1
|
De Leo E, Taranta A, Raso R, Pezzullo M, Piccione M, Matteo V, Vitale A, Bellomo F, Goffredo BM, Diomedi Camassei F, Prencipe G, Rega LR, Emma F. Long-term effects of luteolin in a mouse model of nephropathic cystinosis. Biomed Pharmacother 2024; 178:117236. [PMID: 39096619 DOI: 10.1016/j.biopha.2024.117236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 08/05/2024] Open
Abstract
In infantile nephropathic cystinosis, variants of the CTNS gene cause accumulation of cystine in lysosomes, causing progressive damage to most organs. Patients usually present before 1 year of age with signs of renal Fanconi syndrome. Cysteamine therapy allows cystine clearance from lysosomes and delays kidney damage but does not prevent progression to end-stage kidney disease, suggesting that pathways unrelated to cystine accumulation are also involved. Among these, impaired autophagy, altered endolysosomal trafficking, and increased apoptosis have emerged in recent years as potential targets for new therapies. We previously showed that luteolin, a flavonoid compound, improves these abnormal pathways in cystinotic cells and in zebrafish models of the disease. Herein, we have investigated if prolonged luteolin treatment ameliorates kidney damage in a murine model of cystinosis. To this end, we have treated Ctns-/- mice from 2 to 8 months with 150 mg/kg/day of luteolin. No significant side effects were observed. Compared to untreated animals, analyses of kidney cortex samples obtained after sacrifice showed that luteolin decreased p62/SQSTM1 levels (p <0.001), improved the number, size, and distribution of LAMP1-positive structures (p <0.02), and decreased tissue expression of cleaved caspase 3 (p <0.001). However, we did not observe improvements in renal Fanconi syndrome and kidney inflammation. Kidney function remained normal during the time of the study. These results indicate that luteolin has positive effects on the apoptosis and endo-lysosomal defects of cystinotic proximal tubular cells. However, these beneficial effects did not translate into improvement of renal Fanconi syndrome.
Collapse
Affiliation(s)
- Ester De Leo
- Laboratory of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| | - Anna Taranta
- Laboratory of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Roberto Raso
- Laboratory of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marco Pezzullo
- Core Facilities, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Michela Piccione
- Confocal Microscopy Core Facility, Research Laboratories, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Valentina Matteo
- Laboratory of Immuno-Rheumatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alessia Vitale
- Laboratory of Metabolic Diseases, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesco Bellomo
- Laboratory of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Bianca Maria Goffredo
- Laboratory of Metabolic Diseases, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Giusi Prencipe
- Laboratory of Immuno-Rheumatology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Laura Rita Rega
- Laboratory of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesco Emma
- Division of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
2
|
Medaer L, David D, Smits M, Levtchenko E, Sampaolesi M, Gijsbers R. Residual Cystine Transport Activity for Specific Infantile and Juvenile CTNS Mutations in a PTEC-Based Addback Model. Cells 2024; 13:646. [PMID: 38607085 PMCID: PMC11011962 DOI: 10.3390/cells13070646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Cystinosis is a rare, autosomal recessive, lysosomal storage disease caused by mutations in the gene CTNS, leading to cystine accumulation in the lysosomes. While cysteamine lowers the cystine levels, it does not cure the disease, suggesting that CTNS exerts additional functions besides cystine transport. This study investigated the impact of infantile and juvenile CTNS mutations with discrepant genotype/phenotype correlations on CTNS expression, and subcellular localisation and function in clinically relevant cystinosis cell models to better understand the link between genotype and CTNS function. Using CTNS-depleted proximal tubule epithelial cells and patient-derived fibroblasts, we expressed a selection of CTNSmutants under various promoters. EF1a-driven expression led to substantial overexpression, resulting in CTNS protein levels that localised to the lysosomal compartment. All CTNSmutants tested also reversed cystine accumulation, indicating that CTNSmutants still exert transport activity, possibly due to the overexpression conditions. Surprisingly, even CTNSmutants expression driven by the less potent CTNS and EFS promoters reversed the cystine accumulation, contrary to the CTNSG339R missense mutant. Taken together, our findings shed new light on CTNS mutations, highlighting the need for robust assessment methodologies in clinically relevant cellular models and thus paving the way for better stratification of cystinosis patients, and advocating for the development of more personalized therapy.
Collapse
Affiliation(s)
- Louise Medaer
- Laboratory of Molecular Virology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (L.M.); (M.S.)
| | - Dries David
- Laboratory of Molecular Virology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (L.M.); (M.S.)
| | - Maxime Smits
- Laboratory of Molecular Virology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (L.M.); (M.S.)
- Leuven Viral Vector Core, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Elena Levtchenko
- Department of Paediatric Nephrology & Development and Regeneration, University Hospitals Leuven & KU Leuven, 3000 Leuven, Belgium;
- Department of Paediatric Nephrology, Amsterdam University Medical Centre, 1081 Amsterdam, The Netherlands
| | - Maurilio Sampaolesi
- Translational Cardiology Laboratory, Department of Development and Regeneration, Stem Cell Institute, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium;
| | - Rik Gijsbers
- Laboratory of Molecular Virology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (L.M.); (M.S.)
- Leuven Viral Vector Core, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
3
|
Chen M, Gu X. Emerging roles of proximal tubular endocytosis in renal fibrosis. Front Cell Dev Biol 2023; 11:1235716. [PMID: 37799275 PMCID: PMC10547866 DOI: 10.3389/fcell.2023.1235716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/11/2023] [Indexed: 10/07/2023] Open
Abstract
Endocytosis is a crucial component of many pathological conditions. The proximal tubules are responsible for reabsorbing the majority of filtered water and glucose, as well as all the proteins filtered through the glomerular barrier via endocytosis, indicating an essential role in kidney diseases. Genetic mutations or acquired insults could affect the proximal tubule endocytosis processes, by disturbing or overstressing the endolysosomal system and subsequently activating different pathways, orchestrating renal fibrosis. This paper will review recent studies on proximal tubular endocytosis affected by other diseases and factors. Endocytosis plays a vital role in the development of renal fibrosis, and renal fibrosis could also, in turn, affect tubular endocytosis.
Collapse
Affiliation(s)
- Min Chen
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiangchen Gu
- Department of Nephrology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Nephrology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Medicine, Shanghai Hospital of Civil Aviation Administration of China, Shanghai, China
| |
Collapse
|
4
|
Keidel LF, Schworm B, Hohenfellner K, Kruse F, Priglinger S, Luft N, Priglinger C. Posterior Segment Involvement in Infantile Nephropathic Cystinosis - A Review. Klin Monbl Augenheilkd 2023; 240:266-275. [PMID: 36977427 DOI: 10.1055/s-0037-1599653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Cystinosis is a rare lysosomal storage disease with a prevalence of 1 : 100 000 - 1 : 200 000 cases. It is caused by biallelic mutations in the CTNS gene, which encodes cystinosin, that transport cystine out of the lysosomes. Due to its dysfunction, cystine crystals accumulate in the lysosomes and ultimately cause apoptosis of the cell. Since cystinosin is ubiquitously present in the body, cystine crystals are deposited in every body structure and lead to the dysfunction of various organ systems in the course of time. Cystine crystals deposited in the cornea are a clinical hallmark of the disease, while there is less awareness of concomitant posterior segment alterations. Symmetrical pigment epithelial mottling and patches of depigmentation frequently start in the periphery and progress towards the posterior pole and can be encountered upon fundus biomicroscopy. Spectral-domain optical coherence tomography (SD-OCT) is an elegant tool for visualizing chorioretinal cystine crystals at the posterior pole. An SD-OCT-based clinical grading of the severity of the chorioretinal manifestation can potentially be applied as a biomarker for systemic disease status and for monitoring oral therapy adherence in the future. Along with previous histological examinations, it may also give information about the location of cystine crystals in the choroid and retina. This review aims to increase the awareness of vision-threatening retinal and choroidal changes in cystinosis and the concomitant findings in SD-OCT.
Collapse
Affiliation(s)
| | - Benedikt Schworm
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, Germany
| | | | - Franziska Kruse
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, Germany
| | | | - Nikolaus Luft
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, Germany
| | - Claudia Priglinger
- Department of Ophthalmology, Ludwig-Maximilians-University Munich, Germany
| |
Collapse
|
5
|
Hohenfellner K, Zerell K, Haffner D. Cystinosis. Klin Monbl Augenheilkd 2023; 240:251-259. [PMID: 36977426 DOI: 10.1055/a-2022-8522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Cystinosis is a very rare autosomal recessive lysosomal storage disorder with an incidence of 1 : 150,000 - 1 : 200,000, and is caused by mutations in the CTNS gene encoding the lysosomal membrane protein cystinosin, which transports cystine out of the lysosome into the cytoplasm. As a result, accumulation of cystine occurs in almost all cells and tissues, especially in the kidneys, leading to multiple organ involvement. Introduction of drug therapy with cysteamine in the mid 1980s, along with the availability of renal replacement therapy in childhood, have dramatically improved patient outcome. Whereas patients used to die without therapy with end-stage renal failure during the first decade of life, nowadays most patients live well into adulthood without renal replacement therapy, and several reach 40 years. There is robust evidence that early initiation and sustained lifelong therapy with cysteamine are both essential for morbidity and mortality. The rarity of the disease and the multi-organ involvement present an enormous challenge for those affected and the providers of care for this patient group.
Collapse
Affiliation(s)
- Katharina Hohenfellner
- Klinik für Kinder- und Jugendmedizin/Kindernephrologie, RoMed Kliniken, Rosenheim, Deutschland
| | - Kirstin Zerell
- Rosenheim, kbo-Heckscher-Klinikum gGmbH, München, Deutschland
| | - Dieter Haffner
- Klinik für Pädiatrische Nieren-, Leber- & Stoffwechselerkrankungen, MHH, Hannover, Deutschland
| |
Collapse
|
6
|
Keidel L, Hohenfellner K, Schworm B, Priglinger S, Luft N, Priglinger C. Spectral domain optical coherence tomography-based retinochoroidal cystine crystal score: a window into infantile nephropathic cystinosis. Br J Ophthalmol 2023; 107:234-241. [PMID: 34531199 PMCID: PMC9887385 DOI: 10.1136/bjophthalmol-2021-319612] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/17/2021] [Indexed: 02/03/2023]
Abstract
PRÉCIS: Cystinosis is a lysosomal storage disease leading to an accumulation of cystine crystals in several organs. We aim to comprehensively describe chorioretinal cystine crystals via spectral domain optical coherence tomography (SD-OCT) and elaborate a new biomarker for systemic disease control. BACKGROUND/AIMS Cystinosis is a rare lysosomal storage disease leading to an accumulation of cystine crystals in several organs. This study aims to describe the deposition of retinochoroidal crystals in infantile nephropathic cystinosis and to elucidate their potential value as an objective biomarker for systemic disease control. METHODS This cross-sectional study was carried out by the University Eye Hospital of the Ludwig-Maximilian University (Munich, Germany) in collaboration with the German Cystinosis Study Group. A complete ophthalmologic examination was performed, along with posterior segment SD-OCT (Spectralis; Heidelberg Engineering GmbH, Heidelberg, Germany). Retinochoroidal crystals were graded by employing a novel semiquantitative grading system-the retinochoroidal cystine crystal score (RCCCS). To quantify quality of vision, patients completed a specific questionnaire. A total of 85 eyes of 43 patients with cystinosis were included (mean age 22.3±8.8 years, range 6-39; male:female ratio=23:20). RESULTS Cystine crystals were detectable in all neuroretinal layers and the choroid, most frequently in the choriocapillaris. The RCCCS was negatively correlated with cysteamine intake (r=0.533, p=0.001) and positively with cystatin C, a stable parameter of renal function (r=0.496, p=0.016). Moreover, the value of the RCCCS affected subjective quality of vision. Genetic analysis indicated pronounced crystal deposition in patients with heterozygous mutations containing the 57-kb-deletion allele of the CTNS gene. CONCLUSION Ocular cystinosis leads to retinochoroidal crystal accumulation in every stage of the disease. Crystal deposition may be markedly influenced by oral cysteamine therapy. Therefore, the presented SD-OCT based grading system might serve as an objective biomarker for systemic disease control.
Collapse
Affiliation(s)
- Leonie Keidel
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Bayern, Germany
| | | | - Benedikt Schworm
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Bayern, Germany
| | - Siegfried Priglinger
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Bayern, Germany
| | - Nikolaus Luft
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Bayern, Germany
| | - Claudia Priglinger
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Bayern, Germany
| |
Collapse
|
7
|
Imaging of metabolic and overload disorders in tissues and organs. Jpn J Radiol 2023; 41:571-595. [PMID: 36680702 DOI: 10.1007/s11604-022-01379-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/24/2022] [Indexed: 01/22/2023]
Abstract
Metabolic and overload disorders are a heterogeneous group of relatively uncommon but important diseases. While imaging plays a key role in the early detection and accurate diagnosis in specific organs with a pivotal role in several metabolic pathways, most of these diseases affect different tissues as part of a systemic syndromes. Moreover, since the symptoms are often vague and phenotypes similar, imaging alterations can present as incidental findings, which must be recognized and interpreted in the light of further biochemical and histological investigations. Among imaging modalities, MRI allows, thanks to its multiparametric properties, to obtain numerous information on tissue composition, but many metabolic and accumulation alterations require a multimodal evaluation, possibly using advanced imaging techniques and sequences, not only for the detection but also for accurate characterization and quantification. The purpose of this review is to describe the different alterations resulting from metabolic and overload pathologies in organs and tissues throughout the body, with particular reference to imaging findings.
Collapse
|
8
|
Fernandez RL, Juntunen ND, Brunold TC. Differences in the Second Coordination Sphere Tailor the Substrate Specificity and Reactivity of Thiol Dioxygenases. Acc Chem Res 2022; 55:2480-2490. [PMID: 35994511 PMCID: PMC9583696 DOI: 10.1021/acs.accounts.2c00359] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In recent years, considerable progress has been made toward elucidating the geometric and electronic structures of thiol dioxygenases (TDOs). TDOs catalyze the conversion of substrates with a sulfhydryl group to their sulfinic acid derivatives via the addition of both oxygen atoms from molecular oxygen. All TDOs discovered to date belong to the family of cupin-type mononuclear nonheme Fe(II)-dependent metalloenzymes. While most members of this enzyme family bind the Fe cofactor by two histidines and one carboxylate side chain (2-His-1-carboxylate) to provide a monoanionic binding motif, TDOs feature a neutral three histidine (3-His) facial triad. In this Account, we present a bioinformatics analysis and multiple sequence alignment that highlight the significance of the secondary coordination sphere in tailoring the substrate specificity and reactivity among the different TDOs. These insights provide the framework within which important structural and functional features of the distinct TDOs are discussed.The best studied TDO is cysteine dioxygenase (CDO), which catalyzes the conversion of cysteine to cysteine sulfinic acid in both eukaryotes and prokaryotes. Crystal structures of resting and substrate-bound mammalian CDOs revealed two surprising structural motifs in the first- and second coordination spheres of the Fe center. The first is the presence of the abovementioned neutral 3-His facial triad that coordinates the Fe ion. The second is the existence of a covalent cross-link between the sulfur of Cys93 and an ortho carbon of Tyr157 (mouse CDO numbering scheme). While the exact role of this cross-link remains incompletely understood, various studies established that it is needed for proper substrate Cys positioning and gating solvent access to the active site. Intriguingly, bacterial CDOs lack the Cys-Tyr cross-link; yet, they are as active as cross-linked eukaryotic CDOs.The other known mammalian TDO is cysteamine dioxygenase (ADO). Initially, it was believed that ADO solely catalyzes the oxidation of cysteamine to hypotaurine. However, it has recently been shown that ADO additionally oxidizes N-terminal cysteine (Nt-Cys) peptides, which indicates that ADO may play a much more significant role in mammalian physiology than was originally anticipated. Though predicted on the basis of sequence alignment, site-directed mutagenesis, and spectroscopic studies, it was not until last year that two crystal structures, one of wild-type mouse ADO (solved by us) and the other of a variant of nickel-substituted human ADO, finally provided direct evidence that this enzyme also features a 3-His facial triad. These structures additionally revealed several features that are unique to ADO, including a putative cosubstrate O2 access tunnel that is lined by two Cys residues. Disulfide formation under conditions of high O2 levels may serve as a gating mechanism to prevent ADO from depleting organisms of Nt-Cys-containing molecules.The combination of kinetic and spectroscopic studies in conjunction with structural characterizations of TDOs has furthered our understanding of enzymatic sulfhydryl substrate regulation. In this article, we take advantage of the fact that the ADO X-ray crystal structures provided the final piece needed to compare and contrast key features of TDOs, an essential family of metalloenzymes found across all kingdoms of life.
Collapse
Affiliation(s)
- Rebeca L. Fernandez
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Nicholas D. Juntunen
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Thomas C. Brunold
- Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
9
|
Differences in renal cortex transcriptional profiling of wild-type and novel type B cystinuria model rats. Urolithiasis 2022; 50:279-291. [PMID: 35416493 PMCID: PMC9110498 DOI: 10.1007/s00240-022-01321-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 03/03/2022] [Indexed: 11/20/2022]
Abstract
Cystinuria is a genetic disorder of cystine transport that accounts for 1–2% of all cases of renal lithiasis. It is characterized by hyperexcretion of cystine in urine and recurrent cystine lithiasis. Defective transport of cystine into epithelial cells of renal tubules occurs because of mutations of the transport heterodimer, including protein b0,+AT (encoded by SLC7A9) and rBAT (encoded by SLC3A1) linked through a covalent disulfide bond. Study generated a novel type B cystinuria rat model by artificially deleting 7 bp of Slc7a9 gene exon 3 using the CRISPR-Cas9 system, and those Slc7a9-deficient rats were proved to be similar with cystinuria in terms of genome, transcriptome, translation, and biologic phenotypes with no off-target editing. Subsequent comparisons of renal histopathology indicated model rats gained typical secondary changes as medullary fibrosis with no stone formation. A total of 689 DEGs (383 upregulated and 306 downregulated) were differentially expressed in the renal cortex of cystinuria rats. In accordance with the functional annotation of DEGs, the potential role of glutathione metabolism processes in the kidney of cystinuria rat model was proposed, and KEGG analysis results showed that knock-out of Slc7a9 gene triggered more biological changes which has not been studied. In short, for the first time, a rat model and its transcriptional database that mimics the pathogenesis and clinical consequences of human type B cystinuria were generated.
Collapse
|
10
|
Krohn P, Rega LR, Harvent M, Festa BP, Taranta A, Luciani A, Dewulf J, Cremonesi A, Camassei FD, Hanson JVM, Gerth-Kahlert C, Emma F, Berquez M, Devuyst O. OUP accepted manuscript. Hum Mol Genet 2022; 31:2262-2278. [PMID: 35137071 PMCID: PMC9262394 DOI: 10.1093/hmg/ddac033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/21/2022] [Accepted: 02/02/2022] [Indexed: 11/14/2022] Open
Abstract
Recessive mutations in the CTNS gene encoding the lysosomal transporter cystinosin cause cystinosis, a lysosomal storage disease leading to kidney failure and multisystem manifestations. A Ctns knockout mouse model recapitulates features of cystinosis, but the delayed onset of kidney manifestations, phenotype variability and strain effects limit its use for mechanistic and drug development studies. To provide a better model for cystinosis, we generated a Ctns knockout rat model using CRISPR/Cas9 technology. The Ctns−/− rats display progressive cystine accumulation and crystal formation in multiple tissues including kidney, liver and thyroid. They show an early onset and progressive loss of urinary solutes, indicating generalized proximal tubule dysfunction, with development of typical swan-neck lesions, tubulointerstitial fibrosis and kidney failure, and decreased survival. The Ctns−/− rats also present crystals in the cornea, and bone and liver defects, as observed in patients. Mechanistically, the loss of cystinosin induces a phenotype switch associating abnormal proliferation and dedifferentiation, loss of apical receptors and transporters, and defective lysosomal activity and autophagy in the cells. Primary cultures of proximal tubule cells derived from the Ctns−/− rat kidneys confirmed the key changes caused by cystine overload, including reduced endocytic uptake, increased proliferation and defective lysosomal dynamics and autophagy. The novel Ctns−/− rat model and derived proximal tubule cell system provide invaluable tools to investigate the pathogenesis of cystinosis and to accelerate drug discovery.
Collapse
Affiliation(s)
- Patrick Krohn
- Institute of Physiology, University of Zurich, Zurich 8057, Switzerland
| | - Laura Rita Rega
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
| | - Marianne Harvent
- Institute of Physiology, University of Zurich, Zurich 8057, Switzerland
| | | | - Anna Taranta
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
| | | | - Joseph Dewulf
- Department of Laboratory Medicine, Cliniques universitaires Saint Luc, UCLouvain, Brussels 1200, Belgium
- Department of Biochemistry, de Duve Institute, UCLouvain, Brussels 1200, Belgium
| | - Alessio Cremonesi
- Division of Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich 8032, Switzerland
| | | | - James V M Hanson
- Department of Ophthalmology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Christina Gerth-Kahlert
- Department of Ophthalmology, University Hospital Zurich and University of Zurich, Zurich 8091, Switzerland
| | - Francesco Emma
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Area, Bambino Gesù Children’s Hospital, IRCCS, Rome 00165, Italy
- Department of Pediatric Subspecialties, Division of Nephrology, Children’s Hospital Bambino Gesù, IRCCS, Rome 00165, Italy
| | - Marine Berquez
- To whom correspondence should be addressed at: University of Zurich, Mechanisms of Inherited Kidney Disorders Group, Winterthurerstrasse 190, Zurich 8057, Switzerland. Tel: +41 (0)44 635 51 07; (Marine Berquez); Tel: +41 (0)44 635 50 82; Fax: +41 (0)44 635 68 14; (Olivier Devuyst)
| | - Olivier Devuyst
- To whom correspondence should be addressed at: University of Zurich, Mechanisms of Inherited Kidney Disorders Group, Winterthurerstrasse 190, Zurich 8057, Switzerland. Tel: +41 (0)44 635 51 07; (Marine Berquez); Tel: +41 (0)44 635 50 82; Fax: +41 (0)44 635 68 14; (Olivier Devuyst)
| |
Collapse
|
11
|
Vaisbich MH, Caires Ferreira J, Price H, Young KD, Sile S, Checani G, Langman CB. Cysteamine bitartrate delayed-release capsules control leukocyte cystine levels and promote statural growth and kidney health in an open-label study of treatment-naïve patients <6 years of age with nephropathic cystinosis. JIMD Rep 2022; 63:66-79. [PMID: 35028272 PMCID: PMC8743345 DOI: 10.1002/jmd2.12260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 11/10/2022] Open
Abstract
Nephropathic cystinosis is a rare autosomal recessive lysosomal storage disease that is characterized by accumulation of cysteine and formation of crystals within cells of different organs and tissues causing systemic manifestations in childhood that include poor linear growth, ocular involvement, hypothyroidism, and progressive kidney disease. This study was a long-term, prospective open-label evaluation of twice-daily delayed release (DR) cysteamine capsules in cystinosis patients <6 years of age who were naïve to any form of cysteamine treatment. Fifteen treatment-naïve patients <6 years old (mean age 2.2 ± 1.0 years, 53% male, 73% White) were enrolled and treated with DR-cysteamine capsules for up to 18 months. Patients had clinically meaningful decreases in WBC cysteine concentration during treatment (3.2 ± 3.0 nmol ½ cystine/mg protein at Day 1 to 0.8 ± 0.8 nmol ½ cystine/mg protein at study exit), and anthropometric data improvements were consistently observed in height, weight and body surface area. Additionally, estimated glomerular filtration rate increased from 55.93 ± 22.43 ml/min/1.73 m2 at baseline to 63.79 ± 21.44 ml/min/1.73 m2 at study exit. Pharmacokinetic/Pharmacodynamic results support the use of the same starting, escalation, and maintenance doses according to body surface for children aged <6 years that are currently recommended in adults and older children. All patients experienced ≥1 adverse event(s) with vomiting (80%) and upper respiratory tract infection (53%) most frequently reported. Based on our study, patients <6 years of age with nephropathic cystinosis without prior treatment can safely and effectively initiate treatment with DR-cysteamine, a delayed-release form of cysteamine bitartrate that can be given every 12 h.
Collapse
Affiliation(s)
| | - Juliana Caires Ferreira
- Instituto da Criança do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP)São PauloBrazil
| | - Heather Price
- Ann & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | - Kyleen D. Young
- Ann & Robert H. Lurie Children's Hospital of ChicagoChicagoIllinoisUSA
| | - Saba Sile
- Horizon Therapeutics plcDeerfieldIllinoisUSA
| | | | - Craig B. Langman
- Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
12
|
Das G, Nanda PM, Kaur A, Kumar R. Bartter syndrome and hypothyroidism masquerading cystinosis in a 3-year-old girl: rare manifestation of a rare disease. BMJ Case Rep 2021; 14:14/7/e242954. [PMID: 34312133 PMCID: PMC8314690 DOI: 10.1136/bcr-2021-242954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Cystinosis is a multisystem disorder with varied presentations secondary to deposition of cystine crystals in different organ systems. Children with cystinosis typically present with renal tubular acidosis and failure to thrive. We report a 3-year-old girl, born to a third-degree consanguineous couple, who presented with failure to thrive and polyuria. Laboratory investigations showed metabolic alkalosis suggestive of a Bartter-like syndrome and acquired hypothyroidism. Although metabolic alkalosis is a rare manifestation of cystinosis, the presence of renal tubular dysfunction and hypothyroidism prompted consideration of a probable diagnosis of cystinosis in the index child. Slit-lamp examination revealed cystine crystals in the cornea and genetic analysis showed a mutation in exon 9 of the CTNS (cystinosin, lysosomal cystine transporter) gene on chromosome 17. We highlight the importance of considering cystinosis as a differential diagnosis for Bartter syndrome and hypothyroidism.
Collapse
Affiliation(s)
- Gargi Das
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Pamali Mahasweta Nanda
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Anupriya Kaur
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Rakesh Kumar
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
13
|
Bondue T, Arcolino FO, Veys KRP, Adebayo OC, Levtchenko E, van den Heuvel LP, Elmonem MA. Urine-Derived Epithelial Cells as Models for Genetic Kidney Diseases. Cells 2021; 10:cells10061413. [PMID: 34204173 PMCID: PMC8230018 DOI: 10.3390/cells10061413] [Citation(s) in RCA: 7] [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: 05/04/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/11/2022] Open
Abstract
Epithelial cells exfoliated in human urine can include cells anywhere from the urinary tract and kidneys; however, podocytes and proximal tubular epithelial cells (PTECs) are by far the most relevant cell types for the study of genetic kidney diseases. When maintained in vitro, they have been proven extremely valuable for discovering disease mechanisms and for the development of new therapies. Furthermore, cultured patient cells can individually represent their human sources and their specific variants for personalized medicine studies, which are recently gaining much interest. In this review, we summarize the methodology for establishing human podocyte and PTEC cell lines from urine and highlight their importance as kidney disease cell models. We explore the well-established and recent techniques of cell isolation, quantification, immortalization and characterization, and we describe their current and future applications.
Collapse
Affiliation(s)
- Tjessa Bondue
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
| | - Fanny O. Arcolino
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
| | - Koenraad R. P. Veys
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatrics, Division of Pediatric Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Oyindamola C. Adebayo
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Elena Levtchenko
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatrics, Division of Pediatric Nephrology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Lambertus P. van den Heuvel
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; (T.B.); (F.O.A.); (K.R.P.V.); (O.C.A.); (E.L.); (L.P.v.d.H.)
- Department of Pediatric Nephrology, Radboud University Medical Center, 6500 Nijmegen, The Netherlands
| | - Mohamed A. Elmonem
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo 11628, Egypt
- Correspondence:
| |
Collapse
|
14
|
Lemaire M. Novel Fanconi renotubular syndromes provide insights in proximal tubule pathophysiology. Am J Physiol Renal Physiol 2020; 320:F145-F160. [PMID: 33283647 DOI: 10.1152/ajprenal.00214.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The various forms of Fanconi renotubular syndromes (FRTS) offer significant challenges for clinicians and present unique opportunities for scientists who study proximal tubule physiology. This review will describe the clinical characteristics, genetic underpinnings, and underlying pathophysiology of the major forms of FRST. Although the classic forms of FRTS will be presented (e.g., Dent disease or Lowe syndrome), particular attention will be paid to five of the most recently discovered FRTS subtypes caused by mutations in the genes encoding for L-arginine:glycine amidinotransferase (GATM), solute carrier family 34 (type Ii sodium/phosphate cotransporter), member 1 (SLC34A1), enoyl-CoAhydratase/3-hydroxyacyl CoA dehydrogenase (EHHADH), hepatocyte nuclear factor 4A (HNF4A), or NADH dehydrogenase complex I, assembly factor 6 (NDUFAF6). We will explore how mutations in these genes revealed unexpected mechanisms that led to compromised proximal tubule functions. We will also describe the inherent challenges associated with gene discovery studies based on findings derived from small, single-family studies by focusing the story of FRTS type 2 (SLC34A1). Finally, we will explain how extensive alternative splicing of HNF4A has resulted in confusion with mutation nomenclature for FRTS type 4.
Collapse
Affiliation(s)
- Mathieu Lemaire
- Division of Nephrology and Cell Biology Program, SickKids Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Huizing M, Gahl WA. Inherited disorders of lysosomal membrane transporters. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183336. [PMID: 32389669 PMCID: PMC7508925 DOI: 10.1016/j.bbamem.2020.183336] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/01/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023]
Abstract
Disorders caused by defects in lysosomal membrane transporters form a distinct subgroup of lysosomal storage disorders (LSDs). To date, defects in only 10 lysosomal membrane transporters have been associated with inherited disorders. The clinical presentations of these diseases resemble the phenotypes of other LSDs; they are heterogeneous and often present in children with neurodegenerative manifestations. However, for pathomechanistic and therapeutic studies, lysosomal membrane transport defects should be distinguished from LSDs caused by defective hydrolytic enzymes. The involved proteins differ in function, localization, and lysosomal targeting, and the diseases themselves differ in their stored material and therapeutic approaches. We provide an overview of the small group of disorders of lysosomal membrane transporters, emphasizing discovery, pathomechanism, clinical features, diagnostic methods and therapeutic aspects. We discuss common aspects of lysosomal membrane transporter defects that can provide the basis for preclinical research into these disorders.
Collapse
Affiliation(s)
- Marjan Huizing
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - William A Gahl
- Human Biochemical Genetics Section, Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
De Leo E, Elmonem MA, Berlingerio SP, Berquez M, Festa BP, Raso R, Bellomo F, Starborg T, Janssen MJ, Abbaszadeh Z, Cairoli S, Goffredo BM, Masereeuw R, Devuyst O, Lowe M, Levtchenko E, Luciani A, Emma F, Rega LR. Cell-Based Phenotypic Drug Screening Identifies Luteolin as Candidate Therapeutic for Nephropathic Cystinosis. J Am Soc Nephrol 2020; 31:1522-1537. [PMID: 32503896 PMCID: PMC7351012 DOI: 10.1681/asn.2019090956] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 04/04/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mutations in the gene that encodes the lysosomal cystine transporter cystinosin cause the lysosomal storage disease cystinosis. Defective cystine transport leads to intralysosomal accumulation and crystallization of cystine. The most severe phenotype, nephropathic cystinosis, manifests during the first months of life, as renal Fanconi syndrome. The cystine-depleting agent cysteamine significantly delays symptoms, but it cannot prevent progression to ESKD and does not treat Fanconi syndrome. This suggests the involvement of pathways in nephropathic cystinosis that are unrelated to lysosomal cystine accumulation. Recent data indicate that one such potential pathway, lysosome-mediated degradation of autophagy cargoes, is compromised in cystinosis. METHODS To identify drugs that reduce levels of the autophagy-related protein p62/SQSTM1 in cystinotic proximal tubular epithelial cells, we performed a high-throughput screening on the basis of an in-cell ELISA assay. We then tested a promising candidate in cells derived from patients with, and mouse models of, cystinosis, and in preclinical studies in cystinotic zebrafish. RESULTS Of 46 compounds identified as reducing p62/SQSTM1 levels in cystinotic cells, we selected luteolin on the basis of its efficacy, safety profile, and similarity to genistein, which we previously showed to ameliorate other lysosomal abnormalities of cystinotic cells. Our data show that luteolin improves the autophagy-lysosome degradative pathway, is a powerful antioxidant, and has antiapoptotic properties. Moreover, luteolin stimulates endocytosis and improves the expression of the endocytic receptor megalin. CONCLUSIONS Our data show that luteolin improves defective pathways of cystinosis and has a good safety profile, and thus has potential as a treatment for nephropathic cystinosis and other renal lysosomal storage diseases.
Collapse
Affiliation(s)
- Ester De Leo
- Renal Diseases Research Unit, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Mohamed A. Elmonem
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
- Department of Pediatric Nephrology and Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
| | - Sante Princiero Berlingerio
- Department of Pediatric Nephrology and Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
| | - Marine Berquez
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Roberto Raso
- Renal Diseases Research Unit, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Francesco Bellomo
- Renal Diseases Research Unit, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Tobias Starborg
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health Sciences, University of Manchester, Manchester, UK
| | - Manoe Jacoba Janssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Zeinab Abbaszadeh
- Confocal Microscopy Core Facility, Research Laboratories, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Sara Cairoli
- Department of Pediatric Medicine, Laboratory of Metabolic Biochemistry Unit, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Bianca Maria Goffredo
- Department of Pediatric Medicine, Laboratory of Metabolic Biochemistry Unit, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Martin Lowe
- Division of Molecular and Cellular Function, School of Biological Sciences, Faculty of Biology, Medicine and Health Sciences, University of Manchester, Manchester, UK
| | - Elena Levtchenko
- Department of Pediatric Nephrology and Development and Regeneration, University Hospitals Leuven, Leuven, Belgium
| | | | - Francesco Emma
- Renal Diseases Research Unit, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
- Division of Nephrology, Department of Pediatric Subspecialties, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Laura Rita Rega
- Renal Diseases Research Unit, Bambino Gesù Children’s Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| |
Collapse
|
17
|
Hollywood JA, Przepiorski A, D'Souza RF, Sreebhavan S, Wolvetang EJ, Harrison PT, Davidson AJ, Holm TM. Use of Human Induced Pluripotent Stem Cells and Kidney Organoids To Develop a Cysteamine/mTOR Inhibition Combination Therapy for Cystinosis. J Am Soc Nephrol 2020; 31:962-982. [PMID: 32198276 DOI: 10.1681/asn.2019070712] [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: 07/18/2019] [Accepted: 02/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Mutations in CTNS-a gene encoding the cystine transporter cystinosin-cause the rare, autosomal, recessive, lysosomal-storage disease cystinosis. Research has also implicated cystinosin in modulating the mTORC1 pathway, which serves as a core regulator of cellular metabolism, proliferation, survival, and autophagy. In its severest form, cystinosis is characterized by cystine accumulation, renal proximal tubule dysfunction, and kidney failure. Because treatment with the cystine-depleting drug cysteamine only slows disease progression, there is an urgent need for better treatments. METHODS To address a lack of good human-based cell culture models for studying cystinosis, we generated the first human induced pluripotent stem cell (iPSC) and kidney organoid models of the disorder. We used a variety of techniques to examine hallmarks of cystinosis-including cystine accumulation, lysosome size, the autophagy pathway, and apoptosis-and performed RNA sequencing on isogenic lines to identify differentially expressed genes in the cystinosis models compared with controls. RESULTS Compared with controls, these cystinosis models exhibit elevated cystine levels, increased apoptosis, and defective basal autophagy. Cysteamine treatment ameliorates this phenotype, except for abnormalities in apoptosis and basal autophagy. We found that treatment with everolimus, an inhibitor of the mTOR pathway, reduces the number of large lysosomes, decreases apoptosis, and activates autophagy, but it does not rescue the defect in cystine loading. However, dual treatment of cystinotic iPSCs or kidney organoids with cysteamine and everolimus corrects all of the observed phenotypic abnormalities. CONCLUSIONS These observations suggest that combination therapy with a cystine-depleting drug such as cysteamine and an mTOR pathway inhibitor such as everolimus has potential to improve treatment of cystinosis.
Collapse
Affiliation(s)
- Jennifer A Hollywood
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Aneta Przepiorski
- Department of Developmental Biology, University of Pittsburgh, Pennsylvania
| | - Randall F D'Souza
- Discipline of Nutrition, The University of Auckland, Auckland, New Zealand
| | - Sreevalsan Sreebhavan
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland, Australia
| | - Patrick T Harrison
- Department of Physiology, Biosciences Institute, University College Cork, Cork, Ireland
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Teresa M Holm
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
18
|
De Rasmo D, Signorile A, De Leo E, Polishchuk EV, Ferretta A, Raso R, Russo S, Polishchuk R, Emma F, Bellomo F. Mitochondrial Dynamics of Proximal Tubular Epithelial Cells in Nephropathic Cystinosis. Int J Mol Sci 2019; 21:ijms21010192. [PMID: 31888107 PMCID: PMC6982165 DOI: 10.3390/ijms21010192] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 12/27/2022] Open
Abstract
Nephropathic cystinosis is a rare lysosomal storage disorder caused by mutations in CTNS gene leading to Fanconi syndrome. Independent studies reported defective clearance of damaged mitochondria and mitochondrial fragmentation in cystinosis. Proteins involved in the mitochondrial dynamics and the mitochondrial ultrastructure were analyzed in CTNS-/- cells treated with cysteamine, the only drug currently used in the therapy for cystinosis but ineffective to treat Fanconi syndrome. CTNS-/- cells showed an overexpression of parkin associated with deregulation of ubiquitination of mitofusin 2 and fission 1 proteins, an altered proteolytic processing of optic atrophy 1 (OPA1), and a decreased OPA1 oligomerization. According to molecular findings, the analysis of electron microscopy images showed a decrease of mitochondrial cristae number and an increase of cristae lumen and cristae junction width. Cysteamine treatment restored the fission 1 ubiquitination, the mitochondrial size, number and lumen of cristae, but had no effect on cristae junction width, making CTNS-/- tubular cells more susceptible to apoptotic stimuli.
Collapse
Affiliation(s)
- Domenico De Rasmo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), 70124 Bari, Italy;
- Correspondence: (D.D.R.); (F.B.); Tel.: +39-080-5448516 (D.D.R.); +39-06-68592997 (F.B)
| | - Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.); (S.R.)
| | - Ester De Leo
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (E.D.L.); (R.R.)
| | - Elena V. Polishchuk
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy; (E.V.P.); (R.P.)
| | - Anna Ferretta
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), 70124 Bari, Italy;
| | - Roberto Raso
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (E.D.L.); (R.R.)
| | - Silvia Russo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.S.); (S.R.)
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy; (E.V.P.); (R.P.)
| | - Francesco Emma
- Division of Nephrology, Department of Pediatric Subspecialties, Bambino Gesù Children’s Hospital—IRCCS, 00165 Rome, Italy;
| | - Francesco Bellomo
- Renal Diseases Research Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital—IRCCS, 00146 Rome, Italy; (E.D.L.); (R.R.)
- Correspondence: (D.D.R.); (F.B.); Tel.: +39-080-5448516 (D.D.R.); +39-06-68592997 (F.B)
| |
Collapse
|
19
|
Janssens V, Gaide Chevronnay HP, Marie S, Vincent MF, Van Der Smissen P, Nevo N, Vainio S, Nielsen R, Christensen EI, Jouret F, Antignac C, Pierreux CE, Courtoy PJ. Protection of Cystinotic Mice by Kidney-Specific Megalin Ablation Supports an Endocytosis-Based Mechanism for Nephropathic Cystinosis Progression. J Am Soc Nephrol 2019; 30:2177-2190. [PMID: 31548351 DOI: 10.1681/asn.2019040371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Deletions or inactivating mutations of the cystinosin gene CTNS lead to cystine accumulation and crystals at acidic pH in patients with nephropathic cystinosis, a rare lysosomal storage disease and the main cause of hereditary renal Fanconi syndrome. Early use of oral cysteamine to prevent cystine accumulation slows progression of nephropathic cystinosis but it is a demanding treatment and not a cure. The source of cystine accumulating in kidney proximal tubular cells and cystine's role in disease progression are unknown. METHODS To investigate whether receptor-mediated endocytosis by the megalin/LRP2 pathway of ultrafiltrated, disulfide-rich plasma proteins could be a source of cystine in proximal tubular cells, we used a mouse model of cystinosis in which conditional excision of floxed megalin/LRP2 alleles in proximal tubular cells of cystinotic mice was achieved by a Cre-LoxP strategy using Wnt4-CRE. We evaluated mice aged 6-9 months for kidney cystine levels and crystals; histopathology, with emphasis on swan-neck lesions and proximal-tubular-cell apoptosis and proliferation (turnover); and proximal-tubular-cell expression of the major apical transporters sodium-phosphate cotransporter 2A (NaPi-IIa) and sodium-glucose cotransporter-2 (SGLT-2). RESULTS Wnt4-CRE-driven megalin/LRP2 ablation in cystinotic mice efficiently blocked kidney cystine accumulation, thereby preventing lysosomal deformations and crystal deposition in proximal tubular cells. Swan-neck lesions were largely prevented and proximal-tubular-cell turnover was normalized. Apical expression of the two cotransporters was also preserved. CONCLUSIONS These observations support a key role of the megalin/LRP2 pathway in the progression of nephropathic cystinosis and provide a proof of concept for the pathway as a therapeutic target.
Collapse
Affiliation(s)
- Virginie Janssens
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | | | - Sandrine Marie
- Biochemical Genetics, Academic Hospital Saint-Luc, Brussels, Belgium
| | | | - Patrick Van Der Smissen
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| | - Nathalie Nevo
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Imagine Institute, Paris Descartes University, Paris, France
| | - Seppo Vainio
- Faculty of Biochemistry and Molecular Medicine, Laboratory of Developmental Biology, Oulu Center for Cell-Matrix Research, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Rikke Nielsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and
| | | | - François Jouret
- Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA), Cardiovascular Sciences, University of Liège, Liège, Belgium
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Diseases, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Imagine Institute, Paris Descartes University, Paris, France
| | - Christophe E Pierreux
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium;
| | - Pierre J Courtoy
- Cell Biology Unit, de Duve Institute and Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
20
|
Midgley J. A breakthrough in readthrough? Could geneticin lead the way to effective treatment for cystinosis nonsense mutations? Pediatr Nephrol 2019; 34:917-920. [PMID: 30623245 DOI: 10.1007/s00467-018-4173-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/04/2018] [Accepted: 12/10/2018] [Indexed: 10/27/2022]
Affiliation(s)
- Julian Midgley
- Alberta Children's Hospital, Section of Paediatric Nephrology, Department of Paediatrics, Cumming School of Medicine, University of Calgary, Calgary, Canada.
| |
Collapse
|
21
|
Battafarano G, Rossi M, Rega LR, Di Giovamberardino G, Pastore A, D'Agostini M, Porzio O, Nevo N, Emma F, Taranta A, Del Fattore A. Intrinsic Bone Defects in Cystinotic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1053-1064. [PMID: 30794806 DOI: 10.1016/j.ajpath.2019.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 01/09/2019] [Accepted: 01/24/2019] [Indexed: 01/22/2023]
Abstract
Cystinosis is a rare lysosomal storage disorder caused by loss-of-function mutations of the CTNS gene, encoding cystinosin, a symporter that mediates cystine efflux from lysosomes. Approximately 95% of patients with cystinosis display renal Fanconi syndrome, short stature, osteopenia, and rickets. In this study, we investigated whether the absence of cystinosin primarily affects bone remodeling activity, apart from the influences of the Fanconi syndrome on bone mineral metabolism. Using micro-computed tomography and histomorphometric and bone serum biomarker analysis, we evaluated the bone phenotype of 1-month-old Ctns-/- knockout (KO) male mice without tubulopathy. An in vitro study was performed to characterize the effects of cystinosin deficiency on osteoblasts and osteoclasts. Micro-computed tomography analysis showed a reduction of trabecular bone volume, bone mineral density, and number and thickness in KO mice compared with wild-type animals; histomorphometric analysis revealed a reduction of osteoblast and osteoclast parameters in tibiae of cystinotic mice. Decreased levels of serum procollagen type 1 amino-terminal propeptide and tartrate-resistant acid phosphatase in KO mice confirmed reduced bone remodeling activity. In vitro experiments showed an impairment of Ctns-/- osteoblasts and osteoclasts. In conclusion, cystinosin deficiency primarily affects bone cells, leading to a bone loss phenotype of KO mice, independent from renal failure.
Collapse
Affiliation(s)
- Giulia Battafarano
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy
| | - Michela Rossi
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy
| | - Laura R Rega
- Laboratories of Nephrology, Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Anna Pastore
- Molecular Genetics and Functional Genomics, Division of Genetic and Rare Diseases, Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Ottavia Porzio
- Clinical Laboratory, Bambino Gesù Children's Hospital, Rome, Italy; Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nathalie Nevo
- INSERM U1163, Université Paris Descartes, Institut Imagine, Hôpital Necker Enfants Malades, Paris, France
| | - Francesco Emma
- Laboratories of Nephrology, Bambino Gesù Children's Hospital, Rome, Italy; Division of Nephrology, Department of Pediatric Subspecialities, Bambino Gesù Children's Hospital, Rome, Italy
| | - Anna Taranta
- Laboratories of Nephrology, Bambino Gesù Children's Hospital, Rome, Italy.
| | - Andrea Del Fattore
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy
| |
Collapse
|
22
|
Kovach JL, Isildak H, Sarraf D. Crystalline retinopathy: Unifying pathogenic pathways of disease. Surv Ophthalmol 2019; 64:1-29. [DOI: 10.1016/j.survophthal.2018.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/01/2018] [Accepted: 08/06/2018] [Indexed: 01/02/2023]
|
23
|
Biswas S, Sornalingam K. The Ocular Status of Cystinosis Patients Receiving a Hospital Pharmacy-Made Preparation of Cysteamine Eye Drops: A Case Series. Ophthalmol Ther 2018; 8:125-136. [PMID: 30519924 PMCID: PMC6393247 DOI: 10.1007/s40123-018-0156-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Indexed: 12/01/2022] Open
Abstract
Introduction Infantile nephropathic cystinosis (INC) is an autosomal recessive lysosomal disorder in which patients develop deposits of cystine crystals in their kidneys and corneas from a young age. Methods We conducted a retrospective analysis of children with INC seen by ophthalmologists at the Manchester Royal Eye Hospital between 2002 and 2018, to evaluate clinical findings, symptoms and treatment. Results Twenty-two children diagnosed with INC from age 0 (prenatally) to 11 years were assessed. All evaluable patients had corneal cystine crystal deposits, and 15 had mild to moderate photophobia. Ten patients had other ocular conditions including blepharitis/chalzion (n = 6), swollen optic nerve (n = 3), punctate epitheliopathy (n = 3), corneal scarring (n = 1),and elevated intraocular pressure (n = 2). Confocal imaging identified nerve abnormalities in two patients (enlarged corneal nerve + abnormal-looking tortuous nerves in one patient and beaded nerves in the sub-basal plexus in the other), both of whom had significant crystal deposition in the anterior stroma. Visual acuity was relatively unaffected. All 22 patients were receiving oral cysteamine, and 21 were applying cysteamine eye drops (galenic preparation of 0.55% concentration, compounded by a hospital pharmacy). Recommended application frequency was at least eight times per day in all patients with dosing information available. Conclusions This case series of patients with INC highlights the consistent pattern of corneal cystine crystal deposition, which is universally present from a young age in this condition, and the high incidence of photophobia even in young children. Corneal manifestations of INC persisted despite frequent administration of the hospital pharmacy-made eye drop preparation. Reasons for this lack of efficacy may include the lag period between diagnosis and first prescription of cysteamine eye drops and the difficulty in maintaining rigorous compliance with this treatment. In addition, the challenge for patients of maintaining optimal storage conditions may adversely affect the stability and efficacy of cysteamine within this preparation. Funding Editorial assistance was funded by Orphan Europe Ltd.
Collapse
Affiliation(s)
- Susmito Biswas
- Manchester Royal Eye Hospital, Manchester Academic Health Science Centre, Manchester, UK.
| | - Krishanthy Sornalingam
- Manchester Royal Eye Hospital, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
24
|
Sumayao R, Newsholme P, McMorrow T. The Role of Cystinosin in the Intermediary Thiol Metabolism and Redox Homeostasis in Kidney Proximal Tubular Cells. Antioxidants (Basel) 2018; 7:antiox7120179. [PMID: 30513914 PMCID: PMC6315507 DOI: 10.3390/antiox7120179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 11/22/2018] [Accepted: 11/24/2018] [Indexed: 01/26/2023] Open
Abstract
Cystinosin is a lysosomal transmembrane protein which facilitates transport of the disulphide amino acid cystine (CySS) from the lysosomes of the cell. This protein is encoded by the CTNS gene which is defective in the lysosomal storage disorder, cystinosis. Because of the apparent involvement of cystinosin in the intermediary thiol metabolism, its discovery has fuelled investigations into its role in modulating cellular redox homeostasis. The kidney proximal tubular cells (PTCs) have become the focus of various studies on cystinosin since the protein is highly expressed in these cells and kidney proximal tubular transport dysfunction is the foremost clinical manifestation of cystinosis. The lysosomal CySS pool is a major source of cytosolic cysteine (Cys), the limiting amino acid for the synthesis of an important antioxidant glutathione (GSH) via the γ-glutamyl cycle. Therefore, loss of cystinosin function is presumed to lead to cytosolic deficit of Cys which may impair GSH synthesis. However, studies using in vitro models lacking cystinosin yielded inconsistent results and failed to establish the mechanistic role of cystinosin in modulating GSH synthesis and redox homeostasis. Because of the complexity of the metabolic micro- and macro-environment in vivo, using in vitro models alone may not be able to capture the complete sequence of biochemical and physiological events that occur as a consequence of loss of cystinosin function. The coexistence of pathways for the overall handling and disposition of GSH, the modulation of CTNS gene by intracellular redox status and the existence of a non-canonical isoform of cystinosin may constitute possible rescue mechanisms in vivo to remediate redox perturbations in renal PTCs. Importantly, the mitochondria seem to play a critical role in orchestrating redox imbalances initiated by cystinosin dysfunction. Non-invasive techniques such as in vivo magnetic resonance imaging with the aid of systems biology approaches may provide invaluable mechanistic insights into the role of cystinosin in the essential intermediary thiol metabolism and in the overall regulation cellular redox homeostasis.
Collapse
Affiliation(s)
- Rodolfo Sumayao
- Chemistry Department, De La Salle University, Manila 1004, Philippines.
| | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences and Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth 6845, Australia.
| | - Tara McMorrow
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
25
|
Autonomic Nervous System Dysregulation in Monozygous Twins With Nephropathic Cystinosis. Kidney Int Rep 2018; 3:1489-1496. [PMID: 30450477 PMCID: PMC6224624 DOI: 10.1016/j.ekir.2018.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
26
|
Bretaudeau C, Paredes FS, Sergheraert J, Massa L, Laurence S, Diebold MH, Lefèvre B, Mauprivez C. Dental implant placement in a patient with cystinosis. A case report. JOURNAL OF ORAL MEDICINE AND ORAL SURGERY 2018. [DOI: 10.1051/mbcb/2018019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Introduction: Cystinosis is a rare genetic disease due to a defective transport of cystine out of the lysosomes, caused by a mutation of the gene encoding for the lysosomal carrier protein, the cystinosin. Cystine accumulation results in the formation of intracellular cystine cristals, that causes tissular and multi-organic lesions (kidney, eyes, endocrine glands). Observation: We report a rare case of a patient affected by infantile nephropathic cystinosis, who consulted for an implant placement in a single-tooth gap. Discussion: Accumulation of cystine leads to tissue damage, primarily in the kidney, the liver and the cornea, but other organs, such as the mouth, teeth and jaws may be also involved. The article aimed to present oral manifestations associated with this storage disease and to discuss how oral surgeon can evaluate and manage these patients despite the lack of a standardized protocol.
Collapse
|
27
|
Bellomo F, Signorile A, Tamma G, Ranieri M, Emma F, De Rasmo D. Impact of atypical mitochondrial cyclic-AMP level in nephropathic cystinosis. Cell Mol Life Sci 2018; 75:3411-3422. [PMID: 29549422 PMCID: PMC11105431 DOI: 10.1007/s00018-018-2800-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/20/2018] [Accepted: 03/14/2018] [Indexed: 02/06/2023]
Abstract
Nephropathic cystinosis (NC) is a rare disease caused by mutations in the CTNS gene encoding for cystinosin, a lysosomal transmembrane cystine/H+ symporter, which promotes the efflux of cystine from lysosomes to cytosol. NC is the most frequent cause of Fanconi syndrome (FS) in young children, the molecular basis of which is not well established. Proximal tubular cells have very high metabolic rate due to the active transport of many solutes. Not surprisingly, mitochondrial disorders are often characterized by FS. A similar mechanism may also apply to NC. Because cAMP has regulatory properties on mitochondrial function, we have analyzed cAMP levels and mitochondrial targets in CTNS-/- conditionally immortalized proximal tubular epithelial cells (ciPTEC) carrying the classical homozygous 57-kb deletion (delCTNS-/-) or with compound heterozygous loss-of-function mutations (mutCTNS-/-). Compared to wild-type cells, cystinotic cells had significantly lower mitochondrial cAMP levels (delCTNS-/- ciPTEC by 56% ± 10.5, P < 0.0001; mutCTNS-/- by 26% ± 4.3, P < 0.001), complex I and V activities, mitochondrial membrane potential, and SIRT3 protein levels, which were associated with increased mitochondrial fragmentation. Reduction of complex I and V activities was associated with lower expression of part of their subunits. Treatment with the non-hydrolysable cAMP analog 8-Br-cAMP restored mitochondrial potential and corrected mitochondria morphology. Treatment with cysteamine, which reduces the intra-lysosomal cystine, was able to restore mitochondrial cAMP levels, as well as most other abnormal mitochondrial findings. These observations were validated in CTNS-silenced HK-2 cells, indicating a pivotal role of mitochondrial cAMP in the proximal tubular dysfunction observed in NC.
Collapse
Affiliation(s)
- Francesco Bellomo
- Laboratory of Nephrology, Department of Rare Diseases, Bambino Gesù Children's Hospital, Viale di S. Paolo, 15, 00149, Rome, Italy.
| | - Anna Signorile
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy
| | - Grazia Tamma
- Department of Bioscience, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Marianna Ranieri
- Department of Bioscience, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", Bari, Italy
| | - Francesco Emma
- Laboratory of Nephrology, Department of Rare Diseases, Bambino Gesù Children's Hospital, Viale di S. Paolo, 15, 00149, Rome, Italy
- Division of Nephrology, Department of Pediatric Subspecialties, Bambino Gesù Children's Hospital, Rome, Italy
| | - Domenico De Rasmo
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Policlinico, Piazza G. Cesare, 11, 70124, Bari, Italy.
- Institute of Biomembrane, Bioenergetics and Molecular Biotechnology (IBIOM), National Research Council (CNR), Bari, Italy.
| |
Collapse
|
28
|
Sumayao R, Newsholme P, McMorrow T. Inducible nitric oxide synthase inhibitor 1400W increases Na + ,K + -ATPase levels and activity and ameliorates mitochondrial dysfunction in Ctns null kidney proximal tubular epithelial cells. Clin Exp Pharmacol Physiol 2018; 45:1149-1160. [PMID: 29924417 DOI: 10.1111/1440-1681.12998] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 06/02/2018] [Accepted: 06/11/2018] [Indexed: 02/02/2023]
Abstract
Nitric oxide (NO) has been shown to play an important role in renal physiology and pathophysiology partly through its influence on various transport systems in the kidney proximal tubule. The role of NO in kidney dysfunction associated with lysosomal storage disorder, cystinosis, is largely unknown. In the present study, the effects of inducible nitric oxide synthase (iNOS)-specific inhibitor, 1400W, on Na+ ,K+ -ATPase activity and expression, mitochondrial integrity and function, nutrient metabolism, and apoptosis were investigated in Ctns null proximal tubular epithelial cells (PTECs). Ctns null PTECs exhibited an increase in iNOS expression, augmented NO and nitrite/nitrate production, and reduced Na+ ,K+ -ATPase expression and activity. In addition, these cells displayed depolarized mitochondria, reduced adenosine triphosphate content, altered nutrient metabolism, and elevated apoptosis. Treatment of Ctns null PTECs with 1400W abolished these effects which culminated in the mitigation of apoptosis in these cells. These findings indicate that uncontrolled NO production may constitute the upstream event that leads to the molecular and biochemical alterations observed in Ctns null PTECs and may explain, at least in part, the generalized proximal tubular dysfunction associated with cystinosis. Further studies are needed to realize the potential benefits of anti-nitrosative therapies in improving renal function and/or attenuating renal injury in cystinosis.
Collapse
Affiliation(s)
- Rodolfo Sumayao
- Chemistry Department, De La Salle University, Manila, Philippines
| | - Philip Newsholme
- School of Biomedical Sciences, Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, Western Australia, Australia
| | - Tara McMorrow
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
29
|
Makuloluwa AK, Shams F. Cysteamine hydrochloride eye drop solution for the treatment of corneal cystine crystal deposits in patients with cystinosis: an evidence-based review. Clin Ophthalmol 2018; 12:227-236. [PMID: 29416314 PMCID: PMC5789046 DOI: 10.2147/opth.s133516] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cystinosis is a rare, autosomal recessive disorder leading to defective transport of cystine out of lysosomes. Subsequent cystine crystal accumulation can occur in various tissues, including the ocular surface. This review explores the efficacy of cysteamine hydrochloride eye drops in the treatment of corneal cystine crystal accumulation and its safety profile.
Collapse
Affiliation(s)
| | - Fatemeh Shams
- Tennent Institute of Ophthalmology, Gartnavel General Hospital, Glasgow, UK
| |
Collapse
|
30
|
Bäumner S, Weber LT. Nephropathic Cystinosis: Symptoms, Treatment, and Perspectives of a Systemic Disease. Front Pediatr 2018; 6:58. [PMID: 29594088 PMCID: PMC5861330 DOI: 10.3389/fped.2018.00058] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 02/27/2018] [Indexed: 11/13/2022] Open
Abstract
Cystinosis is a rare autosomal recessive lysosomal storage disorder caused by mutations in the CTNS gene. Main dysfunction is a defective clearance of cystine from lysosomes that leads to accumulation of cystine crystals in every tissue of the body. There are three different forms: infantile nephropathic cystinosis, which is the most common form, juvenile nephropatic, and non-nephropathic cystinosis. Mostly, first symptom in infantile nephropathic cystinosis is renal Fanconi syndrome that occurs within the first year of life. Another prominent symptom is photophobia due to corneal crystal deposition. Cystine depletion therapy with cysteamine delays end-stage renal failure but does not stop progression of the disease. A new cysteamine formulation with delayed-release simplifies the administration schedule but still does not cure cystinosis. Even long-term depletion treatment resulting in bypassing the defective lysosomal transporter cannot reverse Fanconi syndrome. A future perspective offering a curative therapy may be transplantation of CTNS-carrying stem cells that has successfully been performed in mice.
Collapse
Affiliation(s)
- Sören Bäumner
- Pediatric Nephrology, Children's and Adolescents' Hospital, University Hospital Cologne, Cologne, Germany
| | - Lutz T Weber
- Pediatric Nephrology, Children's and Adolescents' Hospital, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
31
|
Wang J, Wen Y, Zhou M, Shi X, Jiang L, Li M, Yu Y, Li X, Li X, Zhang W, Lundquist AL, Chen L. Ectopic germinal center and megalin defect in primary Sjogren syndrome with renal Fanconi syndrome. Arthritis Res Ther 2017; 19:120. [PMID: 28577559 PMCID: PMC5455124 DOI: 10.1186/s13075-017-1317-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 05/02/2017] [Indexed: 01/15/2023] Open
Abstract
Background This study reports the clinical and pathological features of 12 cases of primary Sjogren syndrome (pSS) with renal involvement presenting with proximal tubular dysfunction in a single center, and investigates the possible correlation of ectopic germinal center formation and megalin/cubilin down-expression. Method Clinical and pathological records were reviewed. Immunohistochemistry was carried out to detect megalin, cubilin, CD21 and IL-17 expression. Results Patients presented with different degrees of proximal renal tubule lesion and decreased estimated glomerular filtration rate (eGFR). Renal biopsy revealed tubulointerstitial nephritis, with tubular epithelial cell degeneration, tubular atrophy, interstitial inflammation and focal fibrosis. Immunohistochemistry revealed decreased expression of megalin and cubilin, two important multiligand protein receptors on the brush border of proximal tubular epithelial cells. IL-17 secreted by Th17 subtype effector T cells was diffusely detected in the renal proximal tubule, with a negative correlation of IL-17 and megalin expression. In addition, ectopic germinal centers characterized by CD21+ follicular dendritic cells were present in the renal interstitium. In patients with a decreased eGFR, treatment with 4 weeks of glucocorticoid therapy resulted in an improved eGFR in 75% of patients. Conclusion We report 12 cases of pSS characterized by Fanconi syndrome. The decreased megalin and cubilin expression may contribute to the proximal tubular reabsorption defect, possibly secondary to Th17 infiltration and formation of ectopic germinal centers.
Collapse
Affiliation(s)
- Jing Wang
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Yubing Wen
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Mengyu Zhou
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Xiaoxiao Shi
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Lanping Jiang
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Mingxi Li
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Yang Yu
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China. .,Department of Nephrology, Chinese Academy of Medical Science, Peking Union Medical College Hospital, No 1, Shuaifuyan, Wangfujing St, Beijing, 100730, China.
| | - Xuemei Li
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Xuewang Li
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Wen Zhang
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China
| | - Andrew L Lundquist
- Division of Nephrology, Massachussetts General Hospital, Boston, MA, USA
| | - Limeng Chen
- Nephrology Department, Chinese Academy of Medical Science, Peking Union Medical College Hospital, Tsing Hua University, Beijing, China.
| |
Collapse
|
32
|
Zhang J, Johnson JL, He J, Napolitano G, Ramadass M, Rocca C, Kiosses WB, Bucci C, Xin Q, Gavathiotis E, Cuervo AM, Cherqui S, Catz SD. Cystinosin, the small GTPase Rab11, and the Rab7 effector RILP regulate intracellular trafficking of the chaperone-mediated autophagy receptor LAMP2A. J Biol Chem 2017; 292:10328-10346. [PMID: 28465352 DOI: 10.1074/jbc.m116.764076] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Revised: 04/28/2017] [Indexed: 12/12/2022] Open
Abstract
The lysosomal storage disease cystinosis, caused by cystinosin deficiency, is characterized by cell malfunction, tissue failure, and progressive renal injury despite cystine-depletion therapies. Cystinosis is associated with defects in chaperone-mediated autophagy (CMA), but the molecular mechanisms are incompletely understood. Here, we show CMA substrate accumulation in cystinotic kidney proximal tubule cells. We also found mislocalization of the CMA lysosomal receptor LAMP2A and impaired substrate translocation into the lysosome caused by defective CMA in cystinosis. The impaired LAMP2A trafficking and localization were rescued either by the expression of wild-type cystinosin or by the disease-associated point mutant CTNS-K280R, which has no cystine transporter activity. Defective LAMP2A trafficking in cystinosis was found to associate with decreased expression of the small GTPase Rab11 and the Rab7 effector RILP. Defective Rab11 trafficking in cystinosis was rescued by treatment with small-molecule CMA activators. RILP expression was restored by up-regulation of the transcription factor EB (TFEB), which was down-regulated in cystinosis. Although LAMP2A expression is independent of TFEB, TFEB up-regulation corrected lysosome distribution and lysosomal LAMP2A localization in Ctns-/- cells but not Rab11 defects. The up-regulation of Rab11, Rab7, or RILP, but not its truncated form RILP-C33, rescued LAMP2A-defective trafficking in cystinosis, whereas dominant-negative Rab11 or Rab7 impaired LAMP2A trafficking. Treatment of cystinotic cells with a CMA activator increased LAMP2A localization at the lysosome and increased cell survival. Altogether, we show that LAMP2A trafficking is regulated by cystinosin, Rab11, and RILP and that CMA up-regulation is a potential clinically relevant mechanism to increase cell survival in cystinosis.
Collapse
Affiliation(s)
- Jinzhong Zhang
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Jennifer L Johnson
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Jing He
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Gennaro Napolitano
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Mahalakshmi Ramadass
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Celine Rocca
- the Department of Pediatrics, Division of Genetics, University of California, San Diego, La Jolla, California 92093-0734
| | - William B Kiosses
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Cecilia Bucci
- the Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Provinciale Monteroni n. 165, 73100 Lecce, Italy, and
| | - Qisheng Xin
- the Departments of Biochemistry and.,Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | | | - Ana María Cuervo
- Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York 10461
| | - Stephanie Cherqui
- the Department of Pediatrics, Division of Genetics, University of California, San Diego, La Jolla, California 92093-0734
| | - Sergio D Catz
- From the Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037,
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW Over the past few decades, cystinosis, a rare lysosomal storage disorder, has evolved into a treatable metabolic disease. The increasing understanding of its pathophysiology has made cystinosis a prototype disease, delivering new insights into several fundamental biochemical and cellular processes. RECENT FINDINGS In this review, we aim to provide an overview of the latest advances in the pathogenetic, clinical, and therapeutic aspects of cystinosis. SUMMARY The development of alternative therapeutic monitoring strategies and new systemic and ocular cysteamine formulations might improve outcome of cystinosis patients in the near future. With the dawn of stem cell based therapy and new emerging gene-editing technologies, novel tools have become available in the search for a cure for cystinosis.
Collapse
|
34
|
Cystinosin-LKG rescues cystine accumulation and decreases apoptosis rate in cystinotic proximal tubular epithelial cells. Pediatr Res 2017; 81:113-119. [PMID: 27656773 DOI: 10.1038/pr.2016.184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/10/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Nephropathic cystinosis is a lysosomal storage disease that is caused by mutations in the CTNS gene encoding a cystine/proton symporter cystinosin and an isoform cystinosin-LKG which is generated by an alternative splicing of exon 12. We have investigated the physiological role of the cystinosin-LKG that is widely expressed in epithelial tissues. METHODS We have analyzed the intracellular localization and the function of the cystinosin-LKG conjugated with DsRed (cystinosin-LKG-RFP) in Madin-Darby canine kidney cells (MDCK II) and in proximal tubular epithelial cells carrying a deletion of the CTNS gene (cystinotic PTEC), respectively. RESULTS Cystinosin-LKG-RFP colocalized with markers of lysosomes, late endosomes and was also expressed on the apical surface of polarized MDCK II cells. Moreover, immune-electron microscopy images of MDCK II cells overexpressing cystinosin-LKG-RFP showed stacked lamellar membranes inside perinuclear lysosomal structures. To study the role of LKG-isoform, we have investigated cystine accumulation and apoptosis that have been described in cystinotic cells. Cystinosin-LKG decreased cystine levels by approximately 10-fold similarly to cystinosin-RFP. The levels of TNFα- and actinomycin D-inducted apoptosis dropped in cystinotic cells expressing LKG-isoform. This effect was also similar to the main isoform. CONCLUSION Our results suggest that cystinosin-LKG and cystinosin move similar functional activities in cells.
Collapse
|
35
|
Vu CB, Bridges RJ, Pena-Rasgado C, Lacerda AE, Bordwell C, Sewell A, Nichols AJ, Chandran S, Lonkar P, Picarella D, Ting A, Wensley A, Yeager M, Liu F. Fatty Acid Cysteamine Conjugates as Novel and Potent Autophagy Activators That Enhance the Correction of Misfolded F508del-Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). J Med Chem 2016; 60:458-473. [PMID: 27976892 DOI: 10.1021/acs.jmedchem.6b01539] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A depressed autophagy has previously been reported in cystic fibrosis patients with the common F508del-CFTR mutation. This report describes the synthesis and preliminary biological characterization of a novel series of autophagy activators involving fatty acid cysteamine conjugates. These molecular entities were synthesized by first covalently linking cysteamine to docosahexaenoic acid. The resulting conjugate 1 synergistically activated autophagy in primary homozygous F508del-CFTR human bronchial epithelial (hBE) cells at submicromolar concentrations. When conjugate 1 was used in combination with the corrector lumacaftor and the potentiator ivacaftor, it showed an additive effect, as measured by the increase in the chloride current in a functional assay. In order to obtain a more stable form for oral dosing, the sulfhydryl group in conjugate 1 was converted into a functionalized disulfide moiety. The resulting conjugate 5 is orally bioavailable in the mouse, rat, and dog and allows a sustained delivery of the biologically active conjugate 1.
Collapse
Affiliation(s)
- Chi B Vu
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Robert J Bridges
- Chicago Medical School, Rosalind Franklin University of Medicine and Science , 3333 Green Bay Road, North Chicago, Illinois 60064, United States
| | - Cecilia Pena-Rasgado
- Chicago Medical School, Rosalind Franklin University of Medicine and Science , 3333 Green Bay Road, North Chicago, Illinois 60064, United States
| | - Antonio E Lacerda
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Curtis Bordwell
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Abby Sewell
- Charles River Laboratories , 14656 Neo Parkway, Cleveland, Ohio 44128, United States
| | - Andrew J Nichols
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Sachin Chandran
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Pallavi Lonkar
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Dominic Picarella
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Amal Ting
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Allison Wensley
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Maisy Yeager
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| | - Feng Liu
- Catabasis Pharmaceuticals , One Kendall Square, Suite B14202, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
36
|
Ramazani Y, Levtchenko EN, Van Den Heuvel L, Van Schepdael A, Paul P, Ivanova EA, Pastore A, Hartman TM, Price NPJ. Evaluation of carbohydrate-cysteamine thiazolidines as pro-drugs for the treatment of cystinosis. Carbohydr Res 2016; 439:9-15. [PMID: 28033491 DOI: 10.1016/j.carres.2016.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 10/20/2022]
Abstract
Cystinosis is a genetic disorder caused by malfunction of cystinosin and is characterized by accumulation of cystine. Cysteamine, the medication used in cystinosis, causes halitosis resulting in poor patient compliance. Halitosis is mainly caused by the formation of dimethylsulfide as the final product in the cysteamine metabolism pathway. We have synthesized carbohydrate-cysteamine thiazolidines, and hypothesized that the hydrolytic breakdown of cysteamine-thiazolidines can result in free cysteamine being released in target organs. To examine our hypothesis, we tested these analogs in vitro in patient-derived fibroblasts. Cystinotic fibroblasts were treated with different concentrations of arabinose-cysteamine, glucose-cysteamine and maltose-cysteamine. We demonstrated that the analogs break down into cysteamine extracellularly and might therefore not be fully taken up by the cells under the form of the pro-drug. Potential modifications of the analogs that enable their intracellular rather than extracellular breakdown, is necessary to pursue the potential of these analogs as pro-drugs.
Collapse
Affiliation(s)
- Yasaman Ramazani
- Department of Pediatric Nephrology and Growth and Regeneration, University Hospitals Leuven and University of Leuven, UZ Herestraat 49, Box 817, 3000 Leuven, Belgium
| | - Elena N Levtchenko
- Department of Pediatric Nephrology and Growth and Regeneration, University Hospitals Leuven and University of Leuven, UZ Herestraat 49, Box 817, 3000 Leuven, Belgium
| | - Lambertus Van Den Heuvel
- Department of Pediatric Nephrology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Ann Van Schepdael
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, University of Leuven, O&N II Herestraat 49, Box 923, 3000 Leuven, Belgium
| | - Prasanta Paul
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, University of Leuven, O&N II Herestraat 49, Box 923, 3000 Leuven, Belgium
| | - Ekaterina A Ivanova
- Department of Pediatric Nephrology and Growth and Regeneration, University Hospitals Leuven and University of Leuven, UZ Herestraat 49, Box 817, 3000 Leuven, Belgium
| | - Anna Pastore
- Laboratory of Metabolomics and Proteomics Bambino Gesu Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Trina M Hartman
- Agricultural Research Service, U.S. Department of Agriculture, National Center for Agricultural Utilization Research, Peoria, IL 61604, USA
| | - Neil P J Price
- Agricultural Research Service, U.S. Department of Agriculture, National Center for Agricultural Utilization Research, Peoria, IL 61604, USA.
| |
Collapse
|
37
|
Abstract
Cells lining the proximal tubule (PT) of the kidney are highly specialized for apical endocytosis of filtered proteins and small bioactive molecules from the glomerular ultrafiltrate to maintain essentially protein-free urine. Compromise of this pathway results in low molecular weight (LMW) proteinuria that can progress to end-stage kidney disease. This review describes our current understanding of the endocytic pathway and the multiligand receptors that mediate LMW protein uptake in PT cells, how these are regulated in response to physiologic cues, and the molecular basis of inherited diseases characterized by LMW proteinuria.
Collapse
Affiliation(s)
- Megan L Eshbach
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; ,
| | - Ora A Weisz
- Renal-Electrolyte Division, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; ,
| |
Collapse
|
38
|
Buntinx L, Voets T, Morlion B, Vangeel L, Janssen M, Cornelissen E, Vriens J, de Hoon J, Levtchenko E. TRPV1 dysfunction in cystinosis patients harboring the homozygous 57 kb deletion. Sci Rep 2016; 6:35395. [PMID: 27734949 PMCID: PMC5062165 DOI: 10.1038/srep35395] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 09/27/2016] [Indexed: 11/12/2022] Open
Abstract
Cystinosis is a rare autosomal recessive disorder characterized by lysosomal cystine accumulation due to loss of function of the lysosomal cystine transporter (CTNS). The most common mutation in cystinosis patients of Northern Europe consists of a 57-kb deletion. This deletion not only inactivates the CTNS gene but also extends into the non-coding region upstream of the start codon of the TRPV1 gene, encoding the capsaicin- and heat-sensitive ion channel TRPV1. To evaluate the consequences of the 57-kb deletion on functional TRPV1 expression, we compared thermal, mechanical and chemical sensitivity of cystinosis patients with matched healthy controls. Whereas patients heterozygous for the 57-kb deletion showed normal sensory responses, homozygous subjects exhibited a 60% reduction in vasodilation and pain evoked by capsaicin, as well as an increase in heat detection threshold. Responses to cold, mechanical stimuli or cinnamaldehyde, an agonist of the related nociceptor channel TRPA1, were unaltered. We conclude that cystinosis patients homozygous for the 57-kb deletion exhibit a strong reduction of TRPV1 function, leading to sensory deficiencies akin to the phenotype of TRPV1-deficient mice. These deficits may account for the reported sensory alterations and thermoregulatory deficits in these patients, and provide a paradigm for life-long TRPV1 deficiency in humans.
Collapse
Affiliation(s)
- L Buntinx
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - T Voets
- Department of Cellular and Molecular Medicine, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - B Morlion
- Center for algology and pain management, Department of Cardiovascular Sciences, KULeuven, Weligerveld 1, 3212 Pellenberg, Belgium
| | - L Vangeel
- Department of Cellular and Molecular Medicine, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - M Janssen
- Department of internal medicine, Radboud UMC Nijmegen, Geert Grooteplein-Zuid 22, 6525 GA Nijmegen, The Netherlands
| | - E Cornelissen
- Department of Pediatric Nephrology, Radboud UMC Nijmegen, Geert Grooteplein-Zuid 22, 6525 GA Nijmegen, The Netherlands
| | - J Vriens
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - J de Hoon
- Center for Clinical Pharmacology, Department of Pharmaceutical and Pharmacological Sciences, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| | - E Levtchenko
- Department of Development and Regeneration, KULeuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
39
|
Doğan M, Bulan K, Kaba S, Cesur Y, Ceylaner S, Ustyol L. Cystinosis in Eastern Turkey. J Pediatr Endocrinol Metab 2016; 29:965-9. [PMID: 27269891 DOI: 10.1515/jpem-2014-0477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/21/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND This study was conducted to investigate CTNS (cystinosin, lysosomal cystine transporter) gene mutations and the clinical spectrum of nephropathic cystinosis among patients diagnosed with the disease in a single center in Turkey. METHODS Patients' clinical and laboratory data were extracted from an electronic health registry. Molecular CTNS gene analysis was performed using either next-generation sequencing or Sanger sequencing. RESULTS Eleven patients (age range: 1.5-12 years) from nine families were identified. The presenting complaint was growth retardation in seven patients; polydipsia and polyuria in three patients; and vomiting in two patients. At presentation, electrolyte loss was noted in all patients, of which eight patients presented with metabolic acidosis, and three patients presented with metabolic alkalosis. All patients also presented with proteinuria and glucosuria, and four patients developed varying degrees of renal insufficiency, for which peritoneal dialysis was initiated in one patient. Cystine crystals were detected via ocular examination in one patient at presentation. No cystine crystals were detected among patients who underwent bone marrow aspiration. In the CTNS gene, a p.T7FX7 (c.18-21del4bp) mutation was detected in three patients, whereas a p.E227E (c.681 G>A) (homozygous) mutation was detected in eight patients. CONCLUSIONS We detected two distinct mutations, p.T7FX7 (c.18-21del4bp) and p.E227E (c.681 G>A) (homozygous), in the CTNS gene in 11 patients with cystinosis from the East Anatolian region of Turkey. Patients with a homozygous c.681 G>A (p.E227E) mutation are more likely to develop chronic renal failure and should be monitored closely, whereas patients with a p.T7FX7 (c.18-21del4bp) mutation have a milder phenotype. Additionally, metabolic alkalosis does not exclude cystinosis, although cystinosis is a cause of proximal renal tubular acidosis.
Collapse
|
40
|
Nieskens TTG, Wilmer MJ. Kidney-on-a-chip technology for renal proximal tubule tissue reconstruction. Eur J Pharmacol 2016; 790:46-56. [PMID: 27401035 DOI: 10.1016/j.ejphar.2016.07.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/01/2016] [Accepted: 07/08/2016] [Indexed: 12/11/2022]
Abstract
The renal proximal tubule epithelium is responsible for active secretion of endogenous and exogenous waste products from the body and simultaneous reabsorption of vital compounds from the glomerular filtrate. The complexity of this transport machinery makes investigation of processes such as tubular drug secretion a continuous challenge for researchers. Currently available renal cell culture models often lack sufficient physiological relevance and reliability. Introducing complex biological culture systems in a 3D microfluidic design improves the physiological relevance of in vitro renal proximal tubule epithelium models. Organ-on-a-chip technology provides a promising alternative, as it allows the reconstruction of a renal tubule structure. These microfluidic systems mimic the in vivo microenvironment including multi-compartmentalization and exposure to fluid shear stress. Increasing data supports that fluid shear stress impacts the phenotype and functionality of proximal tubule cultures, for which we provide an extensive background. In this review, we discuss recent developments of kidney-on-a-chip platforms with current and future applications. The improved proximal tubule functionality using 3D microfluidic systems is placed in perspective of investigating cellular signalling that can elucidate mechanistic aberrations involved in drug-induced kidney toxicity.
Collapse
Affiliation(s)
- Tom T G Nieskens
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
41
|
Controversies and research agenda in nephropathic cystinosis: conclusions from a “Kidney Disease: Improving Global Outcomes” (KDIGO) Controversies Conference. Kidney Int 2016; 89:1192-203. [DOI: 10.1016/j.kint.2016.01.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 01/22/2016] [Accepted: 01/28/2016] [Indexed: 01/19/2023]
|
42
|
Ivanova EA, van den Heuvel LP, Elmonem MA, De Smedt H, Missiaen L, Pastore A, Mekahli D, Bultynck G, Levtchenko EN. Altered mTOR signalling in nephropathic cystinosis. J Inherit Metab Dis 2016; 39:457-464. [PMID: 26909499 DOI: 10.1007/s10545-016-9919-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/22/2016] [Accepted: 02/02/2016] [Indexed: 11/27/2022]
Abstract
Lysosomes play a central role in regulating autophagy via activation of mammalian target of rapamycin complex 1 (mTORC1). We examined mTORC1 signalling in the lysosomal storage disease nephropathic cystinosis (MIM 219800), in which accumulation of autophagy markers has been previously demonstrated. Cystinosis is caused by mutations in the lysosomal cystine transporter cystinosin and initially affects kidney proximal tubules causing renal Fanconi syndrome, followed by a gradual development of end-stage renal disease and extrarenal complications. Using proximal tubular kidney cells obtained from healthy donors and from cystinotic patients, we demonstrate that cystinosin deficiency is associated with a perturbed mTORC1 signalling, delayed reactivation of mTORC1 after starvation and abnormal lysosomal retention of mTOR during starvation. These effects could not be reversed by treatment with cystine-depleting drug cysteamine. Altered mTORC1 signalling can contribute to the development of proximal tubular dysfunction in cystinosis and points to new possibilities in therapeutic intervention through modulation of mTORC-dependent signalling cascades.
Collapse
Affiliation(s)
- Ekaterina A Ivanova
- Department of Growth and Regeneration, KU Leuven and University Hospitals Leuven, UZ Herestraat 49, 3000, Leuven, Belgium
| | - Lambertus P van den Heuvel
- Department of Growth and Regeneration, KU Leuven and University Hospitals Leuven, UZ Herestraat 49, 3000, Leuven, Belgium
- Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mohamed A Elmonem
- Department of Growth and Regeneration, KU Leuven and University Hospitals Leuven, UZ Herestraat 49, 3000, Leuven, Belgium
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Humbert De Smedt
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ludwig Missiaen
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Anna Pastore
- Laboratory of Proteomics and Metabolomics, Children's Hospital and Research Institute "Bambino Gesù" IRCCS, Rome, Italy
| | - Djalila Mekahli
- Department of Growth and Regeneration, KU Leuven and University Hospitals Leuven, UZ Herestraat 49, 3000, Leuven, Belgium
| | - Greet Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Elena N Levtchenko
- Department of Growth and Regeneration, KU Leuven and University Hospitals Leuven, UZ Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
43
|
Cheung WW, Cherqui S, Ding W, Esparza M, Zhou P, Shao J, Lieber RL, Mak RH. Muscle wasting and adipose tissue browning in infantile nephropathic cystinosis. J Cachexia Sarcopenia Muscle 2016; 7:152-64. [PMID: 27493869 PMCID: PMC4864942 DOI: 10.1002/jcsm.12056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 04/21/2015] [Accepted: 06/03/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Muscle wasting is a common complication in patients with infantile nephropathic cystinosis, but its mechanism and association with energy metabolism is not known. We define the metabolic phenotype in Ctns(-/-) mice, an established murine model of infantile nephropathic cystinosis, with focus on muscle wasting and energy homeostasis. METHODS Male Ctns(-/-) mice and wild-type (WT) controls were studied at 1, 4, 9, and 12 months of age. As Ctns(-/-) mice started to develop chronic kidney disease (CKD) at 9 months of age, 9- and 12-month-old Ctns(-/-) mice were also compared with age-matched WT mice with CKD. Serum and urine chemistry and energy homeostasis parameters were measured. Skeletal muscle histomorphometry and in vivo muscle function were measured. We studied expression of genes involved in muscle mass regulation, thermogenesis, energy metabolism, adipogenesis, and adipose tissue browning in Ctns(-/-) mice. RESULTS Ctns(-/-) mice showed loss of weight and lean mass and increased energy expenditure. Ctns(-/-) mice exhibited abnormal energy homeostasis before the onset of their CKD. Food intake in Ctns(-/-) mice was comparable with age-matched WT controls. However, significantly lower total body mass starting at 1 month of age and increased energy expenditure at 4 months of age preceded the onset of CKD at 9 months of age in Ctns(-/-) mice. Muscle accept content in 1- and 4-month-old Ctns(-/-) mice was significantly lower than that in age-matched WT controls. At 12 months of age, muscle fibre area and in vivo muscle strength was reduced in Ctns(-/-) mice than that in WT or CKD controls. Muscle wasting in Ctns(-/-) mice was associated with inhibition of myogenesis, activation of muscle proteolysis pathways, and overexpression of pro-inflammatory cytokines. Increased energy expenditure was associated with elevation of thermogenesis in skeletal muscle and adipose tissues. The development of beige adipocytes in Ctns(-/-) mice is a novel finding. Expression of beige adipose cell surface markers (CD137, Tmem26, and Tbx1) and uncoupling protein-1, which is a brown adipose tissue marker, was observed in inguinal white adipose tissue of Ctns(-/-) mice. Expression of key molecules implicated in the pathogenesis of adipose tissue browning (Cox2, cytochrome c oxidase subunit II; PGF2α, prostaglandin F2α; IL-1α, interleukin 1α; IL-6, interleukin 6; TNF-α, tumor necrosis factor α) was significantly increased in inguinal white adipose tissue of Ctns(-/-) mice than that in WT controls. CONCLUSION This study describes a mouse model of nephropathic cystinosis presenting with profound muscle wasting. The mechanism for hypermetabolism in Ctns(-/-) mice may involve up-regulation of thermogenesis pathways in skeletal muscle and adipose tissues. This study demonstrates, for the first time, the development of beige adipocytes in Ctns(-/-) mice. Understanding the underlying mechanisms of adipose tissue browning in cystinosis may lead to novel therapy.
Collapse
Affiliation(s)
- Wai W Cheung
- Department of Pediatrics University of California San Diego CA USA
| | | | - Wei Ding
- Department of Pediatrics University of California San Diego CA USA; Division of Nephrology, The 5th People's Hospital of Shanghai Fudan University Shanghai China
| | - Mary Esparza
- Department of Orthopedic Surgery University of California San Diego CA USA
| | - Ping Zhou
- Department of Pediatrics University of California San Diego CA USA; Department of Pediatrics The 2nd Hospital of Harbin Medical University Harbin China
| | - Jianhua Shao
- Department of Pediatrics University of California San Diego CA USA
| | - Richard L Lieber
- Department of Orthopedic Surgery University of California San Diego CA USA; Rehabilitation Institute of Chicago Chicago
| | - Robert H Mak
- Department of Pediatrics University of California San Diego CA USA
| |
Collapse
|
44
|
Sumayao R, McEvoy B, Newsholme P, McMorrow T. Lysosomal cystine accumulation promotes mitochondrial depolarization and induction of redox-sensitive genes in human kidney proximal tubular cells. J Physiol 2016; 594:3353-70. [PMID: 26915455 DOI: 10.1113/jp271858] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 02/02/2016] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Cystine is a disulphide amino acid that is normally generated in the lysosomes by the breakdown of cystine-containing proteins. Previously, we demonstrated that lysosomal cystine accumulation in kidney proximal tubular epithelial cells (PTECs) dramatically reduced glutathione (GSH) levels, which may result in the disruption of cellular redox balance. In the present study, we show that lysosomal cystine accumulation following CTNS gene silencing in kidney PTECs resulted in elevated intracellular reactive oxygen species production, reduced antioxidant capacity, induction of redox-sensitive proteins, altered mitochondrial integrity and augmented cell death. These alterations may represent different facets of a unique cascade leading to tubular dysfunction initiated by lysosomal cystine accumulation and may present a clear disadvantage for cystinotic PTECs in vivo. Cystine depletion by cysteamine afforded cytoprotection in CTNS knockdown cells by reducing oxidative stress, normalizing intracellular GSH and ATP content, and preserving cell viability. ABSTRACT Cystine is a disulphide amino acid that is normally generated within the lysosomes through lysosomal-based protein degradation and via extracellular uptake of free cystine. In the autosomal recessive disorder, cystinosis, a defect in the CTNS gene results in excessive lysosomal accumulation of cystine, with early kidney failure a hallmark of the disease. Previously, we demonstrated that silencing of the CTNS gene in kidney proximal tubular epithelial cells (PTECs) resulted in an increase in intracellular cystine concentration coupled with a dramatic reduction in the total GSH content. Because of the crucial role of GSH in maintaining the redox status and viability of kidney PTECs, we assessed the effects of CTNS knockdown-induced lysosomal cystine accumulation on intracellular reactive oxygen species (ROS) production, activity of classical redox-sensitive genes, mitochondrial integrity and cell viability. Our results showed that lysosomal cystine accumulation increased ROS production and solicitation to oxidative stress (OS). This was associated with the induction of classical redox-sensitive proteins, NF-κB, NRF2, HSP32 and HSP70. Cystine-loaded PTECs also displayed depolarized mitochondria, reduced ATP content and augmented apoptosis. Treatment of CTNS knockdown PTECs with the cystine-depleting agent cysteamine resulted in the normalization of OS index, increased GSH and ATP content, and preservation of cell viability. Taken together, the alterations observed in cystinotic cells may represent different facets of a cascade leading to tubular dysfunction and, in combination with cysteamine therapy, may offer a novel link for the attenuation of renal injury and preservation of functions of other organs affected in cystinosis.
Collapse
Affiliation(s)
- Rodolfo Sumayao
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Bernadette McEvoy
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Philip Newsholme
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct and Faculty of Health Sciences, Curtin University, Perth, Western Australia
| | - Tara McMorrow
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
45
|
Caetano-Pinto P, Janssen MJ, Gijzen L, Verscheijden L, Wilmer MJ, Masereeuw R. Fluorescence-Based Transport Assays Revisited in a Human Renal Proximal Tubule Cell Line. Mol Pharm 2016; 13:933-44. [DOI: 10.1021/acs.molpharmaceut.5b00821] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Pedro Caetano-Pinto
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, 3508 TB Utrecht, The Netherlands
| | - Manoe J. Janssen
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, 3508 TB Utrecht, The Netherlands
| | - Linda Gijzen
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
| | - Laurens Verscheijden
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
| | - Martijn J.G. Wilmer
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
| | - Rosalinde Masereeuw
- Department
of Pharmacology and Toxicology, Radboud university medical center, Radboud Institute for Molecular Life Sciences, 6500 HB Nijmegen, The Netherlands
- Division
of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, 3508 TB Utrecht, The Netherlands
| |
Collapse
|
46
|
McEvoy B, Sumayao R, Slattery C, McMorrow T, Newsholme P. Cystine accumulation attenuates insulin release from the pancreatic β-cell due to elevated oxidative stress and decreased ATP levels. J Physiol 2015; 593:5167-82. [PMID: 26482480 DOI: 10.1113/jp271237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 10/13/2015] [Indexed: 12/26/2022] Open
Abstract
The pancreatic β-cell has reduced antioxidant defences making it more susceptible to oxidative stress. In cystinosis, a lysosomal storage disorder, an altered redox state may contribute to cellular dysfunction. This rare disease is caused by an abnormal lysosomal cystine transporter, cystinosin, which causes excessive accumulation of cystine in the lysosome. Cystinosis associated kidney damage and dysfunction leads to the Fanconi syndrome and ultimately end-stage renal disease. Following kidney transplant, cystine accumulation in other organs including the pancreas leads to multi-organ dysfunction. In this study, a Ctns gene knockdown model of cystinosis was developed in the BRIN-BD11 rat clonal pancreatic β-cell line using Ctns-targeting siRNA. Additionally there was reduced cystinosin expression, while cell cystine levels were similarly elevated to the cystinotic state. Decreased levels of chronic (24 h) and acute (20 min) nutrient-stimulated insulin secretion were observed. This decrease may be due to depressed ATP generation particularly from glycolysis. Increased ATP production and the ATP/ADP ratio are essential for insulin secretion. Oxidised glutathione levels were augmented, resulting in a lower [glutathione/oxidised glutathione] redox potential. Additionally, the mitochondrial membrane potential was reduced, apoptosis levels were elevated, as were markers of oxidative stress, including reactive oxygen species, superoxide and hydrogen peroxide. Furthermore, the basal and activated phosphorylated forms of the redox-sensitive transcription factor NF-κB were increased in cells with silenced Ctns. From this study, the cystinotic-like pancreatic β-cell model demonstrated that the altered oxidative status of the cell, resulted in depressed mitochondrial function and pathways of ATP production, causing reduced nutrient-stimulated insulin secretion.
Collapse
Affiliation(s)
- Bernadette McEvoy
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Rodolfo Sumayao
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Craig Slattery
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Tara McMorrow
- Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Philip Newsholme
- School of Biomedical Sciences, CHIRI Biosciences Research Precinct and Faculty of Health Sciences, Curtin University, Perth, Australia
| |
Collapse
|
47
|
Conforti A, Taranta A, Biagini S, Starc N, Pitisci A, Bellomo F, Cirillo V, Locatelli F, Bernardo ME, Emma F. Cysteamine treatment restores the in vitro ability to differentiate along the osteoblastic lineage of mesenchymal stromal cells isolated from bone marrow of a cystinotic patient. J Transl Med 2015; 13:143. [PMID: 25947233 PMCID: PMC4428230 DOI: 10.1186/s12967-015-0494-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 04/16/2015] [Indexed: 11/10/2022] Open
Abstract
Background Cystinosis is a rare autosomal recessive disease caused by mutations of the CTNS gene, which encodes for a lysosomal cystine/H+ symporter. In mice, inactivation of the CTNS gene causes intralysosomal cystine accumulation and progressive organ damage that can be reversed, at least in part, by infusion of mesenchymal stromal cells (MSCs). Little is known on the mesenchymal compartment of cystinotic patients. The aim of the study was to test the phenotypical and functional properties of cystinotic MSCs (Cys-MSCs) isolated from bone marrow (BM) aspirate of a patient with nephropathic cystinosis. Methods Morphology, proliferative capacity (measured as population doublings), immunophenotype (by flow-cytometry) and immunomodulatory properties (as phytohemagglutinin-induced peripheral blood mononuclear cell proliferation) were analyzed. The osteogenic differentiation potential of Cys-MSCs was evaluated by histological staining (alkaline phosphatase activity, Alzarin Red and von Kossa staining) spectrophotometry and Quantitative Reverse Transcriptase Polymerase Chain Reaction for osteigenic markers in the presence and in the absence of cysteamine. Cys-MSCs were compared with those isolated and expanded ex vivo from three healthy donors (HD-MSCs). Results Despite a slightly lower proliferative capacity, Cys-MSCs displayed a characteristic spindle-shaped morphology and similar immunephenotype as HD-MSCs. Cys-MSCs and HD-MSCs prevented proliferation of PHA-stimulated allogeneic peripheral blood mononuclear cells to the same extent. After in vitro induction into osteoblasts, Cys-MSCs showed reduced alkaline phosphatase (ALP) activity, calcium depositions and expression of ALP and collagen type 1. When Cys-MSCs were treated in vitro with increasing doses of cysteamine (50-100-200 μM/L) during the differentiation assay, recovery of Cys-MSCs differentiation capacity into osteoblasts was observed. No difference in adipogenic differentiation was found between Cys-MSCs and HD-MSCs. Conclusions Our results indicate that, as compared to HD-MSCs, Cys-MSCs show reduced ability to differentiate into osteoblasts, which can be reverted after cysteamine treatment.
Collapse
Affiliation(s)
- Antonella Conforti
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy.
| | - Anna Taranta
- Department of Nephrology and Urology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy.
| | - Simone Biagini
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy.
| | - Nadia Starc
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy. .,University of Rome, Tor Vergata, Rome.
| | - Angela Pitisci
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy.
| | - Francesco Bellomo
- Department of Nephrology and Urology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy.
| | - Valentina Cirillo
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy.
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy. .,University of Pavia, Pavia, Italy.
| | - Maria Ester Bernardo
- Department of Pediatric Hematology/Oncology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy.
| | - Francesco Emma
- Department of Nephrology and Urology, IRCCS Bambino Gesù Children's Hospital, P.le S. Onofrio, 00165, Rome, Italy.
| |
Collapse
|
48
|
Cistinosis en pacientes adolescentes y adultos: Recomendaciones para la atención integral de la cistinosis. Nefrologia 2015. [DOI: 10.1016/j.nefro.2015.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
49
|
Endo-lysosomal dysfunction in human proximal tubular epithelial cells deficient for lysosomal cystine transporter cystinosin. PLoS One 2015; 10:e0120998. [PMID: 25811383 PMCID: PMC4374958 DOI: 10.1371/journal.pone.0120998] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/09/2015] [Indexed: 12/11/2022] Open
Abstract
Nephropathic cystinosis is a lysosomal storage disorder caused by mutations in the CTNS gene encoding cystine transporter cystinosin that results in accumulation of amino acid cystine in the lysosomes throughout the body and especially affects kidneys. Early manifestations of the disease include renal Fanconi syndrome, a generalized proximal tubular dysfunction. Current therapy of cystinosis is based on cystine-lowering drug cysteamine that postpones the disease progression but offers no cure for the Fanconi syndrome. We studied the mechanisms of impaired reabsorption in human proximal tubular epithelial cells (PTEC) deficient for cystinosin and investigated the endo-lysosomal compartments of cystinosin-deficient PTEC by means of light and electron microscopy. We demonstrate that cystinosin-deficient cells had abnormal shape and distribution of the endo-lysosomal compartments and impaired endocytosis, with decreased surface expression of multiligand receptors and delayed lysosomal cargo processing. Treatment with cysteamine improved surface expression and lysosomal cargo processing but did not lead to a complete restoration and had no effect on the abnormal morphology of endo-lysosomal compartments. The obtained results improve our understanding of the mechanism of proximal tubular dysfunction in cystinosis and indicate that impaired protein reabsorption can, at least partially, be explained by abnormal trafficking of endosomal vesicles.
Collapse
|
50
|
Galarreta CI, Forbes MS, Thornhill BA, Antignac C, Gubler MC, Nevo N, Murphy MP, Chevalier RL. The swan-neck lesion: proximal tubular adaptation to oxidative stress in nephropathic cystinosis. Am J Physiol Renal Physiol 2015; 308:F1155-66. [PMID: 25694483 DOI: 10.1152/ajprenal.00591.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 02/09/2015] [Indexed: 01/14/2023] Open
Abstract
Cystinosis is an inherited disorder resulting from a mutation in the CTNS gene, causing progressive proximal tubular cell flattening, the so-called swan-neck lesion (SNL), and eventual renal failure. To determine the role of oxidative stress in cystinosis, histologic sections of kidneys from C57BL/6 Ctns(-/-) and wild-type mice were examined by immunohistochemistry and morphometry from 1 wk to 20 mo of age. Additional mice were treated from 1 to 6 mo with vehicle or mitoquinone (MitoQ), an antioxidant targeted to mitochondria. The leading edge of the SNL lost mitochondria and superoxide production, and became surrounded by a thickened tubular basement membrane. Progression of the SNL as determined by staining with lectin from Lotus tetragonolobus accelerated after 3 mo, but was delayed by treatment with MitoQ (38 ± 4% vs. 28 ± 1%, P < 0.01). Through 9 mo, glomeruli had retained renin staining and intact macula densa, whereas SNL expressed transgelin, an actin-binding protein, but neither kidney injury molecule-1 (KIM-1) nor cell death was observed. After 9 mo, clusters of proximal tubules exhibited localized oxidative stress (4-hydroxynonenal binding), expressed KIM-1, and underwent apoptosis, leading to the formation of atubular glomeruli and accumulation of interstitial collagen. We conclude that nephron integrity is initially maintained in the Ctns(-/-) mouse by adaptive flattening of cells of the SNL through loss of mitochondria, upregulation of transgelin, and thickened basement membrane. This adaptation ultimately fails in adulthood, with proximal tubular disruption, formation of atubular glomeruli, and renal failure. Antioxidant treatment targeted to mitochondria delays initiation of the SNL, and may provide therapeutic benefit in children with cystinosis.
Collapse
Affiliation(s)
| | - Michael S Forbes
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia
| | | | - Corinne Antignac
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, and Paris Descartes-Sorbonne Paris Cite University, Imagine Institute, Paris, France; and
| | - Marie-Claire Gubler
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, and Paris Descartes-Sorbonne Paris Cite University, Imagine Institute, Paris, France; and
| | - Nathalie Nevo
- Inserm U1163, Laboratory of Hereditary Kidney Diseases, and Paris Descartes-Sorbonne Paris Cite University, Imagine Institute, Paris, France; and
| | | | - Robert L Chevalier
- Department of Pediatrics, University of Virginia, Charlottesville, Virginia;
| |
Collapse
|