51
|
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: 1.9] [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
|
52
|
Abstract
Striking therapeutic advances for lysosomal diseases have harnessed the biology of this organelle and illustrate its central rôle in the dynamic economy of the cell. Further Innovation will require improved protein-targetting or realization of therapeutic gene- and cell transfer stratagems. Rescuing function before irreversible injury, mandates a deep knowledge of clinical behaviour as well as molecular pathology – and frequently requires an understanding of neuropathology. Whether addressing primary causes, or rebalancing the effects of disordered cell function, true therapeutic innovation depends on continuing scientific exploration of the lysosome. Genuine partnerships between biotech and the patients affected by this extraordinary family of disorders continue to drive productive pharmaceutical discovery.
Collapse
Affiliation(s)
- Timothy M Cox
- Department of Medicine, University of Cambridge, UK.
| |
Collapse
|
53
|
Elmonem MA, Makar SH, van den Heuvel L, Abdelaziz H, Abdelrahman SM, Bossuyt X, Janssen MC, Cornelissen EA, Lefeber DJ, Joosten LA, Nabhan MM, Arcolino FO, Hassan FA, Gaide Chevronnay HP, Soliman NA, Levtchenko E. Clinical utility of chitotriosidase enzyme activity in nephropathic cystinosis. Orphanet J Rare Dis 2014; 9:155. [PMID: 25407738 PMCID: PMC4269071 DOI: 10.1186/s13023-014-0155-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/30/2014] [Indexed: 01/02/2023] Open
Abstract
Background Nephropathic cystinosis is an inherited autosomal recessive lysosomal storage disorder characterized by the pathological accumulation and crystallization of cystine inside different cell types. WBC cystine determination forms the basis for the diagnosis and therapeutic monitoring with the cystine depleting drug (cysteamine). The chitotriosidase enzyme is a human chitinase, produced by activated macrophages. Its elevation is documented in several lysosomal storage disorders. Although, about 6% of Caucasians have enzyme deficiency due to homozygosity of 24-bp duplication mutation in the chitotriosidase gene, it is currently established as a screening marker and therapeutic monitor for Gaucher’s disease. Methods Plasma chitotriosidase activity was measured in 45 cystinotic patients, and compared with 87 healthy controls and 54 renal disease patients with different degrees of renal failure (CKD1-5). Chitotriosidase levels were also correlated with WBC cystine in 32 treated patients. Furthermore, we incubated control human macrophages in-vitro with different concentrations of cystine crystals and monitored the response of tumor necrosis factor-alpha (TNF-α) and chitotriosidase activity. We also compared plasma chitotriosidase activity in cystinotic knocked-out (n = 10) versus wild-type mice (n = 10). Results Plasma chitotriosidase activity in cystinotic patients (0–3880, median 163 nmol/ml/h) was significantly elevated compared to healthy controls (0–90, median 18 nmol/ml/h) and to CKD patients (0–321, median 52 nmol/ml/h), P < 0.001 for both groups. Controls with decreased renal function had mild to moderate chitotriosidase elevations; however, their levels were significantly lower than in cystinotic patients with comparable degree of renal insufficiency. Chitotriosidase activity positively correlated with WBC cystine content for patients on cysteamine therapy (r = 0.8), P < 0.001. In culture, human control macrophages engulfed cystine crystals and released TNF-α into culture supernatant in a crystal concentration dependent manner. Chitotriosidase activity was also significantly increased in macrophage supernatant and cell-lysate. Furthermore, chitotriosidase activity was significantly higher in cystinotic knocked-out than in the wild-type mice, P = 0.003. Conclusions This study indicates that cystine crystals are potent activators of human macrophages and that chitotriosidase activity is a useful marker for this activation and a promising clinical biomarker and therapeutic monitor for nephropathic cystinosis.
Collapse
Affiliation(s)
- Mohamed A Elmonem
- Department of Clinical and Chemical Pathology, Inherited Metabolic Disorder Laboratory (IMDL), Cairo University, Cairo, Egypt.
| | - Samuel H Makar
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation (CPNT), Cairo University, Cairo, Egypt. .,EGORD, Egyptian group of orphan renal diseases, Cairo, Egypt.
| | - Lambertus van den Heuvel
- Department of Pediatric Nephrology & Growth and Regeneration, University Hospital Leuven, Catholic University of Leuven, Leuven, Belgium. .,Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Hanan Abdelaziz
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation (CPNT), Cairo University, Cairo, Egypt. .,EGORD, Egyptian group of orphan renal diseases, Cairo, Egypt.
| | - Safaa M Abdelrahman
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation (CPNT), Cairo University, Cairo, Egypt. .,EGORD, Egyptian group of orphan renal diseases, Cairo, Egypt.
| | - Xavier Bossuyt
- University Hospitals Leuven & Department of Microbiology and Immunology, Laboratory Medicine, Catholic University of Leuven, Leuven, Belgium.
| | - Mirian C Janssen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Elisabeth Am Cornelissen
- Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Dirk J Lefeber
- Department of Neurology, Laboratory for Genetic, Endocrine and Metabolic Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Leo Ab Joosten
- Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Marwa M Nabhan
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation (CPNT), Cairo University, Cairo, Egypt. .,EGORD, Egyptian group of orphan renal diseases, Cairo, Egypt.
| | - Fanny O Arcolino
- Department of Pediatric Nephrology & Growth and Regeneration, University Hospital Leuven, Catholic University of Leuven, Leuven, Belgium.
| | - Fayza A Hassan
- Department of Clinical and Chemical Pathology, Inherited Metabolic Disorder Laboratory (IMDL), Cairo University, Cairo, Egypt.
| | | | - Neveen A Soliman
- Department of Pediatrics, Center of Pediatric Nephrology and Transplantation (CPNT), Cairo University, Cairo, Egypt. .,EGORD, Egyptian group of orphan renal diseases, Cairo, Egypt.
| | - Elena Levtchenko
- Department of Pediatric Nephrology & Growth and Regeneration, University Hospital Leuven, Catholic University of Leuven, Leuven, Belgium.
| |
Collapse
|
54
|
Emma F, Nesterova G, Langman C, Labbé A, Cherqui S, Goodyer P, Janssen MC, Greco M, Topaloglu R, Elenberg E, Dohil R, Trauner D, Antignac C, Cochat P, Kaskel F, Servais A, Wühl E, Niaudet P, Van't Hoff W, Gahl W, Levtchenko E. Nephropathic cystinosis: an international consensus document. Nephrol Dial Transplant 2014; 29 Suppl 4:iv87-94. [PMID: 25165189 DOI: 10.1093/ndt/gfu090] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cystinosis is caused by mutations in the CTNS gene (17p13.2), which encodes for a lysosomal cystine/proton symporter termed cystinosin. It is the most common cause of inherited renal Fanconi syndrome in young children. Because of its rarity, the diagnosis and specific treatment of cystinosis are frequently delayed, which has a significant impact on the overall prognosis. In this document, we have summarized expert opinions on several aspects of the disease to improve knowledge and provide guidance for diagnosis and treatment.
Collapse
Affiliation(s)
- Francesco Emma
- Division of Nephrology and Dialysis, Bambino Gesu` Children's Hospital - IRCCS, Rome, Italy
| | - Galina Nesterova
- Section on Human Biochemical Genetics, Human Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-1851, USA
| | - Craig Langman
- Kidney Diseases, Feinberg School of Medicine, Northwestern University and the Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Antoine Labbé
- Quinze-Vingts National Ophthalmology Hospital, Paris and Versailles Saint-Quentin-en-Yvelines University, Versailles, France Clinical Investigations Center, INSERM 503, Quinze-Vingts National Ophthalmology Hospital, Paris, France
| | - Stephanie Cherqui
- Division of Genetics, Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Paul Goodyer
- Department of Pediatrics, McGill University, Montreal Children's Hospital, Montreal, Québec, Canada
| | - Mirian C Janssen
- Department of Internal Medicine, Radboud University Medical Centre Nijmegen, Nijmegen, The Netherlands
| | - Marcella Greco
- Division of Nephrology and Dialysis, Bambino Gesu` Children's Hospital - IRCCS, Rome, Italy
| | - Rezan Topaloglu
- Division of Pediatric Nephrology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ewa Elenberg
- Renal Service, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Ranjan Dohil
- Department of Pediatrics, Rady Children's Hospital, San Diego, University of California San Diego, San Diego, CA, USA
| | - Doris Trauner
- Department of Neurosciences, University of California, San Diego, School of Medicine, San Diego, CA, USA
| | - Corinne Antignac
- Laboratory of Inherited Kidney Diseases, Inserm UMR 1163, Paris, France Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Paris, France APHP, Department of Genetics, Necker Hospital, Paris, France
| | - Pierre Cochat
- Centre de référence des maladies rénales rares, Hospices Civils de Lyon and Université Claude-Bernard Lyon 1, Lyon, France
| | - Frederick Kaskel
- Pediatric Nephrology, Children's Hospital at Montefiore, Bronx, NY, USA
| | - Aude Servais
- Department of Adult Nephrology, Hôpital Necker-Enfants Malades, APHP, Paris Descartes University, Paris, France
| | - Elke Wühl
- Division of Pediatric Nephrology, Center of Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Patrick Niaudet
- Université Paris Descartes, Hôpital Necker-Enfants Malades, Paris 75015, France
| | | | - William Gahl
- Section on Human Biochemical Genetics, Human Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892-1851, USA
| | - Elena Levtchenko
- Department of Pediatric Nephrology and Growth and Regeneration, University Hospitals Leuven, Katholieke Universiteit Leuven, Leuven, Belgium
| |
Collapse
|
55
|
Shams F, Livingstone I, Oladiwura D, Ramaesh K. Treatment of corneal cystine crystal accumulation in patients with cystinosis. Clin Ophthalmol 2014; 8:2077-84. [PMID: 25336909 PMCID: PMC4199850 DOI: 10.2147/opth.s36626] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Cystinosis is a rare autosomal recessive disorder characterized by the accumulation of cystine within the cells of different organs. Infantile nephropathic cystinosis is the most common and severe phenotype. With the success of renal transplantation, these patients are now living longer and thus more long-term complications within different organs are becoming apparent. Ophthalmic manifestations range from corneal deposits of cystine crystals to pigmentary retinopathy. With increasing age, more severe ocular complications have been reported. Photophobia is a prominent symptom for patients. With prolonged survival and increasing age, this symptom, along with corneal erosions and blepharospasm, can become debilitating. This review revisits the basic pathogenesis of cystinosis, the ocular manifestations of the disease, and the treatment of corneal crystals.
Collapse
Affiliation(s)
- Fatemeh Shams
- Department of Ophthalmology, Gartnavel General Hospital, Glasgow, Scotland
| | - Iain Livingstone
- Department of Ophthalmology, Gartnavel General Hospital, Glasgow, Scotland
| | - Dilys Oladiwura
- Department of Ophthalmology, Gartnavel General Hospital, Glasgow, Scotland
| | - Kanna Ramaesh
- Department of Ophthalmology, Gartnavel General Hospital, Glasgow, Scotland
| |
Collapse
|
56
|
Abstract
Cystinosis is an autosomal recessive inherited lysosomal storage disease. It is characterized by generalized proximal tubular dysfunction known as renal Fanconi syndrome and causes end-stage renal disease by the age of about 10 years if left untreated. Extrarenal organs are also affected, including the thyroid gland, gonads, pancreas, liver, muscle, and brain. Treatment consists of administration of cysteamine, resulting in depletion of cystine that is trapped inside the lysosomes. Since cysteamine has a short half-life, it should be administered every 6 hours. Recently, a new delayed-release formulation was marketed, that should be administered every 12 hours. The first studies comparing both cysteamine formulations show comparable results regarding white blood cell cystine depletion (which serves as a measure for cystine accumulation in the body), while a slightly lower daily dose of cysteamine can be used.
Collapse
Affiliation(s)
- Martine Tp Besouw
- Department of Pediatric Nephrology, University Hospitals Leuven, Belgium ; Laboratory of Pediatrics, Catholic University Leuven, Leuven, Belgium
| | - Elena N Levtchenko
- Department of Pediatric Nephrology, University Hospitals Leuven, Belgium ; Laboratory of Pediatrics, Catholic University Leuven, Leuven, Belgium
| |
Collapse
|
57
|
Rocca CJ, Ur SN, Harrison F, Cherqui S. rAAV9 combined with renal vein injection is optimal for kidney-targeted gene delivery: conclusion of a comparative study. Gene Ther 2014; 21:618-28. [PMID: 24784447 PMCID: PMC4047163 DOI: 10.1038/gt.2014.35] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 12/19/2022]
Abstract
Effective gene therapy strategies for the treatment of kidney disorders remain elusive. We report an optimized kidney-targeted gene delivery strategy using recombinant adeno-associated virus (rAAV) administered via retrograde renal vein injection in mice. Renal vein injection of rAAV consistently resulted in superior kidney transduction compared with tail vein injection using as little as half the tail vein dose. We compared rAAV5, 6, 8 and 9, containing either green fluorescent protein (GFP) or luciferase reporter genes driven by the Cytomegalovirus promoter. We demonstrated that although rAAV6 and 8 injected via renal vein transduced the kidney, transgene expression was mainly restricted to the medulla. Transgene expression was systematically low after rAAV5 injection, attributed to T-cell immune response, which could be overcome by transient immunosuppression. However, rAAV9 was the only serotype that permitted high-transduction efficiency of both the cortex and medulla. Moreover, both the glomeruli and tubules were targeted, with a higher efficiency within the glomeruli. To improve the specificity of kidney-targeted gene delivery with rAAV9, we used the parathyroid hormone receptor 'kidney-specific' promoter. We obtained a more efficient transgene expression within the kidney, and a significant reduction in other tissues. Our work represents the first comprehensive and clinically relevant study for kidney gene delivery.
Collapse
Affiliation(s)
- Céline J. Rocca
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| | - Sarah N. Ur
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| | - Frank Harrison
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Stephanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, San Diego, 9500 Gilman drive, MC 0734, La Jolla, California 92093-0734, USA
| |
Collapse
|
58
|
Abstract
At least 10% of adults and nearly all children who receive renal-replacement therapy have an inherited kidney disease. These patients rarely die when their disease progresses and can remain alive for many years because of advances in organ-replacement therapy. However, these disorders substantially decrease their quality of life and have a large effect on health-care systems. Since the kidneys regulate essential homoeostatic processes, inherited kidney disorders have multisystem complications, which add to the usual challenges for rare disorders. In this review, we discuss the nature of rare inherited kidney diseases, the challenges they pose, and opportunities from technological advances, which are well suited to target the kidney. Mechanistic insights from rare disorders are relevant for common disorders such as hypertension, kidney stones, cardiovascular disease, and progression of chronic kidney disease.
Collapse
Affiliation(s)
- Olivier Devuyst
- Division of Nephrology, Université catholique de Louvain, Brussels, Belgium; Institute of Physiology, Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland.
| | - Nine V A M Knoers
- Department of Medical Genetics, Division of Biomedical Genetics, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Giuseppe Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso and Unit of Nephrology and Dialysis, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Franz Schaefer
- Pediatric Nephrology Division, Center for Pediatric and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
59
|
Iglesias DM, Akpa MM, Goodyer P. Priming the renal progenitor cell. Pediatr Nephrol 2014; 29:705-10. [PMID: 24414605 DOI: 10.1007/s00467-013-2685-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 10/21/2013] [Accepted: 10/25/2013] [Indexed: 01/15/2023]
Abstract
The mammalian kidney arises from OSR1(+) progenitor cells in the intermediate mesoderm. However, these cells must acquire unique properties before they can respond to inductive signals that launch the differentiation program. Recent data indicate that the transcription factor, WT1, plays a master role in this transition. Interestingly, some of these embryonic nephron progenitor cells are retained in the adult organ where they may participate in tissue regeneration after acute kidney injury. A better understanding of the biology of these cells may one day allow progenitor cell-based therapeutic strategies to help regenerate damaged adult nephrons.
Collapse
Affiliation(s)
- Diana M Iglesias
- McGill University, Montreal Children's Hospital Research Institute, Montréal, Québec, Canada
| | | | | |
Collapse
|
60
|
Al-Haggar M. Cystinosis as a lysosomal storage disease with multiple mutant alleles: Phenotypic-genotypic correlations. World J Nephrol 2013; 2:94-102. [PMID: 24255892 PMCID: PMC3832870 DOI: 10.5527/wjn.v2.i4.94] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 10/09/2013] [Accepted: 10/17/2013] [Indexed: 02/06/2023] Open
Abstract
Cystinosis is an autosomal recessive lysosomal storage disease with an unclear enzymatic defect causing lysosomal cystine accumulation with no corresponding elevation of plasma cystine levels leading to multisystemic dysfunction. The systemic manifestations include a proximal renal tubular defect (Fanconi-like), endocrinal disturbances, eye involvements, with corneal, conjunctival and retinal depositions, and neurological manifestations in the form of brain and muscle dysfunction. Most of the long-term ill effects of cystinosis are observed particularly in patients with long survival as a result of a renal transplant. Its responsible CTNS gene that encodes the lysosomal cystine carrier protein (cystinosin) has been mapped on the short arm of chromosome 17 (Ch17 p13). There are three clinical forms based on the onset of main symptoms: nephropathic infantile form, nephropathic juvenile form and non-nephropathic adult form with predominant ocular manifestations. Avoidance of eye damage from sun exposure, use of cystine chelators (cysteamine) and finally renal transplantation are the main treatment lines. Pre-implantation genetic diagnosis for carrier parents is pivotal in the prevention of recurrence.
Collapse
|
61
|
Besouw MTP, Emma F, Levtchenko EN. Management of nephropathic cystinosis. Expert Opin Orphan Drugs 2013. [DOI: 10.1517/21678707.2013.855634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
62
|
Cherqui S. Is genetic rescue of cystinosis an achievable treatment goal? Nephrol Dial Transplant 2013; 29:522-8. [PMID: 23861466 DOI: 10.1093/ndt/gft270] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cystinosis is an autosomal recessive metabolic disease that belongs to the family of lysosomal storage disorders. The defective gene is CTNS, which encodes the lysosomal cystine transporter, cystinosin. Cystine accumulates in all tissues and leads to organ damage including end-stage renal disease. In this review, we outline the studies that support that genetic rescue of cystinosis could be an achievable goal, even though cystinosis is a multi-compartmental disease and cystinosin an intracellular transmembrane protein. Using the mouse model of cystinosis, the Ctns(-/-) mice, we showed that transplanted hematopoietic stem cells (HSCs) were able to act as vehicles for the delivery of a functional Ctns gene to the different organs and led to the significant decrease of the tissue cystine content and tissue preservation. Ex vivo gene-modified Ctns(-/-) HSC transplantation using a lentiviral vector containing CTNS complementary DNA (cDNA) was also successful in the Ctns(-/-) mice and built the foundations for a clinical trial for autologous HSC transplantation for cystinosis. The capacity of HSCs for rescuing non-hematopoietic disease is controversial, and new insights into regenerative medicine could be gained from unraveling the underlying mechanism of action.
Collapse
Affiliation(s)
- Stephanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
63
|
Upregulation of the Rab27a-dependent trafficking and secretory mechanisms improves lysosomal transport, alleviates endoplasmic reticulum stress, and reduces lysosome overload in cystinosis. Mol Cell Biol 2013; 33:2950-62. [PMID: 23716592 DOI: 10.1128/mcb.00417-13] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cystinosis is a lysosomal storage disorder caused by the accumulation of the amino acid cystine due to genetic defects in the CTNS gene, which encodes cystinosin, the lysosomal cystine transporter. Although many cellular dysfunctions have been described in cystinosis, the mechanisms leading to these defects are not well understood. Here, we show that increased lysosomal overload induced by accumulated cystine leads to cellular abnormalities, including vesicular transport defects and increased endoplasmic reticulum (ER) stress, and that correction of lysosomal transport improves cellular function in cystinosis. We found that Rab27a was expressed in proximal tubular cells (PTCs) and partially colocalized with the lysosomal marker LAMP-1. The expression of Rab27a but not other small GTPases, including Rab3 and Rab7, was downregulated in kidneys from Ctns-/- mice and in human PTCs from cystinotic patients. Using total internal reflection fluorescence microscopy, we found that lysosomal transport is impaired in Ctns-/- cells. Ctns-/- cells showed significant ER expansion and a marked increase in the unfolded protein response-induced chaperones Grp78 and Grp94. Upregulation of the Rab27a-dependent vesicular trafficking mechanisms rescued the defective lysosomal transport phenotype and reduced ER stress in cystinotic cells. Importantly, reconstitution of lysosomal transport mediated by Rab27a led to decreased lysosomal overload, manifested as reduced cystine cellular content. Our data suggest that upregulation of the Rab27a-dependent lysosomal trafficking and secretory pathways contributes to the correction of some of the cellular defects induced by lysosomal overload in cystinosis, including ER stress.
Collapse
|
64
|
Nesterova G, Gahl WA. Cystinosis: the evolution of a treatable disease. Pediatr Nephrol 2013; 28:51-9. [PMID: 22903658 PMCID: PMC3505515 DOI: 10.1007/s00467-012-2242-5] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/07/2012] [Accepted: 06/08/2012] [Indexed: 11/01/2022]
Abstract
Cystinosis is a rare autosomal recessive disorder involving lysosomal storage of the amino acid cystine due to a defect in the membrane transport protein, cystinosin. Since the introduction of kidney transplants and the availability of cystine-depleting medical therapy, this previously fatal disease was transformed into a treatable disorder. Renal allografts and medical therapy targeting the basic metabolic defect have altered the natural hisotry of cystinosis so drastically that patients have a life expectancy extending past 50 years. Consequently, early diagnosis and appropriate therapy are critically important. In this article, we offer a review of the manifestations of cystinosis, including the proximal tubular dysfunction of renal Fanconi syndrome, and discuss the prevention and treatment of the disorder's systemic complications. We focus on the nephropathic forms of cystinosis, aiming to assist nephrologists and other physicians to develop early recognition and appropriate management of cystinosis patients.
Collapse
Affiliation(s)
- Galina Nesterova
- NHGRI, Medical Biochemical Genetic Section, National Institutes of Health, Bethesda, MD, USA.
| | - William A. Gahl
- NHGRI, Medical Biochemical Genetic Section, National Institutes of Health, Bethesda, MD USA
| |
Collapse
|
65
|
Soylu A, Demirci T, Fırıncı F, Bağrıyanık A, Demir BK, Atmaca S, Türkmen MA, Kavukçu S. Mesenchymal stem cells ameliorate postpyelonephritic renal scarring in rats. Urology 2012; 80:1161.e7-1161.e1.161E12. [PMID: 22921785 DOI: 10.1016/j.urology.2012.06.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 06/24/2012] [Accepted: 06/27/2012] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To evaluate the efficiency of mesenchymal stem cells in ameliorating renal scarring in a rat pyelonephritis model. METHODS Three groups each, including 8 Sprague-Dawley rats were formed: Group 1 = sham operated (4 were given mesenchymal stem cells); group 2 = pyelonephritis induced by Escherichia coli; and group 3 = pyelonephritis and mesenchymal stem cells. Rats not given mesenchymal stem cells in group 1 and 4 rats in groups 2 and 3 were sacrificed on the eighth day for evaluation of inflammation, and the remaining rats were sacrificed at the sixth week to determine renal scarring along with migration of mesenchymal stem cells to renal tubules and differentiation to tubular cells expressing aquaporin-1. RESULTS Rats in group 3 had lower scores of both acute (8th day) and chronic (6th week) histopathological alterations compared with rats in group 2. By contrast, although rats in group 3 were shown to have mesenchymal stem cells expressing aquaporin-1 in their renal tubules, these cells were not detected in kidney tissue of mesenchymal stem cells-treated sham rats. CONCLUSION These results indicate that mesenchymal stem cells migrated to renal tissues and ameliorated renal scarring in this rat model of pyelonephritis.
Collapse
Affiliation(s)
- Alper Soylu
- Department of Pediatrics, Dokuz Eylul University Medical Faculty, İzmir, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
66
|
Harrison F, Yeagy BA, Rocca CJ, Kohn DB, Salomon DR, Cherqui S. Hematopoietic stem cell gene therapy for the multisystemic lysosomal storage disorder cystinosis. Mol Ther 2012; 21:433-44. [PMID: 23089735 DOI: 10.1038/mt.2012.214] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cystinosis is an autosomal recessive metabolic disease that belongs to the family of lysosomal storage disorders (LSDs). The defective gene is CTNS encoding the lysosomal cystine transporter, cystinosin. Cystine accumulates in all tissues and leads to organ damage including end-stage renal disease. Using the Ctns(-/-) murine model for cystinosis, we tested the use of hematopoietic stem and progenitor cells (HSPC) genetically modified to express a functional CTNS transgene using a self-inactivating-lentiviral vector (SIN-LV). We showed that transduced cells were capable of decreasing cystine content in all tissues and improved kidney function. Transduced HSPC retained their differentiative capabilities, populating all tissue compartments examined and allowing long-term expression of the transgene. Direct correlation between the levels of lentiviral DNA present in the peripheral blood and the levels present in tissues were demonstrated, which could be useful to follow future patients. Using a new model of cystinosis, the DsRed Ctns(-/-) mice, and a LV driving the expression of the fusion protein cystinosin-enhanced green fluorescent protein (eGFP), we showed that cystinosin was transferred from CTNS-expressing cells to Ctns-deficient adjacent cells in vitro and in vivo. This transfer led to cystine decreases in Ctns-deficient cells in vitro. These data suggest that the mechanism of cross-correction is possible in cystinosis.
Collapse
Affiliation(s)
- Frank Harrison
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| | | | | | | | | | | |
Collapse
|
67
|
Buchan B, Kay G, Matthews KH, Cairns D. Suppository formulations as a potential treatment for nephropathic cystinosis. J Pharm Sci 2012; 101:3729-38. [DOI: 10.1002/jps.23246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 05/02/2012] [Accepted: 06/08/2012] [Indexed: 11/11/2022]
|
68
|
Verghese E, Johnson C, Bertram JF, Ricardo SD, Deane JA. The fate of bone marrow-derived cells carrying a Polycystic Kidney Disease mutation in the genetically normal kidney. BMC Nephrol 2012; 13:91. [PMID: 22931547 PMCID: PMC3502565 DOI: 10.1186/1471-2369-13-91] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 08/20/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Polycystic Kidney Disease (PKD) is a genetic condition in which dedifferentiated and highly proliferative epithelial cells form renal cysts and is frequently treated by renal transplantation. Studies have reported that bone marrow-derived cells give rise to renal epithelial cells, particularly following renal injury as often occurs during transplantation. This raises the possibility that bone marrow-derived cells from a PKD-afflicted recipient could populate a transplanted kidney and express a disease phenotype. However, for reasons that are not clear the reoccurrence of PKD has not been reported in a genetically normal renal graft. We used a mouse model to examine whether PKD mutant bone marrow-derived cells are capable of expressing a disease phenotype in the kidney. METHODS Wild type female mice were transplanted with bone marrow from male mice homozygous for a PKD-causing mutation and subjected to renal injury. Y chromosome positive, bone marrow-derived cells in the kidney were assessed for epithelial markers. RESULTS Mutant bone marrow-derived cells were present in the kidney. Some mutant cells were within the bounds of the tubule or duct, but none demonstrated convincing evidence of an epithelial phenotype. CONCLUSIONS Bone marrow-derived cells appear incapable of giving rise to genuine epithelial cells and this is the most likely reason cysts do not reoccur in kidneys transplanted into PKD patients.
Collapse
Affiliation(s)
- Elizabeth Verghese
- Biomedical and Health Sciences, Victoria University, St Albans, Australia
| | | | | | | | | |
Collapse
|
69
|
Iglesias DM, El-Kares R, Taranta A, Bellomo F, Emma F, Besouw M, Levtchenko E, Toelen J, van den Heuvel L, Chu L, Zhao J, Young YK, Eliopoulos N, Goodyer P. Stem cell microvesicles transfer cystinosin to human cystinotic cells and reduce cystine accumulation in vitro. PLoS One 2012; 7:e42840. [PMID: 22912749 PMCID: PMC3418268 DOI: 10.1371/journal.pone.0042840] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 07/11/2012] [Indexed: 11/25/2022] Open
Abstract
Cystinosis is a rare disease caused by homozygous mutations of the CTNS gene, encoding a cystine efflux channel in the lysosomal membrane. In Ctns knockout mice, the pathologic intralysosomal accumulation of cystine that drives progressive organ damage can be reversed by infusion of wildtype bone marrow-derived stem cells, but the mechanism involved is unclear since the exogeneous stem cells are rarely integrated into renal tubules. Here we show that human mesenchymal stem cells, from amniotic fluid or bone marrow, reduce pathologic cystine accumulation in co-cultured CTNS mutant fibroblasts or proximal tubular cells from cystinosis patients. This paracrine effect is associated with release into the culture medium of stem cell microvesicles (100–400 nm diameter) containing wildtype cystinosin protein and CTNS mRNA. Isolated stem cell microvesicles reduce target cell cystine accumulation in a dose-dependent, Annexin V-sensitive manner. Microvesicles from stem cells expressing CTNSRed transfer tagged CTNS protein to the lysosome/endosome compartment of cystinotic fibroblasts. Our observations suggest that exogenous stem cells may reprogram the biology of mutant tissues by direct microvesicle transfer of membrane-associated wildtype molecules.
Collapse
Affiliation(s)
- Diana M. Iglesias
- Department of Pediatrics, Montreal Children's Hospital Research Institute, McGill University, Montréal, Québec, Canada
| | - Reyhan El-Kares
- Department of Pediatrics, Montreal Children's Hospital Research Institute, McGill University, Montréal, Québec, Canada
| | - Anna Taranta
- U.O.C. di Nefrologia e Dialisi, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Francesco Bellomo
- U.O.C. di Nefrologia e Dialisi, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Francesco Emma
- U.O.C. di Nefrologia e Dialisi, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Martine Besouw
- Department of Pediatric Nephrology University Hospitals Leuven, Laboratory of Pediatrics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Elena Levtchenko
- Department of Pediatric Nephrology University Hospitals Leuven, Laboratory of Pediatrics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Jaan Toelen
- Department of Pediatric Nephrology University Hospitals Leuven, Laboratory of Pediatrics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Lambertus van den Heuvel
- Department of Pediatric Nephrology University Hospitals Leuven, Laboratory of Pediatrics, Katholieke Universiteit Leuven, Leuven, Belgium
| | - LeeLee Chu
- Department of Pediatrics, Montreal Children's Hospital Research Institute, McGill University, Montréal, Québec, Canada
| | - Jing Zhao
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - Yoon Kow Young
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - Nicoletta Eliopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
- Division of Surgical Research, Department of Surgery, McGill University, Montréal, Québec, Canada
- Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Paul Goodyer
- Department of Pediatrics, Montreal Children's Hospital Research Institute, McGill University, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
70
|
Abstract
Much attention recently has been focused on stem cell technology as a possible alternative modality of treatment of a variety of diseases. Chronic kidney disease is a serious health problem and most chronic kidney diseases share in common the presence of interstitial and glomerular fibrosis, regardless of the underlying cause. To date there are no specific therapies aimed at treating fibrosis in the kidney. In a novel effort to address the underlying pathology in kidney disease, researchers are demonstrating that stem cell therapy can attenuate fibrosis in chronic kidney disease in animal models. This review will focus on the recent developments in stem cell research and their possible implications to treat chronic kidney disease.
Collapse
|
71
|
Abstract
Cystinosis as a clinical entity is a progressive dysfunction of multiple organs caused by the accumulation of cystine in the tissues, leading, for example, to end-stage renal failure, diabetes, hypothyroidism, myopathy, and central nervous system deterioration. Brodin-Sartorius and colleagues present a long-term study on the impact of cysteamine therapy on these complications. The data show that cysteamine improves the outcome and complications of cystinosis but does not prevent them.
Collapse
|
72
|
Goodyer P. The history of cystinosis: lessons for clinical management. Int J Nephrol 2011; 2011:929456. [PMID: 22013525 PMCID: PMC3195959 DOI: 10.4061/2011/929456] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 08/01/2011] [Indexed: 11/20/2022] Open
Abstract
Cystinosis is a rare disorder, and, accordingly, progress on the understanding and treatment of this disease has been relatively slow. Although cystinosis was identified over 100 years ago, the history of cystinosis is marked by a few sudden leaps forward in our understanding rather than by a sustained research effort fuelled by the larger research community. Major conceptual break-throughs include (a) its discovery in 1903, (b) recognition of the renal Fanconi syndrome, (c) realization that tissue accumulation of cystine reflects a defective channel in the lysosomal membrane, (d) translation of this discovery to trials of cysteamine, (e) discovery of the CTNS gene, and (f) report of successful stem cell therapy in the cystinotic mouse. This paper focuses on the importance management lessons from these milestones and the potential new therapeutic strategies which may be looming in the near future.
Collapse
Affiliation(s)
- Paul Goodyer
- Department of Pediatrics, McGill University, Montreal, QC, Canada H3H 1P3
| |
Collapse
|
73
|
Brodin-Sartorius A, Tête MJ, Niaudet P, Antignac C, Guest G, Ottolenghi C, Charbit M, Moyse D, Legendre C, Lesavre P, Cochat P, Servais A. Cysteamine therapy delays the progression of nephropathic cystinosis in late adolescents and adults. Kidney Int 2011; 81:179-89. [PMID: 21900880 DOI: 10.1038/ki.2011.277] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nephropathic cystinosis is a multisystem autosomal recessive disease caused by cystine accumulation, which is usually treated by oral cysteamine. In order to determine long-term effects of this therapy, we enrolled 86 adult patients (mean age 26.7 years) diagnosed with nephropathic cystinosis, 75 of whom received cysteamine. Therapy was initiated at a mean age of 9.9 years with a mean duration of 17.4 years. By last follow-up, 78 patients had end-stage renal disease (mean age 11.1 years), 62 had hypothyroidism (mean age 13.4), 48 developed diabetes (mean age 17.1 years), and 32 had neuromuscular disorders (mean age 23.3 years). Initiating cysteamine therapy before 5 years of age significantly decreased the incidence and delayed the onset of end-stage renal disease, and significantly delayed the onset of hypothyroidism, diabetes, and neuromuscular disorders. The development of diabetes and hypothyroidism was still significantly delayed, however, in patients in whom therapy was initiated after 5 years of age, compared with untreated patients. The life expectancy was significantly improved in cysteamine-treated versus untreated patients. Thus, cysteamine decreases and delays the onset of complications and improves life expectancy in cystinosis. Hence, cysteamine therapy should be introduced as early as possible during childhood and maintained lifelong.
Collapse
|
74
|
Taniguchi N, Caramés B, Hsu E, Cherqui S, Kawakami Y, Lotz M. Expression patterns and function of chromatin protein HMGB2 during mesenchymal stem cell differentiation. J Biol Chem 2011; 286:41489-41498. [PMID: 21890638 DOI: 10.1074/jbc.m111.236984] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The superficial zone (SZ) of articular cartilage is critical in maintaining tissue function and homeostasis and represents the site of the earliest changes in osteoarthritis (OA). The expression of chromatin protein HMGB2 is restricted to the SZ, which contains cells expressing mesenchymal stem cell (MSC) markers. Age-related loss of HMGB2 and gene deletion are associated with reduced SZ cellularity and early onset OA. This study addressed HMGB2 expression patterns in MSC and its role during differentiation. HMGB2 was detected at higher levels in human MSC as compared with human articular chondrocytes, and its expression declined during chondrogenic differentiation of MSC. Lentiviral HMGB2 transduction of MSC suppressed chondrogenesis as reflected by an inhibition of Col2a1 and Col10a1 expression. Conversely, in bone marrow MSC from Hmgb2(-/-) mice, Col10a1 was more strongly expressed than in wild-type MSC. This is consistent with in vivo results from mouse growth plates showing that Hmgb2 is expressed in proliferating and prehypertrophic zones but not in hypertrophic cartilage where Col10a1 is strongly expressed. Osteogenesis was also accelerated in Hmgb2(-/-) MSC. The expression of Runx2, which plays a major role in late stage chondrocyte differentiation, was enhanced in Hmgb2(-/-) MSC, and HMGB2 negatively regulated the stimulatory effect of Wnt/β-catenin signaling on the Runx2 proximal promoter. These results demonstrate that HMGB2 expression is inversely correlated with the differentiation status of MSC and that HMGB2 suppresses chondrogenic differentiation. The age-related loss of HMGB2 in articular cartilage may represent a mechanism responsible for the decline in adult cartilage stem cell populations.
Collapse
Affiliation(s)
- Noboru Taniguchi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Beatriz Caramés
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Emily Hsu
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Stephanie Cherqui
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037
| | - Yasuhiko Kawakami
- Stem Cell Institute and Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455
| | - Martin Lotz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California 92037.
| |
Collapse
|
75
|
Kidney repair and stem cells: a complex and controversial process. Pediatr Nephrol 2011; 26:1427-34. [PMID: 21336814 DOI: 10.1007/s00467-011-1789-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 02/06/2023]
Abstract
Over the last decade, stem cells have been the topic of much debate and investigation for their regenerative potential in the case of renal injury. This review focuses on bone marrow stem cells (BMSC) for renal repair and the potential origins of the controversial results between studies. Some authors have shown that BMSC can differentiate into renal cells and reverse renal dysfunction while others obtained contradictory results. One significant variation between these studies is the choice of BMSC used. According to the literature and our own experience, unfractionated bone marrow cells and hematopoietic stem cells are able to lead to long-term cell tissue engraftment and repair, whereas mesenchymal stem cells have a short-term paracrine effect. Detection of the bone-marrow-derived cells is also an important source of error. However, the major difference between studies is the model of kidney injury used. Two categories of models have to be distinguished: acute and chronic kidney disease. However, variation within these categories also exists. The outcomes of various strategies for BMSC transplantation after injury to the kidney must be compared within a single model and cannot be transposed from one model to another.
Collapse
|
76
|
|
77
|
Cystinosis: practical tools for diagnosis and treatment. Pediatr Nephrol 2011; 26:205-15. [PMID: 20734088 PMCID: PMC3016220 DOI: 10.1007/s00467-010-1627-6] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Revised: 07/05/2010] [Accepted: 07/07/2010] [Indexed: 02/07/2023]
Abstract
Cystinosis is the major cause of inherited Fanconi syndrome, and should be suspected in young children with failure to thrive and signs of renal proximal tubular damage. The diagnosis can be missed in infants, because not all signs of renal Fanconi syndrome are present during the first months of life. In older patients cystinosis can mimic idiopathic nephrotic syndrome due to focal and segmental glomerulosclerosis. Measuring elevated white blood cell cystine content is the corner stone for the diagnosis. The diagnosis is confirmed by molecular analysis of the cystinosin gene. Corneal cystine crystals are invariably present in all patients with cystinosis after the age of 1 year. Treatment with the cystine depleting drug cysteamine should be initiated as soon as possible and continued lifelong to prolong renal function survival and protect extra-renal organs. This educational feature provides practical tools for the diagnosis and treatment of cystinosis.
Collapse
|
78
|
Yeagy BA, Harrison F, Gubler MC, Koziol JA, Salomon DR, Cherqui S. Kidney preservation by bone marrow cell transplantation in hereditary nephropathy. Kidney Int 2011; 79:1198-206. [PMID: 21248718 DOI: 10.1038/ki.2010.537] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The prospect of cell-based therapy for kidney disease remains controversial despite its immense promise. We had previously shown that transplanting bone marrow and hematopoietic stem cells could generate renal cells and lead to the preservation of kidney function in a mouse model for cystinosis (Ctns(-/-)) that develops chronic kidney injury, 4 months post transplantation. Here, we determined the long-term effects of bone marrow stem cell transplantation on the kidney disease of Ctns(-/-) mice 7 to 15 months post transplantation. Transfer of bone marrow stem cells expressing a functional Ctns gene provided long-term protection to the kidney. Effective therapy, however, depended on achieving a relatively high level of donor-derived blood cell engraftment of Ctns-expressing cells, which was directly linked to the quantity of these cells within the kidney. In contrast, kidney preservation was dependent neither on renal cystine content nor on the age of the mice at the time of transplant. Most of the bone marrow-derived cells within the kidney were interstitial and not epithelial, suggesting that the mechanism involved an indirect protection of the tubules. Thus, our model may help in developing strategies to enhance the potential success of cell-based therapy for kidney injury and in understanding some of the discrepancies currently existing in the field.
Collapse
Affiliation(s)
- Brian A Yeagy
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| | | | | | | | | | | |
Collapse
|
79
|
Harding CO, Gibson KM. Therapeutic liver repopulation for phenylketonuria. J Inherit Metab Dis 2010; 33:681-7. [PMID: 20495959 DOI: 10.1007/s10545-010-9099-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2010] [Revised: 03/24/2010] [Accepted: 03/31/2010] [Indexed: 01/22/2023]
Abstract
Problems with long-term dietary compliance in phenylketonuria (PKU) necessitate the development of alternative treatment approaches. Therapeutic liver repopulation with phenylalanine hydroxylase (PAH)-expressing cells following hepatocyte or haematopoietic stem cell transplantation has been investigated as a possible novel treatment approach for PKU. Successful therapeutic liver repopulation requires both a stimulus for liver regeneration at the time of cell transplantation and a selective growth advantage for the PAH+ donor cells. Unfortunately, wild-type PAH+ hepatocytes do not enjoy any growth advantage over PAH- cells. Successful correction of hyperphenylalaninemia following therapeutic liver repopulation has been accomplished only in an animal model that yields a selective advantage for the donor cells. Haematopoietic stem cell (HSC)-mediated therapeutic liver repopulation has not been reported in any hyperphenylalaninemic system, and the success of HSC-mediated liver repopulation for PKU may be limited by the slow kinetics of this approach. If therapeutic liver repopulation is to be employed successfully in humans with PKU, an effective method of providing a selective growth advantage for the donor cells must be developed. If this can be achieved, liver repopulation with 10-20% wild-type hepatocytes will likely completely normalize Phe clearance in individuals with PKU.
Collapse
Affiliation(s)
- Cary O Harding
- Department of Molecular and Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail code L103, Portland, OR 97239, USA.
| | | |
Collapse
|
80
|
Wilmer MJ, Emma F, Levtchenko EN. The pathogenesis of cystinosis: mechanisms beyond cystine accumulation. Am J Physiol Renal Physiol 2010; 299:F905-16. [PMID: 20826575 DOI: 10.1152/ajprenal.00318.2010] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Renal proximal tubules are highly sensitive to ischemic and toxic insults and are affected in diverse genetic disorders, of which nephropathic cystinosis is the most common. The disease is caused by mutations in the CTNS gene, encoding the lysosomal cystine transporter cystinosin, and is characterized by accumulation of cystine in the lysosomes throughout the body. In the majority of the patients, this leads to generalized proximal tubular dysfunction (also called DeToni-Debré-Fanconi syndrome) in the first year and progressive renal failure during the first decade. Extrarenal organs are affected by cystinosis as well, with clinical symptoms manifesting mostly after 10 yr of age. The cystine-depleting agent cysteamine significantly improves life expectancy of patients with cystinosis, but offers no cure, pointing to the complexity of the disease mechanism. In this review, current knowledge on the pathogenesis of cystinosis is described and placed in perspective of future research.
Collapse
Affiliation(s)
- Martijn J Wilmer
- Dept. of Pediatric Nephrology, Univ. Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium
| | | | | |
Collapse
|
81
|
Klintworth GK, Jester JV. Genetic basis of corneal diseases and the role of keratocytes in corneal transparency - a review. Clin Exp Ophthalmol 2010. [DOI: 10.1111/j.1442-9071.2010.02365.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
82
|
Kumar A, Bachhawat AK. A futile cycle, formed between two ATP-dependant gamma-glutamyl cycle enzymes, gamma-glutamyl cysteine synthetase and 5-oxoprolinase: the cause of cellular ATP depletion in nephrotic cystinosis? J Biosci 2010; 35:21-5. [PMID: 20413906 DOI: 10.1007/s12038-010-0004-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cystinosis, an inherited disease caused by a defect in the lysosomal cystine transporter (CTNS), is characterized by renal proximal tubular dysfunction. Adenosine triphosphate (ATP) depletion appears to be a key event in the pathophysiology of the disease, even though the manner in which ATP depletion occurs is still a puzzle. We present a model that explains how a futile cycle that is generated between two ATP-utilizing enzymes of the gamma-glutamyl cycle leads to ATP depletion. The enzyme gamma-glutamyl cysteine synthetase (gamma-GCS), in the absence of cysteine, forms 5-oxoproline (instead of the normal substrate, gamma-glutamyl cysteine) and the 5-oxoproline is converted into glutamate by the ATP-dependant enzyme, 5-oxoprolinase. Thus, in cysteine-limiting conditions, glutamate is cycled back into glutamate via 5-oxoproline at the cost of two ATP molecules without production of glutathione and is the cause of the decreased levels of glutathione synthesis, as well as the ATP depletion observed in these cells. The model is also compatible with the differences seen in the human patients and the mouse model of cystinosis, where renal failure is not observed.
Collapse
Affiliation(s)
- Akhilesh Kumar
- Institute of Microbial Technology, Sector 39-A, Chandigarh 160 036, India
| | | |
Collapse
|
83
|
Duchesneau P, Wong AP, Waddell TK. Optimization of targeted cell replacement therapy: a new approach for lung disease. Mol Ther 2010; 18:1830-6. [PMID: 20647998 DOI: 10.1038/mt.2010.142] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cell replacement therapy is a promising approach for treatment of lung disease such as cystic fibrosis, although rates of engraftment need to be improved. We previously showed improved cell retention in the lung using transtracheal delivery compared to intravenous injection. Here, we optimized other parameters of cell delivery using 7-day cultured bone marrow cells (BMCs). Retention of BMC in the lung was dose-dependent. Naphthalene treatment had maximal effects on BMC retention when given 2 days before cell delivery. Naphthalene treatment of the donor amplified a CCSP(+) population and increased retention efficiency in the recipient. Repeated naphthalene treatment and repeated cell delivery both resulted in greater retention. The contribution of the second cell dose was minimal suggesting that a second delivery of BMC promotes proliferation of the first. Busulfan-induced myelosuppression augmented retention of exogenous BMC by up to 20-fold. These BMC helped CCSP reconstitution. Using the optimal delivery techniques and cytokeratin-18-driven green fluorescent protein (GFP) reporter mice, we detected threefold more GFP suggesting more BMC differentiated to epithelial cells. We propose that improved engraftment in the lung will increase cell replacement and thus be a more efficient therapeutic approach for various lung diseases.
Collapse
Affiliation(s)
- Pascal Duchesneau
- Division of Thoracic Surgery, Latner Thoracic Surgery Research Laboratories, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
84
|
Terryn S, Devuyst O, Antignac C. Cell therapy for cystinosis. Nephrol Dial Transplant 2010; 25:2103-6. [PMID: 20395258 DOI: 10.1093/ndt/gfq198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In the September 2009 issue of Blood, Syres et al. [1] report on syngeneic bone marrow cell (BMC) and haematopoietic stem cell (HSC) therapy as a successful treatment in a mouse model of cystinosis, an autosomal recessive metabolic disease caused by a defect in the transport of cystine across the lysosomal membrane. The accumulation of cystine crystals in lysosomes leads to a multi-organ dysfunction including proximal tubulopathy and renal failure, corneal deposits, myopathy and central nervous system defects. By using Ctns knock-out (Ctns(-/-)) mice as a model for cystinosis, Syres et al. show that BMC transplantation leads to a major reduction of cystine content in all tissues tested, reflected by a significant attenuation of the development and progression of kidney injury and reduction in the number of mice with corneal cystine crystals. These changes were correlated with the engraftment of donor BMC producing a functional cystine transporter in the tissues tested. The transplantation of mouse HSC had the same therapeutic effect than whole BMC in this model, which is important as such HSC can readily be isolated from peripheral blood in humans. This work suggests that BMC or HSC transplantation is a potential treatment for cystinosis and other renal tubular disorders.
Collapse
Affiliation(s)
- Sara Terryn
- Division of Nephrology, Université catholique de Louvain Medical School, B-1200 Brussels, Belgium
| | | | | |
Collapse
|