1
|
Lee S, Ricci B, Tran J, Eul E, Ye J, Ren Q, Clever D, Wang J, Wong P, Haas MS, Stewart SA, Ma CX, Fehniger TA, Faccio R. Stroma-derived Dickkopf-1 contributes to the suppression of NK cell cytotoxicity in breast cancer. Nat Commun 2025; 16:1183. [PMID: 39885132 PMCID: PMC11782527 DOI: 10.1038/s41467-025-56420-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025] Open
Abstract
Mechanisms related to tumor evasion from NK cell-mediated immune surveillance remain enigmatic. Dickkopf-1 (DKK1) is a Wnt/β-catenin inhibitor, whose levels correlate with breast cancer progression. We find DKK1 to be expressed by tumor cells and cancer-associated fibroblasts (CAFs) in patient samples and orthotopic breast tumors, and in bone. By using genetic approaches, we find that bone-derived DKK1 contributes to the systemic DKK1 elevation in tumor-bearing female mice, while CAFs contribute to DKK1 at primary tumor site. Systemic and bone-specific DKK1 targeting reduce tumor growth. Intriguingly, deletion of CAF-derived DKK1 also limits breast cancer progression, without affecting its levels in circulation, and regardless of DKK1 expression in the tumor cells. While not directly supporting tumor proliferation, stromal-DKK1 suppresses NK cell activation and cytotoxicity by downregulating AKT/ERK/S6 phosphorylation. Importantly, increased DKK1 levels and reduced cytotoxic NK cells are detected in women with progressive breast cancer. Our findings indicate that DKK1 represents a barrier to anti-tumor immunity through suppression of NK cells.
Collapse
Affiliation(s)
- Seunghyun Lee
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Biancamaria Ricci
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jennifer Tran
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Emily Eul
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jiayu Ye
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Qihao Ren
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - David Clever
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Julia Wang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO, USA
| | - Pamela Wong
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Sheila A Stewart
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Cynthia X Ma
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Todd A Fehniger
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Roberta Faccio
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
- Shriners Hospitals for Children St Louis, St Louis, MO, USA.
| |
Collapse
|
2
|
Zhao H, Gong H, Zhu P, Sun C, Sun W, Zhou Y, Wu X, Qiu A, Wen X, Zhang J, Luo D, Liu Q, Li Y. Deciphering the cellular and molecular landscapes of Wnt/β-catenin signaling in mouse embryonic kidney development. Comput Struct Biotechnol J 2024; 23:3368-3378. [PMID: 39310276 PMCID: PMC11416353 DOI: 10.1016/j.csbj.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Background The Wnt/β-catenin signaling pathway is critical in kidney development, yet its specific effects on gene expression in different embryonic kidney cell types are not fully understood. Methods Wnt/β-catenin signaling was activated in mouse E12.5 kidneys in vitro using CHIR99021, with RNA sequencing performed afterward, and the results were compared to DMSO controls (dataset GSE131240). Differential gene expression in ureteric buds and cap mesenchyme following pathway activation (datasets GSE20325 and GSE39583) was analyzed. Single-cell RNA-seq data from the Mouse Cell Atlas was used to link differentially expressed genes (DEGs) with kidney cell types. β-catenin ChIP-seq data (GSE39837) identified direct transcriptional targets. Results Activation of Wnt/β-catenin signaling led to 917 significant DEGs, including the upregulation of Notum and Apcdd1 and the downregulation of Crym and Six2. These DEGs were involved in kidney development and immune response. Single-cell analysis identified 787 DEGs across nineteen cell subtypes, with Macrophage_Apoe high cells showing the most pronounced enrichment of Wnt/β-catenin-activated genes. Gene expression profiles in ureteric buds and cap mesenchyme differed significantly upon β-catenin manipulation, with cap mesenchyme showing a unique set of DEGs. Analysis of β-catenin ChIP-seq data revealed 221 potential direct targets, including Dpp6 and Fgf12. Conclusion This study maps the complex gene expression driven by Wnt/β-catenin signaling in embryonic kidney cell types. The identified DEGs and β-catenin targets elucidate the molecular details of kidney development and the pathway's role in immune processes, providing a foundation for further research into Wnt/β-catenin signaling in kidney development and disease.
Collapse
Affiliation(s)
- Hui Zhao
- Guangzhou National Laboratory, Guangzhou International Bio Island, No. 9 Xing Dao Huan Bei Road, Guangzhou 510005, Guangdong Province, China
| | - Hui Gong
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital) and The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong 518052, China
| | - Peide Zhu
- State Key Laboratory of Heavy Oil Processing, China University of Petroleum-Beijing, Beijing 102249, China
| | - Chang Sun
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
| | - Wuping Sun
- Department of Pain Medicine, Shenzhen Municipal Key Laboratory for Pain Medicine, The affiliated Nanshan People's Hospital, The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen 518060, China
| | - Yujin Zhou
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital) and The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong 518052, China
| | - Xiaoxiao Wu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital) and The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong 518052, China
| | - Ailin Qiu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaosha Wen
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital) and The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong 518052, China
| | - Jinde Zhang
- Guangdong Medical University, Zhanjiang 524023, Guangdong China
| | - Dixian Luo
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital) and The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong 518052, China
| | - Quan Liu
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital) and The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong 518052, China
| | - Yifan Li
- Department of Laboratory Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital) and The 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong 518052, China
| |
Collapse
|
3
|
Cheng YJG, Chen CC, Cheng CJ. Postnatal renal tubule development: roles of tubular flow and flux. Curr Opin Nephrol Hypertens 2024; 33:518-525. [PMID: 38913022 PMCID: PMC11290981 DOI: 10.1097/mnh.0000000000001007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
PURPOSE OF REVIEW Postnatal renal tubule development is critical to adult kidney function. Several postnatal changes regulate the differentiation and proliferation of renal tubular cells. Here, we review the literature and our efforts on thick ascending limb (TAL) development in Bartter syndrome (BS). RECENT FINDINGS Glomerular filtrate quickly increases after birth, imposing fluid shear stress and circumferential stretch on immature renal tubules. Recent studies showed that kidney organoids under flow (superfusion) have better development of tubular structures and the expression of cilia and solute transporters. These effects are likely mediated by mechanosensors, such as cilia and the piezo1 channel. Improved renal oxygenation and sodium pump-dependent active transport can stimulate mitochondrial respiration and biogenesis. The functional coupling between transport and mitochondria ensures ATP supply for energy-demanding reactions in tubular cells, including cell cycle progression and proliferation. We recently discovered that postnatal renal medulla maturation and TAL elongation are impaired in Clc-k2-deficient BS mice. Primary cultured Clc-k2-deficient TAL cells have G1-S transition and proliferation delay. These developmental defects could be part of the early pathogenesis of BS and worsen the phenotype. SUMMARY Understanding how tubular flow and transepithelial ion fluxes regulate renal tubule development may improve the treatment of congenital renal tubulopathies.
Collapse
Affiliation(s)
- Yi-Jing G. Cheng
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, U.S.A
| | - Chien-Chou Chen
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chih-Jen Cheng
- Division of Nephrology, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, U.S.A
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
4
|
Maranduca MA, Cozma CT, Clim A, Pinzariu AC, Tudorancea I, Popa IP, Lazar CI, Moscalu R, Filip N, Moscalu M, Constantin M, Scripcariu DV, Serban DN, Serban IL. The Molecular Mechanisms Underlying the Systemic Effects Mediated by Parathormone in the Context of Chronic Kidney Disease. Curr Issues Mol Biol 2024; 46:3877-3905. [PMID: 38785509 PMCID: PMC11120161 DOI: 10.3390/cimb46050241] [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: 03/31/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Chronic kidney disease (CKD) stands as a prominent non-communicable ailment, significantly impacting life expectancy. Physiopathology stands mainly upon the triangle represented by parathormone-Vitamin D-Fibroblast Growth Factor-23. Parathormone (PTH), the key hormone in mineral homeostasis, is one of the less easily modifiable parameters in CKD; however, it stands as a significant marker for assessing the risk of complications. The updated "trade-off hypothesis" reveals that levels of PTH spike out of the normal range as early as stage G2 CKD, advancing it as a possible determinant of systemic damage. The present review aims to review the effects exhibited by PTH on several organs while linking the molecular mechanisms to the observed actions in the context of CKD. From a diagnostic perspective, PTH is the most reliable and accessible biochemical marker in CKD, but its trend bears a higher significance on a patient's prognosis rather than the absolute value. Classically, PTH acts in a dichotomous manner on bone tissue, maintaining a balance between formation and resorption. Under the uremic conditions of advanced CKD, the altered intestinal microbiota majorly tips the balance towards bone lysis. Probiotic treatment has proven reliable in animal models, but in humans, data are limited. Regarding bone status, persistently high levels of PTH determine a reduction in mineral density and a concurrent increase in fracture risk. Pharmacological manipulation of serum PTH requires appropriate patient selection and monitoring since dangerously low levels of PTH may completely inhibit bone turnover. Moreover, the altered mineral balance extends to the cardiovascular system, promoting vascular calcifications. Lastly, the involvement of PTH in the Renin-Angiotensin-Aldosterone axis highlights the importance of opting for the appropriate pharmacological agent should hypertension develop.
Collapse
Affiliation(s)
- Minela Aida Maranduca
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| | - Cristian Tudor Cozma
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| | - Andreea Clim
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| | - Alin Constantin Pinzariu
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| | - Ionut Tudorancea
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| | - Irene Paula Popa
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| | - Cristina Iuliana Lazar
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| | - Roxana Moscalu
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK;
| | - Nina Filip
- Discipline of Biochemistry, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihai Constantin
- Internal Medicine Department, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Dragos Viorel Scripcariu
- Department of Surgery, Grigore T. Popa University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania;
| | - Dragomir Nicolae Serban
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| | - Ionela Lacramioara Serban
- Discipline of Physiology, Department of Morpho-Functional Sciences II, Grigore T. Popa University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.A.M.); (C.T.C.); (A.C.); (A.C.P.); (I.T.); (I.P.P.); (C.I.L.); (D.N.S.); (I.L.S.)
| |
Collapse
|
5
|
Faccio R, Lee S, Ricci B, Tran J, Ye J, Clever D, Eul E, Wang J, Wong P, Ma C, Fehniger T. Cancer-associated fibroblast-derived Dickkopf-1 suppresses NK cell cytotoxicity in breast cancer. RESEARCH SQUARE 2024:rs.3.rs-4202878. [PMID: 38659818 PMCID: PMC11042392 DOI: 10.21203/rs.3.rs-4202878/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Breast cancer is poorly immunogenic, hence able to evade T cell recognition and respond poorly to immune checkpoint blockade. Breast cancer cells can also evade NK cell-mediated immune surveillance, but the mechanism remains enigmatic. Dickkopf-1 (DKK1) is a Wnt/b-catenin inhibitor, whose levels are increased in breast cancer patients and correlate with reduced overall survival. DKK1 is expressed by cancer-associated fibroblasts (CAFs) in orthotopic breast tumors and patient samples, and at higher levels by bone cells. While bone-derived DKK1 contributes to the systemic elevation of DKK1 in tumor-bearing mice, CAFs represent the primary source of DKK1 at the tumor site. Systemic or bone-specific DKK1 targeting reduces primary tumor growth. Intriguingly, specific deletion of CAF-derived DKK1 also limits breast cancer progression, regardless of its elevated levels in circulation and in the bone. DKK1 does not support tumor proliferation directly but rather suppresses the activation and tumoricidal activity of NK cells. Importantly, increased DKK1 levels and reduced number of cytotoxic NK cells are detected in breast cancer patients with progressive bone metastases compared to those with stable disease. Our findings indicate that DKK1 creates a tumor-supporting environment through the suppression of NK cells in breast cancer.
Collapse
Affiliation(s)
| | | | | | | | - Jiayu Ye
- Washington University in St. Louis
| | | | | | | | | | | | | |
Collapse
|
6
|
McCoy MD, Sarasua SM, DeLuca JM, Davis S, Rogers RC, Phelan K, Boccuto L. Genetics of kidney disorders in Phelan-McDermid syndrome: evidence from 357 registry participants. Pediatr Nephrol 2024; 39:749-760. [PMID: 37733098 DOI: 10.1007/s00467-023-06146-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND Phelan-McDermid syndrome (PMS) is a rare genetic disorder caused by SHANK3 pathogenic variants or chromosomal rearrangements affecting the chromosome 22q13 region. Previous research found that kidney disorders, primarily congenital anomalies of the kidney and urinary tract, are common in people with PMS, yet research into candidate genes has been hampered by small study sizes and lack of attention to these problems. METHODS We used a cohort of 357 people from the Phelan-McDermid Syndrome Foundation International Registry to investigate the prevalence of kidney disorders in PMS using a cross-sectional design and to identify 22q13 genes contributing to these disorders. RESULTS Kidney disorders reported included vesicoureteral reflux (n = 37), hydronephrosis (n = 36), dysplastic kidneys (n = 19), increased kidney size (n = 19), polycystic kidneys (15 cases), and kidney stones (n = 4). Out of 315 subjects with a 22q13 deletion, 101 (32%) had at least one kidney disorder, while only one out of 42 (2%) individuals with a SHANK3 pathogenic variant had a kidney disorder (increased kidney size). We identified two genomic regions that were significantly associated with having a kidney disorder with the peak associations observed near positions approximately 5 Mb and 400 Kb from the telomere. CONCLUSIONS The candidate genes for kidney disorders include FBLN1, WNT7B, UPK3A, CELSR1, and PLXNB2. This study demonstrates the utility of patient registries for uncovering genetic contributions to rare diseases. Future work should focus on functional studies for these genes to assess their potential pathogenic contribution to the different subsets of kidney disorders.
Collapse
Affiliation(s)
- Megan D McCoy
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA
| | - Sara M Sarasua
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA.
| | - Jane M DeLuca
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA
| | - Stephanie Davis
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA
| | | | - Katy Phelan
- Genetics Laboratory, Florida Cancer Specialists and Research Institute, Fort Myers, FL, 33916, USA
| | - Luigi Boccuto
- School of Nursing, Healthcare Genetics Program, Clemson University, Clemson, SC, 29634, USA
| |
Collapse
|
7
|
Zhao Z, Dai X, Jiang G, Lin F. ASH2L Controls Ureteric Bud Morphogenesis through the Regulation of RET/GFRA1 Signaling Activity in a Mouse Model. J Am Soc Nephrol 2023; 34:988-1002. [PMID: 36758123 PMCID: PMC10278782 DOI: 10.1681/asn.0000000000000099] [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: 07/30/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
SIGNIFICANCE STATEMENT Causes of congenital anomalies of the kidney and urinary tract (CAKUT) remain unclear. The authors investigated whether and how inactivation of Ash2l -which encodes a subunit of the COMPASS methyltransferase responsible for genome-wide histone H3 lysine K4 (H3K4) methylation-might contribute to CAKUT. In a mouse model, inactivation of Ash2l in the ureteric bud (UB) lineage led to CAKUT-like phenotypes. Removal of ASH2L led to deficient H3K4 trimethylation, which slowed cell proliferation at the UB tip, delaying budding and impairing branching morphogenesis. The absence of ASH2L also downregulated the expression of Ret , Gfra1 , and Wnt11 genes involved in RET/GFRA1 signaling. These findings identify ASH2L-mediated H3K4 methylation as an upstream epigenetic regulator of signaling crucial for UB morphogenesis and indicate that deficiency or dysregulation of these processes may lead to CAKUT. BACKGROUND Ureteric bud (UB) induction and branching morphogenesis are fundamental to the establishment of the renal architecture and are key determinants of nephron number. Defective UB morphogenesis could give rise to a spectrum of malformations associated with congenital anomalies of the kidney and urinary tract (CAKUT). Signaling involving glial cell line-derived neurotrophic factor and its receptor rearranged during transfection (RET) and coreceptor GFRA1 seems to be particularly important in UB development. Recent epigenome profiling studies have uncovered dynamic changes of histone H3 lysine K4 (H3K4) methylation during metanephros development, and dysregulated H3K4 methylation has been associated with a syndromic human CAKUT. METHODS To investigate whether and how inactivation of Ash2l , which encodes a subunit of the COMPASS methyltransferase responsible for genome-wide H3K4 methylation, might contribute to CAKUT, we inactivated Ash2l specifically from the UB lineage in C57BL/6 mice and examined the effects on genome-wide H3K4 methylation and metanephros development. Genes and epigenome changes potentially involved in these effects were screened using RNA-seq combined with Cleavage Under Targets and Tagmentation sequencing. RESULTS UB-specific inactivation of Ash2l caused CAKUT-like phenotypes mainly involving renal dysplasia at birth, which were associated with deficient H3K4 trimethylation. Ash2l inactivation slowed proliferation of cells at the UB tip, delaying budding and impairing UB branching morphogenesis. These effects were associated with downregulation of Ret , Gfra1 , and Wnt11 , which participate in RET/GFRA1 signaling. CONCLUSIONS These experiments identify ASH2L-dependent H3K4 methylation in the UB lineage as an upstream epigenetic regulator of RET/GFRA1 signaling in UB morphogenesis, which, if deficient, may lead to CAKUT.
Collapse
Affiliation(s)
- Ziyi Zhao
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuantong Dai
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gengru Jiang
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Centre for Rare Disease, Shanghai, China
| | - Fujun Lin
- Renal Division, Department of Internal Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Jaschke NP, Pählig S, Sinha A, Adolph TE, Colunga ML, Hofmann M, Wang A, Thiele S, Schwärzler J, Kleymann A, Gentzel M, Tilg H, Wielockx B, Hofbauer LC, Rauner M, Göbel A, Rachner TD. Dickkopf1 fuels inflammatory cytokine responses. Commun Biol 2022; 5:1391. [PMID: 36539532 PMCID: PMC9765382 DOI: 10.1038/s42003-022-04368-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Many human diseases, including cancer, share an inflammatory component but the molecular underpinnings remain incompletely understood. We report that physiological and pathological Dickkopf1 (DKK1) activity fuels inflammatory cytokine responses in cell models, mice and humans. DKK1 maintains the elevated inflammatory tone of cancer cells and is required for mounting cytokine responses following ligation of toll-like and cytokine receptors. DKK1-controlled inflammation derives from cell-autonomous mechanisms, which involve SOCS3-restricted, nuclear RelA (p65) activity. We translate these findings to humans by showing that genetic DKK1 variants are linked to elevated cytokine production across healthy populations. Finally, we find that genetic deletion of DKK1 but not pharmacological neutralization of soluble DKK1 ameliorates inflammation and disease trajectories in a mouse model of endotoxemia. Collectively, our study identifies a cell-autonomous function of DKK1 in the control of the inflammatory response, which is conserved between malignant and non-malignant cells. Additional studies are required to mechanistically dissect cellular DKK1 trafficking and signaling pathways.
Collapse
Affiliation(s)
- Nikolai P Jaschke
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.
| | - Sophie Pählig
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Anupam Sinha
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Innsbruck Medical University, Innsbruck, Austria
| | - Maria Ledesma Colunga
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Maura Hofmann
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Andrew Wang
- Department of Medicine (Rheumatology, Allergy & Immunology), Yale University School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Sylvia Thiele
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Innsbruck Medical University, Innsbruck, Austria
| | - Alexander Kleymann
- Division of Rheumatology, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Marc Gentzel
- Molecular Analysis - Mass Spectrometry, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, and Metabolism, Innsbruck Medical University, Innsbruck, Austria
| | - Ben Wielockx
- Institute of Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Andy Göbel
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Tilman D Rachner
- Department of Medicine III & Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
9
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
10
|
Lehmann J, Thiele S, Baschant U, Rachner TD, Niehrs C, Hofbauer LC, Rauner M. Mice lacking DKK1 in T cells exhibit high bone mass and are protected from estrogen-deficiency-induced bone loss. iScience 2021; 24:102224. [PMID: 33748710 PMCID: PMC7961106 DOI: 10.1016/j.isci.2021.102224] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/16/2021] [Accepted: 02/19/2021] [Indexed: 12/17/2022] Open
Abstract
The Wnt inhibitor Dickkopf-1 (DKK1) is a negative regulator of bone formation and bone mass and is dysregulated in various bone diseases. How DKK1 contributes to postmenopausal osteoporosis, however, remains poorly understood. Here, we show that mice lacking DKK1 in T cells are protected from ovariectomy-induced bone loss. Ovariectomy activated CD4+ and CD8+ T cells and increased their production of DKK1. Co-culture of activated T cells with osteoblasts inhibited Wnt signaling in osteoblasts, leading to impaired differentiation. Importantly, DKK1 expression in T cells also controlled physiological bone remodeling. T-cell-deficient Dkk1 knock-out mice had a higher bone mass with an increased bone formation rate and decreased numbers of osteoclasts compared with controls, a phenotype that was rescued by adoptive transfer of wild-type T cells. Thus, these findings highlight that T cells control bone remodeling in health and disease via their expression of DKK1.
Collapse
Affiliation(s)
- Juliane Lehmann
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Tilman D Rachner
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany.,Institute of Molecular Biology, Mainz, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Division of Endocrinology, Diabetes and Bone Diseases, Technische Universität Dresden, Dresden 01307, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
11
|
Summers ME, Richmond BW, Kropski JA, Majka SA, Bastarache JA, Hatzopoulos AK, Bylund J, Ghosh M, Petrache I, Foronjy RF, Geraghty P, Majka SM. Balanced Wnt/Dickkopf-1 signaling by mesenchymal vascular progenitor cells in the microvascular niche maintains distal lung structure and function. Am J Physiol Cell Physiol 2021; 320:C119-C131. [PMID: 33085496 PMCID: PMC7846975 DOI: 10.1152/ajpcell.00277.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 02/08/2023]
Abstract
The well-described Wnt inhibitor Dickkopf-1 (DKK1) plays a role in angiogenesis as well as in regulation of growth factor signaling cascades in pulmonary remodeling associated with chronic lung diseases (CLDs) including emphysema and fibrosis. However, the specific mechanisms by which DKK1 influences mesenchymal vascular progenitor cells (MVPCs), microvascular endothelial cells (MVECs), and smooth muscle cells (SMCs) within the microvascular niche have not been elucidated. In this study, we show that knockdown of DKK1 in Abcg2pos lung mouse adult tissue resident MVPCs alters lung stiffness, parenchymal collagen deposition, microvessel muscularization and density as well as loss of tissue structure in response to hypoxia exposure. To complement the in vivo mouse modeling, we also identified cell- or disease-specific responses to DKK1, in primary lung chronic obstructive pulmonary disease (COPD) MVPCs, COPD MVECs, and SMCs, supporting a paradoxical disease-specific response of cells to well-characterized factors. Cell responses to DKK1 were dose dependent and correlated with varying expressions of the DKK1 receptor, CKAP4. These data demonstrate that DKK1 expression is necessary to maintain the microvascular niche whereas its effects are context specific. They also highlight DKK1 as a regulatory candidate to understand the role of Wnt and DKK1 signaling between cells of the microvascular niche during tissue homeostasis and during the development of chronic lung diseases.
Collapse
Affiliation(s)
- Megan E Summers
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Bradley W Richmond
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Sarah A Majka
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Julie A Bastarache
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Antonis K Hatzopoulos
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Jeffery Bylund
- Division of Allergy, Pulmonary and Critical Care Medicine or Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs Medical Center, Nashville, Tennessee
| | - Moumita Ghosh
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Irina Petrache
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
| | - Robert F Foronjy
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York
| | - Patrick Geraghty
- Division of Pulmonary and Critical Care Medicine, SUNY Downstate Medical Center, Brooklyn, New York
| | - Susan M Majka
- Division of Pulmonary, Critical Care & Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado
- Department of Medicine, Pulmonary & Critical Care Medicine, Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
| |
Collapse
|
12
|
Meng P, Zhu M, Ling X, Zhou L. Wnt signaling in kidney: the initiator or terminator? J Mol Med (Berl) 2020; 98:1511-1523. [PMID: 32939578 PMCID: PMC7591426 DOI: 10.1007/s00109-020-01978-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
The kidney is a key organ in the human body that excretes toxins and sustains the water-electrolyte balance. During embryonic development and disease progression, the kidney undergoes enormous changes in macrostructure, accompanied by a variety of microstructural histological changes, such as glomerular formation and sclerosis, tubule elongation and atrophy, interstitial establishment, and fibrosis progression. All of these rely on the frequent occurrence of cell death and growth. Notably, to overcome disease, some cells regenerate through self-repair or progenitor cell differentiation. However, the signaling mechanisms underlying kidney development and regeneration have not been elucidated. Recently, Wnt signaling has been noted to play an important role. Although it is a well-known developmental signal, the role of Wnt signaling in kidney development and regeneration is not well recognized. In this review, we review the role of Wnt signaling in kidney embryonic development, tissue repair, cell division, and progenitor cell differentiation after injury. Moreover, we briefly highlight advances in our understanding of the pathogenic mechanisms of Wnt signaling in mediating cellular senescence in kidney parenchymal and stem cells, an irreversible arrest of cell proliferation blocking tissue repair and regeneration. We also highlight the therapeutic targets of Wnt signaling in kidney diseases and provide important clues for clinical strategies.
Collapse
Affiliation(s)
- Ping Meng
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, China
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Mingsheng Zhu
- Department of Nephrology, The People's Hospital of Gaozhou, Maoming, China
| | - Xian Ling
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave, Guangzhou, 510515, 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, 1838 North Guangzhou Ave, Guangzhou, 510515, China.
- Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.
| |
Collapse
|
13
|
de las Fuentes L, Sung YJ, Sitlani CM, Avery CL, Bartz TM, Keyser CD, Evans DS, Li X, Musani SK, Ruiter R, Smith AV, Sun F, Trompet S, Xu H, Arnett DK, Bis JC, Broeckel U, Busch EL, Chen YDI, Correa A, Cummings SR, Floyd JS, Ford I, Guo X, Harris TB, Ikram MA, Lange L, Launer LJ, Reiner AP, Schwander K, Smith NL, Sotoodehnia N, Stewart JD, Stott DJ, Stürmer T, Taylor KD, Uitterlinden A, Vasan RS, Wiggins KL, Cupples LA, Gudnason V, Heckbert SR, Jukema JW, Liu Y, Psaty BM, Rao DC, Rotter JI, Stricker B, Wilson JG, Whitsel EA. Genome-wide meta-analysis of variant-by-diuretic interactions as modulators of lipid traits in persons of European and African ancestry. THE PHARMACOGENOMICS JOURNAL 2020; 20:482-493. [PMID: 31806883 PMCID: PMC7260079 DOI: 10.1038/s41397-019-0132-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/13/2019] [Accepted: 11/20/2019] [Indexed: 01/11/2023]
Abstract
Hypertension (HTN) is a significant risk factor for cardiovascular morbidity and mortality. Metabolic abnormalities, including adverse cholesterol and triglycerides (TG) profiles, are frequent comorbid findings with HTN and contribute to cardiovascular disease. Diuretics, which are used to treat HTN and heart failure, have been associated with worsening of fasting lipid concentrations. Genome-wide meta-analyses with 39,710 European-ancestry (EA) individuals and 9925 African-ancestry (AA) individuals were performed to identify genetic variants that modify the effect of loop or thiazide diuretic use on blood lipid concentrations. Both longitudinal and cross sectional data were used to compute cohort-specific interaction results, which were then combined through meta-analysis in each ancestry. These ancestry-specific results were further combined through trans-ancestry meta-analysis. Analysis of EA data identified two genome-wide significant (p < 5 × 10-8) loci with single nucleotide variant (SNV)-loop diuretic interaction on TG concentrations (including COL11A1). Analysis of AA data identified one genome-wide significant locus adjacent to BMP2 with SNV-loop diuretic interaction on TG concentrations. Trans-ancestry analysis strengthened evidence of association for SNV-loop diuretic interaction at two loci (KIAA1217 and BAALC). There were few significant SNV-thiazide diuretic interaction associations on TG concentrations and for either diuretic on cholesterol concentrations. Several promising loci were identified that may implicate biologic pathways that contribute to adverse metabolic side effects from diuretic therapy.
Collapse
Affiliation(s)
- Lisa de las Fuentes
- Cardiovascular Division, Department of Medicine, Washington University, St. Louis, MO, USA.
| | - Y J Sung
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - C M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - C L Avery
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - T M Bartz
- Cardiovascular Health Research Unit, Departments of Medicine and Biostatistics, University of Washington, Seattle, WA, USA
| | - C de Keyser
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - D S Evans
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | - X Li
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - S K Musani
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - R Ruiter
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - A V Smith
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - F Sun
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - S Trompet
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - H Xu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - D K Arnett
- Dean's Office, University of Kentucky College of Public Health, Lexington, KY, USA
| | - J C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - U Broeckel
- Section of Genomic Pediatrics, Department of Pediatrics, Medicine and Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - E L Busch
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Y-D I Chen
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - A Correa
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - S R Cummings
- Research Institute, California Pacific Medical Center, San Francisco, CA, USA
| | - J S Floyd
- Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology, University of Washington, Seattle, WA, USA
| | - I Ford
- Robertson Center for biostatistics, University of Glasgow, Glasgow, UK
| | - X Guo
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - T B Harris
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - M A Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - L Lange
- Department of Genetics, University of Colorado, Denver, Denver, CO, USA
| | - L J Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - A P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- School of Public Health, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - K Schwander
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - N L Smith
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center (ERIC), VA Cooperative Studies Program, VA Puget Sound Health Care System, Seattle, WA, USA
| | - N Sotoodehnia
- Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology, University of Washington, Seattle, WA, USA
- Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - J D Stewart
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Carolina Population Center, University of North Carolina, Chapel Hill, NC, USA
| | - D J Stott
- Institute of cardiovascular and medical sciences, Faculty of Medicine, University of Glasgow, Glasgow, United Kingdom
| | - T Stürmer
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Center for Pharmacoepidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - K D Taylor
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - A Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - R S Vasan
- The Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - K L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - L A Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - V Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - S R Heckbert
- Cardiovascular Health Research Unit, Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - J W Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands, Utrecht, The Netherlands
| | - Y Liu
- Division of Public Health Sciences, Department of Epidemiology and Prevention, Wake Forest University, Winston-, Salem, NC, USA
| | - B M Psaty
- Cardiovascular Health Research Unit, Departments of Epidemiology, Medicine, and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - D C Rao
- Division of Biostatistics, Washington University, St. Louis, MO, USA
| | - J I Rotter
- Institute for Translational Genomics and Population Sciences, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - B Stricker
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - J G Wilson
- Biophysics and Physiology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - E A Whitsel
- Gillings School of Global Public Health, Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- School of Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
14
|
Colditz J, Picke AK, Hofbauer LC, Rauner M. Contributions of Dickkopf-1 to Obesity-Induced Bone Loss and Marrow Adiposity. JBMR Plus 2020; 4:e10364. [PMID: 32537550 PMCID: PMC7285751 DOI: 10.1002/jbm4.10364] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/08/2020] [Accepted: 04/05/2020] [Indexed: 12/20/2022] Open
Abstract
Low bone strength in overweight individuals is a significant medical problem. One important determinant of mesenchymal stem cell fate into osteoblasts or adipocytes is the Wnt signaling pathway. We recently showed that Dickkopf‐1 (DKK1), a potent Wnt inhibitor, is upregulated in obese mice. In this study, we investigated the role of DKK1 in the pathogenesis of obesity‐induced bone loss using global and tissue‐specific KO mice. Obesity was induced in 8‐week‐old male mice with an inducible global (Rosa26‐CreERT2) or osteoprogenitor‐ (Osx–Cre‐) specific deletion of Dkk1 with a high‐fat diet (HFD) containing 60% fat. After 12 weeks, body weight, bone volume, bone fat mass, and bone turnover were assessed. Dkk1fl/fl;Rosa26‐CreERT2 mice experienced a similar increase in body weight and white fat pads as control mice. A HFD significantly reduced trabecular bone mass and the bone formation rate in Cre‐ mice and Dkk1fl/fl;Rosa26‐CreERT2 mice. Interestingly, Dkk1fl/fl;Rosa26‐CreERT2 mice were protected from HFD‐induced cortical bone loss. Furthermore, a HFD was associated with increased bone marrow fat in the femur, which was less pronounced in Dkk1fl/fl;Rosa26‐CreERT2 mice. Mice with an osteoprogenitor‐specific Dkk1 deletion showed similar results as the global knockout, showing a protection against HFD‐induced cortical bone loss and an accumulation of bone marrow fat, but a similar decrease in trabecular bone volume. In summary, DKK1 appears to contribute distinctly to cortical, but not trabecular bone loss in obesity. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Juliane Colditz
- Department of Medicine III, Center for Healthy Aging Technische Universität Dresden Dresden Germany
| | - Ann-Kristin Picke
- Department of Medicine III, Center for Healthy Aging Technische Universität Dresden Dresden Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Center for Healthy Aging Technische Universität Dresden Dresden Germany
| | - Martina Rauner
- Department of Medicine III, Center for Healthy Aging Technische Universität Dresden Dresden Germany
| |
Collapse
|
15
|
Deacon P, Concodora CW, Chung E, Park JS. β-catenin regulates the formation of multiple nephron segments in the mouse kidney. Sci Rep 2019; 9:15915. [PMID: 31685872 PMCID: PMC6828815 DOI: 10.1038/s41598-019-52255-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/13/2019] [Indexed: 01/15/2023] Open
Abstract
The nephron is composed of distinct segments that perform unique physiological functions. Little is known about how multipotent nephron progenitor cells differentiate into different nephron segments. It is well known that β-catenin signaling regulates the maintenance and commitment of mesenchymal nephron progenitors during kidney development. However, it is not fully understood how it regulates nephron segmentation after nephron progenitors undergo mesenchymal-to-epithelial transition. To address this, we performed β-catenin loss-of-function and gain-of-function studies in epithelial nephron progenitors in the mouse kidney. Consistent with a previous report, the formation of the renal corpuscle was defective in the absence of β-catenin. Interestingly, we found that epithelial nephron progenitors lacking β-catenin were able to form presumptive proximal tubules but that they failed to further develop into differentiated proximal tubules, suggesting that β-catenin signaling plays a critical role in proximal tubule development. We also found that epithelial nephron progenitors lacking β-catenin failed to form the distal tubules. Expression of a stable form of β-catenin in epithelial nephron progenitors blocked the proper formation of all nephron segments, suggesting tight regulation of β-catenin signaling during nephron segmentation. This work shows that β-catenin regulates the formation of multiple nephron segments along the proximo-distal axis of the mammalian nephron.
Collapse
Affiliation(s)
- Patrick Deacon
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Charles W Concodora
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.,Urology for Children, 200 Bowman Drive, Voorhees, NJ, 08043, USA
| | - Eunah Chung
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Joo-Seop Park
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA. .,University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
16
|
Colditz J, Thiele S, Baschant U, Garbe AI, Niehrs C, Hofbauer LC, Rauner M. Osteogenic Dkk1 Mediates Glucocorticoid-Induced but Not Arthritis-Induced Bone Loss. J Bone Miner Res 2019; 34:1314-1323. [PMID: 30779862 DOI: 10.1002/jbmr.3702] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/01/2019] [Accepted: 02/05/2019] [Indexed: 12/17/2022]
Abstract
Dickkopf-1 (Dkk1) is a negative regulator of bone formation and bone mass and is deregulated in bone loss induced by arthritis and glucocorticoid (GC) exposure. However, the role of Dkk1 in these pathological processes is still unknown. Here, we used conditional Dkk1 knock-out mice to determine the role of Dkk1 produced by osteolineage cells in the development of arthritis and GC-induced bone loss. Osteoprogenitor (Osx-Cre)- and osteocyte (Dmp1-Cre)-specific knock-out mice and their Cre-negative controls were subjected to two arthritis models, K/BxN and antigen-induced arthritis. Disease induction and progression were assessed. GC-induced bone loss was induced in 25-week-old female mice by implanting prednisolone (7.5 mg) slow-release pellets for 4 weeks. Dkk1fl/fl ;Osx-Cre mice subjected to K/BxN arthritis showed mildly reduced disease severity with reduced infiltration of neutrophils and T cells into affected joints and reduced bone erosions compared with Cre-negative controls. Osteocyte-specific Dkk1 deletion did not affect disease severity or local bone erosions. However, systemic bone loss at the spine was less severe in both mouse lines. In contrast to arthritis, both lines were protected from GC-induced bone loss. Although the Cre-negative controls lost about 26% and 31% bone volume potentially caused by decreased bone formation, Cre-positive mice did not exhibit such alterations. Dkk-1 deficiency in osteolineage cells protects against GC-induced bone loss, whereas it had only minor effects in arthritis. Therefore, Dkk1 may be a promising therapeutic target especially for bone diseases in which inhibition of bone formation represents the predominant mechanism. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Juliane Colditz
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Annette I Garbe
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany.,Institute of Molecular Biology, Mainz, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
17
|
Chang CH, Davies JA. In developing mouse kidneys, orientation of loop of Henle growth is adaptive and guided by long-range cues from medullary collecting ducts. J Anat 2019; 235:262-270. [PMID: 31099428 PMCID: PMC6637448 DOI: 10.1111/joa.13012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2019] [Indexed: 11/28/2022] Open
Abstract
The path taken by the loop of Henle, from renal cortex to medulla and back, is critical to the ability of the kidney to concentrate urine and recover water. Unlike most developing tubules, which navigate as blind‐ended cylinders, the loop of Henle extends as a sharply bent loop, the apex of which leads the double tubes behind it in a ‘V’ shape. Here, we show that, in normal kidney development, loops of Henle extend towards the centroid of the kidney with an accuracy that increases the longer they extend. Using cultured kidney rudiments, and manipulations that rotate or remove portions of the organ, we show that loop orientation depends on long‐range cues from the medulla rather than either the orientation of the parent nephron or local cues in the cortex. The loops appear to be attracted to the most mature branch point of the collecting duct system but, if this is removed, they will head towards the most mature collecting duct branch available to them. Our results demonstrate the adaptive nature of guidance of this unusual example of a growing epithelium, and set the stage for later work devoted to understanding the molecules and mechanisms that underlie it.
Collapse
Affiliation(s)
- C-Hong Chang
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK.,Yale University School of Medicine, Medicine, New Haven, CT, USA
| | - Jamie A Davies
- Deanery of Biomedical Science, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
18
|
Kamei CN, Gallegos TF, Liu Y, Hukriede N, Drummond IA. Wnt signaling mediates new nephron formation during zebrafish kidney regeneration. Development 2019; 146:dev.168294. [PMID: 31036548 PMCID: PMC6503981 DOI: 10.1242/dev.168294] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 04/05/2019] [Indexed: 12/24/2022]
Abstract
Zebrafish kidneys use resident kidney stem cells to replace damaged tubules with new nephrons: the filtration units of the kidney. What stimulates kidney progenitor cells to form new nephrons is not known. Here, we show that wnt9a and wnt9b are induced in the injured kidney at sites where frizzled9b- and lef1-expressing progenitor cells form new nephrons. New nephron aggregates are patterned by Wnt signaling, with high canonical Wnt-signaling cells forming a single cell thick rosette that demarcates: domains of cell proliferation in the elongating nephron; and tubule fusion where the new nephron plumbs into the distal tubule and establishes blood filtrate drainage. Pharmacological blockade of canonical Wnt signaling inhibited new nephron formation after injury by inhibiting cell proliferation, and resulted in loss of polarized rosette structures in the aggregates. Mutation in frizzled9b reduced total kidney nephron number, caused defects in tubule morphology and reduced regeneration of new nephrons after injury. Our results demonstrate an essential role for Wnt/frizzled signaling in adult zebrafish kidney development and regeneration, highlighting conserved mechanisms underlying both mammalian kidney development and kidney stem cell-directed neonephrogenesis in zebrafish. Summary: Adult zebrafish kidneys induce Wnt signaling to generate new nephrons from resident kidney progenitor cells, highlighting how embryonic morphogens are reactivated in adult organs to drive regeneration.
Collapse
Affiliation(s)
- Caramai N Kamei
- Massachusetts General Hospital, Department of Medicine, Nephrology Division, 149 13th Street, Charlestown, MA 02129, USA
| | - Thomas F Gallegos
- Massachusetts General Hospital, Department of Medicine, Nephrology Division, 149 13th Street, Charlestown, MA 02129, USA
| | - Yan Liu
- Massachusetts General Hospital, Department of Medicine, Nephrology Division, 149 13th Street, Charlestown, MA 02129, USA
| | - Neil Hukriede
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Iain A Drummond
- Massachusetts General Hospital, Department of Medicine, Nephrology Division, 149 13th Street, Charlestown, MA 02129, USA .,Harvard Medical School Department of Genetics, Boston, MA 02115, USA
| |
Collapse
|
19
|
Tsourdi E, Colditz J, Lademann F, Rijntjes E, Köhrle J, Niehrs C, Hofbauer LC, Rauner M. The Role of Dickkopf-1 in Thyroid Hormone-Induced Changes of Bone Remodeling in Male Mice. Endocrinology 2019; 160:664-674. [PMID: 30689850 DOI: 10.1210/en.2018-00998] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/21/2019] [Indexed: 12/17/2022]
Abstract
Thyroid hormones regulate bone homeostasis, and exogenously induced hyperthyroidism and hypothyroidism in mice was recently found to be associated with an altered expression of the Wnt inhibitor Dickkopf-1 (Dkk1), a determinant of bone mass. Here, we assessed the role of Dkk1 in thyroid hormone-induced changes in bone using conditional Dkk1 knockout mice. Male mice with a global (Dkk1fl/fl;Rosa26-CreERT2) or osteocyte-specific (Dkk1fl/fl;Dmp1:Cre) deletion of Dkk1 were pharmacologically rendered hypothyroid or hyperthyroid. The bone phenotype was analyzed using micro-CT analysis, dynamic histomorphometry, and serum concentrations of bone turnover markers. Hypothyroid and hyperthyroid Cre-negative mice of either Cre line revealed the expected changes in bone volume with hypothyroid mice displaying a 40% to 60% increase in vertebral trabecular bone volume, while hyperthyroid mice lost 45% to 60% of bone volume. Similar changes were observed at the spine. Interestingly, Cre-positive mice of both lines did not gain or lose as much bone at the femur when rendered hypothyroid or hyperthyroid. While Cre-negative hypothyroid mice gained 80% to 100% bone volume, Cre-positive hypothyroid mice only increased their bone volume by 55% to 90%. Similarly, Cre-negative hyperthyroid mice lost 74% to 79% bone, while Cre-positive hyperthyroid mice merely lost 40% to 54%. Despite these site-specific differences, both global and osteocyte-specific Dkk1 knockout mice displayed similar changes in bone turnover as their Cre-negative controls in the hypothyroid and hyperthyroid states. While osteoblast and osteoclast parameters were increased in hyperthyroidism, hypothyroidism potently suppressed bone cell activities. Loss of Dkk1 is not sufficient to fully reverse thyroid hormone-induced changes in bone mass and bone turnover.
Collapse
Affiliation(s)
- Elena Tsourdi
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| | - Juliane Colditz
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| | - Franziska Lademann
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| | - Eddy Rijntjes
- Charité-Universitätsmedizin Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | - Josef Köhrle
- Charité-Universitätsmedizin Berlin, Institut für Experimentelle Endokrinologie, Berlin, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany
- Institute of Molecular Biology, Mainz, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
- Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden Medical Center, Dresden, Germany
- Center for Healthy Aging, Technische Universität Dresden Medical Center, Dresden, Germany
| |
Collapse
|
20
|
Colditz J, Thiele S, Baschant U, Niehrs C, Bonewald LF, Hofbauer LC, Rauner M. Postnatal Skeletal Deletion of Dickkopf-1 Increases Bone Formation and Bone Volume in Male and Female Mice, Despite Increased Sclerostin Expression. J Bone Miner Res 2018; 33:1698-1707. [PMID: 29734465 DOI: 10.1002/jbmr.3463] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/16/2018] [Accepted: 04/28/2018] [Indexed: 12/14/2022]
Abstract
The Wnt antagonist Dickkopf-1 (Dkk1) is a negative regulator of osteoblast function and bone mass. However, because of the lack of appropriate models, many aspects of its role in the regulation of postnatal bone turnover and its cellular source have remained unknown. In this study, we deleted Dkk1 postnatally and in different cell types using various Cre-drivers (Rosa26-ERT2-Cre, Osx-cre, Dmp1-Cre) and assessed to which extent cells of the osteoblastic lineage contribute to the effects of Dkk1 on bone turnover and homeostasis. Female and male mice were examined at 12 weeks of age. Mice with a global or cell type-specific deletion of Dkk1 showed a two- to threefold higher bone volume compared with their Cre-negative littermates. The mineral apposition rate and the bone formation rate were increased two- to fourfold in all three mouse lines, despite a significant increase in systemic and skeletal levels of sclerostin. Dkk1 deletion further reduced the number of osteoclasts about twofold, which was accompanied by a strong decrease in the receptor activator of nuclear factor-κB ligand/osteoprotegerin mRNA ratio in femoral bone. Despite similar increases in bone mass, the deletion of Dkk1 in osterix-expressing cells reduced circulating Dkk1 significantly (males, -79%; females, -77%), whereas they were not changed in Dkk1fl/fl ;Dmp1-Cre mice. However, both lines showed significantly reduced Dkk1 mRNA levels in bone. In summary, we show that lack of Dkk1 in cells of the osteoblastic lineage leads to high bone mass with increased bone formation, despite increased levels of sclerostin. Moreover, the majority of systemic Dkk1 appears to originate from osteoprogenitors but not from mature osteoblasts or osteocytes. Nevertheless, the amount of Dkk1 produced locally by more mature osteogenic cells is sufficient to modulate bone mass. Thus, this study highlights the importance of local Wnt signaling on postnatal bone homeostasis. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Juliane Colditz
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany.,Institute of Molecular Biology, Mainz, Germany
| | - Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, Departments of Anatomy and Cell Biology and Orthopaedic Surgery, School of Medicine, Indianapolis, IN, USA
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.,Center for Regenerative Therapies Dresden, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
21
|
Witcher PC, Miner SE, Horan DJ, Bullock WA, Lim KE, Kang KS, Adaniya AL, Ross RD, Loots GG, Robling AG. Sclerostin neutralization unleashes the osteoanabolic effects of Dkk1 inhibition. JCI Insight 2018; 3:98673. [PMID: 29875318 DOI: 10.1172/jci.insight.98673] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/26/2018] [Indexed: 12/12/2022] Open
Abstract
The WNT pathway has become an attractive target for skeletal therapies. High-bone-mass phenotypes in patients with loss-of-function mutations in the LRP5/6 inhibitor Sost (sclerosteosis), or in its downstream enhancer region (van Buchem disease), highlight the utility of targeting Sost/sclerostin to improve bone properties. Sclerostin-neutralizing antibody is highly osteoanabolic in animal models and in human clinical trials, but antibody-based inhibition of another potent LRP5/6 antagonist, Dkk1, is largely inefficacious for building bone in the unperturbed adult skeleton. Here, we show that conditional deletion of Dkk1 from bone also has negligible effects on bone mass. Dkk1 inhibition increases Sost expression, suggesting a potential compensatory mechanism that might explain why Dkk1 suppression lacks anabolic action. To test this concept, we deleted Sost from osteocytes in, or administered sclerostin neutralizing antibody to, mice with a Dkk1-deficient skeleton. A robust anabolic response to Dkk1 deletion was manifest only when Sost/sclerostin was impaired. Whole-body DXA scans, μCT measurements of the femur and spine, histomorphometric measures of femoral bone formation rates, and biomechanical properties of whole bones confirmed the anabolic potential of Dkk1 inhibition in the absence of sclerostin. Further, combined administration of sclerostin and Dkk1 antibody in WT mice produced a synergistic effect on bone gain that greatly exceeded individual or additive effects of the therapies, confirming the therapeutic potential of inhibiting multiple WNT antagonists for skeletal health. In conclusion, the osteoanabolic effects of Dkk1 inhibition can be realized if sclerostin upregulation is prevented. Anabolic therapies for patients with low bone mass might benefit from a strategy that accounts for the compensatory milieu of WNT inhibitors in bone tissue.
Collapse
Affiliation(s)
- Phillip C Witcher
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Sara E Miner
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Daniel J Horan
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Whitney A Bullock
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kyung-Eun Lim
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kyung Shin Kang
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Physical Sciences & Engineering, Anderson University, Anderson, Indiana, USA
| | - Alison L Adaniya
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ryan D Ross
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Gabriela G Loots
- Biology and Biotechnology Division, Lawrence Livermore National Laboratory, Livermore, California, USA.,School of Natural Sciences, University of California, Merced, California, USA
| | - Alexander G Robling
- Department of Anatomy & Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA.,Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana, USA
| |
Collapse
|
22
|
Renal Tubule Repair: Is Wnt/β-Catenin a Friend or Foe? Genes (Basel) 2018; 9:genes9020058. [PMID: 29364168 PMCID: PMC5852554 DOI: 10.3390/genes9020058] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/14/2018] [Accepted: 01/16/2018] [Indexed: 12/15/2022] Open
Abstract
Wnt/β-catenin signaling is extremely important for proper kidney development. This pathway is also upregulated in injured renal tubular epithelia, both in acute kidney injury and chronic kidney disease. The renal tubular epithelium is an important target of kidney injury, and its response (repair versus persistent injury) is critical for determining whether tubulointerstitial fibrosis, the hallmark of chronic kidney disease, develops. This review discusses how Wnt/β-catenin signaling in the injured tubular epithelia promotes either repair or fibrosis after kidney injury. There is data suggesting that epithelial Wnt/β-catenin signaling is beneficial in acute kidney injury and important in tubular progenitors responsible for epithelial repair. The role of Wnt/β-catenin signaling in chronically injured epithelia is less clear. There is convincing data that Wnt/β-catenin signaling in interstitial fibroblasts and pericytes contributes to the extracellular matrix accumulation that defines fibrosis. However, some recent studies question whether Wnt/β-catenin signaling in chronically injured epithelia actually promotes fibrosis or repair.
Collapse
|
23
|
Leonard JL, Leonard DM, Wolfe SA, Liu J, Rivera J, Yang M, Leonard RT, Johnson JPS, Kumar P, Liebmann KL, Tutto AA, Mou Z, Simin KJ. The Dkk3 gene encodes a vital intracellular regulator of cell proliferation. PLoS One 2017; 12:e0181724. [PMID: 28738084 PMCID: PMC5524345 DOI: 10.1371/journal.pone.0181724] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 07/06/2017] [Indexed: 11/18/2022] Open
Abstract
Members of the Dickkopf (Dkk) family of Wnt antagonists interrupt Wnt-induced receptor assembly and participate in axial patterning and cell fate determination. One family member, DKK3, does not block Wnt receptor activation. Loss of Dkk3 expression in cancer is associated with hyperproliferation and dysregulated ß-catenin signaling, and ectopic expression of Dkk3 halts cancer growth. The molecular events mediating the DKK3-dependent arrest of ß-catenin-driven cell proliferation in cancer cells are unknown. Here we report the identification of a new intracellular gene product originating from the Dkk3 locus. This Dkk3b transcript originates from a second transcriptional start site located in intron 2 of the Dkk3 gene. It is essential for early mouse development and is a newly recognized regulator of ß-catenin signaling and cell proliferation. Dkk3b interrupts nuclear translocation ß-catenin by capturing cytoplasmic, unphosphorylated ß-catenin in an extra-nuclear complex with ß-TrCP. These data reveal a new regulator of one of the most studied signal transduction pathways in metazoans and provides a novel, completely untapped therapeutic target for silencing the aberrant ß-catenin signaling that drives hyperproliferation in many cancers.
Collapse
Affiliation(s)
- Jack L. Leonard
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| | - Deborah M. Leonard
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Scot A. Wolfe
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jilin Liu
- Department of Cell and Molecular Physiology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jaime Rivera
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Michelle Yang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Ryan T. Leonard
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Jacob P. S. Johnson
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Prashant Kumar
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Kate L. Liebmann
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Amanda A. Tutto
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Zhongming Mou
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Karl J. Simin
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
24
|
Hruska KA, Sugatani T, Agapova O, Fang Y. The chronic kidney disease - Mineral bone disorder (CKD-MBD): Advances in pathophysiology. Bone 2017; 100:80-86. [PMID: 28119179 PMCID: PMC5502716 DOI: 10.1016/j.bone.2017.01.023] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 01/01/2023]
Abstract
The causes of excess cardiovascular mortality associated with chronic kidney disease (CKD) have been attributed in part to the CKD-mineral bone disorder syndrome (CKD-MBD), wherein, novel cardiovascular risk factors have been identified. New advances in the causes of the CKD-MBD are discussed in this review. They demonstrate that repair and disease processes in the kidneys release factors to the circulation that cause the systemic complications of CKD. The discovery of WNT inhibitors, especially Dickkopf 1 (Dkk1), produced during renal repair as participating in the pathogenesis of the vascular and skeletal components of the CKD-MBD implied that additional pathogenic factors are critical. This lead to the discovery that activin A is a second renal repair factor circulating in increased levels during CKD. Activin A derives from peritubular myofibroblasts of diseased kidneys, wherein it stimulates fibrosis, and decreases tubular klotho expression. Activin A binds to the type 2 activin A receptor, ActRIIA, which is variably affected by CKD in the vasculature. In diabetic/atherosclerotic aortas, specifically in vascular smooth muscle cells (VSMC), ActRIIA signaling is inhibited and contributes to CKD induced VSMC dedifferentiation, osteogenic transition and neointimal atherosclerotic calcification. In nondiabetic/nonatherosclerotic aortas, CKD increases VSMC ActRIIA signaling, and vascular fibroblast signaling causing the latter to undergo osteogenic transition and stimulate vascular calcification. In both vascular situations, a ligand trap for ActRIIA prevented vascular calcification. In the skeleton, activin A is responsible for CKD stimulation of osteoclastogenesis and bone remodeling increasing bone turnover. These studies demonstrate that circulating renal repair and injury factors are causal of the CKD-MBD and CKD associated cardiovascular disease.
Collapse
Affiliation(s)
- Keith A Hruska
- Department of Pediatrics, Nephrology, Washington University Saint Louis, MO, United States; Departments of Medicine, Washington University Saint Louis, MO, United States; Department of Cell Biology, Washington University Saint Louis, MO, United States.
| | - Toshifumi Sugatani
- Department of Pediatrics, Nephrology, Washington University Saint Louis, MO, United States
| | - Olga Agapova
- Department of Pediatrics, Nephrology, Washington University Saint Louis, MO, United States
| | - Yifu Fang
- Department of Pediatrics, Nephrology, Washington University Saint Louis, MO, United States
| |
Collapse
|
25
|
Dickkopf-1 promotes hematopoietic regeneration via direct and niche-mediated mechanisms. Nat Med 2016; 23:91-99. [PMID: 27918563 DOI: 10.1038/nm.4251] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/11/2016] [Indexed: 12/13/2022]
Abstract
The role of osteolineage cells in regulating hematopoietic stem cell (HSC) regeneration following myelosuppression is not well understood. Here we show that deletion of the pro-apoptotic genes Bak and Bax in osterix (Osx, also known as Sp7 transcription factor 7)-expressing cells in mice promotes HSC regeneration and hematopoietic radioprotection following total body irradiation. These mice showed increased bone marrow (BM) levels of the protein dickkopf-1 (Dkk1), which was produced in Osx-expressing BM cells. Treatment of irradiated HSCs with Dkk1 in vitro increased the recovery of both long-term repopulating HSCs and progenitor cells, and systemic administration of Dkk1 to irradiated mice increased hematopoietic recovery and improved survival. Conversely, inducible deletion of one allele of Dkk1 in Osx-expressing cells in adult mice inhibited the recovery of BM stem and progenitor cells and of complete blood counts following irradiation. Dkk1 promoted hematopoietic regeneration via both direct effects on HSCs, in which treatment with Dkk1 decreased the levels of mitochondrial reactive oxygen species and suppressed senescence, and indirect effects on BM endothelial cells, in which treatment with Dkk1 induced epidermal growth factor (EGF) secretion. Accordingly, blockade of the EGF receptor partially abrogated Dkk1-mediated hematopoietic recovery. These data identify Dkk1 as a regulator of hematopoietic regeneration and demonstrate paracrine cross-talk between BM osteolineage cells and endothelial cells in regulating hematopoietic reconstitution following injury.
Collapse
|
26
|
Roker LA, Nemri K, Yu J. Wnt7b Signaling from the Ureteric Bud Epithelium Regulates Medullary Capillary Development. J Am Soc Nephrol 2016; 28:250-259. [PMID: 27432740 DOI: 10.1681/asn.2015111205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 05/26/2016] [Indexed: 01/29/2023] Open
Abstract
The renal vasculature is integral to the physiologic function of the kidneys in regulating hemodynamics of the body and maintaining organ health. The close inter-relationship of capillaries and the renal epithelium is key to renal physiology, but how renal tubules regulate capillary development remains unclear. Our previous work showed that Wnt7b is expressed in the ureteric trunk epithelium and activates canonical Wnt signaling in the surrounding medullary interstitium, where the capillaries reside. In this study, we showed by immunofluorescence that the target interstitial cells of Wnt7b/canonical Wnt signaling are mural cells of periureteric bud capillaries in the nascent renal medulla of embryonic mice. Genetic ablation of Wnt7b enhanced the proliferation of Wnt7b target mural cells, an effect that associated with decreased expression of PDGFRβ and p57kip2, a cyclin-dependent kinase inhibitor, in these cells. Furthermore, Wnt7b regulated lumen formation of the capillary endothelium in the renal medulla. In the absence of Wnt7b signaling, the periureteric bud medullary capillaries displayed narrower lumens lined with less flattened endothelial cells and a significantly increased presence of luminal endothelial cell-cell junctions, a transient configuration in the forming blood vessels in the controls. Moreover, the absence of Wnt7b led to greatly diminished levels of vascular endothelial (VE)-cadherin at the cell surface in these blood vessels. VE-cadherin is essential for blood vessel lumen formation; thus, Wnt7b may regulate lumen formation through modulation of VE-cadherin localization. Overall, these results indicate a novel role of Wnt7b signaling and the ureteric bud epithelium in renal medullary capillary development.
Collapse
Affiliation(s)
| | | | - Jing Yu
- Department of Cell Biology, .,Child Health Research Center, University of Virginia School of Medicine, Charlottesville, Virginia.,Center for Immunity, Inflammation and Regenerative Medicine and
| |
Collapse
|
27
|
Denby L, Conway BR. Wnt6: another player in the yin and yang of renal Wnt signaling. Am J Physiol Renal Physiol 2016; 311:F404-5. [PMID: 27279489 DOI: 10.1152/ajprenal.00296.2016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/02/2016] [Indexed: 11/22/2022] Open
Affiliation(s)
- Laura Denby
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, United Kingdom
| | - Bryan R Conway
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, United Kingdom
| |
Collapse
|
28
|
Beaton H, Andrews D, Parsons M, Murphy M, Gaffney A, Kavanagh D, McKay GJ, Maxwell AP, Taylor CT, Cummins EP, Godson C, Higgins DF, Murphy P, Crean J. Wnt6 regulates epithelial cell differentiation and is dysregulated in renal fibrosis. Am J Physiol Renal Physiol 2016; 311:F35-45. [PMID: 27122540 DOI: 10.1152/ajprenal.00136.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 04/22/2016] [Indexed: 02/07/2023] Open
Abstract
Diabetic nephropathy is the most common microvascular complication of diabetes mellitus, manifesting as mesangial expansion, glomerular basement membrane thickening, glomerular sclerosis, and progressive tubulointerstitial fibrosis leading to end-stage renal disease. Here we describe the functional characterization of Wnt6, whose expression is progressively lost in diabetic nephropathy and animal models of acute tubular injury and renal fibrosis. We have shown prominent Wnt6 and frizzled 7 (FzD7) expression in the mesonephros of the developing mouse kidney, suggesting a role for Wnt6 in epithelialization. Importantly, TCF/Lef reporter activity is also prominent in the mesonephros. Analysis of Wnt family members in human renal biopsies identified differential expression of Wnt6, correlating with severity of the disease. In animal models of tubular injury and fibrosis, loss of Wnt6 was evident. Wnt6 signals through the canonical pathway in renal epithelial cells as evidenced by increased phosphorylation of GSK3β (Ser9), nuclear accumulation of β-catenin and increased TCF/Lef transcriptional activity. FzD7 was identified as a putative receptor of Wnt6. In vitro Wnt6 expression leads to de novo tubulogenesis in renal epithelial cells grown in three-dimensional culture. Importantly, Wnt6 rescued epithelial cell dedifferentiation in response to transforming growth factor-β (TGF-β); Wnt6 reversed TGF-β-mediated increases in vimentin and loss of epithelial phenotype. Wnt6 inhibited TGF-β-mediated p65-NF-κB nuclear translocation, highlighting cross talk between the two pathways. The critical role of NF-κB in the regulation of vimentin expression was confirmed in both p65(-/-) and IKKα/β(-/-) embryonic fibroblasts. We propose that Wnt6 is involved in epithelialization and loss of Wnt6 expression contributes to the pathogenesis of renal fibrosis.
Collapse
Affiliation(s)
- Hayley Beaton
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Darrell Andrews
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Martin Parsons
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Mary Murphy
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland
| | - Andrew Gaffney
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - David Kavanagh
- Nephrology Research Group, Centre for Public Health, Queens University Belfast, Royal Victoria Hospital, Belfast, United Kingdom; and
| | - Gareth J McKay
- Nephrology Research Group, Centre for Public Health, Queens University Belfast, Royal Victoria Hospital, Belfast, United Kingdom; and
| | - Alexander P Maxwell
- Nephrology Research Group, Centre for Public Health, Queens University Belfast, Royal Victoria Hospital, Belfast, United Kingdom; and
| | - Cormac T Taylor
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Eoin P Cummins
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Catherine Godson
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Debra F Higgins
- UCD School of Medicine and Medical Science, University College Dublin, Belfield, Dublin, Ireland
| | - Paula Murphy
- Zoology Department, School of Natural Sciences, Trinity College Dublin, Dublin, Ireland
| | - John Crean
- Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Research, UCD School of Biomolecular and Biomedical Science, Belfield, Dublin, Ireland;
| |
Collapse
|
29
|
Kimura-Yoshida C, Mochida K, Ellwanger K, Niehrs C, Matsuo I. Fate Specification of Neural Plate Border by Canonical Wnt Signaling and Grhl3 is Crucial for Neural Tube Closure. EBioMedicine 2015; 2:513-27. [PMID: 26288816 PMCID: PMC4535158 DOI: 10.1016/j.ebiom.2015.04.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/17/2015] [Accepted: 04/17/2015] [Indexed: 12/05/2022] Open
Abstract
During primary neurulation, the separation of a single-layered ectodermal sheet into the surface ectoderm (SE) and neural tube specifies SE and neural ectoderm (NE) cell fates. The mechanisms underlying fate specification in conjunction with neural tube closure are poorly understood. Here, by comparing expression profiles between SE and NE lineages, we observed that uncommitted progenitor cells, expressing stem cell markers, are present in the neural plate border/neural fold prior to neural tube closure. Our results also demonstrated that canonical Wnt and its antagonists, DKK1/KREMEN1, progressively specify these progenitors into SE or NE fates in accord with the progress of neural tube closure. Additionally, SE specification of the neural plate border via canonical Wnt signaling is directed by the grainyhead-like 3 (Grhl3) transcription factor. Thus, we propose that the fate specification of uncommitted progenitors in the neural plate border by canonical Wnt signaling and its downstream effector Grhl3 is crucial for neural tube closure. This study implicates that failure in critical genetic factors controlling fate specification of progenitor cells in the neural plate border/neural fold coordinated with neural tube closure may be potential causes of human neural tube defects. Neural plate border/neural fold possesses stem cell-like characters during primary neurulation. Canonical Wnt and its antagonists progressively specify progenitors into surface or neural fates upon neural tube closure. Fate specification into surface ectoderm in the neural fold is directed by the Grhl3 transcription factor. Fate specification of uncommitted progenitors in the neural plate border is intimately coupled to neural tube closure.
Collapse
Affiliation(s)
- Chiharu Kimura-Yoshida
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Kyoko Mochida
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| | - Kristina Ellwanger
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany ; Institute of Molecular Biology, 55128 Mainz, Germany
| | - Isao Matsuo
- Department of Molecular Embryology, Osaka Medical Center, Research Institute for Maternal and Child Health, Osaka Prefectural Hospital Organization, 840 Murodo-cho, Izumi, Osaka 594-1101, Japan
| |
Collapse
|
30
|
Nagalakshmi VK, Yu J. The ureteric bud epithelium: morphogenesis and roles in metanephric kidney patterning. Mol Reprod Dev 2015; 82:151-66. [PMID: 25783232 DOI: 10.1002/mrd.22462] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/12/2015] [Indexed: 01/03/2023]
Abstract
The mammalian metanephric kidney is composed of two epithelial components, the collecting duct system and the nephron epithelium, that differentiate from two different tissues -the ureteric bud epithelium and the nephron progenitors, respectively-of intermediate mesoderm origin. The collecting duct system is generated through reiterative ureteric bud branching morphogenesis, whereas the nephron epithelium is formed in a process termed nephrogenesis, which is initiated with the mesenchymal-epithelial transition of the nephron progenitors. Ureteric bud branching morphogenesis is regulated by nephron progenitors, and in return, the ureteric bud epithelium regulates nephrogenesis. The metanephric kidney is physiologically divided along the corticomedullary axis into subcompartments that are enriched with specific segments of these two epithelial structures. Here, we provide an overview of the major molecular and cellular processes underlying the morphogenesis and patterning of the ureteric bud epithelium and its roles in the cortico-medullary patterning of the metanephric kidney.
Collapse
Affiliation(s)
- Vidya K Nagalakshmi
- Department of Cell Biology and Division of Center of Immunity, Inflammation and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | | |
Collapse
|
31
|
Moura RS, Carvalho-Correia E, daMota P, Correia-Pinto J. Canonical Wnt signaling activity in early stages of chick lung development. PLoS One 2014; 9:e112388. [PMID: 25460002 PMCID: PMC4251901 DOI: 10.1371/journal.pone.0112388] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 10/15/2014] [Indexed: 01/08/2023] Open
Abstract
Wnt signaling pathway is an essential player during vertebrate embryonic development which has been associated with several developmental processes such as gastrulation, body axis formation and morphogenesis of numerous organs, namely the lung. Wnt proteins act through specific transmembrane receptors, which activate intracellular pathways that regulate cellular processes such as cell proliferation, differentiation and death. Morphogenesis of the fetal lung depends on epithelial-mesenchymal interactions that are governed by several growth and transcription factors that regulate cell proliferation, fate, migration and differentiation. This process is controlled by different signaling pathways such as FGF, Shh and Wnt among others. Wnt signaling is recognized as a key molecular player in mammalian pulmonary development but little is known about its function in avian lung development. The present work characterizes, for the first time, the expression pattern of several Wnt signaling members, such as wnt-1, wnt-2b, wnt-3a, wnt-5a, wnt-7b, wnt-8b, wnt-9a, lrp5, lrp6, sfrp1, dkk1, β-catenin and axin2 at early stages of chick lung development. In general, their expression is similar to their mammalian counterparts. By assessing protein expression levels of active/total β-catenin and phospho-LRP6/LRP6 it is revealed that canonical Wnt signaling is active in this embryonic tissue. In vitro inhibition studies were performed in order to evaluate the function of Wnt signaling pathway in lung branching. Lung explants treated with canonical Wnt signaling inhibitors (FH535 and PK115-584) presented an impairment of secondary branch formation after 48 h of culture along with a decrease in axin2 expression levels. Branching analysis confirmed this inhibition. Wnt-FGF crosstalk assessment revealed that this interaction is preserved in the chick lung. This study demonstrates that Wnt signaling is crucial for precise chick lung branching and further supports the avian lung as a good model for branching studies since it recapitulates early mammalian pulmonary development.
Collapse
Affiliation(s)
- Rute Silva Moura
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Biology Department, School of Sciences, University of Minho, Braga, Portugal
| | - Eduarda Carvalho-Correia
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paulo daMota
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Urology, Hospital de Braga, Braga, Portugal
| | - Jorge Correia-Pinto
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Pediatric Surgery, Hospital de Braga, Braga, Portugal
| |
Collapse
|
32
|
Halt K, Vainio S. Coordination of kidney organogenesis by Wnt signaling. Pediatr Nephrol 2014; 29:737-44. [PMID: 24445433 PMCID: PMC3928513 DOI: 10.1007/s00467-013-2733-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 10/25/2013] [Accepted: 12/12/2013] [Indexed: 11/28/2022]
Abstract
Several Wnt proteins are expressed in the embryonic kidney during various stages of development. Gene knockout models and ex vivo studies have provided strong evidence that Wnt-mediated signals are essential in renal ontogeny. Perhaps the most critical factors, Wnt9b and Wnt4, function during the early phase when the cap mesenchyme is induced to undergo morphogenesis into a nephron. Wnt11 controls early ureteric bud branching and contributes to the final kidney size. In addition to its inductive role, later on Wnt9b plays a significant role in the convergent extension of the tubular epithelial cells, while Wnt4 signaling controls smooth muscle cell fates in the medulla. Wnt7b has a specific function together with its likely antagonist Dkk1 in controlling the morphogenesis of the renal medulla. The signal-transduction mechanisms of the Wnts in kidney ontogeny have not been resolved, but studies characterizing the downstream signaling pathways are emerging. Aberrant Wnt signaling may lead to kidney diseases ranging from fatal kidney agenesis to more benign phenotypes. Wnt-mediated signaling regulates several critical aspects of kidney development from the early inductive stages to later steps of tubular epithelial maturation.
Collapse
Affiliation(s)
- Kimmo Halt
- The Centre of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
- Biocenter Oulu, Oulu, Finland
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Institute of Biomedicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland
| | - Seppo Vainio
- The Centre of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
- Biocenter Oulu, Oulu, Finland
- Laboratory of Developmental Biology, Department of Medical Biochemistry and Molecular Biology, Institute of Biomedicine, University of Oulu, PO Box 5000, 90014 Oulu, Finland
| |
Collapse
|
33
|
Fang Y, Ginsberg C, Seifert M, Agapova O, Sugatani T, Register TC, Freedman BI, Monier-Faugere MC, Malluche H, Hruska KA. CKD-induced wingless/integration1 inhibitors and phosphorus cause the CKD-mineral and bone disorder. J Am Soc Nephrol 2014; 25:1760-73. [PMID: 24578135 DOI: 10.1681/asn.2013080818] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
In chronic kidney disease, vascular calcification, renal osteodystrophy, and phosphate contribute substantially to cardiovascular risk and are components of CKD-mineral and bone disorder (CKD-MBD). The cause of this syndrome is unknown. Additionally, no therapy addresses cardiovascular risk in CKD. In its inception, CKD-MBD is characterized by osteodystrophy, vascular calcification, and stimulation of osteocyte secretion. We tested the hypothesis that increased production of circulating factors by diseased kidneys causes the CKD-MBD in diabetic mice subjected to renal injury to induce stage 2 CKD (CKD-2 mice). Compared with non-CKD diabetic controls, CKD-2 mice showed increased renal production of Wnt inhibitor family members and higher levels of circulating Dickkopf-1 (Dkk1), sclerostin, and secreted klotho. Neutralization of Dkk1 in CKD-2 mice by administration of a monoclonal antibody after renal injury stimulated bone formation rates, corrected the osteodystrophy, and prevented CKD-stimulated vascular calcification. Mechanistically, neutralization of Dkk1 suppressed aortic expression of the osteoblastic transcription factor Runx2, increased expression of vascular smooth muscle protein 22-α, and restored aortic expression of klotho. Neutralization of Dkk1 did not affect the elevated plasma levels of osteocytic fibroblast growth factor 23 but decreased the elevated levels of sclerostin. Phosphate binder therapy restored plasma fibroblast growth factor 23 levels but had no effect on vascular calcification or osteodystrophy. The combination of the Dkk1 antibody and phosphate binder therapy completely treated the CKD-MBD. These results show that circulating Wnt inhibitors are involved in the pathogenesis of CKD-MBD and that the combination of Dkk1 neutralization and phosphate binding may have therapeutic potential for this disorder.
Collapse
Affiliation(s)
- Yifu Fang
- Departments of Pediatrics/Nephrology and
| | | | - Michael Seifert
- Departments of Pediatrics/Nephrology and Department of Pediatric Nephrology, Southern Illinois School of Medicine, Springfield, Illinois
| | | | | | | | - Barry I Freedman
- Internal Medicine/Nephrology, Wake Forest School of Medicine, Winston-Salem, North Carolina; and
| | | | - Hartmut Malluche
- Department of Medicine/Nephrology, University of Kentucky, Lexington, Kentucky
| | - Keith A Hruska
- Departments of Pediatrics/Nephrology and Medicine, Washington University, St. Louis, Missouri;
| |
Collapse
|
34
|
Seib DRM, Corsini NS, Ellwanger K, Plaas C, Mateos A, Pitzer C, Niehrs C, Celikel T, Martin-Villalba A. Loss of Dickkopf-1 restores neurogenesis in old age and counteracts cognitive decline. Cell Stem Cell 2013; 12:204-14. [PMID: 23395445 DOI: 10.1016/j.stem.2012.11.010] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 09/11/2012] [Accepted: 11/15/2012] [Indexed: 12/19/2022]
Abstract
Memory impairment has been associated with age-related decline in adult hippocampal neurogenesis. Although Notch, bone morphogenetic protein, and Wnt signaling pathways are known to regulate multiple aspects of adult neural stem cell function, the molecular basis of declining neurogenesis in the aging hippocampus remains unknown. Here, we show that expression of the Wnt antagonist Dickkopf-1 (Dkk1) increases with age and that its loss enhances neurogenesis in the hippocampus. Neural progenitors with inducible loss of Dkk1 increase their Wnt activity, which leads to enhanced self-renewal and increased generation of immature neurons. This Wnt-expanded progeny subsequently matures into glutamatergic granule neurons with increased dendritic complexity. As a result, mice deficient in Dkk1 exhibit enhanced spatial working memory and memory consolidation and also show improvements in affective behavior. Taken together, our findings show that upregulating Wnt signaling by reducing Dkk1 expression can counteract age-related decrease in neurogenesis and its associated cognitive decline.
Collapse
Affiliation(s)
- Désirée R M Seib
- Division of Molecular Neurobiology, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Maupin KA, Droscha CJ, Williams BO. A Comprehensive Overview of Skeletal Phenotypes Associated with Alterations in Wnt/β-catenin Signaling in Humans and Mice. Bone Res 2013; 1:27-71. [PMID: 26273492 DOI: 10.4248/br201301004] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 01/20/2013] [Indexed: 12/23/2022] Open
Abstract
The Wnt signaling pathway plays key roles in differentiation and development and alterations in this signaling pathway are causally associated with numerous human diseases. While several laboratories were examining roles for Wnt signaling in skeletal development during the 1990s, interest in the pathway rose exponentially when three key papers were published in 2001-2002. One report found that loss of the Wnt co-receptor, Low-density lipoprotein related protein-5 (LRP5), was the underlying genetic cause of the syndrome Osteoporosis pseudoglioma (OPPG). OPPG is characterized by early-onset osteoporosis causing increased susceptibility to debilitating fractures. Shortly thereafter, two groups reported that individuals carrying a specific point mutation in LRP5 (G171V) develop high-bone mass. Subsequent to this, the causative mechanisms for these observations heightened the need to understand the mechanisms by which Wnt signaling controlled bone development and homeostasis and encouraged significant investment from biotechnology and pharmaceutical companies to develop methods to activate Wnt signaling to increase bone mass to treat osteoporosis and other bone disease. In this review, we will briefly summarize the cellular mechanisms underlying Wnt signaling and discuss the observations related to OPPG and the high-bone mass disorders that heightened the appreciation of the role of Wnt signaling in normal bone development and homeostasis. We will then present a comprehensive overview of the core components of the pathway with an emphasis on the phenotypes associated with mice carrying genetically engineered mutations in these genes and clinical observations that further link alterations in the pathway to changes in human bone.
Collapse
Affiliation(s)
- Kevin A Maupin
- Program for Skeletal Pathobiology and Center for Tumor Metastasis, Van Andel Research Institute , 333 Bostwick NE, Grand Rapids, MI 49503, USA
| | - Casey J Droscha
- Program for Skeletal Pathobiology and Center for Tumor Metastasis, Van Andel Research Institute , 333 Bostwick NE, Grand Rapids, MI 49503, USA
| | - Bart O Williams
- Program for Skeletal Pathobiology and Center for Tumor Metastasis, Van Andel Research Institute , 333 Bostwick NE, Grand Rapids, MI 49503, USA
| |
Collapse
|
36
|
Kawakami T, Ren S, Duffield JS. Wnt signalling in kidney diseases: dual roles in renal injury and repair. J Pathol 2012; 229:221-31. [DOI: 10.1002/path.4121] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 09/24/2012] [Accepted: 09/30/2012] [Indexed: 01/25/2023]
Affiliation(s)
- Takahisa Kawakami
- Division of Nephrology, Center for Lung Biology, Department of Medicine, and Institute of Stem Cell and Regenerative Medicine; University of Washington; Seattle WA USA
| | - Shuyu Ren
- Division of Nephrology, Center for Lung Biology, Department of Medicine, and Institute of Stem Cell and Regenerative Medicine; University of Washington; Seattle WA USA
| | - Jeremy S Duffield
- Division of Nephrology, Center for Lung Biology, Department of Medicine, and Institute of Stem Cell and Regenerative Medicine; University of Washington; Seattle WA USA
| |
Collapse
|
37
|
Maezawa Y, Binnie M, Li C, Thorner P, Hui CC, Alman B, Taketo MM, Quaggin SE. A new Cre driver mouse line, Tcf21/Pod1-Cre, targets metanephric mesenchyme. PLoS One 2012; 7:e40547. [PMID: 22792366 PMCID: PMC3391250 DOI: 10.1371/journal.pone.0040547] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 06/08/2012] [Indexed: 11/19/2022] Open
Abstract
Conditional gene targeting in mice has provided great insight into the role of gene function in kidney development and disease. Although a number of Cre-driver mouse strains already exist for the kidney, development of additional strains with unique expression patterns is needed. Here we report the generation and validation of a Tcf21/Pod1-Cre driver strain that expresses Cre recombinase throughout the condensing and stromal mesenchyme of developing kidneys and in their derivatives including epithelial components of the nephron and interstitial cells. To test the efficiency of this line, we crossed it to mice transgenic for either loss or gain of function β-catenin conditional alleles. Mice with deletion of β-catenin from Tcf21-expressing cells are born with hypoplastic kidneys, hydroureters and hydronephrosis. By contrast, Tcf21-Cre driven gain of function for β-catenin in mice results in fused midline kidneys and hypoplastic kidneys. Finally, we report the first renal mesenchymal deletion of Patched1 (Ptch1), the receptor for sonic hedgehog (Shh), which results in renal cysts demonstrating a functional role of Shh signaling pathway in renal cystogensis. In summary, we report the generation and validation of a new Cre driver strain that provides robust excision in metanephric mesenchyme.
Collapse
Affiliation(s)
- Yoshiro Maezawa
- The Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Matthew Binnie
- Division of Respirology, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Chengjin Li
- The Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Paul Thorner
- Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chi-Chung Hui
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Benjamin Alman
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Orthopaedic Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Makoto Mark Taketo
- Department of Pharmacology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Susan E. Quaggin
- The Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
- Division of Nephrology, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
38
|
Wilkinson L, Kurniawan ND, Phua YL, Nguyen MJ, Li J, Galloway GJ, Hashitani H, Lang RJ, Little MH. Association between congenital defects in papillary outgrowth and functional obstruction in Crim1 mutant mice. J Pathol 2012; 227:499-510. [PMID: 22488641 DOI: 10.1002/path.4036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 03/09/2012] [Accepted: 03/29/2012] [Indexed: 12/25/2022]
Abstract
Crim1 hypomorphic (Crim1(KST264/KST264)) mice display progressive renal disease characterized by glomerular defects, leaky peritubular vasculature, and progressive interstitial fibrosis. Here we show that 27% of these mice also present with hydronephrosis, suggesting obstructive nephropathy. Dynamic magnetic resonance imaging using Magnevist showed fast development of hypo-intense signal in the kidneys of Crim1(KST264/KST264) mice, suggesting pooling of filtrate within the renal parenchyma. Rhodamine dextran (10 kDa) clearance was also delayed in Crim1(KST264/KST264) mice. Pyeloureteric peristalsis, while present, was less co-ordinated in Crim1(KST264/KST264) mice. However, isolated renal pelvis preparations suggest normal pelvic smooth muscle contractile responses. An analysis of maturation during the immediate postnatal period [postnatal day (P) 0-15] revealed defects in papillary extension in Crim1({KST264/KST264) mice. While Crim1 expression is weak in pelvic smooth muscle, strong expression is seen in the interstitium and loops of Henle of the extending papilla, commencing at the tip of the P1 papilla and disseminating throughout the papilla by P15. These results, as well as implicating Crim1 in papillary extension and pelvic smooth muscle contractility, highlight the previously unrecognized association between defects in papillary development and progression to chronic kidney disease later in life.
Collapse
Affiliation(s)
- Lorine Wilkinson
- Institute for Molecular Bioscience, The University of Queensland, QLD 4072, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Song R, Preston G, Khalili A, El-Dahr SS, Yosypiv IV. Angiotensin II regulates growth of the developing papillas ex vivo. Am J Physiol Renal Physiol 2012; 302:F1112-20. [PMID: 22301625 DOI: 10.1152/ajprenal.00435.2011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We tested the hypothesis that lack of angiotensin (ANG) II production in angiotensinogen (AGT)-deficient mice or pharmacologic antagonism of ANG II AT(1) receptor (AT(1)R) impairs growth of the developing papillas ex vivo, thus contributing to the hypoplastic renal medulla phenotype observed in AGT- or AT(1)R-null mice. Papillas were dissected from Hoxb7(GFP+) or AGT(+/+), (+/-), (-/-) mouse metanephroi on postnatal day P3 and grown in three-dimentional collagen matrix gels in the presence of media (control), ANG II (10(-5) M), or the specific AT(1)R antagonist candesartan (10(-6) M) for 24 h. Percent reduction in papillary length was attenuated in AGT(+/+) and in AGT(+/-) compared with AGT(-/-) (-18.4 ± 1.3 vs. -32.2 ± 1.6%, P < 0.05, -22.8 ± 1.3 vs. -32.2 ± 1.6%, P < 0.05, respectively). ANG II blunted the decrease in papilla length observed in respective media-treated controls in Hoxb7(GFP+) (-1.5 ± 0.3 vs. -10.0 ± 1.4%, P < 0.05) or AGT(+/+), (+/-), and (-/-) papillas (-12.8 ± 0.7 vs. -18.4 ± 1.3%, P < 0.05, -16.8 ± 1.1 vs. -23 ± 1.2%, P < 0.05; -26.2 ± 1.6 vs. -32.2 ± 1.6%, P < 0.05, respectively). In contrast, percent decrease in the length of Hoxb7(GFP+) papillas in the presence of the AT(1)R antagonist candesartan was higher compared with control (-24.3 ± 2.1 vs. -10.5 ± 1.8%, P < 0.05). The number of proliferating phospho-histone H3 (pH3)-positive collecting duct cells was lower, whereas the number of caspase 3-positive cells undergoing apoptosis was higher in candesartan- vs. media-treated papillas (pH3: 12 ± 1.4 vs. 21 ± 2.1, P < 0.01; caspase 3: 3.8 ± 0.5 vs. 1.7 ± 0.2, P < 0.01). Using quantitative RT-PCR, we demonstrate that AT(1)R signaling regulates the expression of genes implicated in morphogenesis of the renal medulla. We conclude that AT(1)R prevents shrinkage of the developing papillas observed ex vivo via control of Wnt7b, FGF7, β-catenin, calcineurin B1, and α3 integrin gene expression, collecting duct cell proliferation, and survival.
Collapse
Affiliation(s)
- Renfang Song
- Division of Pediatric Nephrology, Department of Pediatrics, Hypertension, and Renal Center of Excellence, Tulane University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
40
|
Abstract
The mature renal medulla, the inner part of the kidney, consists of the medullary collecting ducts, loops of Henle, vasa recta and the interstitium. The unique spatial arrangement of these components is essential for the regulation of urine concentration and other specialized kidney functions. Thus, the proper and timely assembly of medulla constituents is a crucial morphogenetic event leading to the formation of a functioning metanephric kidney. Mechanisms that direct renal medulla formation are poorly understood. This review describes the current understanding of the key molecular and cellular mechanisms underlying morphological aspects of medulla formation. Given that hypoplasia of the renal medulla is a common manifestation of congenital obstructive nephropathy and other types of congenital anomalies of the kidney and urinary tract (CAKUT), better understanding of how disruptions in medulla formation are linked to CAKUT will enable improved diagnosis, treatment and prevention of CAKUT and their associated morbidity.
Collapse
Affiliation(s)
- Renfang Song
- Section of Pediatric Nephrology, Department of Pediatrics, Hypertension and Renal Center of Excellence, Tulane University School of Medicine, New Orleans, LA, USA
| | | |
Collapse
|
41
|
Abstract
Renal cell carcinoma (RCC) is the most lethal of all the genitourinary cancers, as it is generally refractory to current treatment regimens, including chemotherapy and radiation therapy. Targeted therapies against critical signaling pathways associated with RCC pathogenesis, such as vascular endothelial growth factor, von Hippel-Lindau tumor suppressor and mammalian target of rapamycin, have shown limited efficacy so far. Thus, Wnt signaling, which is known to be intricately involved in the pathogenesis of RCC, has attracted much interest. Several Wnt signaling components have been examined in RCC, and, while studies suggest that Wnt signaling is constitutively active in RCC, the molecular mechanisms differ considerably from other human carcinomas. Increasing evidence indicates that secreted Wnt antagonists have important roles in RCC pathogenesis. Considering these vital roles, it has been postulated--and supported by experimental evidence--that the functional loss of Wnt antagonists, for example by promoter hypermethylation, can contribute to constitutive activation of the Wnt pathway, resulting in carcinogenesis through dysregulation of cell proliferation and differentiation. However, subsequent functional studies of these Wnt antagonists have demonstrated the inherent complexities underlying their role in RCC pathogenesis.
Collapse
|
42
|
Grouls S, Iglesias DM, Wentzensen N, Moeller MJ, Bouchard M, Kemler R, Goodyer P, Niggli F, Gröne HJ, Kriz W, Koesters R. Lineage specification of parietal epithelial cells requires β-catenin/Wnt signaling. J Am Soc Nephrol 2011; 23:63-72. [PMID: 22021707 DOI: 10.1681/asn.2010121257] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
β-Catenin/Wnt signaling is essential during early inductive stages of kidney development, but its role during postinductive stages of nephron development and maturation is not well understood. In this study, we used Pax8Cre mice to target β-catenin deficiency to renal epithelial cells at the late S-shaped body stage and the developing collecting ducts. The conditional β-catenin knockout mice formed abnormal kidneys and had reduced renal function. The kidneys were hypoplastic with a thin cortex; a superficial layer of tubules was missing. A high proportion of glomeruli had small, underdeveloped capillary tufts. In these glomeruli, well differentiated podocytes replaced parietal epithelial cells in Bowman's capsule; capillaries toward the outer aspect of these podocytes mimicked the formation of glomerular capillaries. Tracing nephrogenesis in embryonic conditional β-catenin knockout mice revealed that these "parietal podocytes" derived from precursor cells in the parietal layer of the S-shaped body by direct lineage switch. Taken together, these findings demonstrate that β-catenin/Wnt signaling is important during the late stages of nephrogenesis and for the lineage specification of parietal epithelial cells.
Collapse
Affiliation(s)
- Stephan Grouls
- University of Heidelberg, Department of Nephrology, Im Neuenheimer Feld 162, 69120 Heidelberg, Baden-Württemberg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yu J. Wnt signaling and renal medulla formation. Pediatr Nephrol 2011; 26:1553-7. [PMID: 21533626 DOI: 10.1007/s00467-011-1888-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 03/07/2011] [Accepted: 03/23/2011] [Indexed: 11/29/2022]
Abstract
The renal medulla, the inner compartment of the metanephric kidney, plays vital roles in the regulation of body water, electrolyte homeostasis, and systemic blood pressure. It is composed of the loops-of-Henle, the medullary collecting ducts, the vasa recta, and the medullary interstitium. Its epithelial and endothelial components display ordered spatial organization. This organization serves as the structural basis for its function in urine concentration. The urine concentration ability of a renal medulla is also related to its length among species. In this review, the current understanding of the molecular and cellular mechanisms underlying renal medulla formation (elongation) is summarized, with a focus on the role of Wnt signaling in this developmental process. Renal medulla blunting and effacement is a common symptom of many renal and urological destructions. The knowledge in renal medulla formation should assist efforts in repair and regeneration of a damaged renal medulla, so to improve renal physiology in diseased situations.
Collapse
Affiliation(s)
- Jing Yu
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|