyte foot processes with no evidence of glomerulosclerosis. Steroid-resistant proteinuria progressed to the nephrotic range at the age of 10 years, which responded to once-daily administration of cyclosporine A with low two-hour post-dose cyclosporine A (C2) levels; she then achieved stable partial remission in combination with renin-angiotensin system (RAS) blockade. At the age of 12 years, examinations for delayed puberty confirmed the diagnosis of Frasier syndrome. The second renal biopsy showed widespread foot process effacement and a minor lesion of segmental glomerulosclerosis without findings suggestive of cyclosporine A nephropathy. She maintained partial remission and normal renal function with the continuation of once-daily low-dose cyclosporine A. The C2 levels required for the remission were between 212 and 520 ng/ml. Cyclosporine A dosages sufficient for maintaining the C2 levels were 1.1-1.2 mg/kg per day. In conclusion, the long-lasting treatment of once-daily low-dose cyclosporine A with RAS inhibition was effective for induction and maintenance of partial remission in Frasier syndrome.
Collapse
Affiliation(s)
- Yasushi Chiba
- Department of Pediatrics, Red Cross Sendai Hospital.,Department of Pediatrics, Tohoku Rosai Hospital
| | | |
Collapse
|
41
|
|
42
|
Tan W, Zhang X, Wang Z, Chen Y, Wang A, Chu M, Tang B, Li Z. Differential expression of Wilms' tumour 1 gene in porcine urogenital organs during development. Anat Histol Embryol 2018; 48:102-109. [PMID: 30450614 DOI: 10.1111/ahe.12415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 09/07/2018] [Accepted: 10/28/2018] [Indexed: 11/28/2022]
Abstract
Wilms' tumour 1 gene (WT1) is essential for the development of mammalian urogenital system. However, the expression pattern of WT1 in the development of porcine urogenital organs is still unclear. Here, we examined the expression of WT1 mRNA and protein in porcine kidneys, ovaries and testes from embryonic days 35 and 60 (E35d, E60d, n = 3) to the newborn (0d, n = 4) and adult (210d, n = 3) stages, using real-time PCR and immunofluorescent staining. Real-time PCR analysis showed that porcine kidneys, ovaries and testes all expressed high level of WT1 mRNAs, especially in adult testes (p < 0.05 or 0.01 vs. kidney and ovary, respectively). Morphologically, characteristic microstructures of the kidneys, ovaries and testes were observed and discerned at all four stages. Immunofluorescently, WT1 expression was detected in a dynamic and context-specific pattern during the development of these organs. Taken together, porcine urogenital organs express relatively high levels of WT1 mRNA. Dynamical and context-specific expression profile of WT1 in these organs occurs during their development, implying its close association with the development and function of porcine kidney, ovary and testis.
Collapse
Affiliation(s)
- Wentao Tan
- The First Bethune Hospital, Jilin University, Changchun, China
| | - Xueming Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhengzhu Wang
- The First Bethune Hospital, Jilin University, Changchun, China
| | - Yue Chen
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Aibing Wang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Meiran Chu
- The First Bethune Hospital, Jilin University, Changchun, China
| | - Bo Tang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ziyi Li
- The First Bethune Hospital, Jilin University, Changchun, China
| |
Collapse
|
43
|
Yu SMW, Nissaisorakarn P, Husain I, Jim B. Proteinuric Kidney Diseases: A Podocyte's Slit Diaphragm and Cytoskeleton Approach. Front Med (Lausanne) 2018; 5:221. [PMID: 30255020 PMCID: PMC6141722 DOI: 10.3389/fmed.2018.00221] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/18/2018] [Indexed: 01/19/2023] Open
Abstract
Proteinuric kidney diseases are a group of disorders with diverse pathological mechanisms associated with significant losses of protein in the urine. The glomerular filtration barrier (GFB), comprised of the three important layers, the fenestrated glomerular endothelium, the glomerular basement membrane (GBM), and the podocyte, dictates that disruption of any one of these structures should lead to proteinuric disease. Podocytes, in particular, have long been considered as the final gatekeeper of the GFB. This specialized visceral epithelial cell contains a complex framework of cytoskeletons forming foot processes and mediate important cell signaling to maintain podocyte health. In this review, we will focus on slit diaphragm proteins such as nephrin, podocin, TRPC6/5, as well as cytoskeletal proteins Rho/small GTPases and synaptopodin and their respective roles in participating in the pathogenesis of proteinuric kidney diseases. Furthermore, we will summarize the potential therapeutic options targeting the podocyte to treat this group of kidney diseases.
Collapse
Affiliation(s)
- Samuel Mon-Wei Yu
- Department of Medicine, Jacobi Medical Center, Bronx, NY, United States
| | | | - Irma Husain
- Department of Medicine, James J. Peters VA Medical Center, Bronx, NY, United States
| | - Belinda Jim
- Department of Medicine, Jacobi Medical Center, Bronx, NY, United States.,Renal Division, Jacobi Medical Center, Bronx, NY, United States
| |
Collapse
|
44
|
Souza PVS, Badia BML, Silva LHL, Teixeira CAC, Seneor DD, Marin VDGB, Farias IB, Dias RB, Oliveira ASB, Pinto WBVR. Leukodystrophy with disorders of sex development due to WT1 mutations. J Neurol Sci 2018; 390:94-98. [DOI: 10.1016/j.jns.2018.04.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 04/12/2018] [Accepted: 04/12/2018] [Indexed: 11/28/2022]
|
45
|
Li N, Zhang J, Yan X, Zhang C, Liu H, Shan X, Li J, Yang Y, Huang C, Zhang P, Zhang Y, Bu P. SIRT3-KLF15 signaling ameliorates kidney injury induced by hypertension. Oncotarget 2018; 8:39592-39604. [PMID: 28465484 PMCID: PMC5503635 DOI: 10.18632/oncotarget.17165] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/27/2017] [Indexed: 12/11/2022] Open
Abstract
Renal fibrosis participates in the progression of hypertension-induced kidney injury. The effect of SIRT3, a member of the NAD+-dependent deacetylase family, in hypertensive nephropathy remains unclear. In this study, we found that SIRT3 was reduced after angiotensin II (AngII) treatment both in vivo and in vitro. Furthermore, SIRT3-knockout mice aggravated hypertension-induced renal dysfunction and renal fibrosis via chronic AngII infusion (2000 ng/kg per minute for 42 days). On the contrary, SIRT3-overexpression mice attenuated AngII-induced kidney injury compared with wild-type mice. Remarkably, a co-localization of SIRT3 and KLF15, a kidney-enriched nuclear transcription factor, led to SIRT3 directly deacetylating KLF15, followed by decreased expression of fibronectin and collagen type IV in cultured MPC-5 podocytes. In addition, honokiol (HKL), a major bioactive compound isolated from Magnolia officinalis (Houpo), suppressed AngII-induced renal fibrosis through activating SIRT3-KLF15 signaling. Taken together, our findings implicate that a novel SIRT3-KLF15 signaling may prevent kidney injury from hypertension and HKL can act as a SIRT3-KLF15 signaling activator to protect against hypertensive nephropathy.
Collapse
Affiliation(s)
- Na Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jie Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xuefang Yan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chen Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hui Liu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiaolan Shan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jingyuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yi Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Chengmin Huang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Peili Bu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
46
|
Abstract
Ultimately, the common final pathway of any glomerular disease is podocyte effacement, podocyte loss, and, eventually, glomerular scarring. There has been a long-standing debate on the underlying mechanisms for podocyte depletion, ranging from necrosis and apoptosis to detachment of viable cells from the glomerular basement membrane. However, this debate still continues because additional pathways of programmed cell death have been reported in recent years. Interestingly, viable podocytes can be isolated out of the urine of proteinuric patients easily, emphasizing the importance of podocyte detachment in glomerular diseases. In contrast, detection of apoptosis and other pathways of programmed cell death in podocytes is technically challenging. In fact, we still are lacking direct evidence showing, for example, the presence of apoptotic bodies in podocytes, leaving the question unanswered as to whether podocytes undergo mechanisms of programmed cell death. However, understanding the mechanisms leading to podocyte depletion is of particular interest because future therapeutic strategies might interfere with these to prevent glomerular scarring. In this review, we summarize our current knowledge on podocyte cell death, the different molecular pathways and experimental approaches to study these, and, finally, focus on the mechanisms that prevent the onset of programmed cell death.
Collapse
Affiliation(s)
- Fabian Braun
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, University of Cologne, Cologne, Germany
| | - Jan U Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases, University of Cologne, Cologne, Germany.
| |
Collapse
|
47
|
Miao F, Zhang X, Cao Y, Wang Y, Zhang X. Effect of siRNA-silencing of SALL2 gene on growth, migration and invasion of human ovarian carcinoma A2780 cells. BMC Cancer 2017; 17:838. [PMID: 29228922 PMCID: PMC5725831 DOI: 10.1186/s12885-017-3843-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 11/24/2017] [Indexed: 12/28/2022] Open
Abstract
Background The role of Spalt-like gene-2 (SALL2) in tumorigenesis remains incompletely elucidated. This study investigated the effects of SALL2 on human ovarian carcinoma (OC) A2780 cells and the probable mechanism. Methods Expression of SALL2 in human OC cell lines were detected by reverse transcription PCR (RT-PCR) and Western blot analysis. A2780 cells were transfected with small-interfering ribonucleic acid (siRNA) to silence SALL2. SALL2 expression was detected by RT-PCR, Western blot analysis and immunofluorescence assay. Cell proliferation was measured by CCK-8 assay and flow cytometry (FCM). Apoptosis was measured by FCM. Cell migration was detected by real-time cell analysis. Cell invasion was detected by transwell assay. mRNA expression of p21 was detected by quantitative real-time PCR. Western blot analysis was used to determine the expression of matrix metalloproteinase (MMP)2, MMP9, protein kinase B (PKB, also called Akt), and phosphorylated-Akt (p-Akt). Results SALL2 was expressed in six OC cell lines, and the expression was the highest in A2780 cells. Compared with that in the Scramble group, SALL2 expression in A2780 was downregulated after transfection with siRNA-2 and siRNA-3 for 48 h. Compared with that in the Scramble group, proliferation of A2780 cells in the siRNA-2 group increased after transfection for 24, 48 and 72 h. In the siRNA-2 group, the proportion of A2780 cells decreased in the G0/G1 phase, and cell apoptosis decreased after transfection for 48 h. Compared with that in the Scramble group, the cell migration and invasion abilities of A2780 cells increased. Compared with that in the Scramble group, p21 mRNA expression in A2780 cells decreased after transfection with siRNA2. When SALL2 was silenced, the expression of MMP2/9 and p-Akt in A2780 cells increased. Furthermore, the PI3K inhibitor LY294002 could effectively reversed SALL2 siRNA-induced phosphorylation of Akt, migration and invasion of A2780 cells. Conclusion Transient silencing of SALL2 promotes cell proliferation, migration, and invasion, and inhibits apoptosis of A2780 cells. In SALL2 siRNA-silenced cells, p21 expression was decreased. SALL2 knockdown by siRNA induces the migration and invasion of A2780 cells; this phenomenon is possibly associated with the increased expression of MMP2/9 and the activation of the PI3K/Akt signalling pathway. Electronic supplementary material The online version of this article (10.1186/s12885-017-3843-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fang Miao
- School of Basic Medical Sciences, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong, People's Republic of China
| | - Xueshan Zhang
- School of Basic Medical Sciences, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong, People's Republic of China
| | - Yanning Cao
- School of Basic Medical Sciences, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong, People's Republic of China
| | - Yue Wang
- School of Basic Medical Sciences, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong, People's Republic of China
| | - Xiaoshu Zhang
- School of Basic Medical Sciences, Binzhou Medical University, 346 Guanhai Road, Yantai, Shandong, People's Republic of China.
| |
Collapse
|
48
|
Glucocorticoid therapy regulates podocyte motility by inhibition of Rac1. Sci Rep 2017; 7:6725. [PMID: 28751734 PMCID: PMC5532274 DOI: 10.1038/s41598-017-06810-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/19/2017] [Indexed: 02/03/2023] Open
Abstract
Nephrotic syndrome (NS) occurs when the glomerular filtration barrier becomes excessively permeable leading to massive proteinuria. In childhood NS, immune system dysregulation has been implicated and increasing evidence points to the central role of podocytes in the pathogenesis. Children with NS are typically treated with an empiric course of glucocorticoid (Gc) therapy; a class of steroids that are activating ligands for the glucocorticoid receptor (GR) transcription factor. Although Gc-therapy has been the cornerstone of NS management for decades, the mechanism of action, and target cell, remain poorly understood. We tested the hypothesis that Gc acts directly on the podocyte to produce clinically useful effects without involvement of the immune system. In human podocytes, we demonstrated that the basic GR-signalling mechanism is intact and that Gc induced an increase in podocyte barrier function. Defining the GR-cistrome identified Gc regulation of motility genes. These findings were functionally validated with live-cell imaging. We demonstrated that treatment with Gc reduced the activity of the pro-migratory small GTPase regulator Rac1. Furthermore, Rac1 inhibition had a direct, protective effect on podocyte barrier function. Our studies reveal a new mechanism for Gc action directly on the podocyte, with translational relevance to designing new selective synthetic Gc molecules.
Collapse
|
49
|
Sugano Y, Cianciolo Cosentino C, Loffing-Cueni D, Neuhauss SCF, Loffing J. Comparative transcriptomic analysis identifies evolutionarily conserved gene products in the vertebrate renal distal convoluted tubule. Pflugers Arch 2017; 469:859-867. [PMID: 28656378 DOI: 10.1007/s00424-017-2009-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/29/2017] [Accepted: 05/31/2017] [Indexed: 12/11/2022]
Abstract
Understanding the molecular basis of the complex regulatory networks controlling renal ion transports is of major physiological and clinical importance. In this study, we aimed to identify evolutionarily conserved critical players in the function of the renal distal convoluted tubule (DCT) by a comparative transcriptomic approach. We generated a transgenic zebrafish line with expression of the red fluorescent mCherry protein under the control of the zebrafish DCT-specific promoter of the thiazide-sensitive NaCl cotransporter (NCC). The mCherry expression was then used to isolate from the zebrafish mesonephric kidneys the distal late (DL) segments, the equivalent of the mammalian DCT, for subsequent RNA-seq analysis. We next compared this zebrafish DL transcriptome to the previously established mouse DCT transcriptome and identified a subset of gene products significantly enriched in both the teleost DL and the mammalian DCT, including SLCs and nuclear transcription factors. Surprisingly, several of the previously described regulators of NCC (e.g., SPAK, KLHL3, ppp1r1a) in the mouse were not found enriched in the zebrafish DL. Nevertheless, the zebrafish DL expressed enriched levels of related homologues. Functional knockdown of one of these genes, ppp1r1b, reduced the phosphorylation of NCC in the zebrafish pronephros, similar to what was seen previously in knockout mice for its homologue, Ppp1r1a. The present work is the first report on global gene expression profiling in a specific nephron portion of the zebrafish kidney, an increasingly used model system for kidney research. Our study suggests that comparative analysis of gene expression between phylogenetically distant species may be an effective approach to identify novel regulators of renal function.
Collapse
Affiliation(s)
- Yuya Sugano
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Chiara Cianciolo Cosentino
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Dominique Loffing-Cueni
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Stephan C F Neuhauss
- Institute of Molecular Life Sciences, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
| | - Johannes Loffing
- Institute of Anatomy, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland.
- Swiss National Center of Competence in Research "Kidney.CH", Zurich, Switzerland.
| |
Collapse
|
50
|
Poelmann RE, Gittenberger-de Groot AC, Biermans MWM, Dolfing AI, Jagessar A, van Hattum S, Hoogenboom A, Wisse LJ, Vicente-Steijn R, de Bakker MAG, Vonk FJ, Hirasawa T, Kuratani S, Richardson MK. Outflow tract septation and the aortic arch system in reptiles: lessons for understanding the mammalian heart. EvoDevo 2017; 8:9. [PMID: 28491275 PMCID: PMC5424407 DOI: 10.1186/s13227-017-0072-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/03/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cardiac outflow tract patterning and cell contribution are studied using an evo-devo approach to reveal insight into the development of aorto-pulmonary septation. RESULTS We studied embryonic stages of reptile hearts (lizard, turtle and crocodile) and compared these to avian and mammalian development. Immunohistochemistry allowed us to indicate where the essential cell components in the outflow tract and aortic sac were deployed, more specifically endocardial, neural crest and second heart field cells. The neural crest-derived aorto-pulmonary septum separates the pulmonary trunk from both aortae in reptiles, presenting with a left visceral and a right systemic aorta arising from the unseptated ventricle. Second heart field-derived cells function as flow dividers between both aortae and between the two pulmonary arteries. In birds, the left visceral aorta disappears early in development, while the right systemic aorta persists. This leads to a fusion of the aorto-pulmonary septum and the aortic flow divider (second heart field population) forming an avian aorto-pulmonary septal complex. In mammals, there is also a second heart field-derived aortic flow divider, albeit at a more distal site, while the aorto-pulmonary septum separates the aortic trunk from the pulmonary trunk. As in birds there is fusion with second heart field-derived cells albeit from the pulmonary flow divider as the right 6th pharyngeal arch artery disappears, resulting in a mammalian aorto-pulmonary septal complex. In crocodiles, birds and mammals, the main septal and parietal endocardial cushions receive neural crest cells that are functional in fusion and myocardialization of the outflow tract septum. Longer-lasting septation in crocodiles demonstrates a heterochrony in development. In other reptiles with no indication of incursion of neural crest cells, there is either no myocardialized outflow tract septum (lizard) or it is vestigial (turtle). Crocodiles are unique in bearing a central shunt, the foramen of Panizza, between the roots of both aortae. Finally, the soft-shell turtle investigated here exhibits a spongy histology of the developing carotid arteries supposedly related to regulation of blood flow during pharyngeal excretion in this species. CONCLUSIONS This is the first time that is shown that an interplay of second heart field-derived flow dividers with a neural crest-derived cell population is a variable but common, denominator across all species studied for vascular patterning and outflow tract septation. The observed differences in normal development of reptiles may have impact on the understanding of development of human congenital outflow tract malformations.
Collapse
Affiliation(s)
- Robert E Poelmann
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands.,Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | | | - Marcel W M Biermans
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Anne I Dolfing
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Armand Jagessar
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Sam van Hattum
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Amanda Hoogenboom
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Lambertus J Wisse
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, Leiden, The Netherlands
| | - Rebecca Vicente-Steijn
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands.,Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, Leiden, The Netherlands
| | - Merijn A G de Bakker
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| | - Freek J Vonk
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands.,Naturalis Biodiversity Center, Darwinweg 2, Leiden, The Netherlands
| | - Tatsuya Hirasawa
- Laboratory for Evolutionary Morphology, RIKEN, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Shigeru Kuratani
- Laboratory for Evolutionary Morphology, RIKEN, 2-2-3 Minatojima-minami, Chuo-ku, Kobe, Hyogo 650-0047 Japan
| | - Michael K Richardson
- Animal Sciences and Health, Sylvius Laboratories, University of Leiden, Sylviusweg 72, Leiden, The Netherlands
| |
Collapse
|