1
|
Wu Y, Lee TH, Cheng OH, Peden EK, Li Q, Wang J, Huang F, Melancon MP, Sheikh-Hamad D, Wang T, Truong L, Mitch WE, Liang M, Cheng J. Interplay between Skeletal Muscle Catabolism and Remodeling of Arteriovenous Fistula by Yes-Associated Protein 1 (YAP1) Signaling. J Am Soc Nephrol 2025; 36:845-858. [PMID: 39883520 PMCID: PMC12059102 DOI: 10.1681/asn.0000000605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
Key Points Atrophied muscle–derived myostatin stimulated mesenchymal stem cell differentiation and adverse arteriovenous (AV) fistula remodeling through yes-associated protein 1 (YAP1) activation. Treatment with myostatin peptibody inhibited muscle wasting and blocked mesenchymal stem cell activation and AV fistula fibrosis. A light-sensitive drug-release strategy was engineered for the periadventitial delivery of verteporfin to improve AV fistula patency. Background Arteriovenous (AV) fistulas are the preferred access for dialysis but have a high incidence of failure. The aim of this study was to understand the crosstalk between skeletal muscle catabolism and AV fistula maturation failure. Methods Skeletal muscle metabolism and AV fistula maturation were evaluated in mice with CKD. The roles of myostatin and yes-associated protein 1 (YAP1) in regulating the transdifferentiation of adventitial mesenchymal stem cells (MSCs) and intima hyperplasia in AV fistula were investigated. Nanoparticles carrying a YAP1 inhibitor, verteporfin, with light irradiation–controlled release were synthesized and applied to AV fistula. Results Increased trichrome signals and stenosis were observed in AV fistulas from mice treated with myostatin and from mice with CKD. By contrast, blocking myostatin function with an anti-myostatin peptibody not only improved body weight and muscle size in CKD mice but also decreased neointima formation in AV fistulas. In cultured MSCs, myostatin induced YAP1 expression, promoting the differentiation of MSCs into myofibroblasts and inducing extracellular matrix deposition. Red light irradiation–controlled release of verteporfin from nanoparticles blocked YAP1 activation and alleviated myostatin-induced MSC activation. Periadventitial application and red light irradiation of nanoparticles carrying verteporfin significantly suppressed stiffening and neointima formation in AV fistula. Conclusions CKD induced muscle wasting, leading to increased production of myostatin, which stimulated MSC activation and vascular fibrosis linked to AV fistula stenosis. YAP1 signaling was activated in these processes. Red light irradiation–controlled release of verteporfin offered a feasible approach for local vascular drug intervention to improve AV fistula maturation.
Collapse
Affiliation(s)
- Yongdong Wu
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Tae Hoon Lee
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Owen H. Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Eric K. Peden
- Department of Surgery, Houston Methodist Hospital, Houston, Texas
| | - Qingtian Li
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas
| | - Fengzhang Huang
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Marites P. Melancon
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Sheikh-Hamad
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Tao Wang
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Luan Truong
- Department of Pathology and Genomic medicine, Houston Methodist Hospital, Houston, Texas
| | - William E. Mitch
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| | - Ming Liang
- Department of Nephrology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jizhong Cheng
- Section of Nephrology, Department of Medicine, Selzman Institute for Kidney Health, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
2
|
Williams MJ, Patel HM, Halling CB, Hruska KA. The Impact of a Western Diet High in Phosphate on the CKD-MBD in an Alport Syndrome Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633378. [PMID: 39896481 PMCID: PMC11785106 DOI: 10.1101/2025.01.17.633378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Background Chronic kidney disease - mineral bone disorder (CKD-MBD) is a syndrome that begins early in CKD, contributes to CKD-associated mortality, and includes components of FGF23 elevation, αklotho deficiency, CKD-stimulated vascular disease, and renal osteodystrophy. Hyperphosphatemia, occurring in later stages of CKD, is also driven by mechanisms of CKD-MBD, and has been shown to stimulate vascular calcification. In a mouse model of Alport CKD that is resistant to vascular calcification, we examine the effects of a high-phosphate Western-type diet on the CKD-MBD, and test whether the diet promotes induction of vascular calcification. Methods An X-linked Col4a5 deficient murine homolog of Alport Syndrome (CKD) and wild type (WT) littermates were fed an animal protein 1.2% high phosphate diet or a standard vegetable protein diet. At disease progression equivalent to CKD stage 4-5, we examined kidney histology for fibrosis, blood for BUN (marker of CKD), and markers of CKD-MBD disease progression, kidney tissue for klotho production, and aorta histology and tissue mRNA and protein analysis for vascular calcification. Results The Western high Pi diet produced hyperphosphatemia in the CKD animals compared to WT and increased plasma PTH (1880 from 110 pg / ml), FGF23 c-term (670 from 120 pg / ml), and FGF23 intact (3780 from 280 pg / ml), and reduced kidney klotho mRNA and protein (57-67% reduction) (all p < 0.01). Referenced against the CKD animals fed vegetable-based diet, the Western high phosphate-fed CKD animals showed higher levels of plasma PTH and FGF23s. In the wild-type control mice with normal renal function, Western diet produced increased PTH, intact FGF23, and reduced renal klotho (all p <0.01). Vascular smooth muscle transdifferentiation and vascular calcification was not induced by Western high phosphate diet in this model of CKD. Conclusions Our results show that a Western-style high-phosphate diet advances elements of the CKD-MBD. Renal klotho, FGF23 and PTH are affected by diet even with normal kidney function, suggesting a need for early intervention in the management of phosphate homeostasis as a component of CKD therapy. Additionally, CKD, klotho, and FGF23 all are associated with early aging. Therefore, our findings suggest that a Western high Pi diet accelerates aging and would contribute to the systemic complications of CKD - cardiac disease, osteodystrophy, and vascular disease.
Collapse
|
3
|
Deng AF, Wang FX, Wang SC, Zhang YZ, Bai L, Su JC. Bone-organ axes: bidirectional crosstalk. Mil Med Res 2024; 11:37. [PMID: 38867330 PMCID: PMC11167910 DOI: 10.1186/s40779-024-00540-9] [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: 10/09/2023] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
In addition to its recognized role in providing structural support, bone plays a crucial role in maintaining the functionality and balance of various organs by secreting specific cytokines (also known as osteokines). This reciprocal influence extends to these organs modulating bone homeostasis and development, although this aspect has yet to be systematically reviewed. This review aims to elucidate this bidirectional crosstalk, with a particular focus on the role of osteokines. Additionally, it presents a unique compilation of evidence highlighting the critical function of extracellular vesicles (EVs) within bone-organ axes for the first time. Moreover, it explores the implications of this crosstalk for designing and implementing bone-on-chips and assembloids, underscoring the importance of comprehending these interactions for advancing physiologically relevant in vitro models. Consequently, this review establishes a robust theoretical foundation for preventing, diagnosing, and treating diseases related to the bone-organ axis from the perspective of cytokines, EVs, hormones, and metabolites.
Collapse
Affiliation(s)
- An-Fu Deng
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Fu-Xiao Wang
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
| | - Si-Cheng Wang
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200444, China
| | - Ying-Ze Zhang
- Department of Orthopaedics, the Third Hospital of Hebei Medical University, Orthopaedic Research Institution of Hebei Province, NHC Key Laboratory of Intelligent Orthopaedic Equipment, Shijiazhuang, 050051, China.
| | - Long Bai
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- School of Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, 325000, Zhejiang, China.
| | - Jia-Can Su
- Institute of Translational Medicine, Organoid Research Center, Shanghai University, Shanghai, 200444, China.
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
4
|
Williams MJ, Halabi CM, Patel HM, Joseph Z, McCommis K, Weinheimer C, Kovacs A, Lima F, Finck B, Malluche H, Hruska KA. In chronic kidney disease altered cardiac metabolism precedes cardiac hypertrophy. Am J Physiol Renal Physiol 2024; 326:F751-F767. [PMID: 38385175 PMCID: PMC11386984 DOI: 10.1152/ajprenal.00416.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/15/2024] [Accepted: 02/15/2024] [Indexed: 02/23/2024] Open
Abstract
Conduit arterial disease in chronic kidney disease (CKD) is an important cause of cardiac complications. Cardiac function in CKD has not been studied in the absence of arterial disease. In an Alport syndrome model bred not to have conduit arterial disease, mice at 225 days of life (dol) had CKD equivalent to humans with CKD stage 4-5. Parathyroid hormone (PTH) and FGF23 levels were one log order elevated, circulating sclerostin was elevated, and renal activin A was strongly induced. Aortic Ca levels were not increased, and vascular smooth muscle cell (VSMC) transdifferentiation was absent. The CKD mice were not hypertensive, and cardiac hypertrophy was absent. Freshly excised cardiac tissue respirometry (Oroboros) showed that ADP-stimulated O2 flux was diminished from 52 to 22 pmol/mg (P = 0.022). RNA-Seq of cardiac tissue from CKD mice revealed significantly decreased levels of cardiac mitochondrial oxidative phosphorylation genes. To examine the effect of activin A signaling, some Alport mice were treated with a monoclonal Ab to activin A or an isotype-matched IgG beginning at 75 days of life until euthanasia. Treatment with the activin A antibody (Ab) did not affect cardiac oxidative phosphorylation. However, the activin A antibody was active in the skeleton, disrupting the effect of CKD to stimulate osteoclast number, eroded surfaces, and the stimulation of osteoclast-driven remodeling. The data reported here show that cardiac mitochondrial respiration is impaired in CKD in the absence of conduit arterial disease. This is the first report of the direct effect of CKD on cardiac respiration.NEW & NOTEWORTHY Heart disease is an important morbidity of chronic kidney disease (CKD). Hypertension, vascular stiffness, and vascular calcification all contribute to cardiac pathophysiology. However, cardiac function in CKD devoid of vascular disease has not been studied. Here, in an animal model of human CKD without conduit arterial disease, we analyze cardiac respiration and discover that CKD directly impairs cardiac mitochondrial function by decreasing oxidative phosphorylation. Protection of cardiac oxidative phosphorylation may be a therapeutic target in CKD.
Collapse
Affiliation(s)
- Matthew J Williams
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Carmen M Halabi
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Hiral M Patel
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Zachary Joseph
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Kyle McCommis
- Geriatrics and Nutritional Science Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Carla Weinheimer
- Cardiology Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Attila Kovacs
- Cardiology Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Florence Lima
- Renal Division, Department of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Brian Finck
- Geriatrics and Nutritional Science Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
| | - Hartmut Malluche
- Renal Division, Department of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Keith A Hruska
- Renal Division, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri, United States
- Renal Division, Department of Medicine, Washington University in St. Louis, St. Louis, Missouri, United States
- Department of Cell Biology, Washington University in St. Louis, St. Louis, Missouri, United States
| |
Collapse
|
5
|
Zheng Y, Feng J, Yu Y, Ling M, Wang X. Advances in sarcopenia: mechanisms, therapeutic targets, and intervention strategies. Arch Pharm Res 2024; 47:301-324. [PMID: 38592582 DOI: 10.1007/s12272-024-01493-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/25/2024] [Indexed: 04/10/2024]
Abstract
Sarcopenia is a multifactorial condition characterized by loss of muscle mass. It poses significant health risks in older adults worldwide. Both pharmacological and non-pharmacological approaches are reported to address this disease. Certain dietary patterns, such as adequate energy intake and essential amino acids, have shown positive outcomes in preserving muscle function. Various medications, including myostatin inhibitors, growth hormones, and activin type II receptor inhibitors, have been evaluated for their effectiveness in managing sarcopenia. However, it is important to consider the variable efficacy and potential side effects associated with these treatments. There are currently no drugs approved by the Food and Drug Administration for sarcopenia. The ongoing research aims to develop more effective strategies in the future. Our review of research on disease mechanisms and drug development will be a valuable contribution to future research endeavors.
Collapse
Affiliation(s)
- Youle Zheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Yixin Yu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Min Ling
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
- MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
| |
Collapse
|
6
|
Nordholm A, Sørensen IMH, Bjergfelt SS, Fuchs A, Kofoed KF, Landler NE, Biering-Sørensen T, Carlson N, Feldt-Rasmussen B, Christoffersen C, Bro S. Plasma activin A rises with declining kidney function and is independently associated with mortality in patients with chronic kidney disease. Clin Kidney J 2023; 16:2712-2720. [PMID: 38046005 PMCID: PMC10689128 DOI: 10.1093/ckj/sfad238] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Indexed: 12/05/2023] Open
Abstract
Background Plasma (p-)activin A is elevated in chronic kidney disease-mineral and bone disorder (CKD-MBD). Activin A inhibition ameliorates CKD-MBD complications (vascular calcification and bone disease) in rodent CKD models. We examined whether p-activin A was associated with major adverse cardiovascular events (MACE), all-cause mortality and CKD-MBD complications in CKD patients. Methods The study included 916 participants (741 patients and 175 controls) from the prospective Copenhagen CKD cohort. Comparisons of p-activin A with estimated glomerular filtration rate (eGFR), coronary and thoracic aorta Agatston scores, and bone mineral density (BMD) were evaluated by univariable linear regression using Spearman's rank correlation, analysis of covariance and ordinal logistic regression with adjustments. Association of p-activin A with rates of MACE and all-cause mortality was evaluated by the Aalen-Johansen or Kaplan-Meier estimator, with subsequent multiple Cox regression analyses. Results P-activin A was increased by CKD stage 3 (124-225 pg/mL, P < .001) and correlated inversely with eGFR (r = -0.53, P < 0.01). P-activin A was associated with all-cause mortality [97 events, hazard ratio 1.55 (95% confidence interval 1.04; 2.32), P < 0.05] after adjusting for age, sex, diabetes mellitus (DM) and eGFR. Median follow-up was 4.36 (interquartile range 3.64-4.75) years. The association with MACE was not significant after eGFR adjustment. Agatston scores and BMD were not associated with p-activin A. Conclusion P-activin A increased with declining kidney function and was associated with all-cause mortality independently of age, sex, DM and eGFR. No association with MACE, vascular calcification or BMD was demonstrated.
Collapse
Affiliation(s)
- Anders Nordholm
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
- Department of Nephrology, Herlev & Gentofte Hospital, Copenhagen, Denmark
| | | | - Sasha S Bjergfelt
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Fuchs
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark
| | - Klaus F Kofoed
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nino E Landler
- Department of Cardiology, Herlev & Gentofte Hospital, Copenhagen, Denmark
| | - Tor Biering-Sørensen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Herlev & Gentofte Hospital, Copenhagen, Denmark
| | | | - Bo Feldt-Rasmussen
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Christina Christoffersen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Susanne Bro
- Department of Nephrology, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
7
|
Williams MJ, Hruska KA. Acidosis in CKD May Affect Mineralization of Newly Formed Bone According to HR-pQCT and Quantitative Back Scatter Electron Imaging. J Am Soc Nephrol 2023; 34:520-523. [PMID: 37000951 PMCID: PMC10103222 DOI: 10.1681/asn.0000000000000086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Affiliation(s)
- Matthew J. Williams
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri
| | - Keith A. Hruska
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University in St. Louis, St. Louis, Missouri
- Departments of Medicine and Cell Biology, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
8
|
Mouse Models of Mineral Bone Disorders Associated with Chronic Kidney Disease. Int J Mol Sci 2023; 24:ijms24065325. [PMID: 36982400 PMCID: PMC10048881 DOI: 10.3390/ijms24065325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
Patients with chronic kidney disease (CKD) inevitably develop mineral and bone disorders (CKD–MBD), which negatively impact their survival and quality of life. For a better understanding of underlying pathophysiology and identification of novel therapeutic approaches, mouse models are essential. CKD can be induced by surgical reduction of a functional kidney mass, by nephrotoxic compounds and by genetic engineering specifically interfering with kidney development. These models develop a large range of bone diseases, recapitulating different types of human CKD–MBD and associated sequelae, including vascular calcifications. Bones are usually studied by quantitative histomorphometry, immunohistochemistry and micro-CT, but alternative strategies have emerged, such as longitudinal in vivo osteoblast activity quantification by tracer scintigraphy. The results gained from the CKD–MBD mouse models are consistent with clinical observations and have provided significant knowledge on specific pathomechanisms, bone properties and potential novel therapeutic strategies. This review discusses available mouse models to study bone disease in CKD.
Collapse
|
9
|
Williams MJ, White SC, Joseph Z, Hruska KA. Updates in the chronic kidney disease-mineral bone disorder show the role of osteocytic proteins, a potential mechanism of the bone-Vascular paradox, a therapeutic target, and a biomarker. Front Physiol 2023; 14:1120308. [PMID: 36776982 PMCID: PMC9909112 DOI: 10.3389/fphys.2023.1120308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023] Open
Abstract
The chronic kidney disease-mineral bone disorder (CKD-MBD) is a complex multi-component syndrome occurring during kidney disease and its progression. Here, we update progress in the components of the syndrome, and synthesize recent investigations, which suggest a potential mechanism of the bone-vascular paradox. The discovery that calcified arteries in chronic kidney disease inhibit bone remodeling lead to the identification of factors produced by the vasculature that inhibit the skeleton, thus providing a potential explanation for the bone-vascular paradox. Among the factors produced by calcifying arteries, sclerostin secretion is especially enlightening. Sclerostin is a potent inhibitor of bone remodeling and an osteocyte specific protein. Its production by the vasculature in chronic kidney disease identifies the key role of vascular cell osteoblastic/osteocytic transdifferentiation in vascular calcification and renal osteodystrophy. Subsequent studies showing that inhibition of sclerostin activity by a monoclonal antibody improved bone remodeling as expected, but stimulated vascular calcification, demonstrate that vascular sclerostin functions to brake the Wnt stimulation of the calcification milieu. Thus, the target of therapy in the chronic kidney disease-mineral bone disorder is not inhibition of sclerostin function, which would intensify vascular calcification. Rather, decreasing sclerostin production by decreasing the vascular osteoblastic/osteocytic transdifferentiation is the goal. This might decrease vascular calcification, decrease vascular stiffness, decrease cardiac hypertrophy, decrease sclerostin production, reduce serum sclerostin and improve skeletal remodeling. Thus, the therapeutic target of the chronic kidney disease-mineral bone disorder may be vascular osteoblastic transdifferentiation, and sclerostin levels may be a useful biomarker for the diagnosis of the chronic kidney disease-mineral bone disorder and the progress of its therapy.
Collapse
Affiliation(s)
- Matthew J. Williams
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Sarah C. White
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Zachary Joseph
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
| | - Keith A. Hruska
- Division of Pediatric Nephrology, Department of Pediatrics, Washington University, Saint Louis, MO, United States
- Departments of Medicine and Cell Biology, Washington University, Saint Louis, MO, United States
| |
Collapse
|
10
|
Tang W, Zhang Y, Cui S, Yi F. The Growth Factors: Potential Biomarkers and Therapeutic Targets in Kidney Diseases. KIDNEY DISEASES (BASEL, SWITZERLAND) 2022; 8:368-380. [PMID: 36466071 PMCID: PMC9710479 DOI: 10.1159/000526208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/17/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Kidney diseases are a prevalent health problem worldwide. Although substantial progress has been made in understanding the pathophysiology of kidney disease, currently there is no satisfactory clinical treatment available to prevent or treat kidney disease. Therefore, strategies to establish early diagnosis, identify the key molecules, and develop novel therapeutic interventions to slow the progression of kidney diseases and reduce their complications are encouraged. SUMMARY The growth factors play a crucial role in the development of kidney diseases. The altered levels of growth factors are usually detected in circulation and urine in the disease course. A growing body of studies has suggested that growth factors, receptors, and related regulators are promising biomarkers for the diagnosis and/or prognosis and potential therapeutic targets for the treatment of kidney diseases. In this review, we summarize recent advances in the potential applications of growth factors for diagnostic biomarkers and therapeutic targets in kidney diseases and highlight their performances in clinical trials. KEY MESSAGES Most diagnostic and therapeutic strategies targeting growth factors are still far from clinical implementation. The better understanding of growth factor-regulated pathophysiology and the progress of new intervention approaches are expected to facilitate the clinical translation of growth factor-based diagnosis and therapy of kidney diseases.
Collapse
Affiliation(s)
- Wei Tang
- Department of Pharmacology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yufeng Zhang
- Department of Pharmacology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Sijia Cui
- Department of Pharmacology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, The Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, China
- Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, and The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
11
|
Mao M, Labelle-Dumais C, Tufa SF, Keene DR, Gould DB. Elevated TGFβ signaling contributes to ocular anterior segment dysgenesis in Col4a1 mutant mice. Matrix Biol 2022; 110:151-173. [PMID: 35525525 PMCID: PMC10410753 DOI: 10.1016/j.matbio.2022.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/08/2022] [Accepted: 05/02/2022] [Indexed: 10/18/2022]
Abstract
Ocular anterior segment dysgenesis (ASD) refers to a collection of developmental disorders affecting the anterior structures of the eye. Although a number of genes have been implicated in the etiology of ASD, the underlying pathogenetic mechanisms remain unclear. Mutations in genes encoding collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) cause Gould syndrome, a multi-system disorder that often includes ocular manifestations such as ASD and glaucoma. COL4A1 and COL4A2 are abundant basement membrane proteins that provide structural support to tissues and modulate signaling through interactions with other extracellular matrix proteins, growth factors, and cell surface receptors. In this study, we used a combination of histological, molecular, genetic and pharmacological approaches to demonstrate that altered TGFβ signaling contributes to ASD in mouse models of Gould syndrome. We show that TGFβ signaling was elevated in anterior segments from Col4a1 mutant mice and that genetically reducing TGFβ signaling partially prevented ASD. Notably, we identified distinct roles for TGFβ1 and TGFβ2 in ocular defects observed in Col4a1 mutant mice. Importantly, we show that pharmacologically promoting type IV collagen secretion or reducing TGFβ signaling ameliorated ocular pathology in Col4a1 mutant mice. Overall, our findings demonstrate that altered TGFβ signaling contributes to COL4A1-related ocular dysgenesis and implicate this pathway as a potential therapeutic target for the treatment of Gould syndrome.
Collapse
Affiliation(s)
- Mao Mao
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Sara F Tufa
- Shriners Children's, Micro-Imaging Center, Portland, Oregon 97239, United States
| | - Douglas R Keene
- Shriners Children's, Micro-Imaging Center, Portland, Oregon 97239, United States
| | - Douglas B Gould
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, United States; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, United States; Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, United States; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143, United States; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94143, United States.
| |
Collapse
|
12
|
Yamada S, Tsuruya K, Kitazono T, Nakano T. Emerging cross-talks between chronic kidney disease-mineral and bone disorder (CKD-MBD) and malnutrition-inflammation complex syndrome (MICS) in patients receiving dialysis. Clin Exp Nephrol 2022; 26:613-629. [PMID: 35353283 PMCID: PMC9203392 DOI: 10.1007/s10157-022-02216-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/22/2022] [Indexed: 12/14/2022]
Abstract
Chronic kidney disease–mineral and bone disorder (CKD–MBD) is a systemic disorder that affects multiple organs and systems and increases the risk of morbidity and mortality in patients with CKD, especially those receiving dialysis therapy. CKD–MBD is highly prevalent in CKD patients, and its treatment is gaining attention from healthcare providers who manage these patients. Additional important pathologies often observed in CKD patients are chronic inflammation and malnutrition/protein-energy wasting (PEW). These two pathologies coexist to form a vicious cycle that accelerates the progression of various other pathologies in CKD patients. This concept is integrated into the term “malnutrition–inflammation–atherosclerosis syndrome” or “malnutrition–inflammation complex syndrome (MICS)”. Recent basic and clinical studies have shown that CKD–MBD directly induces inflammation as well as malnutrition/PEW. Indeed, higher circulating levels of inorganic phosphate, fibroblast growth factor 23, parathyroid hormone, and calciprotein particles, as markers for critical components and effectors of CKD–MBD, were shown to directly induce inflammatory responses, thereby leading to malnutrition/PEW, cardiovascular diseases, and clinically relevant complications. In this short review, we discuss the close interplay between CKD–MBD and MICS and emphasize the significance of simultaneous control of these two seemingly distinct pathologies in patients with CKD, especially those receiving dialysis therapy, for better management of the CKD/hemodialysis population.
Collapse
Affiliation(s)
- Shunsuke Yamada
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan.
| | | | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan
| | - Toshiaki Nakano
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 8128582, Japan
| |
Collapse
|
13
|
Cosgrove D, Madison J. Molecular and Cellular Mechanisms Underlying the Initiation and Progression of Alport Glomerular Pathology. Front Med (Lausanne) 2022; 9:846152. [PMID: 35223933 PMCID: PMC8863674 DOI: 10.3389/fmed.2022.846152] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 01/17/2022] [Indexed: 01/11/2023] Open
Abstract
Alport syndrome results from a myriad of variants in the COL4A3, COL4A4, or COL4A5 genes that encode type IV (basement membrane) collagens. Unlike type IV collagen α1(IV)2α2(IV)1 heterotrimers, which are ubiquitous in basement membranes, α3/α4/α5 have a limited tissue distribution. The absence of these basement membrane networks causes pathologies in some, but not all these tissues. Primarily the kidney glomerulus, the stria vascularis of the inner ear, the lens, and the retina as well as a rare link with aortic aneurisms. Defects in the glomerular basement membranes results in delayed onset and progressive focal segmental glomerulosclerosis ultimately requiring the patient to undergo dialysis and if accessible, kidney transplant. The lifespan of patients with Alport syndrome is ultimately significantly shortened. This review addresses the consequences of the altered glomerular basement membrane composition in Alport syndrome with specific emphasis on the mechanisms underlying initiation and progression of glomerular pathology.
Collapse
Affiliation(s)
| | - Jacob Madison
- Boys Town National Research Hospital, Omaha, NE, United States
| |
Collapse
|
14
|
Guo FH, Guan YN, Guo JJ, Zhang LJ, Qiu JJ, Ji Y, Chen AF, Jing Q. Single-Cell Transcriptome Analysis Reveals Embryonic Endothelial Heterogeneity at Spatiotemporal Level and Multifunctions of MicroRNA-126 in Mice. Arterioscler Thromb Vasc Biol 2022; 42:326-342. [PMID: 35021856 PMCID: PMC8860216 DOI: 10.1161/atvbaha.121.317093] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Supplemental Digital Content is available in the text. Endothelial cells (ECs) play a critical role in angiogenesis and vascular remodeling. The heterogeneity of ECs has been reported at adult stages, yet it has not been fully investigated. This study aims to assess the transcriptional heterogeneity of developmental ECs at spatiotemporal level and to reveal the changes of embryonic ECs clustering when endothelium-enriched microRNA-126 (miR-126) was specifically knocked out.
Collapse
Affiliation(s)
- Fang-Hao Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (F.-H.G., Y.-N.G., J.-J.G., J.J.Q., Q.J.)
| | - Ya-Na Guan
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (F.-H.G., Y.-N.G., J.-J.G., J.J.Q., Q.J.)
| | - Jun-Jun Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (F.-H.G., Y.-N.G., J.-J.G., J.J.Q., Q.J.)
| | - Lu-Jun Zhang
- Department of Cardiology, Changhai Hospital, Shanghai, China (L.-J.Z.)
| | - Jing-Jing Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (F.-H.G., Y.-N.G., J.-J.G., J.J.Q., Q.J.)
| | - Yong Ji
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Jiangsu, China (Y.J.)
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, China (A.F.C.)
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Innovation Center for Intervention of Chronic Disease and Promotion of Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China (F.-H.G., Y.-N.G., J.-J.G., J.J.Q., Q.J.)
| |
Collapse
|
15
|
Mace ML, Gravesen E, Nordholm A, Egstrand S, Morevati M, Olgaard K, Lewin E. The calcified vasculature in chronic kidney disease secretes factors that inhibit bone mineralization. JBMR Plus 2022; 6:e10610. [PMID: 35434452 PMCID: PMC9009125 DOI: 10.1002/jbm4.10610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/03/2022] [Accepted: 01/26/2022] [Indexed: 11/07/2022] Open
Affiliation(s)
| | | | - Anders Nordholm
- Department of Nephrology Rigshospitalet
- Department of Nephrology Herlev Hospital University of Copenhagen Denmark
| | - Soeren Egstrand
- Department of Nephrology Rigshospitalet
- Department of Nephrology Herlev Hospital University of Copenhagen Denmark
| | | | | | - Ewa Lewin
- Department of Nephrology Rigshospitalet
- Department of Nephrology Herlev Hospital University of Copenhagen Denmark
| |
Collapse
|
16
|
New Insights to the Crosstalk between Vascular and Bone Tissue in Chronic Kidney Disease-Mineral and Bone Disorder. Metabolites 2021; 11:metabo11120849. [PMID: 34940607 PMCID: PMC8708186 DOI: 10.3390/metabo11120849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/30/2021] [Accepted: 12/02/2021] [Indexed: 12/18/2022] Open
Abstract
Vasculature plays a key role in bone development and the maintenance of bone tissue throughout life. The two organ systems are not only linked in normal physiology, but also in pathophysiological conditions. The chronic kidney disease–mineral and bone disorder (CKD-MBD) is still the most serious complication to CKD, resulting in increased morbidity and mortality. Current treatment therapies aimed at the phosphate retention and parathyroid hormone disturbances fail to reduce the high cardiovascular mortality in CKD patients, underlining the importance of other factors in the complex syndrome. This review will focus on vascular disease and its interplay with bone disorders in CKD. It will present the very late data showing a direct effect of vascular calcification on bone metabolism, indicating a vascular-bone tissue crosstalk in CKD. The calcified vasculature not only suffers from the systemic effects of CKD but seems to be an active player in the CKD-MBD syndrome impairing bone metabolism and might be a novel target for treatment and prevention.
Collapse
|
17
|
Perens EA, Hoffman HM, Mak RH. Activin A Signaling Provides an Interorgan Link Between Kidney and Muscle in CKD-Associated Muscle Wasting. Am J Kidney Dis 2021; 79:302-304. [PMID: 34653537 DOI: 10.1053/j.ajkd.2021.09.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Elliot A Perens
- Department of Pediatrics, Rady Children's Hospital, University of California-San Diego, San Diego, California
| | - Hal M Hoffman
- Department of Pediatrics, Rady Children's Hospital, University of California-San Diego, San Diego, California
| | - Robert H Mak
- Department of Pediatrics, Rady Children's Hospital, University of California-San Diego, San Diego, California.
| |
Collapse
|
18
|
Wang C, Dong L, Wang Y, Jiang Z, Zhang J, Yang G. Bioinformatics Analysis Identified miR-584-5p and Key miRNA-mRNA Networks Involved in the Osteogenic Differentiation of Human Periodontal Ligament Stem Cells. Front Genet 2021; 12:750827. [PMID: 34646313 PMCID: PMC8503254 DOI: 10.3389/fgene.2021.750827] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 08/30/2021] [Indexed: 12/29/2022] Open
Abstract
Human periodontal ligament cells (PDLCs) play an important role in periodontal tissue stabilization and function. In the process of osteogenic differentiation of PDLSCs, the regulation of molecular signal pathways are complicated. In this study, the sequencing results of three datasets on GEO were used to comprehensively analyze the miRNA-mRNA network during the osteogenic differentiation of PDLSCs. Using the GSE99958 and GSE159507, a total of 114 common differentially expressed genes (DEGs) were identified, including 62 up-regulated genes and 52 down-regulated genes. GO enrichment analysis was performed. The up-regulated 10 hub genes and down-regulated 10 hub genes were screened out by protein-protein interaction network (PPI) analysis and STRING in Cytoscape. Similarly, differentially expressed miRNAs (DEMs) were selected by limma package from GSE159508. Then, using the miRwalk website, we further selected 11 miRNAs from 16 DEMs that may have a negative regulatory relationship with hub genes. In vitro RT-PCR verification revealed that nine DEMs and 18 hub genes showed the same trend as the RNA-seq results during the osteogenic differentiation of PDLSCs. Finally, using miR-584-5p inhibitor and mimics, it was found that miR-584-5p negatively regulates the osteogenic differentiation of PDLSCs in vitro. In summary, the present results found several potential osteogenic-related genes and identified candidate miRNA-mRNA networks for the further study of osteogenic differentiation of PDLSCs.
Collapse
Affiliation(s)
| | | | | | | | | | - Guoli Yang
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, The Affiliated Hospital of Stomatology, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
19
|
Esposito P, Verzola D, Picciotto D, Cipriani L, Viazzi F, Garibotto G. Myostatin/Activin-A Signaling in the Vessel Wall and Vascular Calcification. Cells 2021; 10:2070. [PMID: 34440838 PMCID: PMC8393536 DOI: 10.3390/cells10082070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 02/07/2023] Open
Abstract
A current hypothesis is that transforming growth factor-β signaling ligands, such as activin-A and myostatin, play a role in vascular damage in atherosclerosis and chronic kidney disease (CKD). Myostatin and activin-A bind with different affinity the activin receptors (type I or II), activating distinct intracellular signaling pathways and finally leading to modulation of gene expression. Myostatin and activin-A are expressed by different cell types and tissues, including muscle, kidney, reproductive system, immune cells, heart, and vessels, where they exert pleiotropic effects. In arterial vessels, experimental evidence indicates that myostatin may mostly promote vascular inflammation and premature aging, while activin-A is involved in the pathogenesis of vascular calcification and CKD-related mineral bone disorders. In this review, we discuss novel insights into the biology and physiology of the role played by myostatin and activin in the vascular wall, focusing on the experimental and clinical data, which suggest the involvement of these molecules in vascular remodeling and calcification processes. Moreover, we describe the strategies that have been used to modulate the activin downward signal. Understanding the role of myostatin/activin signaling in vascular disease and bone metabolism may provide novel therapeutic opportunities to improve the treatment of conditions still associated with high morbidity and mortality.
Collapse
Affiliation(s)
- Pasquale Esposito
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
- IRCCS Ospedale Policlinico San Martino, Clinica Nefrologica, Dialisi, Trapianto, 16132 Genova, Italy;
| | - Daniela Verzola
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
| | - Daniela Picciotto
- IRCCS Ospedale Policlinico San Martino, Clinica Nefrologica, Dialisi, Trapianto, 16132 Genova, Italy;
| | - Leda Cipriani
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
| | - Francesca Viazzi
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
- IRCCS Ospedale Policlinico San Martino, Clinica Nefrologica, Dialisi, Trapianto, 16132 Genova, Italy;
| | - Giacomo Garibotto
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy; (P.E.); (D.V.); (L.C.); (F.V.)
| |
Collapse
|
20
|
The Influence of Dietary Interventions on Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Nutrients 2021; 13:nu13062065. [PMID: 34208727 PMCID: PMC8235119 DOI: 10.3390/nu13062065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease is a health problem whose prevalence is increasing worldwide. The kidney plays an important role in the metabolism of minerals and bone health and therefore, even at the early stages of CKD, disturbances in bone metabolism are observed. In the course of CKD, various bone turnover or mineralization disturbances can develop including adynamic hyperparathyroid, mixed renal bone disease, osteomalacia. The increased risk of fragility fractures is present at any age in these patients. Nutritional treatment of patients with advanced stages of CKD is aiming at prevention or correction of signs, symptoms of renal failure, avoidance of protein-energy wasting (PEW), delaying or prevention of the occurrence of mineral/bone disturbances, and delaying the start of dialysis. The results of studies suggest that progressive protein restriction is beneficial with the progression of renal insufficiency; however, other aspects of dietary management of CKD patients, including changes in sodium, phosphorus, and energy intake, as well as the source of protein and lipids (animal or plant origin) should also be considered carefully. Energy intake must cover patients' energy requirement, in order to enable correct metabolic adaptation in the course of protein-restricted regimens and prevent negative nitrogen balance and protein-energy wasting.
Collapse
|
21
|
Differentiating the causes of adynamic bone in advanced chronic kidney disease informs osteoporosis treatment. Kidney Int 2021; 100:546-558. [PMID: 34102219 DOI: 10.1016/j.kint.2021.04.043] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/15/2022]
Abstract
Patients with chronic kidney disease (CKD) have an increased fracture risk because of impaired bone quality and quantity. Low bone mineral density predicts fracture risk in all CKD stages, including advanced CKD (CKD G4-5D). Pharmacological therapy improves bone mineral density and reduces fracture risk in moderate CKD. Its efficacy in advanced CKD remains to be determined, although pilot studies suggest a positive effect on bone mineral density. Currently, antiresorptive agents are the most commonly prescribed drugs for the prevention and therapy of osteoporosis. Their use in advanced CKD has been limited by the lack of large clinical trials and fear of causing kidney dysfunction and adynamic bone disease. In recent decades, adynamic bone disease has evolved as the most predominant form of renal osteodystrophy, commonly associated with poor outcomes, including premature mortality and progression of vascular calcification. Evolving evidence indicates that reduction of bone turnover by parathyroidectomy or pharmacological therapies, such as calcimimetics and antiresorptive agents, are not associated with premature mortality or accelerated vascular calcification in CKD. In contrast, chronic inflammation, oxidative stress, malnutrition, and diabetes can induce low bone turnover and associate with poor prognosis. Thus, the conditions causing suppression of bone turnover rather than the low bone turnover per se may account for the perceived association with outcomes. Anabolic treatment, in contrast, has been suggested to improve turnover and bone mass in patients with advanced CKD and low bone turnover; however, uncertainty about safety even exceeds that of antiresorptive agents. Here, we critically review the pathophysiological concept of adynamic bone disease and discuss the effect of low bone turnover on the safety and efficacy of anti-osteoporosis pharmacotherapy in advanced CKD.
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW This review highlights recent discoveries and advances that have been made in understanding the role of the TGFβ superfamily members activins, and in particular, activin A (ActA), in renal disease. RECENT FINDINGS A deleterious role for ActA in renal disease and its complications has begun to emerge. We summarize data supporting an important contribution of ActA to kidney fibrosis and inflammation of varying causes, as well as its role in the development of a particular bone mineral disorder seen in chronic kidney disease (CKD) called mineral bone disorder (MBD), including vascular calcification. Finally, we discuss ActA in the context of anemia associated with chronic kidney disease and review potential approaches to treatment based on ActA blockade. SUMMARY ActA is an important contributor to the pathogenesis of acute and chronic kidney disease of varying causes. Preclinical studies show that ActA inhibition, through various approaches, is protective in rodent models of kidney disease. The potential adverse effects of some of these approaches can be attributed to their targeting of other TGFβ family ligands. Further preclinical and clinical investigations testing the therapeutic efficacy of more selective ActA inhibition on the progression of acute and chronic kidney disease and its impact on bone-mineral disorder would more definitively establish its role in renal disease.
Collapse
|
23
|
Circadian rhythms of mineral metabolism in chronic kidney disease-mineral bone disorder. Curr Opin Nephrol Hypertens 2021; 29:367-377. [PMID: 32452917 DOI: 10.1097/mnh.0000000000000611] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW The circadian rhythms have a systemic impact on all aspects of physiology. Kidney diseases are associated with extremely high-cardiovascular mortality, related to chronic kidney disease-mineral bone disorder (CKD-MBD), involving bone, parathyroids and vascular calcification. Disruption of circadian rhythms may cause serious health problems, contributing to development of cardiovascular diseases, metabolic syndrome, cancer, organ fibrosis, osteopenia and aging. Evidence of disturbed circadian rhythms in CKD-MBD parameters and organs involved is emerging and will be discussed in this review. RECENT FINDINGS Kidney injury induces unstable behavioral circadian rhythm. Potentially, uremic toxins may affect the master-pacemaker of circadian rhythm in hypothalamus. In CKD disturbances in the circadian rhythms of CKD-MBD plasma-parameters, activin A, fibroblast growth factor 23, parathyroid hormone, phosphate have been demonstrated. A molecular circadian clock is also expressed in peripheral tissues, involved in CKD-MBD; vasculature, parathyroids and bone. Expression of the core circadian clock genes in the different tissues is disrupted in CKD-MBD. SUMMARY Disturbed circadian rhythms is a novel feature of CKD-MBD. There is a need to establish which specific input determines the phase of the local molecular clock and to characterize its regulation and deregulation in tissues involved in CKD-MBD. Finally, it is important to establish what are the implications for treatment including the potential applications for chronotherapy.
Collapse
|
24
|
Hashimoto M, Ho G, Sugama S, Takenouchi T, Waragai M, Sugino H, Inoue S, Masliah E. Possible Role of Activin in the Adiponectin Paradox-Induced Progress of Alzheimer's Disease. J Alzheimers Dis 2021; 81:451-458. [PMID: 33814453 PMCID: PMC8203218 DOI: 10.3233/jad-210206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 12/14/2022]
Abstract
Accumulating evidence suggests that the adiponectin (APN) paradox might be involved in promoting aging-associated chronic diseases such as Alzheimer's disease (AD). In human brain, APN regulation of the evolvability of amyloidogenic proteins (APs), including amyloid-β (Aβ) and tau, in developmental/reproductive stages, might be paradoxically manifest as APN stimulation of AD through antagonistic pleiotropy in aging. The unique mechanisms underlying APN activity remain unclear, a better understanding of which might provide clues for AD therapy. In this paper, we discuss the possible relevance of activin, a member of transforming growth factor β (TGFβ) superfamily of peptides, to antagonistic pleiotropy effects of APN. Notably, activin, a multiple regulator of cell proliferation and differentiation, as well as an endocrine modulator in reproduction and an organizer in early development, might promote aging-associated disorders, such as inflammation and cancer. Indeed, serum activin, but not serum TGFβ increases during aging. Also, activin/TGFβ signal through type II and type I receptors, both of which are transmembrane serine/threonine kinases, and the serine/threonine phosphorylation of APs, including Aβ42 serine 8 and αS serine 129, may confer pathological significance in neurodegenerative diseases. Moreover, activin expression is induced by APN in monocytes and hepatocytes, suggesting that activin might be situated downstream of the APN paradox. Finally, a meta-analysis of genome-wide association studies demonstrated that two SNPs relevant to the activin/TGFβ receptor signaling pathways conferred risk for major aging-associated disease. Collectively, activin might be involved in the APN paradox of AD and could be a significant therapeutic target.
Collapse
Affiliation(s)
| | - Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, USA
| | - Shuei Sugama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiromu Sugino
- Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Satoshi Inoue
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
- Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Eliezer Masliah
- Division of Neuroscience, National Institute on Aging, Bethesda, MD, USA
| |
Collapse
|
25
|
The Association between the Activin A Serum Level and Carotid Intima-Media Thickness in Chronic Kidney Disease Patients. Int J Nephrol 2020; 2020:8893653. [PMID: 33294228 PMCID: PMC7700056 DOI: 10.1155/2020/8893653] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/18/2020] [Accepted: 10/25/2020] [Indexed: 12/23/2022] Open
Abstract
Introduction Chronic kidney disease (CKD) is associated with high mortality rates, mainly as a result of cardiovascular complications. Meanwhile, recent studies have suggested a role of a homodimer protein called activin A in chronic kidney disease-mineral and bone disorder (CKD-MBD) conditions that may exist in the vascular calcification and osteolytic process. Ultrasound examination of the carotid intima-media thickness (cIMT) is a noninvasive method to assess vascular calcification. This study aimed to analyze the relationship between the activin A serum level and cIMT in patients with CKD at Mohammad Hoesin Hospital, Palembang, Indonesia. Methods We conducted a hospital-based, cross-sectional study of consecutive CKD patients at the Department of Internal Medicine, Mohammad Hoesin Hospital, from July to November 2019. The level of activin A was measured by enzyme-linked immunosorbent assay. Meanwhile, cIMT measurements were collected by B-mode ultrasound imaging. Results A total of 55 patients with CKD were included in this investigation. The median serum activin A level in these patients was 236.17 (116.33–283) pg/mL, while the median cIMT was 0.8 (0.6–1.45) mm. A relationship between the serum activin A level and cIMT (r = 0.449; p = 0.001) was observed. During multivariate analysis with linear regression, triglyceride (p = 0.049), phosphate (p = 0.005), and activin A (p = 0.020) serum levels were factors associated with cIMT. Conclusion In this study, a relationship between the activin A serum level and cIMT in patients with CKD was identified. Vascular calcification should be screened for in all CKD patients by the measurement of cIMT.
Collapse
|
26
|
Yang P, Troncone L, Augur ZM, Kim SSJ, McNeil ME, Yu PB. The role of bone morphogenetic protein signaling in vascular calcification. Bone 2020; 141:115542. [PMID: 32736145 PMCID: PMC8185454 DOI: 10.1016/j.bone.2020.115542] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/02/2020] [Accepted: 07/04/2020] [Indexed: 01/10/2023]
Abstract
Vascular calcification is associated with atherosclerosis, chronic kidney disease, and diabetes, and results from processes resembling endochondral or intramembranous ossification, or from processes that are distinct from ossification. Bone morphogenetic proteins (BMP), as well as other ligands, receptors, and regulators of the transforming growth factor beta (TGFβ) family regulate vascular and valvular calcification by modulating the phenotypic plasticity of multipotent progenitor lineages associated with the vasculature or valves. While osteogenic ligands BMP2 and BMP4 appear to be both markers and drivers of vascular calcification, particularly in atherosclerosis, BMP7 may serve to protect against calcification in chronic kidney disease. BMP signaling regulators such as matrix Gla protein and BMP-binding endothelial regulator protein (BMPER) play protective roles in vascular calcification. The effects of BMP signaling molecules in vascular calcification are context-dependent, tissue-dependent, and cell-type specific. Here we review the current knowledge on mechanisms by which BMP signaling regulates vascular calcification and the potential therapeutic implications.
Collapse
Affiliation(s)
- Peiran Yang
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Luca Troncone
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Zachary M Augur
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stephanie S J Kim
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Megan E McNeil
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Hauge SC, Frost M, Hansen D. Understanding Bone Disease in Patients with Diabetic Kidney Disease: a Narrative Review. Curr Osteoporos Rep 2020; 18:727-736. [PMID: 33048275 DOI: 10.1007/s11914-020-00630-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Both diabetes and kidney disease associate with the development of bone disease and an increased risk of fragility fractures. The etiologies of bone disease in patients with diabetic kidney disease (DKD) are multiple and complex. This review explores the association between DKD and bone disease and discusses how the presence of both diabetes and kidney disease may impair bone quality and increase fracture risk. Diagnostic tools as well as future research areas are also discussed. RECENT FINDINGS Patients with DKD have an increased risk of fragility fracture, most pronounced in patients with type 1 diabetes, and in DKD a high prevalence of adynamic bone disease is found. Recent studies have demonstrated disturbances in the interplay between bone regulating factors in DKD, such as relative hypoparathyroidism and alterations of bone-derived hormones including fibroblast growth factor-23 (FGF-23), sclerostin and klotho, which lead to bone disease. This review examines the current knowledge on bone disease in patients with DKD, clinical considerations for patient care, as well as subjects for future research.
Collapse
Affiliation(s)
- Sabina Chaudhary Hauge
- Department of Nephrology, Herlev Hospital, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark.
| | - Morten Frost
- Department of Endocrinology, Odense University Hospital, Kløvervænget 6, 5000, Odense C, Denmark
| | - Ditte Hansen
- Department of Nephrology, Herlev Hospital, Borgmester Ib Juuls Vej 1, 2730, Herlev, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen N, Denmark
| |
Collapse
|
28
|
Mace ML, Olgaard K, Lewin E. New Aspects of the Kidney in the Regulation of Fibroblast Growth Factor 23 (FGF23) and Mineral Homeostasis. Int J Mol Sci 2020; 21:E8810. [PMID: 33233840 PMCID: PMC7699902 DOI: 10.3390/ijms21228810] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
The bone-derived hormone fibroblast growth factor 23 (FGF23) acts in concert with parathyroid hormone (PTH) and the active vitamin D metabolite calcitriol in the regulation of calcium (Ca) and phosphate (P) homeostasis. More factors are being identified to regulate FGF23 levels and the endocrine loops between the three hormones. The present review summarizes the complex regulation of FGF23 and the disturbed FGF23/Klotho system in chronic kidney disease (CKD). In addition to the reduced ability of the injured kidney to regulate plasma levels of FGF23, several CKD-related factors have been shown to stimulate FGF23 production. The high circulating FGF23 levels have detrimental effects on erythropoiesis, the cardio-vascular system and the immune system, all contributing to the disturbed system biology in CKD. Moreover, new factors secreted by the injured kidney and the uremic calcified vasculature play a role in the mineral and bone disorder in CKD and create a vicious pathological crosstalk.
Collapse
Affiliation(s)
- Maria L. Mace
- Department of Nephrology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark; (K.O.); (E.L.)
| | - Klaus Olgaard
- Department of Nephrology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark; (K.O.); (E.L.)
| | - Ewa Lewin
- Department of Nephrology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark; (K.O.); (E.L.)
- Department of Nephrology, Herlev Hospital, University of Copenhagen, 2730 Herlev, Denmark
| |
Collapse
|
29
|
Guan YN, Li Y, Roosan M, Jing Q. Single-cell transcriptomics of murine mural cells reveals cellular heterogeneity. SCIENCE CHINA-LIFE SCIENCES 2020; 64:1077-1086. [PMID: 33165809 PMCID: PMC7649565 DOI: 10.1007/s11427-020-1823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/23/2020] [Indexed: 10/28/2022]
Abstract
Mural cells (MCs) wrap around the endothelium, and participate in the development and homeostasis of vasculature. MCs have been reported as heterogeneous population morphologically and functionally. However, the transcriptional heterogeneity of MCs was rarely studied. In this study, we illustrated the transcriptional heterogeneity of MCs with different perspectives by using publicly available single-cell dataset GSE109774. Specifically, MCs are transcriptionally different from other cell types, and ligand-receptor interactions of different cells with MCs vary. Re-clustering of MCs identified five distinct subclusters. The heterogeneity of MCs in tissues was reflected by MC coverage, various distribution of MC subclusters, and ligand-receptor interactions of MCs and parenchymal cells. The transcriptomic diversity of MCs revealed in this article will help facilitate further research into MCs.
Collapse
Affiliation(s)
- Ya-Na Guan
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), Shanghai Jiao Tong University School of Medicine (SJTUSM) & CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai, 200031, China
| | - Yue Li
- Chapman University, Irvine, CA, 92618, USA
| | | | - Qing Jing
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences (CAS), Shanghai Jiao Tong University School of Medicine (SJTUSM) & CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai, 200031, China.
| |
Collapse
|
30
|
Esposito P, Verzola D, La Porta E, Milanesi S, Grignano MA, Avella A, Gregorini M, Abelli M, Ticozzelli E, Rampino T, Garibotto G. Myostatin in the Arterial Wall of Patients with End-Stage Renal Disease. J Atheroscler Thromb 2020; 27:1039-1052. [PMID: 32173683 PMCID: PMC7585912 DOI: 10.5551/jat.51144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 12/10/2019] [Indexed: 02/05/2023] Open
Abstract
AIM Myostatin (Mstn) has been described as a trigger for the progression of atherosclerosis. In this study, we evaluated the role of Mstn in arterial remodeling in patients with end-stage renal disease (ESRD). METHODS Vascular specimens were collected from 16 ESRD patients (56.4±7.9 years) undergoing renal transplant (recipients) and 15 deceased kidney non-uremic donors (55.4±12.1 years). We studied gene and protein expression of Mstn, ubiquitin ligases, Atrogin-1, and muscle ring finger protein-1 (MuRF-1), inflammatory marker CCL2, cytoskeleton components, and Klotho by reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. Moreover, we assessed vascular calcification and collagen deposition. Finally, we studied the effects of recombinant Mstn on rat vascular smooth muscle cells (VSMCs, A7r5) and evaluated the effects of uremic serum (US) on primary human VSMCs. RESULTS Myostatin mRNA was upregulated in the arterial vascular wall of recipients compared with donors (~15- folds, p<0.05). This response was accompanied by the upregulation of gene expression of Atrogin-1 and MuRF-1 (+2.5- and +10-fold) and CCL2 (+3-fold). Conversely, we found downregulation of protein expression of Smoothelin, α-smooth muscle actin (α-SMA), vimentin, and Klotho (-85%, -50%, -70%, and -80%, respectively; p<0.05) and gene expression of vimentin and Klotho. Exposition of A7r5 to Mstn induced a time-dependent SMAD 2/SMAD 3 phosphorylation and expression of collagen-1 and transforming growth factor β (TGFβ) mRNA, while US induced overexpression of Mstn and Atrogin-1 and downregulation of Smoothelin and Klotho. CONCLUSIONS Our data suggest that uremia might induce vascular Mstn gene expression together with a complex pathway of molecular and structural changes in the vascular wall. Myostatin, in turn, can translate the metabolic alterations of uremia into profibrotic and stiffness inducing signals.
Collapse
Affiliation(s)
- Pasquale Esposito
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, and University of Pavia, Pavia, Italy
- Department of Internal Medicine, Nephrology, Dialysis and Transplantation Clinics, Genoa University and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniela Verzola
- Department of Internal Medicine, Nephrology, Dialysis and Transplantation Clinics, Genoa University and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Edoardo La Porta
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, and University of Pavia, Pavia, Italy
- Department of Internal Medicine, Nephrology, Dialysis and Transplantation Clinics, Genoa University and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Samantha Milanesi
- Department of Internal Medicine, Nephrology, Dialysis and Transplantation Clinics, Genoa University and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Maria Antonietta Grignano
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, and University of Pavia, Pavia, Italy
| | - Alessandro Avella
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, and University of Pavia, Pavia, Italy
| | - Marilena Gregorini
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, and University of Pavia, Pavia, Italy
| | - Massimo Abelli
- Service of Surgery, University of Pavia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elena Ticozzelli
- Service of Surgery, University of Pavia, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Teresa Rampino
- Department of Nephrology, Dialysis and Transplantation, Fondazione IRCCS Policlinico San Matteo, and University of Pavia, Pavia, Italy
| | - Giacomo Garibotto
- Department of Internal Medicine, Nephrology, Dialysis and Transplantation Clinics, Genoa University and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
31
|
Cianciolo G, La Manna G, Capelli I, Gasperoni L, Galassi A, Ciceri P, Cozzolino M. The role of activin: the other side of chronic kidney disease-mineral bone disorder? Nephrol Dial Transplant 2020; 36:966-974. [PMID: 32940690 DOI: 10.1093/ndt/gfaa203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Indexed: 12/21/2022] Open
Abstract
Chronic kidney disease-mineral bone disorder (CKD-MBD) plays a pivotal role in the excess of cardiovascular morbidity and mortality associated with CKD. There is now a growing awareness that pathways involved in CKD-MBD, like canonical Wnt signalling, are activated from the earliest stages of CKD, playing a role in the development of adynamic bone disease with unknown consequences on vasculature. These changes occur before the classic changes in mineral metabolism: secondary hyperparathyroidism, calcitriol deficiency and hyperphosphataemia. Furthermore, vascular calcification is frequently associated and evolves with decreased bone mineral density and deranged bone turnover, while bone and arterial mineralization share common pathways. Therefore, results of clinical trials focused on mineral bone disorder, aimed at preserving bone and cardiovascular health, are considered unsatisfactory. In order to identify more effective therapeutic strategies, it is necessary to clarify the pathways modulating the cross-talk between bone and vasculature and identify new mediators involved in the pathogenesis of CKD-MBD. Much attention has been paid recently to the role of the transforming growth factor-beta superfamily members in renal disease, and in particular of activin A (ActA). Preclinical studies demonstrate an upgrade of ActA signalling in kidney, skeleton, vasculature and heart during CKD. This supports the idea that an endocrine factor produced in the kidney during renal disease, in addition to promoting the progression of kidney damage, deranges other organs' homoeostasis and participates in CKD-MBD. In this review, we analyse the contribution of ActA to kidney fibrosis and inflammation as well as its role in the development of CKD-MBD.
Collapse
Affiliation(s)
- Giuseppe Cianciolo
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Gaetano La Manna
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Irene Capelli
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Lorenzo Gasperoni
- Department of Experimental Diagnostic and Specialty Medicine (DIMES), Nephrology, Dialysis and Renal Transplant Unit, S. Orsola Hospital, University of Bologna, Bologna, Italy
| | - Andrea Galassi
- Department of Health Sciences, Renal Division, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Paola Ciceri
- Department of Nephrology, Dialysis and Renal Transplant, Renal Research Laboratory, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Mario Cozzolino
- Department of Health Sciences, Renal Division, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| |
Collapse
|
32
|
Razzaque MS, Atfi A. Regulatory Role of the Transcription Factor Twist1 in Cancer-Associated Muscle Cachexia. Front Physiol 2020; 11:662. [PMID: 32655411 PMCID: PMC7324683 DOI: 10.3389/fphys.2020.00662] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/25/2020] [Indexed: 12/19/2022] Open
Abstract
Muscle cachexia is a catabolic response, usually takes place in various fatal diseases, such as sepsis, burn injury, and chronic kidney disease. Muscle cachexia is also a common co-morbidity seen in the vast majority of advanced cancer patients, often associated with low quality of life and death due to general organ dysfunction. The triggering events and underlying molecular mechanisms of muscle wasting are not yet clearly defined. Our recent study has shown that the ectopic expression of Twist1 in muscle progenitor cells is sufficient to drive muscle structural protein breakdown and attendant muscle atrophy, reminiscent of muscle cachexia. Intriguingly, muscle Twist1 expression is highly induced in cachectic muscles from several mouse models of pancreatic ductal adenocarcinoma (PDAC), raising the interesting possibility that Twist1 may mediate PDAC-driven muscle cachexia. Along these lines, both genetic and pharmacological inactivation of Twist1 function was highly significant at protecting against cancer cachexia, which translated into a significant survival benefit in the experimental PDAC animals. From a translational perspective, elevated expression of Twist1 is also detected in cancer patients with severe muscle wasting, implicating a role of Twist1 in cancer cachexia, and further providing a possible target for therapeutic attenuation of cachexia to improve cancer patient survival. In this article, we will briefly summarize how Twist1 acts as a master regulator of tumor-induced cachexia, and discuss the relevance of our findings to muscle wasting diseases in general. The mechanism of decreased muscle mass in various catabolic conditions is thought to rely on similar pathways, and, therefore, Twist1-induced cancer cachexia may benefit diverse groups of patients with clinical complications associated with loss of muscle mass and functions, beyond the expected benefits for cancer patients.
Collapse
Affiliation(s)
- Mohammed S Razzaque
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA, United States
| | - Azeddine Atfi
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The molecular mechanisms of the bone disease associated with chronic kidney disease (CKD), called renal osteodystrophy (ROD), are poorly understood. New transcriptomics technologies may provide clinically relevant insights into the pathogenesis of ROD. This review summarizes current progress and limitations in the study and treatment of ROD, and in transcriptomics analyses of skeletal tissues. RECENT FINDINGS ROD is characterized by poor bone quality and strength leading to increased risk of fracture. Recent studies indicate permanent alterations in bone cell populations during ROD. Single-cell transcriptomics analyses, successful at identifying specialized cell subpopulations in bone, have not yet been performed in ROD. ROD is a widespread poorly understood bone disease with limited treatment options. Transcriptomics analyses of bone are needed to identify the bone cell subtypes and their role in the pathogenesis of ROD, and to develop adequate diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Aline Martin
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health and Feinberg Cardiovascular and Renal Research Institute, Northwestern University, 320 East Superior Street, Chicago, IL, 60611, USA.
| | - Valentin David
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health and Feinberg Cardiovascular and Renal Research Institute, Northwestern University, 320 East Superior Street, Chicago, IL, 60611, USA.
| |
Collapse
|
34
|
Verzola D, Milanesi S, Viazzi F, Ansaldo F, Saio M, Garibaldi S, Carta A, Costigliolo F, Salvidio G, Barisione C, Esposito P, Garibotto G, Picciotto D. Enhanced myostatin expression and signalling promote tubulointerstitial inflammation in diabetic nephropathy. Sci Rep 2020; 10:6343. [PMID: 32286342 PMCID: PMC7156449 DOI: 10.1038/s41598-020-62875-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 03/12/2020] [Indexed: 02/07/2023] Open
Abstract
Myostatin (MSTN), a family member of the transforming growth factor (TGF)-β super family, has been detected in the tubuli of pig kidney, but its role in the human kidney is not known. In this study we observed upregulation of MSTN mRNA (~8 to 10-fold increase) both in the glomeruli and tubulointerstitium in diabetic nephropathy (DN). In DN, immunoreactive MSTN was mainly localized in the tubuli and interstitium (∼4-8 fold increase), where it colocalized in CD45+ cells. MSTN was also upregulated in the glomeruli and the arterial vessels. Tubulointerstitial MSTN expression was directly related to interstitial fibrosis (r = 0.54, p < 0.01). In HK-2 tubular epithelial cells, both high (30 mmol) glucose and glycated albumin upregulated MSTN mRNA and its protein (p < 0.05-0.01). MSTN-treated HK-2 cells underwent decreased proliferation, together with NF-kB activation and CCL-2 and SMAD 2,3 overexpression. In addition, MSTN induced intracellular ROS release and upregulated NADPH oxidase, effects which were mediated by ERK activation. In conclusion, our data show that MSTN is expressed in the human kidney and overexpressed in DN, mainly in the tubulointerstitial compartment. Our results also show that MSTN is a strong inducer of proximal tubule activation and suggest that MSTN overexpression contributes to kidney interstitial fibrosis in DN.
Collapse
Affiliation(s)
- Daniela Verzola
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Samantha Milanesi
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Viazzi
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Ansaldo
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Michela Saio
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Silvano Garibaldi
- Division of Cardiology, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Annalisa Carta
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Francesca Costigliolo
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Gennaro Salvidio
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Chiara Barisione
- Division of Cardiology, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Pasquale Esposito
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Giacomo Garibotto
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Daniela Picciotto
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Department of Internal Medicine and IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
35
|
Tsai YL, Chou RH, Lu YW, Liu CT, Huang PH, Lin SJ. Serum Activin A Levels and Renal Outcomes After Coronary Angiography. Sci Rep 2020; 10:3365. [PMID: 32099067 PMCID: PMC7042345 DOI: 10.1038/s41598-020-60359-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/04/2020] [Indexed: 01/09/2023] Open
Abstract
Prevention for contrast-induced nephropathy (CIN) is limited by the lack of a single predictor. As activin A is upregulated in heart failure and chronic kidney disease, we aimed to clarify the association between activin A levels and renal outcomes after coronary angiography (CAG). This prospective observational study included 267 patients who received CAG between 2009 and 2015. CIN was defined as elevation of serum creatinine to >0.5 mg/dL or to >25% above baseline within 48 hours after CAG. During follow-up, laboratory parameters were measured every 3–6 months. Renal decline was defined as>2-fold increase in serum creatinine or initiation of dialysis. The patients were stratified into tertiles according to serum activin A levels at baseline. High activin A tertile was significantly associated more CIN and renal function decline compared to low activin A tertile (all p < 0.001). After adjusting potential confounding factors, high serum activin A tertiles was associated to CIN (Odds ratio 4.49, 95% CI 1.07–18.86, p = 0.040) and renal function decline (Hazard ratio 4.49, 95% CI 1.27–11.41, p = 0.017) after CAG.
Collapse
Affiliation(s)
- Yi-Lin Tsai
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Ruey-Hsing Chou
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ya-Wen Lu
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Chung-Te Liu
- Division of Nephrology, Department of Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan. .,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan. .,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan. .,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Shing-Jong Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Healthcare and Management Center, Taipei Veterans General Hospital, Taipei, Taiwan.,Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
36
|
Garibotto G, Esposito P, Picciotto D, Verzola D. Activin/myostatin receptor signaling and vascular calcifications in chronic kidney disease: A "liaison dangereuse"? Kidney Res Clin Pract 2019; 38:407-410. [PMID: 31640302 PMCID: PMC6913597 DOI: 10.23876/j.krcp.19.085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 02/05/2023] Open
Affiliation(s)
- Giacomo Garibotto
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Genova,
Italy
- Department of Internal Medicine, IRCCS Ospedale Policlinico San Martino, Genova,
Italy
| | - Pasquale Esposito
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Genova,
Italy
- Department of Internal Medicine, IRCCS Ospedale Policlinico San Martino, Genova,
Italy
| | - Daniela Picciotto
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Genova,
Italy
- Department of Internal Medicine, IRCCS Ospedale Policlinico San Martino, Genova,
Italy
| | - Daniela Verzola
- Division of Nephrology, Dialysis and Transplantation, University of Genova, Genova,
Italy
- Department of Internal Medicine, IRCCS Ospedale Policlinico San Martino, Genova,
Italy
| |
Collapse
|
37
|
Waziri B, Duarte R, Naicker S. Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD): Current Perspectives. Int J Nephrol Renovasc Dis 2019; 12:263-276. [PMID: 31920363 PMCID: PMC6935280 DOI: 10.2147/ijnrd.s191156] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
Despite the availability of global and regional guidelines to curtail the adverse clinical outcomes associated with chronic kidney disease–mineral and bone disorder (CKD-MBD), most CKD patients are still affected by the consequences of abnormalities of CKD-MBD. This important clinical complication of CKD continues to be studied, in order to improve the understanding and management of CKD-MBD. Some notable discoveries include the role of fibroblast growth factor 23 (FGF23) in the pathogenesis of CKD-MBD, leading to a shift from the previous well-established classic trade-off hypothesis to the updated trade-off hypothesis. More recently, there has been a shift from the treatment of CKD-MBD based on a single level of biomarkers to serial measurements of calcium, phosphate and parathyroid hormone (PTH). Furthermore, some clinical trials have emerged after the 2009 Kidney Disease-Improving Global Outcomes (KDIGO) Guidelines, leading to the 2017 KDIGO updated recommendations. Hence, this review gives an overview of the rapidly evolving trends in CKD-MBD, linking the past and current concepts of CKD-MBD.
Collapse
Affiliation(s)
- Bala Waziri
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Department of Medicine, Ibrahim Badamasi Babangida Specialist Hospital, Minna, Nigeria
| | - Raquel Duarte
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Saraladevi Naicker
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
38
|
Delitsikou V, Jarad G, Rajaram RD, Ino F, Rutkowski JM, Chen CD, Santos CXC, Scherer PE, Abraham CR, Shah AM, Feraille E, Miner JH, de Seigneux S. Klotho regulation by albuminuria is dependent on ATF3 and endoplasmic reticulum stress. FASEB J 2019; 34:2087-2104. [PMID: 31907991 DOI: 10.1096/fj.201900893r] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 10/31/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022]
Abstract
Proteinuria is associated with renal function decline and cardiovascular mortality. This association may be attributed in part to alterations of Klotho expression induced by albuminuria, yet the underlying mechanisms are unclear. The presence of albumin decreased Klotho expression in the POD-ATTAC mouse model of proteinuric kidney disease as well as in kidney epithelial cell lines. This downregulation was related to both decreased Klotho transcription and diminished protein half-life, whereas cleavage by ADAM proteases was not modified. The regulation was albumin specific since it was neither observed in the analbuminemic Col4α3-/- Alport mice nor induced by exposure of kidney epithelial cells to purified immunoglobulins. Albumin induced features of ER stress in renal tubular cells with ATF3/ATF4 activation. ATF3 and ATF4 induction downregulated Klotho through altered transcription mediated by their binding on the Klotho promoter. Inhibiting ER stress with 4-PBA decreased the effect of albumin on Klotho protein levels without altering mRNA levels, thus mainly abrogating the increased protein degradation. Taken together, albuminuria decreases Klotho expression through increased protein degradation and decreased transcription mediated by ER stress induction. This implies that modulating ER stress may improve proteinuria-induced alterations of Klotho expression, and hence renal and extrarenal complications associated with Klotho loss.
Collapse
Affiliation(s)
- Vasiliki Delitsikou
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland.,Laboratory of Nephrology, Department of Internal Medicine Specialties, HUG, Geneva, Switzerland
| | - George Jarad
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Renuga Devi Rajaram
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland.,Laboratory of Nephrology, Department of Internal Medicine Specialties, HUG, Geneva, Switzerland
| | - Frédérique Ino
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland.,Laboratory of Nephrology, Department of Internal Medicine Specialties, HUG, Geneva, Switzerland
| | - Joseph M Rutkowski
- Touchstone Diabetes Centre, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Medical Physiology, Texas A&M College of Medicine, College Station, Texas
| | - Ci-Di Chen
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Celio X C Santos
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Philipp E Scherer
- Touchstone Diabetes Centre, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Carmela R Abraham
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Eric Feraille
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Sophie de Seigneux
- Department of Cell Physiology and Metabolism, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland.,Laboratory of Nephrology, Department of Internal Medicine Specialties, HUG, Geneva, Switzerland
| |
Collapse
|
39
|
Kakitapalli Y, Ampolu J, Madasu SD, Sai Kumar MLS. Detailed Review of Chronic Kidney Disease. KIDNEY DISEASES 2019; 6:85-91. [PMID: 32309290 DOI: 10.1159/000504622] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 10/30/2019] [Indexed: 01/09/2023]
Abstract
Background Nephropathy problems in the Udhanam region of Andhra Pradesh in India have motivated researchers to investigate the various factors related to chronic kidney disease (CKD). Initially, studies came across the markers of identification of CKD, i.e., glomerular filtration rate (GFR) and albumin creatinine rate, as global markers of identification. Cystatin C (Cys C) and its reciprocal (1/Cys C) are used to calculate GFR. This is a very easy method compared to the more accurate methods such as radiolabelled tracer clearances, which are invasive, may involve radiation, and require several hours to perform, e.g., 99-diethylene triamine penta-acetic acid (<sup>99m</sup>Tc-DTPA) and <sup>51</sup>Cr-EDTA. This article provides the causes (or risk factors), symptoms, and complications of CKD in a clear manner such that even common people can easily understand. Once a patient is detected and proved to be affected by CKD then the patient as well as the caretakers, including doctors, must follow some constraints. Thereby it is possible to prevent CKD progression in the patient. Modern methods are needed to prevent the pathogens which are responsible for CKD. Summary With the help of various engineering techniques one can easily design controllers to assess as well as to prevent CKD permanently. The easiest procedure for identifying CKD is to screen people. Current recommendations suggest screening of individuals with diabetes, hypertension, cardiovascular disease, and family history of kidney diseases in the course of routine health check-ups. Much work has been done in medical sciences in the area of CKD, but there is still scope for further research. From the recent studies, advanced tools such as data mining, etc., are considered to be the current trend in the area of CKD. Key Message From this article, the authors propose that patients who are already affected by urinary tract infection, acute kidney injury, and a family history of CKD should be examined via some basic tests for the presence of CKD.
Collapse
Affiliation(s)
- Yesubabu Kakitapalli
- Department of Electrical and Electronics Engineering, Aditya Institute of Technology and Management, Tekkali, India
| | - Janakiram Ampolu
- Department of Electrical and Electronics Engineering, Aditya Institute of Technology and Management, Tekkali, India
| | - Satya Dinesh Madasu
- Department of Electrical and Electronics Engineering, Bapatla Engineering College, Bapatla, India
| | - M L S Sai Kumar
- Department of Electrical and Electronics Engineering, National Institute of Technology Jamshedpur, Jamshedpur, India
| |
Collapse
|
40
|
Bian X, Griffin TP, Zhu X, Islam MN, Conley SM, Eirin A, Tang H, O’Shea PM, Palmer AK, McCoy RG, Herrmann SM, Mehta RA, Woollard JR, Rule AD, Kirkland JL, Tchkonia T, Textor SC, Griffin MD, Lerman LO, Hickson LJ. Senescence marker activin A is increased in human diabetic kidney disease: association with kidney function and potential implications for therapy. BMJ Open Diabetes Res Care 2019; 7:e000720. [PMID: 31908790 PMCID: PMC6936543 DOI: 10.1136/bmjdrc-2019-000720] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/28/2019] [Accepted: 10/23/2019] [Indexed: 12/31/2022] Open
Abstract
Objective Activin A, an inflammatory mediator implicated in cellular senescence-induced adipose tissue dysfunction and profibrotic kidney injury, may become a new target for the treatment of diabetic kidney disease (DKD) and chronic kidney diseases. We tested the hypothesis that human DKD-related injury leads to upregulation of activin A in blood and urine and in a human kidney cell model. We further hypothesized that circulating activin A parallels kidney injury markers in DKD. Research design and methods In two adult diabetes cohorts and controls (Minnesota, USA; Galway, Ireland), the relationships between plasma (or urine) activin A, estimated glomerular filtration rate (eGFR) and DKD injury biomarkers were tested with logistic regression and correlation coefficients. Activin A, inflammatory, epithelial-mesenchymal-transition (EMT) and senescence markers were assayed in human kidney (HK-2) cells incubated in high glucose plus transforming growth factor-β1 or albumin. Results Plasma activin A levels were elevated in diabetes (n=206) compared with controls (n=76; 418.1 vs 259.3 pg/mL; p<0.001) and correlated inversely with eGFR (rs=-0.61; p<0.001; diabetes). After eGFR adjustment, only albuminuria (OR 1.56, 95% CI 1.16 to 2.09) and tumor necrosis factor receptor-1 (OR 6.40, 95% CI 1.08 to 38.00) associated with the highest activin tertile. Albuminuria also related to urinary activin (rs=0.65; p<0.001). Following in vitro HK-2 injury, activin, inflammatory, EMT genes and supernatant activin levels were increased. Conclusions Circulating activin A is increased in human DKD and correlates with reduced kidney function and kidney injury markers. DKD-injured human renal tubule cells develop a profibrotic and inflammatory phenotype with activin A upregulation. These findings underscore the role of inflammation and provide a basis for further exploration of activin A as a diagnostic marker and therapeutic target in DKD.
Collapse
Affiliation(s)
- Xiaohui Bian
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tomás P Griffin
- Centre for Endocrinology, Diabetes and Metabolism, Saolta University Health Care Group, Galway University Hospitals, Galway, Ireland
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Xiangyang Zhu
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Md Nahidul Islam
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Clinical Biochemistry, Saolta University Health Care Group, Galway University Hospitals, Galway, Ireland
| | - Sabena M Conley
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Alfonso Eirin
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Paula M O’Shea
- Department of Clinical Biochemistry, Saolta University Health Care Group, Galway University Hospitals, Galway, Ireland
| | - Allyson K Palmer
- Division of Community Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Rozalina G McCoy
- Division of Community Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ramila A Mehta
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - John R Woollard
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrew D Rule
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - James L Kirkland
- Division of Community Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM SFI Research Centre, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Department of Nephrology, Saolta University Health Care Group, Galway University Hospitals, Galway, Ireland
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - LaTonya J Hickson
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
41
|
The caveolin-1 regulated protein follistatin protects against diabetic kidney disease. Kidney Int 2019; 96:1134-1149. [DOI: 10.1016/j.kint.2019.05.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/27/2019] [Accepted: 05/30/2019] [Indexed: 01/30/2023]
|
42
|
Coyne DW, Singh HN, Smith WT, Giuseppi AC, Connarn JN, Sherman ML, Dellanna F, Malluche HH, Hruska KA. Sotatercept Safety and Effects on Hemoglobin, Bone, and Vascular Calcification. Kidney Int Rep 2019; 4:1585-1597. [PMID: 31891000 PMCID: PMC6933454 DOI: 10.1016/j.ekir.2019.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/29/2019] [Accepted: 08/03/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Patients with end-stage kidney disease (ESKD) exhibit anemia, chronic kidney disease‒mineral bone disorder (CKD-MBD), and cardiovascular disease. The REN-001 and REN-002 phase II, multicenter, randomized studies examined safety, tolerability, and effects of sotatercept, an ActRIIA-IgG1 fusion protein trap, on hemoglobin concentration; REN-001 also explored effects on bone mineral density (BMD) and abdominal aortic vascular calcification. METHODS Forty-three patients were treated in REN-001 (dose range: sotatercept 0.3‒0.7 mg/kg or placebo subcutaneously [s.c.] for 200 days) and 50 in REN-002 (dose range: 0.1‒0.4 mg/kg i.v. and 0.13‒0.5 mg/kg s.c. for 99 days). RESULTS In REN-001, frequency of achieving target hemoglobin response (>10 g/dl [6.21 mmol/l]) with sotatercept was dose-related and greater than placebo (0.3 mg/kg: 33.3%; 0.5 mg/kg: 62.5%; 0.7 mg/kg: 77.8%; 0.7 mg/kg [doses 1 and 2]/0.4 mg/kg [doses 3‒15]: 33.3%; placebo: 27.3%). REN-002 hemoglobin findings were similar (i.v.: 16.7%-57.1%; s.c.: 11.1%‒42.9%). Dose-related achievement of ≥2% increase in femoral neck cortical BMD was seen among only REN-001 patients receiving sotatercept (0.3‒0.7 mg/kg: 20.0%‒57.1%; placebo: 0.0%). Abdominal aortic vascular calcification was slowed in a dose-related manner, with a ≤15% increase in Agatston score achieved by more REN-001 sotatercept versus placebo patients (60%‒100% vs. 16.7%). The most common adverse events during treatment were hypertension, muscle spasm, headache, arteriovenous fistula site complication, and influenza observed in both treatment and placebo groups. CONCLUSION In patients with ESKD, sotatercept exhibited a favorable safety profile and was associated with trends in dose-related slowing of vascular calcification. Less-consistent trends in improved hemoglobin concentration and BMD were observed.
Collapse
Affiliation(s)
- Daniel W. Coyne
- Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | | | | - Hartmut H. Malluche
- Division of Nephrology, Bone and Mineral Metabolism, University of Kentucky, Lexington, Kentucky, USA
| | - Keith A. Hruska
- Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Cell Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
43
|
Nordholm A, Egstrand S, Gravesen E, Mace ML, Morevati M, Olgaard K, Lewin E. Circadian rhythm of activin A and related parameters of mineral metabolism in normal and uremic rats. Pflugers Arch 2019; 471:1079-1094. [PMID: 31236663 PMCID: PMC6614158 DOI: 10.1007/s00424-019-02291-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
Activin A is a new fascinating player in chronic kidney disease-mineral and bone disorder (CKD-MBD), which is implicated in progressive renal disease, vascular calcification, and osteodystrophy. Plasma activin A rises early in the progression of renal disease. Disruption of circadian rhythms is related to increased risk of several diseases and circadian rhythms are observed in mineral homeostasis, bone parameters, and plasma levels of phosphate and PTH. Therefore, we examined the circadian rhythm of activin A and CKD-MBD-related parameters (phosphate, PTH, FGF23, and klotho) in healthy controls and CKD rats (5/6 nephrectomy) on high-, standard- and low-dietary phosphate contents as well as during fasting conditions. Plasma activin A exhibited circadian rhythmicity in healthy control rats with fourfold higher values at acrophase compared with nadir. The rhythm was obliterated in CKD. Activin A was higher in CKD rats compared with controls when measured at daytime but not significantly when measured at evening/nighttime, stressing the importance of time-specific reference intervals when interpreting plasma values. Plasma phosphate, PTH, and FGF23 all showed circadian rhythms in control rats, which were abolished or disrupted in CKD. Plasma klotho did not show circadian rhythm. Thus, the present investigation shows, for the first time, circadian rhythm of plasma activin A. The rhythmicity is severely disturbed by CKD and is associated with disturbed rhythms of phosphate and phosphate-regulating hormones PTH and FGF23, indicating that disturbed circadian rhythmicity is an important feature of CKD-MBD.
Collapse
Affiliation(s)
- Anders Nordholm
- Nephrological Department, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark.,Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Søren Egstrand
- Nephrological Department, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark.,Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Eva Gravesen
- Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Maria L Mace
- Nephrological Department, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark.,Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Marya Morevati
- Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Klaus Olgaard
- Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Ewa Lewin
- Nephrological Department, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark. .,Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark.
| |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Patients with chronic kidney disease have a high risk of fractures and no established treatments that have been shown to prevent the bone disease. The physiology of renal osteodystrophy is complex and recently more factors have been found that complicate the mineral metabolism. The recognition that vascular calcifications are related to bone disease has made treatment even more challenging. RECENT FINDINGS The most exciting new findings relate to the signaling pathways that are seen in kidney disease and how they cause abnormalities in bone physiology. In particular, wnt and activin signaling pathways are seen early in the course of renal disease. The bones react by increasing FGF-23, which targets both renal phosphate secretion and a variety of other systemic effects. Secreted klotho is another newly described hormone with effects on several systems.Clinical studies have focused on treatments for hyperparathyroidism and phosphate, and frustrating limitations of the treatments used for ordinary osteoporosis. SUMMARY Treatment of bone disease in patients with chronic kidney disease is challenging, and understanding the physiological pathways could lead to novel therapies.
Collapse
|
45
|
Serum bone markers in ROD patients across the spectrum of decreases in GFR: Activin A increases before all other markers
. Clin Nephrol 2019; 91:222-230. [PMID: 30862350 PMCID: PMC6595397 DOI: 10.5414/cn109650] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2019] [Indexed: 12/26/2022] Open
Abstract
Introduction: Renal osteodystrophy (ROD) develops early in chronic kidney disease (CKD) and progresses with loss of kidney function. While intact parathyroid hormone (PTH), 1,25-dihydroxyvitamin D3 (1,25D), and fibroblast growth factor-23 (FGF-23) levels are usually considered the primary abnormalities in ROD development, the role of serum activin A elevations in CKD and its relationships to ROD have not been explored. The aims of this study were to evaluate serum activin A at different CKD stages, and to establish the relationships between activin A, bone biomarkers, and bone histomorphometric parameters. Materials and methods: 104 patients with CKD stages 2 – 5D underwent bone biopsies. We measured in the serum activin A, BSAP, DKK1, FGF-23, α-Klotho, intact PTH, sclerostin, TRAP-5b, and 1,25D. Biochemical results were compared across CKD stages and with 19 age-matched controls with normal kidney function. Results: Median activin A levels were increased in all stages of CKD compared to controls from 544 pg/mL in CKD 2 (431 – 628) to 1,135 pg/mL in CKD 5D (816 – 1,456), compared to 369 pg/mL in controls (316 – 453, p < 0.01). The increase of activin A in CKD 2 (p = 0.016) occurred before changes in the other measured biomarkers. Activin A correlated with intact PTH and FGF-23 (r = 0.65 and 0.61; p < 0.01) and with histomorphometric parameters of bone turnover (BFR/BS, Acf, ObS/BS and OcS/BS; r = 0.47 – 0.52; p < 0.01). These correlations were comparable to those found with intact PTH and FGF-23. Conclusion: Serum activin A levels increase starting at CKD 2 before elevations in intact PTH and FGF-23. Activin A correlates with bone turnover similar to intact PTH and FGF-23. These findings suggest a role for activin A in early development of ROD.
Collapse
|
46
|
Gu TT, Chen TY, Yang YZ, Zhao XJ, Sun Y, Li TS, Zhang DM, Kong LD. Pterostilbene alleviates fructose-induced renal fibrosis by suppressing TGF-β1/TGF-β type I receptor/Smads signaling in proximal tubular epithelial cells. Eur J Pharmacol 2019; 842:70-78. [DOI: 10.1016/j.ejphar.2018.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/30/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022]
|
47
|
Emerging role of myostatin and its inhibition in the setting of chronic kidney disease. Kidney Int 2018; 95:506-517. [PMID: 30598193 DOI: 10.1016/j.kint.2018.10.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/13/2018] [Accepted: 10/02/2018] [Indexed: 12/25/2022]
Abstract
The past two decades have witnessed tremendous progress in our understanding of the mechanisms underlying wasting and cachexia in chronic kidney disease (CKD) and in other chronic illnesses, such as cancer and heart failure. In all these conditions wasting is an effect of the activation of protein degradation in muscle, a response that increases the risk of morbidity and mortality. Major recent advances in our knowledge on how CKD and inflammation affect cellular signaling include the identification of the myostatin (MSTN)/activin system, and its related transcriptional program that promotes protein degradation. In addition, the identification of the role of MSTN/activin in the vascular wall shows premise that its inhibition can better control or prevent some effects of CKD on vessels, such as accelerated atherosclerosis and vascular calcifications. In this review, we summarize the expanding role of MSTN activation in promoting muscle atrophy and the recent clinical studies that investigated the efficacy of MSTN/activin pathway antagonism in sarcopenic patients. Moreover, we also review the utility of MSTN inhibition in the experimental models of CKD and its potential advantages in CKD patients. Lessons learned from clinical studies on MSTN antagonism in sarcopenic patients tell us that the anabolic intervention is likely better if we use a block of the two ActRII receptors. At the same time, however, it is becoming clear that MSTN-targeted therapies should not be seen as a substitute for physical activity and nutritional supplementation which are mandatory to successfully manage patients with wasting.
Collapse
|
48
|
Systemic Activation of Activin A Signaling Causes Chronic Kidney Disease-Mineral Bone Disorder. Int J Mol Sci 2018; 19:ijms19092490. [PMID: 30142896 PMCID: PMC6163495 DOI: 10.3390/ijms19092490] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/20/2018] [Accepted: 08/21/2018] [Indexed: 11/19/2022] Open
Abstract
The high cardiovascular mortality associated with chronic kidney disease (CKD) is caused in part by the CKD-mineral bone disorder (CKD-MBD) syndrome. The CKD-MBD consists of skeletal, vascular and cardiac pathology caused by metabolic derangements produced by kidney disease. The prevalence of osteopenia/osteoporosis resulting from the skeletal component of the CKD-MBD, renal osteodystrophy (ROD), in patients with CKD exceeds that of the general population and is a major public health concern. That CKD is associated with compromised bone health is widely accepted, yet the mechanisms underlying impaired bone metabolism in CKD are not fully understood. Therefore, clarification of the molecular mechanisms by which CKD produces ROD is of crucial significance. We have shown that activin A, a member of the transforming growth factor (TGF)-β super family, is an important positive regulator of receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis with Smad-mediated signaling being crucial for inducing osteoclast development and function. Recently, we have demonstrated systemic activation of activin receptors and activin A levels in CKD mouse models, such as diabetic CKD and Alport (AL) syndrome. In these CKD mouse models, bone remodeling caused by increased osteoclast numbers and activated osteoclastic bone resorption was observed and treatment with an activin receptor ligand trap repaired CKD-induced-osteoclastic bone resorption and stimulated individual osteoblastic bone formation, irrespective of parathyroid hormone (PTH) elevation. These findings have opened a new field for exploring mechanisms of activin A-enhanced osteoclast formation and function in CKD. Activin A appears to be a strong candidate for CKD-induced high-turnover ROD. Therefore, the treatment with the decoy receptor for activin A might be a good candidate for treatment for CKD-induced osteopenia or osteoporosis, indicating that the new findings from in these studies will lead to the identification of novel therapeutic targets for CKD-related and osteopenia and osteoporosis in general. In this review, we describe the impact of CKD-induced Smad signaling in osteoclasts, osteoblasts and vascular cells in CKD.
Collapse
|
49
|
Iwasaki Y, Yamato H, Fukagawa M. TGF-Beta Signaling in Bone with Chronic Kidney Disease. Int J Mol Sci 2018; 19:E2352. [PMID: 30103389 PMCID: PMC6121599 DOI: 10.3390/ijms19082352] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/30/2018] [Accepted: 08/08/2018] [Indexed: 01/05/2023] Open
Abstract
Transforming growth factor (TGF)-β signaling is not only important in skeletal development, but also essential in bone remodeling in adult bone. The bone remodeling process involves integrated cell activities induced by multiple stimuli to balance bone resorption and bone formation. TGF-β plays a role in bone remodeling by coordinating cell activities to maintain bone homeostasis. However, mineral metabolism disturbance in chronic kidney disease (CKD) results in abnormal bone remodeling, which leads to ectopic calcification in CKD. High circulating levels of humoral factors such as parathyroid hormone, fibroblast growth factor 23, and Wnt inhibitors modulate bone remodeling in CKD. Several reports have revealed that TGF-β is involved in the production and functions of these factors in bone. TGF-β may act as a factor that mediates abnormal bone remodeling in CKD.
Collapse
Affiliation(s)
- Yoshiko Iwasaki
- Department of Health Sciences, Oita University of Nursing and Health Sciences, Oita 870-1163, Japan.
| | - Hideyuki Yamato
- Division of Nephrology and Metabolism, Tokai University School of Medicine, Kanagawa 259-119, Japan.
| | - Masafumi Fukagawa
- Division of Nephrology and Metabolism, Tokai University School of Medicine, Kanagawa 259-119, Japan.
| |
Collapse
|