51
|
AlTaisan A, Magliyah M, Abouammoh MA, Taskintuna I, Alzahrani Y, Chang E, Alsulaiman SM. Posterior Segment Characterization in Children With Pierson Syndrome. Ophthalmic Surg Lasers Imaging Retina 2020; 51:618-627. [DOI: 10.3928/23258160-20201104-03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/10/2020] [Indexed: 01/25/2023]
|
52
|
Pierson Syndrome in an Infant With Congenital Nephrotic Syndrome and Unique Brain Pathology. Kidney Int Rep 2020; 5:2371-2374. [PMID: 33305134 PMCID: PMC7710838 DOI: 10.1016/j.ekir.2020.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 11/23/2022] Open
|
53
|
Abstract
The glomerular basement membrane (GBM) is a key component of the glomerular capillary wall and is essential for kidney filtration. The major components of the GBM include laminins, type IV collagen, nidogens and heparan sulfate proteoglycans. In addition, the GBM harbours a number of other structural and regulatory components and provides a reservoir for growth factors. New technologies have improved our ability to study the composition and assembly of basement membranes. We now know that the GBM is a complex macromolecular structure that undergoes key transitions during glomerular development. Defects in GBM components are associated with a range of hereditary human diseases such as Alport syndrome, which is caused by defects in the genes COL4A3, COL4A4 and COL4A5, and Pierson syndrome, which is caused by variants in LAMB2. In addition, the GBM is affected by acquired autoimmune disorders and metabolic diseases such as diabetes mellitus. Current treatments for diseases associated with GBM involvement aim to reduce intraglomerular pressure and to treat the underlying cause where possible. As our understanding about the maintenance and turnover of the GBM improves, therapies to replace GBM components or to stimulate GBM repair could translate into new therapies for patients with GBM-associated disease.
Collapse
|
54
|
Park E, Lee C, Kim NKD, Ahn YH, Park YS, Lee JH, Kim SH, Cho MH, Cho H, Yoo KH, Shin JI, Kang HG, Ha IS, Park WY, Cheong HI. Genetic Study in Korean Pediatric Patients with Steroid-Resistant Nephrotic Syndrome or Focal Segmental Glomerulosclerosis. J Clin Med 2020; 9:jcm9062013. [PMID: 32604935 PMCID: PMC7355646 DOI: 10.3390/jcm9062013] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/16/2020] [Accepted: 06/19/2020] [Indexed: 11/17/2022] Open
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is one of the major causes of end-stage renal disease (ESRD) in childhood and is mostly associated with focal segmental glomerulosclerosis (FSGS). More than 50 monogenic causes of SRNS or FSGS have been identified. Recently, the mutation detection rate in pediatric patients with SRNS has been reported to be approximately 30%. In this study, genotype-phenotype correlations in a cohort of 291 Korean pediatric patients with SRNS/FSGS were analyzed. The overall mutation detection rate was 43.6% (127 of 291 patients). WT1 was the most common causative gene (23.6%), followed by COQ6 (8.7%), NPHS1 (8.7%), NUP107 (7.1%), and COQ8B (6.3%). Mutations in COQ6, NUP107, and COQ8B were more frequently detected, and mutations in NPHS2 were less commonly detected in this cohort than in study cohorts from Western countries. The mutation detection rate was higher in patients with congenital onset, those who presented with proteinuria or chronic kidney disease/ESRD, and those who did not receive steroid treatment. Genetic diagnosis in patients with SRNS provides not only definitive diagnosis but also valuable information for decisions on treatment policy and prediction of prognosis. Therefore, further genotype-phenotype correlation studies are required.
Collapse
Affiliation(s)
- Eujin Park
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
- Department of Pediatrics, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07441, Korea
| | - Chung Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; (C.L.); (N.K.D.K.); (W.-Y.P.)
- GENINUS Inc., Seoul 05836, Korea
| | - Nayoung K. D. Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; (C.L.); (N.K.D.K.); (W.-Y.P.)
- GENINUS Inc., Seoul 05836, Korea
| | - Yo Han Ahn
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
| | - Young Seo Park
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul 05505, Korea; (Y.S.P.); (J.H.L.)
| | - Joo Hoon Lee
- Department of Pediatrics, Asan Medical Center Children’s Hospital, University of Ulsan College of Medicine, Seoul 05505, Korea; (Y.S.P.); (J.H.L.)
| | - Seong Heon Kim
- Department of Pediatrics, Pusan National University Children’s Hospital, Yangsan 50612, Korea;
| | - Min Hyun Cho
- Department of Pediatrics, Kyungpook National University School of Medicine, Daegu 41944, Korea;
| | - Heeyeon Cho
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Kee Hwan Yoo
- Department of Pediatrics, Korea University Guro Hospital, Seoul 02841, Korea;
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 03722, Korea;
- Division of Pediatric Nephrology, Severance Children’s Hospital, Seoul 03722, Korea
| | - Hee Gyung Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
| | - Il-Soo Ha
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Korea; (C.L.); (N.K.D.K.); (W.-Y.P.)
- GENINUS Inc., Seoul 05836, Korea
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Korea
| | - Hae Il Cheong
- Department of Pediatrics, Seoul National University College of Medicine, Seoul 03080, Korea; (E.P.); (Y.H.A.); (H.G.K.); (I.-S.H.)
- Correspondence: ; Tel.: +82-2-2072-2810
| |
Collapse
|
55
|
Jones LK, Lam R, McKee KK, Aleksandrova M, Dowling J, Alexander SI, Mallawaarachchi A, Cottle DL, Short KM, Pais L, Miner JH, Mallett AJ, Simons C, McCarthy H, Yurchenco PD, Smyth IM. A mutation affecting laminin alpha 5 polymerisation gives rise to a syndromic developmental disorder. Development 2020; 147:dev189183. [PMID: 32439764 PMCID: PMC7540250 DOI: 10.1242/dev.189183] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/30/2020] [Indexed: 12/15/2022]
Abstract
Laminin alpha 5 (LAMA5) is a member of a large family of proteins that trimerise and then polymerise to form a central component of all basement membranes. Consequently, the protein plays an instrumental role in shaping the normal development of the kidney, skin, neural tube, lung and limb, and many other organs and tissues. Pathogenic mutations in some laminins have been shown to cause a range of largely syndromic conditions affecting the competency of the basement membranes to which they contribute. We report the identification of a mutation in the polymerisation domain of LAMA5 in a patient with a complex syndromic disease characterised by defects in kidney, craniofacial and limb development, and by a range of other congenital defects. Using CRISPR-generated mouse models and biochemical assays, we demonstrate the pathogenicity of this variant, showing that the change results in a failure of the polymerisation of α/β/γ laminin trimers. Comparing these in vivo phenotypes with those apparent upon gene deletion in mice provides insights into the specific functional importance of laminin polymerisation during development and tissue homeostasis.
Collapse
Affiliation(s)
- Lynelle K Jones
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Rachel Lam
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Karen K McKee
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08901, USA
| | - Maya Aleksandrova
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08901, USA
| | | | - Stephen I Alexander
- Nephrology Department, Centre for Kidney Research, The Children's Hospital at Westmead, Sydney 2145, New South Wales, Australia
| | - Amali Mallawaarachchi
- Department of Medical Genomics, Royal Prince Alfred Hospital; Garvan Institute of Medical Research, Sydney 2010, New South Wales, Australia
| | - Denny L Cottle
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Kieran M Short
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| | - Lynn Pais
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffery H Miner
- Division of Nephrology, Department of Medicine and Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Andrew J Mallett
- Kidney Health Service, Royal Brisbane and Women's Hospital and the Institute for Molecular Bioscience and Faculty of Medicine, The University of Queensland, Brisbane 4072, Queensland, Australia
| | - Cas Simons
- Murdoch Children's Research Institute, The Royal Children's Hospital Melbourne, Melbourne 3052, Victoria, Australia
| | - Hugh McCarthy
- The Sydney Children's Hospitals Network and the Children's Hospital Westmead Clinical School, University of Sydney, Sydney 2145, New South Wales, Australia
| | - Peter D Yurchenco
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08901, USA
| | - Ian M Smyth
- Department of Anatomy and Developmental Biology, Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia
| |
Collapse
|
56
|
Little MH, Quinlan C. Advances in our understanding of genetic kidney disease using kidney organoids. Pediatr Nephrol 2020; 35:915-926. [PMID: 31065797 DOI: 10.1007/s00467-019-04259-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/27/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022]
Abstract
A significant proportion of kidney disease presenting in childhood is likely genetic in origin with a growing number of genes implicated in its development. However, many children may have changes in previously undescribed or unrecognised genes. The recent development of methods for generating human kidney organoids from human pluripotent stem cells has the potential to substantially change the rate of diagnosis and the development of new treatments for some forms of genetic kidney disease. In this review, we discuss how accurately a kidney organoid models the human kidney, identifying the strengths and weaknesses of these potentially patient-derived models of renal disease.
Collapse
Affiliation(s)
- Melissa H Little
- Murdoch Children's Research Institute, Flemington Rd., Parkville, VIC, Australia. .,Department of Anatomy and Neuroscience, The University of Melbourne, Melbourne, VIC, Australia. .,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.
| | - Catherine Quinlan
- Murdoch Children's Research Institute, Flemington Rd., Parkville, VIC, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia.,Department of Nephrology, Royal Children's Hospital, Flemington Rd., Parkville, VIC, Australia
| |
Collapse
|
57
|
Dufek S, Holtta T, Trautmann A, Ylinen E, Alpay H, Ariceta G, Aufricht C, Bacchetta J, Bakkaloglu SA, Bayazit A, Cicek RY, Dursun I, Duzova A, Ekim M, Iancu D, Jankauskiene A, Klaus G, Paglialonga F, Pasini A, Printza N, Said Conti V, do Sameiro Faria M, Schmitt CP, Stefanidis CJ, Verrina E, Vidal E, Vondrak K, Webb H, Zampetoglou A, Bockenhauer D, Edefonti A, Shroff R. Management of children with congenital nephrotic syndrome: challenging treatment paradigms. Nephrol Dial Transplant 2020; 34:1369-1377. [PMID: 30215773 DOI: 10.1093/ndt/gfy165] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/24/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Management of children with congenital nephrotic syndrome (CNS) is challenging. Bilateral nephrectomies followed by dialysis and transplantation are practiced in most centres, but conservative treatment may also be effective. METHODS We conducted a 6-year review across members of the European Society for Paediatric Nephrology Dialysis Working Group to compare management strategies and their outcomes in children with CNS. RESULTS Eighty children (50% male) across 17 tertiary nephrology units in Europe were included (mutations in NPHS1, n = 55; NPHS2, n = 1; WT1, n = 9; others, n = 15). Excluding patients with mutations in WT1, antiproteinuric treatment was given in 42 (59%) with an increase in S-albumin in 70% by median 6 (interquartile range: 3-8) g/L (P < 0.001). Following unilateral nephrectomy, S-albumin increased by 4 (1-8) g/L (P = 0.03) with a reduction in albumin infusion dose by 5 (2-9) g/kg/week (P = 0.02). Median age at bilateral nephrectomies (n = 29) was 9 (7-16) months. Outcomes were compared between two groups of NPHS1 patients: those who underwent bilateral nephrectomies (n = 25) versus those on conservative management (n = 17). The number of septic or thrombotic episodes and growth were comparable between the groups. The response to antiproteinuric treatment, as well as renal and patient survival, was independent of NPHS1 mutation type. At final follow-up (median age 34 months) 20 (80%) children in the nephrectomy group were transplanted and 1 died. In the conservative group, 9 (53%) remained without dialysis, 4 (24%; P < 0.001) were transplanted and 2 died. CONCLUSION An individualized, stepwise approach with prolonged conservative management may be a reasonable alternative to early bilateral nephrectomies and dialysis in children with CNS and NPHS1 mutations. Further prospective studies are needed to define indications for unilateral nephrectomy.
Collapse
Affiliation(s)
- Stephanie Dufek
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Tuula Holtta
- Department of Pediatric Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Agnes Trautmann
- Center for Pediatric & Adolescent Medicine, Heidelberg, Germany
| | - Elisa Ylinen
- Department of Pediatric Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harika Alpay
- School of Medicine, Marmara University, Istanbul, Turkey
| | - Gema Ariceta
- Hospital MaternoInfantil de la Vall d'Hebron, Barcelona, Spain
| | | | | | - Sevcan A Bakkaloglu
- Department of Pediatric Nephrology, Gazi University Hospital, Ankara, Turkey
| | - Aysun Bayazit
- Department of Pediatric Nephrology, Cukurova University, Adana, Turkey
| | | | - Ismail Dursun
- Department of Pediatric Nephrology, Erciyes University, Kayseri, Turkey
| | - Ali Duzova
- Division of Pediatric Nephrology, Hacettepe University Faculty of Medicine, Sihhiye, Ankara, Turkey
| | | | - Daniela Iancu
- Center for Nephrology, University College London, London, UK
| | | | | | - Fabio Paglialonga
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Pasini
- Azienda Ospedaliero-Universitaria Sant'Orsola-Malpighi, Bologna, Italy
| | - Nikoleta Printza
- Hippokratio General Hospital, Aristotle University, Thessaloniki, Greece
| | | | | | | | | | | | - Enrico Vidal
- Department of Pediatrics, University Hospital of Padova, Padova, Italy
| | - Karel Vondrak
- Pediatric Nephrology, University Hospital Motol, Prague, Czech Republic
| | - Hazel Webb
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | | | - Detlef Bockenhauer
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Alberto Edefonti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Rukshana Shroff
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| |
Collapse
|
58
|
Butt L, Unnersjö-Jess D, Höhne M, Edwards A, Binz-Lotter J, Reilly D, Hahnfeldt R, Ziegler V, Fremter K, Rinschen MM, Helmstädter M, Ebert LK, Castrop H, Hackl MJ, Walz G, Brinkkoetter PT, Liebau MC, Tory K, Hoyer PF, Beck BB, Brismar H, Blom H, Schermer B, Benzing T. A molecular mechanism explaining albuminuria in kidney disease. Nat Metab 2020; 2:461-474. [PMID: 32694662 DOI: 10.1038/s42255-020-0204-y] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/07/2020] [Indexed: 01/18/2023]
Abstract
Mammalian kidneys constantly filter large amounts of liquid, with almost complete retention of albumin and other macromolecules in the plasma. Breakdown of the three-layered renal filtration barrier results in loss of albumin into urine (albuminuria) across the wall of small renal capillaries, and is a leading cause of chronic kidney disease. However, exactly how the renal filter works and why its permeability is altered in kidney diseases is poorly understood. Here we show that the permeability of the renal filter is modulated through compression of the capillary wall. We collect morphometric data prior to and after onset of albuminuria in a mouse model equivalent to a human genetic disease affecting the renal filtration barrier. Combining quantitative analyses with mathematical modelling, we demonstrate that morphological alterations of the glomerular filtration barrier lead to reduced compressive forces that counteract filtration pressure, thereby resulting in capillary dilatation, and ultimately albuminuria. Our results reveal distinct functions of the different layers of the filtration barrier and expand the molecular understanding of defective renal filtration in chronic kidney disease.
Collapse
Affiliation(s)
- Linus Butt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - David Unnersjö-Jess
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Royal Institute of Technology, Stockholm, Sweden
| | - Martin Höhne
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Aurelie Edwards
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Julia Binz-Lotter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Dervla Reilly
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Robert Hahnfeldt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Vera Ziegler
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Katharina Fremter
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Center for Mass Spectrometry and Metabolomics, The Scripps Research Institute, La Jolla, CA, USA
| | - Martin Helmstädter
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, and Signalling Research Centres BIOSS and CIBSS, University of Freiburg,, Freiburg, Germany
| | - Lena K Ebert
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Hayo Castrop
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Matthias J Hackl
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Gerd Walz
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, and Signalling Research Centres BIOSS and CIBSS, University of Freiburg,, Freiburg, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Max C Liebau
- Department of Pediatrics and Center for Molecular Medicine, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Kálmán Tory
- MTA-SE Lendület Nephrogenetic Laboratory, 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Peter F Hoyer
- University Children's Hospital, Clinic for Pediatrics II, University of Duisburg-Essen, Essen, Germany
| | - Bodo B Beck
- Institute of Human Genetics and Center for Molecular Medicine, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | | | - Hans Blom
- Royal Institute of Technology, Stockholm, Sweden
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
59
|
Nishiyama K, Kurokawa M, Torio M, Sakai Y, Arima M, Tsukamoto S, Obata S, Minamikawa S, Nozu K, Kaku N, Maehara Y, Sonoda KH, Taguchi T, Ohga S. Gastrointestinal symptoms as an extended clinical feature of Pierson syndrome: a case report and review of the literature. BMC MEDICAL GENETICS 2020; 21:80. [PMID: 32295525 PMCID: PMC7160948 DOI: 10.1186/s12881-020-01019-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 04/01/2020] [Indexed: 11/17/2022]
Abstract
Background Pierson syndrome (PS) is a rare autosomal recessive disorder, characterized by congenital nephrotic syndrome and microcoria. Advances in renal replacement therapies have extended the lifespan of patients, whereas the full clinical spectrum of PS in infancy and beyond remains elusive. Case presentation We present the case of a 12-month-old boy with PS, manifesting as the bilateral microcoria and congenital nephrotic syndrome. He was born without asphyxia, and was neurologically intact from birth through the neonatal period. Generalized muscle weakness and hypotonia were recognized from 3 months of age. The infant showed recurrent vomiting at age 5 months of age, and was diagnosed with gastroesophageal reflux and intestinal malrotation. Despite the successful surgical treatment, vomiting persisted and led to severely impaired growth. Tulobuterol treatment was effective in reducing the frequency of vomiting. Targeted sequencing confirmed that he had a compound heterozygous mutation in LAMB2 (NM_002292.3: p.Arg550X and p.Glu1507X). A search of the relevant literature identified 19 patients with severe neuro-muscular phenotypes. Among these, only 8 survived the first 12 months of life, and one had feeding difficulty with similar gastrointestinal problems. Conclusions This report demonstrated that severe neurological deficits and gastrointestinal dysfunction may emerge in PS patients after the first few months of life.
Collapse
Affiliation(s)
- Kei Nishiyama
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mari Kurokawa
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Michiko Torio
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Mitsuru Arima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoko Tsukamoto
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Satoshi Obata
- Department of Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shogo Minamikawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Noriyuki Kaku
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.,Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshihiko Maehara
- Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomoaki Taguchi
- Department of Pediatric Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
60
|
Gribouval O, Boyer O, Knebelmann B, Karras A, Dantal J, Fourrage C, Alibeu O, Hogan J, Dossier C, Tête MJ, Antignac C, Servais A. APOL1 risk genotype in European steroid-resistant nephrotic syndrome and/or focal segmental glomerulosclerosis patients of different African ancestries. Nephrol Dial Transplant 2020; 34:1885-1893. [PMID: 29992269 DOI: 10.1093/ndt/gfy176] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/03/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Apolipoprotein L1 (APOL1) risk variants are strongly associated with sporadic focal segmental glomerulosclerosis (FSGS) in populations with African ancestry. We determined the frequency of G1/G2 variants in steroid-resistant nephrotic syndrome (SRNS)/FSGS patients with African or French West Indies ancestry in France and its relationships with other SRNS genes. METHODS In a cohort of 152 patients (139 families), the APOL1 risk variants were genotyped by direct Sanger sequencing and pathogenic mutations were screened by next-generation sequencing with a panel including 35 SRNS genes. RESULTS The two risk allele [high-risk (HR)] genotypes were found in 43.1% (66/152) of subjects compared with 18.9% (106/562) in a control population (P < 0.0001): 33 patients homozygous for APOL1 G1 alleles, 4 homozygous for G2 and 29 compound heterozygous for G1 and G2. Compared with patients in the low-risk (LR) group, patients in the HR group were more likely to originate from the French West Indies than from Africa [45/66 (68.2%) versus 30/86 (34.9%); P < 0.0001]. There were more familial cases in the HR group [27 (41.5%) versus 8 (11.4%); P < 0.0001]. However, causative mutations in monogenic SRNS genes were found in only 1 patient in the HR group compared with 16 patients (14 families) in the LR group (P = 0.0006). At diagnosis, patients in the HR group without other mutations were more often adults [35 (53.8%) versus 19 (27.1%); P = 0.003] and had a lower estimated glomerular filtration rate (78.9 versus 98.8 mL/min/1.73 m2; P = 0.02). CONCLUSIONS The HR genotype is frequent in FSGS patients with African ancestry in our cohort, especially in those originating from the West Indies, and confer a poor renal prognosis. It is usually not associated with other causative mutations in monogenic SRNS genes.
Collapse
Affiliation(s)
- Olivier Gribouval
- Inserm U1163, Institut Imagine, University Paris Descartes, Paris, France
| | - Olivia Boyer
- Inserm U1163, Institut Imagine, University Paris Descartes, Paris, France.,Pediatric Nephrology Department, Necker Hospital, APHP, Paris, France
| | - Bertrand Knebelmann
- Nephrology and Transplantation Department, Necker Hospital, APHP, Paris, France
| | - Alexandre Karras
- Nephrology Department, European Georges Pompidou Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Jacques Dantal
- Nephrology Department, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Cécile Fourrage
- Bioinformatic Platform, Paris Descartes Sorbonne Paris Cité University, Imagine Institute, Paris, France
| | - Olivier Alibeu
- Genomic Platform, Inserm UMR1163, Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Julien Hogan
- Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Claire Dossier
- Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marie Josèphe Tête
- Inserm U1163, Institut Imagine, University Paris Descartes, Paris, France
| | - Corinne Antignac
- Inserm U1163, Institut Imagine, University Paris Descartes, Paris, France.,Genetic Department, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Aude Servais
- Inserm U1163, Institut Imagine, University Paris Descartes, Paris, France.,Nephrology and Transplantation Department, Necker Hospital, APHP, Paris, France
| |
Collapse
|
61
|
Pozza E, Verdin H, Deconinck H, Dheedene A, Menten B, De Baere E, Balikova I. Microcoria due to first duplication of 13q32.1 including the GPR180 gene and maternal mosaicism. Eur J Med Genet 2020; 63:103918. [PMID: 32200002 DOI: 10.1016/j.ejmg.2020.103918] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/16/2020] [Indexed: 11/28/2022]
Abstract
Congenital microcoria (MCOR) is an eye anomaly characterized by a pupil with diameter below 2 mm, and is caused by underdevelopment or absence of the dilator muscle of the pupil. Two types have been described: a recessive, syndromic (Pierson syndrome OMIM 609049) and a dominant, isolated form (MCOR syndrome OMIM 156600). Fares-Taie and colleagues described inherited microdeletions in chromosome band 13q32.1 segregating with dominant microcoria in several families. The GPR180 gene is located within the smallest commonly deleted region and encodes a G protein-coupled receptor involved in smooth muscle cells growth. We here describe a patient with isolated, non-syndromic MCOR. The patient presented with a blue iris and small pupils, non-reactive to cycloplegic agents. Her mother had a milder ocular phenotype, namely a blue iris with hypoplastic crypts and mild myopia. We present a detailed clinical examination and follow up. DNA from the index patient was analyzed for the presence of chromosomal imbalances using molecular karyotyping. The genetic test revealed a small duplication of chromosome band 13q32.1. The duplication affected a 289 kb region, encompassing 11 genes including GPR180. Interestingly, the patient displays only MCOR in contrast to patients with the reciprocal deletion who present with MCOR and iridocorneal angle dysgenesis. This genetic anomaly was inherited from the mother who carries the duplication in mosaic form, which should be considered when offering genetic counselling. In summary, we describe the first 13q32.1 duplication encompassing GPR180 associated with MCOR.
Collapse
Affiliation(s)
- Elise Pozza
- Department of Ophthalmology, Children Hospital Queen Fabiola, Brussels, Belgium
| | - Hannah Verdin
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium University of Ghent, Ghent, Belgium
| | | | - Annelies Dheedene
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium University of Ghent, Ghent, Belgium
| | - Björn Menten
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium University of Ghent, Ghent, Belgium
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium University of Ghent, Ghent, Belgium
| | - Irina Balikova
- Department of Ophthalmology, Children Hospital Queen Fabiola, Brussels, Belgium; Department of Ophthalmology, Ghent University Hospital, Belgium; Department of Ophthalmology, Leuven University Hospital, Belgium.
| |
Collapse
|
62
|
Tahoun M, Chandler JC, Ashton E, Haston S, Hannan A, Kim JS, D’Arco F, Bockenhauer D, Anderson G, Lin MH, Marzouk S, Saied MH, Miner JH, Dattani MT, Waters AM. Mutations in LAMB2 Are Associated With Albuminuria and Optic Nerve Hypoplasia With Hypopituitarism. J Clin Endocrinol Metab 2020; 105:5643661. [PMID: 31769495 PMCID: PMC7048679 DOI: 10.1210/clinem/dgz216] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 11/25/2019] [Indexed: 01/15/2023]
Abstract
CONTEXT Mutations in LAMB2, encoding the basement membrane protein, laminin β2, are associated with an autosomal recessive disorder characterized by congenital nephrotic syndrome, ocular abnormalities, and neurodevelopmental delay (Pierson syndrome). CASE DESCRIPTION This report describes a 12-year-old boy with short stature, visual impairment, and developmental delay who presented with macroscopic hematuria and albuminuria. He had isolated growth hormone deficiency, optic nerve hypoplasia, and a small anterior pituitary with corpus callosum dysgenesis on his cranial magnetic resonance imaging, thereby supporting a diagnosis of optic nerve hypoplasia syndrome. Renal histopathology revealed focal segmental glomerulosclerosis. Using next-generation sequencing on a targeted gene panel for steroid-resistant nephrotic syndrome, compound heterozygous missense mutations were identified in LAMB2 (c.737G>A p.Arg246Gln, c.3982G>C p.Gly1328Arg). Immunohistochemical analysis revealed reduced glomerular laminin β2 expression compared to control kidney and a thin basement membrane on electron microscopy. Laminin β2 is expressed during pituitary development and Lamb2-/- mice exhibit stunted growth, abnormal neural retinae, and here we show, abnormal parenchyma of the anterior pituitary gland. CONCLUSION We propose that patients with genetically undefined optic nerve hypoplasia syndrome should be screened for albuminuria and, if present, screened for mutations in LAMB2.
Collapse
Affiliation(s)
- Mona Tahoun
- Clinical and Chemical Pathology Department, Faculty of Medicine, Alexandria University, Egypt
| | - Jennifer C Chandler
- UCL Great Ormond Street Institute of Child Health, University College London, UK
| | - Emma Ashton
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Scott Haston
- UCL Great Ormond Street Institute of Child Health, University College London, UK
| | - Athia Hannan
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Ji Soo Kim
- UCL Great Ormond Street Institute of Child Health, University College London, UK
| | - Felipe D’Arco
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - D Bockenhauer
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - G Anderson
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Meei-Hua Lin
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Salah Marzouk
- Clinical and Chemical Pathology Department, Faculty of Medicine, Alexandria University, Egypt
| | - Marwa H Saied
- Clinical and Chemical Pathology Department, Faculty of Medicine, Alexandria University, Egypt
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Mehul T Dattani
- UCL Great Ormond Street Institute of Child Health, University College London, UK
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Aoife M Waters
- UCL Great Ormond Street Institute of Child Health, University College London, UK
- Great Ormond Street Hospital NHS Foundation Trust, London, UK
- Correspondence: Aoife Waters, MB, BAO, MSc, PhD, FRCPCH, Programme of Developmental Biology of Birth Defects, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; Consultant Paediatric Nephrologist, Great Ormond Street Hospital NHS Foundation Trust, London, UK. E-mail: ;
| |
Collapse
|
63
|
Funk SD, Bayer RH, McKee KK, Okada K, Nishimune H, Yurchenco PD, Miner JH. A deletion in the N-terminal polymerizing domain of laminin β2 is a new mouse model of chronic nephrotic syndrome. Kidney Int 2020; 98:133-146. [PMID: 32456966 DOI: 10.1016/j.kint.2020.01.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 01/14/2020] [Accepted: 01/21/2020] [Indexed: 10/25/2022]
Abstract
The importance of the glomerular basement membrane (GBM) in glomerular filtration is underscored by the manifestations of Alport and Pierson syndromes, caused by defects in type IV collagen α3α4α5 and the laminin β2 chain, respectively. Lamb2 null mice, which model the most severe form of Pierson syndrome, exhibit proteinuria prior to podocyte foot process effacement and are therefore useful for studying GBM permselectivity. We hypothesize that some LAMB2 missense mutations that cause mild forms of Pierson syndrome induce GBM destabilization with delayed effects on podocytes. While generating a CRISPR/Cas9-mediated analogue of a human LAMB2 missense mutation in mice, we identified a 44-amino acid deletion (LAMB2-Del44) within the laminin N-terminal domain, a domain mediating laminin polymerization. Laminin heterotrimers containing LAMB2-Del44 exhibited a 90% reduction in polymerization in vitro that was partially rescued by type IV collagen and nidogen. Del44 mice showed albuminuria at 1.8-6.0 g/g creatinine (ACR) at one to two months, plateauing at an average 200 g/g ACR at 3.7 months, when GBM thickening and hallmarks of nephrotic syndrome were first observed. Despite the massive albuminuria, some Del44 mice survived for up to 15 months. Blood urea nitrogen was modestly elevated at seven-nine months. Eight to nine-month-old Del44 mice exhibited glomerulosclerosis and interstitial fibrosis. Similar to Lamb2-/- mice, proteinuria preceded foot process effacement. Foot processes were widened but not effaced at one-two months despite the high ACRs. At three months some individual foot processes were still observed amid widespread effacement. Thus, our chronic model of nephrotic syndrome may prove useful to study filtration mechanisms, long-term proteinuria with preserved kidney function, and to test therapeutics.
Collapse
Affiliation(s)
- Steven D Funk
- Department of Internal Medicine, Division of Nephrology, Washington University, St. Louis, Missouri, USA
| | - Raymond H Bayer
- Department of Internal Medicine, Division of Nephrology, Washington University, St. Louis, Missouri, USA
| | - Karen K McKee
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Kazushi Okada
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Hiroshi Nishimune
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Peter D Yurchenco
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA
| | - Jeffrey H Miner
- Department of Internal Medicine, Division of Nephrology, Washington University, St. Louis, Missouri, USA.
| |
Collapse
|
64
|
Minamikawa S, Miwa S, Inagaki T, Nishiyama K, Kaito H, Ninchoji T, Yamamura T, Nagano C, Sakakibara N, Ishimori S, Hara S, Yoshikawa N, Hirano D, Harada R, Hamada R, Matsunoshita N, Nagata M, Shima Y, Nakanishi K, Nagase H, Takeda H, Morisada N, Iijima K, Nozu K. Molecular mechanisms determining severity in patients with Pierson syndrome. J Hum Genet 2020; 65:355-362. [PMID: 31959872 DOI: 10.1038/s10038-019-0715-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 01/15/2023]
Abstract
Null variants in LAMB2 cause Pierson syndrome (PS), a severe congenital nephrotic syndrome with ocular and neurological defects. Patients' kidney specimens show complete negativity for laminin β2 expression on glomerular basement membrane (GBM). In contrast, missense variants outside the laminin N-terminal (LN) domain in LAMB2 lead to milder phenotypes. However, we experienced cases not showing these typical genotype-phenotype correlations. In this paper, we report six PS patients: four with mild phenotypes and two with severe phenotypes. We conducted molecular studies including protein expression and transcript analyses. The results revealed that three of the four cases with milder phenotypes had missense variants located outside the LN domain and one of the two severe PS cases had a homozygous missense variant located in the LN domain; these variant positions could explain their phenotypes. However, one mild case possessed a splicing site variant (c.3797 + 5G>A) that should be associated with a severe phenotype. Upon transcript analysis, this variant generated some differently sized transcripts, including completely normal transcript, which could have conferred the milder phenotype. In one severe case, we detected the single-nucleotide substitution of c.4616G>A located outside the LN domain, which should be associated with a milder phenotype. However, we detected aberrant splicing caused by the creation of a novel splice site by this single-base substitution. These are novel mechanisms leading to an atypical genotype-phenotype correlation. In addition, all four cases with milder phenotypes showed laminin β2 expression on GBM. We identified novel mechanisms leading to atypical genotype-phenotype correlation in PS.
Collapse
Affiliation(s)
- Shogo Minamikawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Saori Miwa
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
| | - Tetsuji Inagaki
- Department of Pediatric Nephrology, Miyagi Children's Hospital, Sendai, Japan
| | - Kei Nishiyama
- Department of Pediatrics, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Hiroshi Kaito
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takeshi Ninchoji
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomohiko Yamamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - China Nagano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Nana Sakakibara
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shingo Ishimori
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shigeo Hara
- Department of Diagnostic Pathology, Kobe City Medical Center General Hospital, Kobe, Japan
| | | | - Daishi Hirano
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan
| | - Ryoko Harada
- Department of Nephrology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | - Riku Hamada
- Department of Nephrology, Tokyo Metropolitan Children's Medical Center, Tokyo, Japan
| | | | - Michio Nagata
- Department of Kidney and Vascular Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuko Shima
- Department of Pediatrics, Wakayama Medical University, Wakayama, Japan
| | - Koichi Nakanishi
- Department of Child Health and Welfare (Pediatrics), Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| | - Hiroaki Nagase
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroki Takeda
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoya Morisada
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
65
|
Congenital nephrotic syndrome: is early aggressive treatment needed? Yes. Pediatr Nephrol 2020; 35:1985-1990. [PMID: 32377865 PMCID: PMC7501131 DOI: 10.1007/s00467-020-04578-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 04/05/2020] [Accepted: 04/16/2020] [Indexed: 12/24/2022]
Abstract
Congenital nephrotic syndrome (CNS) was primarily considered one disease entity. Hence, one treatment protocol was proposed in the beginning to all CNS patients. Today, with the help of gene diagnostics, we know that CNS is a heterogeneous group of disorders and therefore, different treatment protocols are needed. The most important gene defects causing CNS are NPHS1, NPHS2, WT1, LAMB2, and PLCE1. Before active treatment, all infants with CNS died. It was stated already in the mid-1980s that intensive medical therapy followed by kidney transplantation (KTx) should be the choice of treatment for infants with severe CNS. In Finland, early aggressive treatment protocol was adopted from the USA and further developed for treatment of children with the Finnish type of CNS. The aim of this review is to state reasons for "early aggressive treatment" including daily albumin infusions, intensified nutrition, and timely bilateral nephrectomy followed by KTx at the age of 1-2 years.
Collapse
|
66
|
Kuure S, Sariola H. Mouse Models of Congenital Kidney Anomalies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1236:109-136. [PMID: 32304071 DOI: 10.1007/978-981-15-2389-2_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are common birth defects, which cause the majority of chronic kidney diseases in children. CAKUT covers a wide range of malformations that derive from deficiencies in embryonic kidney and lower urinary tract development, including renal aplasia, hypodysplasia, hypoplasia, ectopia, and different forms of ureter abnormalities. The majority of the genetic causes of CAKUT remain unknown. Research on mutant mice has identified multiple genes that critically regulate renal differentiation. The data generated from this research have served as an excellent resource to identify the genetic bases of human kidney defects and have led to significantly improved diagnostics. Furthermore, genetic data from human CAKUT studies have also revealed novel genes regulating kidney differentiation.
Collapse
Affiliation(s)
- Satu Kuure
- GM-Unit, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland. .,Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland. .,Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Hannu Sariola
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Paediatric Pathology, HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|
67
|
Schapiro D, Daga A, Lawson JA, Majmundar AJ, Lovric S, Tan W, Warejko JK, Fessi I, Rao J, Airik M, Gee HY, Schneider R, Widmeier E, Hermle T, Ashraf S, Jobst-Schwan T, van der Ven AT, Nakayama M, Shril S, Braun DA, Hildebrandt F. Panel sequencing distinguishes monogenic forms of nephritis from nephrosis in children. Nephrol Dial Transplant 2019; 34:474-485. [PMID: 30295827 DOI: 10.1093/ndt/gfy050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/21/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Alport syndrome (AS) and atypical hemolytic-uremic syndrome (aHUS) are rare forms of chronic kidney disease (CKD) that can lead to a severe decline of renal function. Steroid-resistant nephrotic syndrome (SRNS) is more common than AS and aHUS and causes 10% of childhood-onset CKD. In recent years, multiple monogenic causes of AS, aHUS and SRNS have been identified, but their relative prevalence has yet to be studied together in a typical pediatric cohort of children with proteinuria and hematuria. We hypothesized that identification of causative mutations by whole exome sequencing (WES) in known monogenic nephritis and nephrosis genes would allow distinguishing nephritis from nephrosis in a typical pediatric group of patients with both proteinuria and hematuria at any level. METHODS We therefore conducted an exon sequencing (WES) analysis for 11 AS, aHUS and thrombotic thrombocytopenic purpura-causing genes in an international cohort of 371 patients from 362 families presenting with both proteinuria and hematuria before age 25 years. In parallel, we conducted either WES or high-throughput exon sequencing for 23 SRNS-causing genes in all patients. RESULTS We detected pathogenic mutations in 18 of the 34 genes analyzed, leading to a molecular diagnosis in 14.1% of families (51 of 362). Disease-causing mutations were detected in 3 AS-causing genes (4.7%), 3 aHUS-causing genes (1.4%) and 12 NS-causing genes (8.0%). We observed a much higher mutation detection rate for monogenic forms of CKD in consanguineous families (35.7% versus 10.1%). CONCLUSIONS We present the first estimate of relative frequency of inherited AS, aHUS and NS in a typical pediatric cohort with proteinuria and hematuria. Important therapeutic and preventative measures may result from mutational analysis in individuals with proteinuria and hematuria.
Collapse
Affiliation(s)
- David Schapiro
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ankana Daga
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer A Lawson
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amar J Majmundar
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Svjetlana Lovric
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Weizhen Tan
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jillian K Warejko
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Inés Fessi
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jia Rao
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Merlin Airik
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heon Yung Gee
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ronen Schneider
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eugen Widmeier
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tobias Hermle
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shazia Ashraf
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tilman Jobst-Schwan
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amelie T van der Ven
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Makiko Nakayama
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shirlee Shril
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniela A Braun
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Friedhelm Hildebrandt
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
68
|
Cinà DP, Ketela T, Brown KR, Chandrashekhar M, Mero P, Li C, Onay T, Fu Y, Han Z, Saleem M, Moffat J, Quaggin SE. Forward genetic screen in human podocytes identifies diphthamide biosynthesis genes as regulators of adhesion. Am J Physiol Renal Physiol 2019; 317:F1593-F1604. [PMID: 31566424 PMCID: PMC6962514 DOI: 10.1152/ajprenal.00195.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/28/2019] [Accepted: 09/25/2019] [Indexed: 02/06/2023] Open
Abstract
Podocyte function is tightly linked to the complex organization of its cytoskeleton and adhesion to the underlying glomerular basement membrane. Adhesion of cultured podocytes to a variety of substrates is reported to correlate with podocyte health. To identify novel genes that are important for podocyte function, we designed an in vitro genetic screen based on podocyte adhesion to plates coated with either fibronectin or soluble Fms-like tyrosine kinase-1 (sFLT1)/Fc. A genome-scale pooled RNA interference screen on immortalized human podocytes identified 77 genes that increased adhesion to fibronectin, 101 genes that increased adhesion to sFLT1/Fc, and 44 genes that increased adhesion to both substrates when knocked down. Multiple shRNAs against diphthamide biosynthesis protein 1-4 (DPH1-DPH4) were top hits for increased adhesion. Immortalized human podocyte cells stably expressing these hairpins displayed increased adhesion to both substrates. We then used CRISPR-Cas9 to generate podocyte knockout cells for DPH1, DPH2, or DPH3, which also displayed increased adhesion to both fibronectin and sFLT1/Fc, as well as a spreading defect. Finally, we showed that Drosophila nephrocyte-specific knockdown of Dph1, Dph2, and Dph4 resulted in altered nephrocyte function. In summary, we report here a novel high-throughput method to identify genes important for podocyte function. Given the central role of podocyte adhesion as a marker of podocyte health, these data are a rich source of candidate regulators of glomerular disease.
Collapse
Affiliation(s)
- Davide P Cinà
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Troy Ketela
- Donnelly Centre, Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Kevin R Brown
- Donnelly Centre, Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Megha Chandrashekhar
- Donnelly Centre, Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Patricia Mero
- Donnelly Centre, Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Chengjin Li
- Tanenbaum-Lunenfeld Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Tuncer Onay
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Yulong Fu
- Center for Genetic Medicine Research, Children's National Health System, Washington, District of Columbia
| | - Zhe Han
- Center for Genetic Medicine Research, Children's National Health System, Washington, District of Columbia
| | - Moin Saleem
- School of Clinical Sciences, Children's Renal Unit and Academic Renal Unit, University of Bristol, Bristol, United Kingdom
| | - Jason Moffat
- Donnelly Centre, Banting and Best Department of Medical Research, University of Toronto, Toronto, Ontario, Canada
| | - Susan E Quaggin
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
69
|
Kampf LL, Schneider R, Gerstner L, Thünauer R, Chen M, Helmstädter M, Amar A, Onuchic-Whitford AC, Loza Munarriz R, Berdeli A, Müller D, Schrezenmeier E, Budde K, Mane S, Laricchia KM, Rehm HL, MacArthur DG, Lifton RP, Walz G, Römer W, Bergmann C, Hildebrandt F, Hermle T. TBC1D8B Mutations Implicate RAB11-Dependent Vesicular Trafficking in the Pathogenesis of Nephrotic Syndrome. J Am Soc Nephrol 2019; 30:2338-2353. [PMID: 31732614 DOI: 10.1681/asn.2019040414] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Mutations in about 50 genes have been identified as monogenic causes of nephrotic syndrome, a frequent cause of CKD. These genes delineated the pathogenetic pathways and rendered significant insight into podocyte biology. METHODS We used whole-exome sequencing to identify novel monogenic causes of steroid-resistant nephrotic syndrome (SRNS). We analyzed the functional significance of an SRNS-associated gene in vitro and in podocyte-like Drosophila nephrocytes. RESULTS We identified hemizygous missense mutations in the gene TBC1D8B in five families with nephrotic syndrome. Coimmunoprecipitation assays indicated interactions between TBC1D8B and active forms of RAB11. Silencing TBC1D8B in HEK293T cells increased basal autophagy and exocytosis, two cellular functions that are independently regulated by RAB11. This suggests that TBC1D8B plays a regulatory role by inhibiting endogenous RAB11. Coimmunoprecipitation assays showed TBC1D8B also interacts with the slit diaphragm protein nephrin, and colocalizes with it in immortalized cell lines. Overexpressed murine Tbc1d8b with patient-derived mutations had lower affinity for endogenous RAB11 and nephrin compared with wild-type Tbc1d8b protein. Knockdown of Tbc1d8b in Drosophila impaired function of the podocyte-like nephrocytes, and caused mistrafficking of Sns, the Drosophila ortholog of nephrin. Expression of Rab11 RNAi in nephrocytes entailed defective delivery of slit diaphragm protein to the membrane, whereas RAB11 overexpression revealed a partial phenotypic overlap to Tbc1d8b loss of function. CONCLUSIONS Novel mutations in TBC1D8B are monogenic causes of SRNS. This gene inhibits RAB11. Our findings suggest that RAB11-dependent vesicular nephrin trafficking plays a role in the pathogenesis of nephrotic syndrome.
Collapse
Affiliation(s)
- Lina L Kampf
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Ronen Schneider
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lea Gerstner
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Roland Thünauer
- Signalling Research Centres BIOSS and CIBSS and Faculty of Biology, University of Freiburg, Freiburg, Germany.,Advanced Light and Fluorescence Microscopy Facility, Centre for Structural Systems Biology (CSSB) and University of Hamburg, Hamburg, Germany
| | - Mengmeng Chen
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Martin Helmstädter
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Ali Amar
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ana C Onuchic-Whitford
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts.,Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Afig Berdeli
- Department of Pediatrics, Molecular Medicine Laboratory, Ege University, Izmir, Turkey
| | - Dominik Müller
- Department of Pediatric Nephrology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Shrikant Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | - Kristen M Laricchia
- Broad Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge
| | - Heidi L Rehm
- Broad Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge
| | - Daniel G MacArthur
- Broad Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.,Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York
| | - Gerd Walz
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Winfried Römer
- Signalling Research Centres BIOSS and CIBSS and Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Carsten Bergmann
- Center for Human Genetics, Mainz, Germany.,Center for Human Genetics, Bioscientia, Ingelheim, Germany; and.,Department of Medicine, University Hospital Freiburg, Freiburg, Germany
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts;
| | - Tobias Hermle
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany;
| |
Collapse
|
70
|
Hall TE, Wood AJ, Ehrlich O, Li M, Sonntag CS, Cole NJ, Huttner IG, Sztal TE, Currie PD. Cellular rescue in a zebrafish model of congenital muscular dystrophy type 1A. NPJ Regen Med 2019; 4:21. [PMID: 31754462 PMCID: PMC6858319 DOI: 10.1038/s41536-019-0084-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 10/11/2019] [Indexed: 01/11/2023] Open
Abstract
Laminins comprise structural components of basement membranes, critical in the regulation of differentiation, survival and migration of a diverse range of cell types, including skeletal muscle. Mutations in one muscle enriched Laminin isoform, Laminin alpha2 (Lama2), results in the most common form of congenital muscular dystrophy, congenital muscular dystrophy type 1A (MDC1A). However, the exact cellular mechanism by which Laminin loss results in the pathological spectrum associated with MDC1A remains elusive. Here we show, via live tracking of individual muscle fibres, that dystrophic myofibres in the zebrafish model of MDC1A maintain sarcolemmal integrity and undergo dynamic remodelling behaviours post detachment, including focal sarcolemmal reattachment, cell extension and hyper-fusion with surrounding myoblasts. These observations imply the existence of a window of therapeutic opportunity, where detached cells may be “re-functionalised” prior to their delayed entry into the cell death program, a process we show can be achieved by muscle specific or systemic Laminin delivery. We further reveal that Laminin also acts as a pro-regenerative factor that stimulates muscle stem cell-mediated repair in lama2-deficient animals in vivo. The potential multi-mode of action of Laminin replacement therapy suggests it may provide a potent therapeutic axis for the treatment for MDC1A.
Collapse
Affiliation(s)
- T E Hall
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia.,2Institute for Molecular Bioscience, University of Queensland, 306 Carmody Road, St Lucia, 4067 Australia
| | - A J Wood
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia
| | - O Ehrlich
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia
| | - M Li
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia
| | - C S Sonntag
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia
| | - N J Cole
- 3Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, New South Wales 2010 Australia.,4Anatomy & Histology, School of Medical Science, Anderson Stuart Building, Eastern Avenue, The University of Sydney, Sydney, New South Wales 2006 Australia
| | - I G Huttner
- 3Victor Chang Cardiac Research Institute, 405 Liverpool Street, Darlinghurst, Sydney, New South Wales 2010 Australia
| | - T E Sztal
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia.,5Department of Biological Sciences, Monash University, Victoria, 3800 Australia
| | - P D Currie
- 1Australian Regenerative Medicine Institute, Monash University, Level 1, 15 Innovation Walk, Victoria, 3800 Australia.,6EMBL Australia, Victorian Node, Monash University, Clayton, VIC 3800 Australia
| |
Collapse
|
71
|
Kalantari S, Naji M, Nafar M, Yazdani-Kachooei H, Borumandnia N, Parvin M. Chondroitin sulfate degradation and eicosanoid metabolism pathways are impaired in focal segmental glomerulosclerosis: Experimental confirmation of an in silico prediction. ACTA ACUST UNITED AC 2019; 9:89-95. [PMID: 31334040 PMCID: PMC6637215 DOI: 10.15171/bi.2019.12] [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: 10/22/2018] [Revised: 12/16/2018] [Accepted: 12/22/2018] [Indexed: 11/09/2022]
Abstract
Introduction: Focal segmental glomerulosclerosis (FSGS), the most common primary glomerular disease, is a diverse clinical entity that occurs after podocyte injury. Although numerous studies have suggested molecular pathways responsible for the development of FSGS, many still remain unknown about its pathogenic mechanisms. Two important pathways were predicted as candidates for the pathogenesis of FSGS in our previous in silico analysis, whom we aim to confirm experimentally in the present study. Methods: The expression levels of 4 enzyme genes that are representative of "chondroitin sulfate degradation" and "eicosanoid metabolism" pathways were investigated in the urinary sediments of biopsy-proven FSGS patients and healthy subjects using real-time polymerase chain reaction (RT-PCR). These target genes were arylsulfatase, hexosaminidase, cyclooxygenase-2 (COX-2), and prostaglandin I2 synthase. The patients were sub-divided into 2 groups based on the range of proteinuria and glomerular filtration rate and were compared for variation in the expression of target genes. Correlation of target genes with clinical and pathological characteristics of the disease was calculated and receiver operating characteristic (ROC) analysis was performed. Results: A combined panel of arylsulfatase, hexosaminidase, and COX-2 improved the diagnosis of FSGS by 76%. Hexosaminidase was correlated with the level of proteinuria, while COX-2 was correlated with interstitial inflammation and serum creatinine level in the disease group. Conclusion: Our data supported the implication of these target genes and pathways in the pathogenesis of FSGS. In addition, these genes can be considered as non-invasive biomarkers for FSGS.
Collapse
Affiliation(s)
- Shiva Kalantari
- Chronic Kidney Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Naji
- Urology-Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Nafar
- Urology-Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hootan Yazdani-Kachooei
- Department of Biology, Faculty of Basic Sciences, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Nasrin Borumandnia
- Urology-Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Parvin
- Department of Pathology, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
72
|
Bülow RD, Boor P. Extracellular Matrix in Kidney Fibrosis: More Than Just a Scaffold. J Histochem Cytochem 2019; 67:643-661. [PMID: 31116062 DOI: 10.1369/0022155419849388] [Citation(s) in RCA: 243] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kidney fibrosis is the common histological end-point of progressive, chronic kidney diseases (CKDs) regardless of the underlying etiology. The hallmark of renal fibrosis, similar to all other organs, is pathological deposition of extracellular matrix (ECM). Renal ECM is a complex network of collagens, elastin, and several glycoproteins and proteoglycans forming basal membranes and interstitial space. Several ECM functions beyond providing a scaffold and organ stability are being increasingly recognized, for example, in inflammation. ECM composition is determined by the function of each of the histological compartments of the kidney, that is, glomeruli, tubulo-interstitium, and vessels. Renal ECM is a dynamic structure undergoing remodeling, particularly during fibrosis. From a clinical perspective, ECM proteins are directly involved in several rare renal diseases and indirectly in CKD progression during renal fibrosis. ECM proteins could serve as specific non-invasive biomarkers of fibrosis and scaffolds in regenerative medicine. The gold standard and currently only specific means to measure renal fibrosis is renal biopsy, but new diagnostic approaches are appearing. Here, we discuss the localization, function, and remodeling of major renal ECM components in healthy and diseased, fibrotic kidneys and the potential use of ECM in diagnostics of renal fibrosis and in tissue engineering.
Collapse
Affiliation(s)
- Roman David Bülow
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany.,Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
73
|
Kovel ES, Sachkova AS, Vnukova NG, Churilov GN, Knyazeva EM, Kudryasheva NS. Antioxidant Activity and Toxicity of Fullerenols via Bioluminescence Signaling: Role of Oxygen Substituents. Int J Mol Sci 2019; 20:ijms20092324. [PMID: 31083407 PMCID: PMC6539272 DOI: 10.3390/ijms20092324] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/04/2019] [Accepted: 05/09/2019] [Indexed: 12/17/2022] Open
Abstract
Fullerenols are nanosized water-soluble polyhydroxylated derivatives of fullerenes, a specific allotropic form of carbon, bioactive compounds, and perspective basis for drug development. Our paper analyzes the antioxidant activity and toxicity of a series of fullerenols with different number of oxygen substituents. Two groups of fullerenols were under investigation: (1) C60Oy(OH)x, C60,70Oy(OH)x, where x+y = 24–28 and (2) C60,70Oy(OH)x, Fe0,5C60Oy(OH)x, Gd@C82Oy(OH)x, where x+y = 40–42. Bioluminescent cellular and enzymatic assays (luminous marine bacteria and their enzymatic reactions, respectively) were applied to monitor toxicity in the model fullerenol solutions and bioluminescence was applied as a signaling physiological parameter. The inhibiting concentrations of the fullerenols were determined, revealing the fullerenols’ toxic effects. Antioxidant fullerenol’ ability was studied in solutions of model oxidizer, 1,4-benzoquinone, and detoxification coefficients of general and oxidative types (DGT and DOxT) were calculated. All fullerenols produced toxic effect at high concentrations (>0.01 g L−1), while their antioxidant activity was demonstrated at low and ultralow concentrations (<0.001 g L−1). Quantitative toxic and antioxidant characteristics of the fullerenols (effective concentrations, concentration ranges, DGT, and DOxT) were found to depend on the number of oxygen substituents. Lower toxicity and higher antioxidant activity were determined in solutions of fullerenols with fewer oxygen substituents (x+y = 24–28). The differences in fullerenol properties were attributed to their catalytic activity due to reversible electron acceptance, radical trapping, and balance of reactive oxygen species in aqueous solutions. The results provide pharmaceutical sciences with a basis for selection of carbon nanoparticles with appropriate toxic and antioxidant characteristics. Based on the results, we recommend, to reduce the toxicity of prospective endohedral gadolinium-fullerenol preparations Gd@C82Oy(OH)x, decreasing the number of oxygen groups to x+y = 24–28. The potential of bioluminescence methods to compare toxic and antioxidant characteristics of carbon nanostructures were demonstrated.
Collapse
Affiliation(s)
- Ekaterina S Kovel
- Institute of Biophysics SB RAS, FRC KSC SB RAS, 660036 Krasnoyarsk, Russia.
- Institute of Physics SB RAS, FRC KSC SB RAS, 660036 Krasnoyarsk, Russia.
| | - Anna S Sachkova
- National Research Tomsk Polytechnic University, 634050 Tomsk, Russia.
| | - Natalia G Vnukova
- Institute of Physics SB RAS, FRC KSC SB RAS, 660036 Krasnoyarsk, Russia.
- Siberian Federal University, 660041 Krasnoyarsk, Russia.
| | - Grigoriy N Churilov
- Institute of Physics SB RAS, FRC KSC SB RAS, 660036 Krasnoyarsk, Russia.
- Siberian Federal University, 660041 Krasnoyarsk, Russia.
| | - Elena M Knyazeva
- National Research Tomsk Polytechnic University, 634050 Tomsk, Russia.
| | - Nadezhda S Kudryasheva
- Institute of Biophysics SB RAS, FRC KSC SB RAS, 660036 Krasnoyarsk, Russia.
- Siberian Federal University, 660041 Krasnoyarsk, Russia.
| |
Collapse
|
74
|
Eissa L, Ismail HI, Elhassan MMO, Ali HA. Basement membranes in the kidney of the dromedary camel (Camelus dromedarius): An immunohistochemical and ultrastructural study. Acta Histochem 2019; 121:419-429. [PMID: 30904316 DOI: 10.1016/j.acthis.2019.03.005] [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: 11/16/2018] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 11/16/2022]
Abstract
Basement membranes consist of various proteins, the major ones being laminin and collagen type IV. Primary defects in these two proteins have been extensively associated with kidney pathologies. This study aimed to establish baseline information on the immunohistochemical distribution of laminin and collagen type IV, and to corelate these with the ultrastructure of basal laminae in the uriniferous tubules of the dromedary camel. Tissue samples were taken from the kidneys of eight adult female camels, and processed for immunohistochemical and ultrastructural investigations. Strong intensity of collagen type IV was observed within the basement membranes of Bowman's capsule. The thickness of the basal lamina of the parietal layer of Bowman's capsule varied extensively depending on the region of the renal corpuscle; the thicker areas were always associated with cuboidal epithelial cells. The glomerular basement membrane revealed strong immunostaining of laminin, whereas the mesangial matrix was strongly immunoreactive to collagen type IV. Abundant amount of laminin was found in the basement membranes of proximal convoluted tubules, thin limbs of the loop of Henle, and collecting ducts. Dense immunostainings of laminin and collagen type IV were observed in the medullary regions of uriniferous tubule, in which numerous projections extended from the basal laminae into the subjacent connective tissue. Overall, the present study revealed marked variations in the distribution of the basement membrane markers laminin and collagen type IV in the uriniferous tubules of camel kidney. The results have also shown difference in the thickness of basal laminae. This variation in thickness, however, was unlikely to be influenced by the amount of laminin and collagen type IV.
Collapse
Affiliation(s)
- Lemiaa Eissa
- Department of Anatomy, College of Veterinary Medicine, University of Bahri, Khartoum, Sudan
| | - Haider I Ismail
- Department of Anatomy, College of Veterinary Medicine, University of Bahri, Khartoum, Sudan
| | - Mortada M O Elhassan
- Department of Anatomy, College of Veterinary Medicine, University of Bahri, Khartoum, Sudan.
| | - Hassan A Ali
- Department of Biomedical Sciences, College of Veterinary Medicine, Sudan University of Science and Technology, Khartoum, Sudan
| |
Collapse
|
75
|
Widmeier E, Airik M, Hugo H, Schapiro D, Wedel J, Ghosh CC, Nakayama M, Schneider R, Awad AM, Nag A, Cho J, Schueler M, Clarke CF, Airik R, Hildebrandt F. Treatment with 2,4-Dihydroxybenzoic Acid Prevents FSGS Progression and Renal Fibrosis in Podocyte-Specific Coq6 Knockout Mice. J Am Soc Nephrol 2019; 30:393-405. [PMID: 30737270 PMCID: PMC6405149 DOI: 10.1681/asn.2018060625] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 12/29/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Although studies have identified >55 genes as causing steroid-resistant nephrotic syndrome (SRNS) and localized its pathogenesis to glomerular podocytes, the disease mechanisms of SRNS remain largely enigmatic. We recently reported that individuals with mutations in COQ6, a coenzyme Q (also called CoQ10, CoQ, or ubiquinone) biosynthesis pathway enzyme, develop SRNS with sensorineural deafness, and demonstrated the beneficial effect of CoQ for maintenace of kidney function. METHODS To study COQ6 function in podocytes, we generated a podocyte-specific Coq6 knockout mouse (Coq6podKO ) model and a transient siRNA-based COQ6 knockdown in a human podocyte cell line. Mice were monitored for development of proteinuria and assessed for development of glomerular sclerosis. Using a podocyte migration assay, we compared motility in COQ6 knockdown podocytes and control podocytes. We also randomly assigned 5-month-old Coq6podKO mice and controls to receive no treatment or 2,4-dihydroxybenzoic acid (2,4-diHB), an analog of a CoQ precursor molecule that is classified as a food additive by health authorities in Europe and the United States. RESULTS Abrogation of Coq6 in mouse podocytes caused FSGS and proteinuria (>46-fold increases in albuminuria). In vitro studies revealed an impaired podocyte migration rate in COQ6 knockdown human podocytes. Treating Coq6podKO mice or cells with 2,4-diHB prevented renal dysfunction and reversed podocyte migration rate impairment. Survival of Coq6podKO mice given 2,4diHB was comparable to that of control mice and significantly higher than that of untreated Coq6podKO mice, half of which died by 10 months of age. CONCLUSIONS These findings reveal a potential novel treatment strategy for those cases of human nephrotic syndrome that are caused by a primary dysfunction in the CoQ10 biosynthesis pathway.
Collapse
Affiliation(s)
- Eugen Widmeier
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts;,Department of Medicine IV, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Merlin Airik
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Hannah Hugo
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - David Schapiro
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Johannes Wedel
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chandra C. Ghosh
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Makiko Nakayama
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ronen Schneider
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Agape M. Awad
- Department of Chemistry and Biochemistry and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Anish Nag
- Department of Chemistry and Biochemistry and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Jang Cho
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Markus Schueler
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Catherine F. Clarke
- Department of Chemistry and Biochemistry and Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Rannar Airik
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Pennsylvania; and
| | - Friedhelm Hildebrandt
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
76
|
Song CC, Hong Q, Geng XD, Wang X, Wang SQ, Cui SY, Guo MD, Li O, Cai GY, Chen XM, Wu D. New Mutation of Coenzyme Q 10 Monooxygenase 6 Causing Podocyte Injury in a Focal Segmental Glomerulosclerosis Patient. Chin Med J (Engl) 2019; 131:2666-2675. [PMID: 30425193 PMCID: PMC6247592 DOI: 10.4103/0366-6999.245158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Background: Focal segmental glomerulosclerosis (FSGS) is a kidney disease that is commonly associated with proteinuria and the progressive loss of renal function, which is characterized by podocyte injury and the depletion and collapse of glomerular capillary segments. The pathogenesis of FSGS has not been completely elucidated; however, recent advances in molecular genetics have provided increasing evidence that podocyte structural and functional disruption is central to FSGS pathogenesis. Here, we identified a patient with FSGS and aimed to characterize the pathogenic gene and verify its mechanism. Methods: Using next-generation sequencing and Sanger sequencing, we screened the causative gene that was linked to FSGS in this study. The patient's total blood RNA was extracted to validate the messenger RNA (mRNA) expression of coenzyme Q10 monooxygenase 6 (COQ6) and validated it by immunohistochemistry. COQ6 knockdown in podocytes was performed in vitro with small interfering RNA, and then, F-actin was determined using immunofluorescence staining. Cell apoptosis was evaluated by flow cytometry, the expression of active caspase-3 was determined by Western blot, and mitochondrial function was detected by MitoSOX. Results: Using whole-exome sequencing and Sanger sequencing, we screened a new causative gene, COQ6, NM_182480: exon1: c.G41A: p.W14X. The mRNA expression of COQ6 in the proband showed decreased. Moreover, the expression of COQ6, which was validated by immunohistochemistry, also had the same change in the proband. Finally, we focused on the COQ6 gene to clarify the mechanism of podocyte injury. Flow cytometry showed significantly increased in apoptotic podocytes, and Western blotting showed increases in active caspase-3 in si-COQ6 podocytes. Meanwhile, reactive oxygen species (ROS) levels were increased and F-actin immunofluorescence was irregularly distributed in the si-COQ6 group. Conclusions: This study reported a possible mechanism for FSGS and suggested that a new mutation in COQ6, which could cause respiratory chain defect, increase the generation of ROS, destroy the podocyte cytoskeleton, and induce apoptosis. It provides basic theoretical basis for the screening of FSGS in the future.
Collapse
Affiliation(s)
- Cheng-Cheng Song
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Quan Hong
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Xiao-Dong Geng
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Xu Wang
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Shu-Qiang Wang
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Shao-Yuan Cui
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Man-Di Guo
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Ou Li
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Guang-Yan Cai
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Xiang-Mei Chen
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Di Wu
- Department of Nephrology, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Institute of Nephrology, State Key Laboratory of Kidney Diseases (2011DAV00088), National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing 100853, China
| |
Collapse
|
77
|
Ho CPS, Lai TYY. Pharmacotherapy for Choroidal Neovascularization Due to Uncommon Causes. Curr Pharm Des 2019; 24:4882-4895. [PMID: 30727875 DOI: 10.2174/1381612825666190206105943] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/18/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Choroidal neovascularization (CNV) in adults is most commonly associated with neovascular age-related macular degeneration (AMD) and pathologic myopia. Though less common, CNV can also develop from other conditions such as uveitis, central serous chorioretinopathy, angioid streaks, intraocular tumors, hereditary chorioretinal dystrophies, or can be idiopathic in origin. If left untreated, CNV may cause visual loss because of exudation of intraretinal or subretinal fluid, retinal or subretinal hemorrhage, or fibrosis involving the macula. It is well known that one of the main drivers of angiogenesis in CNV development is vascular endothelial growth factor (VEGF) and therefore inhibitors of VEGF might be an effective treatment for CNV. METHODS The goal of this review is to provide an overview and summary in the use of pharmacotherapy especially anti-VEGF therapy, in the treatment of CNV due to uncommon causes. RESULTS Results from uncontrolled case series and controlled clinical trials have reported good efficacy and safety in using anti-VEGF agents including bevacizumab, ranibizumab, aflibercept and ziv-aflibercept in the treatment of CNV due to uncommon causes. Anti-VEGF has also been used in combination with verteporfin PDT and anti-inflammatory agents for treating CNV of various causes. CONCLUSION Pharmacotherapy with anti-VEGF agents is an effective treatment option for CNV due to uncommon etiologies.
Collapse
Affiliation(s)
- Christine P S Ho
- Faculty of Medicine, The University of Hong Kong, Kowloon, Hong Kong
| | - Timothy Y Y Lai
- Hong Kong Eye Hospital, Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong.,2010 Retina & Macula Centre, Kowloon, Hong Kong
| |
Collapse
|
78
|
Zhu HT, Maimaiti M, Cao C, Luo YF, Julaiti D, Liang L, Abudureheman A. A Novel Homozygous Truncating Mutation in LAMB2 Gene in a Chinese Uyghur Patient With Severe Phenotype Pierson Syndrome. Front Med (Lausanne) 2019; 6:12. [PMID: 30778388 PMCID: PMC6369156 DOI: 10.3389/fmed.2019.00012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 01/16/2019] [Indexed: 11/13/2022] Open
|
79
|
Stone H, Magella B, Bennett MR. The Search for Biomarkers to Aid in Diagnosis, Differentiation, and Prognosis of Childhood Idiopathic Nephrotic Syndrome. Front Pediatr 2019; 7:404. [PMID: 31681707 PMCID: PMC6805718 DOI: 10.3389/fped.2019.00404] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
Identification of genes associated with childhood-onset nephrotic syndrome has significantly advanced our understanding of the pathogenesis of this complex disease over the past two decades, however the precise etiology in many cases remains unclear. At this time, we still rely on invasive kidney biopsy to determine the underlying cause of nephrotic syndrome in adults. In children, response to steroid therapy has been shown to be the best indicator of prognosis, and therefore all children are treated initially with corticosteroids. Because this strategy exposes a large number of children to the toxicities of steroids without providing any benefit, many researchers have sought to find a marker that could predict a patient's response to steroids at the time of diagnosis. Additionally, the identification of such a marker could provide prognostic information about a patient's response to medications, progression to end stage renal disease, and risk of disease recurrence following transplantation. Major advances have been made in understanding how genetic biomarkers can be used to predict a patient's response to therapies and disease course, especially after transplantation. Research attempting to identify urine- and serum-based biomarkers which could be used for the diagnosis, differentiation, and prognosis of nephrotic syndrome has become an area of emphasis. In this review, we explore the most exciting biomarkers and their potential clinical applications.
Collapse
Affiliation(s)
- Hillarey Stone
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Bliss Magella
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Michael R Bennett
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
80
|
Genetic testing in steroid-resistant nephrotic syndrome: why, who, when and how? Pediatr Nephrol 2019; 34:195-210. [PMID: 29181713 PMCID: PMC6311200 DOI: 10.1007/s00467-017-3838-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 10/24/2017] [Accepted: 10/25/2017] [Indexed: 12/23/2022]
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is a common cause of chronic kidney disease in childhood and has a significant risk of rapid progression to end-stage renal disease. The identification of over 50 monogenic causes of SRNS has revealed dysfunction in podocyte-associated proteins in the pathogenesis of proteinuria, highlighting their essential role in glomerular function. Recent technological advances in high-throughput sequencing have enabled indication-driven genetic panel testing for patients with SRNS. The availability of genetic testing, combined with the significant phenotypic variability of monogenic SRNS, poses unique challenges for clinicians when directing genetic testing. This highlights the need for clear clinical guidelines that provide a systematic approach for mutational screening in SRNS. The likelihood of identifying a causative mutation is inversely related to age at disease onset and is increased with a positive family history or the presence of extra-renal manifestations. An unequivocal molecular diagnosis could allow for a personalised treatment approach with weaning of immunosuppressive therapy, avoidance of renal biopsy and provision of accurate, well-informed genetic counselling. Identification of novel causative mutations will continue to unravel the pathogenic mechanisms of glomerular disease and provide new insights into podocyte biology and glomerular function.
Collapse
|
81
|
Li GM, Cao Q, Shen Q, Sun L, Zhai YH, Liu HM, An Y, Xu H. Gene mutation analysis in 12 Chinese children with congenital nephrotic syndrome. BMC Nephrol 2018; 19:382. [PMID: 30594156 PMCID: PMC6311020 DOI: 10.1186/s12882-018-1184-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 12/11/2018] [Indexed: 11/13/2022] Open
Abstract
Background Congenital nephrotic syndrome (CNS) is characterised by increased proteinuria, hypoproteinemia, and edema beginning in the first 3 months of life. Recently, molecular genetic studies have identified several genes involved in the pathogenesis of CNS. A systematic investigation of the genes for CNS in China has never been performed; therefore, we conducted a mutational analysis in 12 children with CNS,with the children coming from 10 provinces and autonomous regions in China. Methods Twelve children with CNS were enrolled from 2009 to 2016. A mutational analysis was performed in six children by Sanger sequencing in eight genes (NPHS1, NPHS2, PLCE1, WT1, LAMB2, LMXIB, COQ6 and COQ2) before 2014, and whole-exome sequencing was used from 2014 to 2016 in another six children. Significant variants that were detected by next generation sequencing were confirmed by conventional Sanger sequencing in the patients’ families. Results Of the 12 children, eight patients had a compound heterozygous NPHS1 mutation, one patient had a de novo mutation in the WT1 gene, and another patient with extrarenal symptoms had a homozygous mutation in the COQ6 gene. No mutations were detected in genes NPHS2, PLCE1, LAMB2, LMXIB, and COQ2 in the 12 patients. Conclusions This study demonstrates that the majority of CNS cases (67%, 8/12 patients) are caused by genetic defects, and the NPHS1 mutation is the most common cause of CNS in Chinese patients. A mutational analysis of NPHS1 should be recommended in Chinese patients with CNS in all exons of NPHS1 and in the intron-exon boundaries.
Collapse
Affiliation(s)
- Guo-Min Li
- Children's Hospital of Fudan University, 399 Wanyuanlu, Shanghai, 201102, China
| | - Qi Cao
- Children's Hospital of Fudan University, 399 Wanyuanlu, Shanghai, 201102, China
| | - Qian Shen
- Children's Hospital of Fudan University, 399 Wanyuanlu, Shanghai, 201102, China
| | - Li Sun
- Children's Hospital of Fudan University, 399 Wanyuanlu, Shanghai, 201102, China
| | - Yi-Hui Zhai
- Children's Hospital of Fudan University, 399 Wanyuanlu, Shanghai, 201102, China
| | - Hai-Mei Liu
- Children's Hospital of Fudan University, 399 Wanyuanlu, Shanghai, 201102, China
| | - Yu An
- Institutes of Biomedical Sciences of Fudan University, 220 Handanlu, Shanghai, 200433, China
| | - Hong Xu
- Children's Hospital of Fudan University, 399 Wanyuanlu, Shanghai, 201102, China.
| |
Collapse
|
82
|
Abstract
Zusammenfassung
Das steroid-resistente nephrotische Syndrom (SRNS) mit dem histomorphologischen Korrelat der fokal-segmentalen Glomerulosklerose (FSGS) stellt eine bedeutende Ursache für eine terminale Niereninsuffizienz im Kindesalter, aber auch bei erwachsenen Patienten dar. Das Erkrankungsspektrum zeichnet sich durch eine große genetische Heterogenität aus, wobei auch nicht genetische Ursachen bei der FSGS beobachtet werden. Die genetische Grundlage des SRNS/FSGS-Komplexes ist v. a. für ältere Kinder/Jugendliche und Erwachsene bisher noch unzureichend verstanden. Die eindeutige Abgrenzung genetischer SRNS/FSGS-Ursachen ist unerlässlich, da sich bereits heute hieraus eine Vielzahl an klinischen Implikationen ergeben. Die Identifikation unbekannter Erkrankungsallele oder Erkrankungsgene kann zudem Erkenntnisse bringen, die ein gänzlich neues Verständnis der Pathomechanismen ermöglichen. Durch umfassende genetische Untersuchungen besteht die Möglichkeit, die ungelöste genetische Basis der Rekurrenz der FSGS-Erkrankung bei bislang Varianten-negativen Patienten zu finden.
Collapse
Affiliation(s)
- Julia Hoefele
- Aff1 Institut für Humangenetik Klinikum rechts der Isar, Technische Universität München Trogerstr. 32 81675 München Deutschland
| | - Bodo B. Beck
- Aff2 0000 0000 8852 305X grid.411097.a Institut für Humangenetik Uniklinik Köln Kerpener Str. 34 50937 Köln Deutschland
| | - Lutz T. Weber
- Aff3 0000 0000 8852 305X grid.411097.a Klinik und Poliklinik für Kinder- und Jugendmedizin Uniklinik Köln Kerpener Str. 62 50937 Köln Deutschland
| | - Paul Brinkkötter
- Aff4 0000 0000 8852 305X grid.411097.a Klinik II für Innere Medizin Uniklinik Köln Kerpener Str. 62 50937 Köln Deutschland
| |
Collapse
|
83
|
Arima M, Tsukamoto S, Akiyama R, Nishiyama K, Kohno RI, Tachibana T, Hayashida A, Murayama M, Hisatomi T, Nozu K, Iijima K, Ohga S, Sonoda KH. Ocular findings in a case of Pierson syndrome with a novel mutation in laminin ß2 gene. J AAPOS 2018; 22:401-403.e1. [PMID: 30120985 DOI: 10.1016/j.jaapos.2018.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 11/16/2022]
Abstract
Pierson syndrome, an autosomal recessive disorder caused by a mutation in laminin ß2 (LAMB2) gene, is characterized by congenital nephrotic syndrome and various ocular abnormalities. The ocular findings in Pierson syndrome are not well understood, because the incidence of this syndrome is very rare. We report ocular findings in a 5-month-old boy with Pierson syndrome with a novel mutation in LAMB2. We performed a pupilloplasty for his microcoria. Ophthalmic examinations after surgery revealed that he had cataract, severe retinal degeneration, and high myopia. Optical coherence tomography showed the collapse of retinal layer structures and a marked decrease of choroidal thickness. Immunohistochemistry and electron microscopy examinations revealed abnormal iris differentiation and thinning or defect of basal membranes. These results suggest that the development of the iris, lens, retina, and choroid are affected in this type of mutation.
Collapse
Affiliation(s)
- Mitsuru Arima
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan.
| | - Shoko Tsukamoto
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Rumi Akiyama
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Kei Nishiyama
- Department of Pediatrics, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Ri-Ichiro Kohno
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Takashi Tachibana
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Akira Hayashida
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Miwa Murayama
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Toshio Hisatomi
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe City, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe City, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Kyushu University Graduate School of Medical Sciences, Fukuoka City, Japan
| |
Collapse
|
84
|
Funk SD, Lin MH, Miner JH. Alport syndrome and Pierson syndrome: Diseases of the glomerular basement membrane. Matrix Biol 2018; 71-72:250-261. [PMID: 29673759 PMCID: PMC6146048 DOI: 10.1016/j.matbio.2018.04.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 12/17/2022]
Abstract
The glomerular basement membrane (GBM) is an important component of the kidney's glomerular filtration barrier. Like all basement membranes, the GBM contains type IV collagen, laminin, nidogen, and heparan sulfate proteoglycan. It is flanked by the podocytes and glomerular endothelial cells that both synthesize it and adhere to it. Mutations that affect the GBM's collagen α3α4α5(IV) components cause Alport syndrome (kidney disease with variable ear and eye defects) and its variants, including thin basement membrane nephropathy. Mutations in LAMB2 that impact the synthesis or function of laminin α5β2γ1 (LM-521) cause Pierson syndrome (congenital nephrotic syndrome with eye and neurological defects) and its less severe variants, including isolated congenital nephrotic syndrome. The very different types of kidney diseases that result from mutations in collagen IV vs. laminin are likely due to very different pathogenic mechanisms. A better understanding of these mechanisms should lead to targeted therapeutic approaches that can help people with these rare but important diseases.
Collapse
Affiliation(s)
- Steven D Funk
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Meei-Hua Lin
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
85
|
Hermle T, Schneider R, Schapiro D, Braun DA, van der Ven AT, Warejko JK, Daga A, Widmeier E, Nakayama M, Jobst-Schwan T, Majmundar AJ, Ashraf S, Rao J, Finn LS, Tasic V, Hernandez JD, Bagga A, Jalalah SM, El Desoky S, Kari JA, Laricchia KM, Lek M, Rehm HL, MacArthur DG, Mane S, Lifton RP, Shril S, Hildebrandt F. GAPVD1 and ANKFY1 Mutations Implicate RAB5 Regulation in Nephrotic Syndrome. J Am Soc Nephrol 2018; 29:2123-2138. [PMID: 29959197 PMCID: PMC6065084 DOI: 10.1681/asn.2017121312] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/24/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of CKD. The discovery of monogenic causes of SRNS has revealed specific pathogenetic pathways, but these monogenic causes do not explain all cases of SRNS. METHODS To identify novel monogenic causes of SRNS, we screened 665 patients by whole-exome sequencing. We then evaluated the in vitro functional significance of two genes and the mutations therein that we discovered through this sequencing and conducted complementary studies in podocyte-like Drosophila nephrocytes. RESULTS We identified conserved, homozygous missense mutations of GAPVD1 in two families with early-onset NS and a homozygous missense mutation of ANKFY1 in two siblings with SRNS. GAPVD1 and ANKFY1 interact with the endosomal regulator RAB5. Coimmunoprecipitation assays indicated interaction between GAPVD1 and ANKFY1 proteins, which also colocalized when expressed in HEK293T cells. Silencing either protein diminished the podocyte migration rate. Compared with wild-type GAPVD1 and ANKFY1, the mutated proteins produced upon ectopic expression of GAPVD1 or ANKFY1 bearing the patient-derived mutations exhibited altered binding affinity for active RAB5 and reduced ability to rescue the knockout-induced defect in podocyte migration. Coimmunoprecipitation assays further demonstrated a physical interaction between nephrin and GAPVD1, and immunofluorescence revealed partial colocalization of these proteins in rat glomeruli. The patient-derived GAPVD1 mutations reduced nephrin-GAPVD1 binding affinity. In Drosophila, silencing Gapvd1 impaired endocytosis and caused mistrafficking of the nephrin ortholog. CONCLUSIONS Mutations in GAPVD1 and probably in ANKFY1 are novel monogenic causes of NS. The discovery of these genes implicates RAB5 regulation in the pathogenesis of human NS.
Collapse
Affiliation(s)
- Tobias Hermle
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
- Renal Division, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ronen Schneider
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David Schapiro
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniela A Braun
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amelie T van der Ven
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jillian K Warejko
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ankana Daga
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eugen Widmeier
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Makiko Nakayama
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Tilman Jobst-Schwan
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amar J Majmundar
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shazia Ashraf
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jia Rao
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Laura S Finn
- Department of Pathology, Seattle Children's Hospital, University of Washington, Seattle, Washington
| | - Velibor Tasic
- Department of Pediatric Nephrology, Medical Faculty Skopje, University Children's Hospital, Skopje, Macedonia
| | - Joel D Hernandez
- Department of Pediatric Nephrology, Providence Sacred Heart Medical Center and Children's Hospital, Spokane, Washington
| | - Arvind Bagga
- Division of Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Sherif El Desoky
- Pediatric Nephrology Center of Excellence and Pediatric Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Jameela A Kari
- Pediatric Nephrology Center of Excellence and Pediatric Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Kristen M Laricchia
- Broad Center for Mendelian Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Monkol Lek
- Broad Center for Mendelian Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Heidi L Rehm
- Broad Center for Mendelian Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Daniel G MacArthur
- Broad Center for Mendelian Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Shrikant Mane
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut; and
| | - Richard P Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut; and
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, New York
| | - Shirlee Shril
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts;
| |
Collapse
|
86
|
Rodríguez Cruz PM, Palace J, Beeson D. The Neuromuscular Junction and Wide Heterogeneity of Congenital Myasthenic Syndromes. Int J Mol Sci 2018; 19:ijms19061677. [PMID: 29874875 PMCID: PMC6032286 DOI: 10.3390/ijms19061677] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/17/2018] [Accepted: 05/21/2018] [Indexed: 01/16/2023] Open
Abstract
Congenital myasthenic syndromes (CMS) are genetic disorders characterised by impaired neuromuscular transmission. This review provides an overview on CMS and highlights recent advances in the field, including novel CMS causative genes and improved therapeutic strategies. CMS due to mutations in SLC5A7 and SLC18A3, impairing the synthesis and recycling of acetylcholine, have recently been described. In addition, a novel group of CMS due to mutations in SNAP25B, SYT2, VAMP1, and UNC13A1 encoding molecules implicated in synaptic vesicles exocytosis has been characterised. The increasing number of presynaptic CMS exhibiting CNS manifestations along with neuromuscular weakness demonstrate that the myasthenia can be only a small part of a much more extensive disease phenotype. Moreover, the spectrum of glycosylation abnormalities has been increased with the report that GMPPB mutations can cause CMS, thus bridging myasthenic disorders with dystroglycanopathies. Finally, the discovery of COL13A1 mutations and laminin α5 deficiency has helped to draw attention to the role of extracellular matrix proteins for the formation and maintenance of muscle endplates. The benefit of β2-adrenergic agonists alone or combined with pyridostigmine or 3,4-Dyaminopiridine is increasingly being reported for different subtypes of CMS including AChR-deficiency and glycosylation abnormalities, thus expanding the therapeutic repertoire available.
Collapse
Affiliation(s)
- Pedro M Rodríguez Cruz
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford OX3 9DS, UK.
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.
| | - David Beeson
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX3 9DU, UK.
- Neurosciences Group, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford OX3 9DS, UK.
| |
Collapse
|
87
|
Toffoli B, Zennaro C, Winkler C, Giordano Attianese GMP, Bernardi S, Carraro M, Gilardi F, Desvergne B. Hemicentin 1 influences podocyte dynamic changes in glomerular diseases. Am J Physiol Renal Physiol 2018; 314:F1154-F1165. [DOI: 10.1152/ajprenal.00198.2017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022] Open
Abstract
Different complex mechanisms control the morphology of podocyte foot processes and their interactions with the underlying basement membrane. Injuries to this system often cause glomerular dysfunction and albuminuria. The present study aimed at identifying early markers of glomerular damage in diabetic nephropathy. For this purpose, we performed a microarray analysis on kidneys of 3-wk-old peroxisome proliferator-activated receptor-γ (PPARγ)-null and AZIP/F1 mice, which are two models of diabetic nephropathy due to lipodystrophy. This was followed by functional annotation of the enriched clusters of genes. One of the significant changes in the early stages of glomerular damage was the increase of hemicentin 1 (HMCN1). Its expression and distribution were then studied by real-time PCR and immunofluorescence in various models of glomerular damage and on podocyte cell cultures. HMCN1 progressively increased in the glomeruli of diabetic mice, according to disease severity, as well as in puromycin aminonucleoside (PA)-treated rats. Studies on murine and human podocytes showed an increased HMCN1 deposition upon different pathological stimuli, such as hyperglycemia, transforming growth factor-β (TGF-β), and PA. In vitro silencing studies showed that HMCN1 mediated the rearrangements of podocyte cytoskeleton induced by TGF-β. Finally, we demonstrated an increased expression of HMCN1 in the kidneys of patients with proteinuric nephropathies. In summary, our studies identified HMCN1 as a new molecule involved in the dynamic changes of podocyte foot processes. Its increased expression associated with podocyte dysfunction points to HMCN1 as a possible marker for the early glomerular damage occurring in different proteinuric nephropathies.
Collapse
Affiliation(s)
- Barbara Toffoli
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Cristina Zennaro
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Carine Winkler
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | | | - Stella Bernardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Michele Carraro
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Federica Gilardi
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Béatrice Desvergne
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
88
|
Lin MH, Miller JB, Kikkawa Y, Suleiman HY, Tryggvason K, Hodges BL, Miner JH. Laminin-521 Protein Therapy for Glomerular Basement Membrane and Podocyte Abnormalities in a Model of Pierson Syndrome. J Am Soc Nephrol 2018; 29:1426-1436. [PMID: 29472414 PMCID: PMC5967757 DOI: 10.1681/asn.2017060690] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 01/14/2018] [Indexed: 12/22/2022] Open
Abstract
Background Laminin α5β2γ1 (LM-521) is a major component of the GBM. Mutations in LAMB2 that prevent LM-521 synthesis and/or secretion cause Pierson syndrome, a rare congenital nephrotic syndrome with diffuse mesangial sclerosis and ocular and neurologic defects. Because the GBM is uniquely accessible to plasma, which permeates endothelial cell fenestrae, we hypothesized that intravenous delivery of LM-521 could replace the missing LM-521 in the GBM of Lamb2 mutant mice and restore glomerular permselectivity.Methods We injected human LM-521 (hLM-521), a macromolecule of approximately 800 kD, into the retro-orbital sinus of Lamb2-/- pups daily. Deposition of hLM-521 into the GBM was investigated by fluorescence microscopy. We assayed the effects of hLM-521 on glomerular permselectivity by urinalysis and the effects on podocytes by desmin immunostaining and ultrastructural analysis of podocyte architecture.Results Injected hLM-521 rapidly and stably accumulated in the GBM of all glomeruli. Super-resolution imaging showed that hLM-521 accumulated in the correct orientation in the GBM, primarily on the endothelial aspect. Treatment with hLM-521 greatly reduced the expression of the podocyte injury marker desmin and attenuated the foot process effacement observed in untreated pups. Moreover, treatment with hLM-521 delayed the onset of proteinuria but did not prevent nephrotic syndrome, perhaps due to its absence from the podocyte aspect of the GBM.Conclusions These studies show that GBM composition and function can be altered in vivovia vascular delivery of even very large proteins, which may advance therapeutic options for patients with abnormal GBM composition, whether genetic or acquired.
Collapse
Affiliation(s)
- Meei-Hua Lin
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Joseph B Miller
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Yamato Kikkawa
- Department of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hani Y Suleiman
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Karl Tryggvason
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore, Singapore
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden; and
| | | | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri;
| |
Collapse
|
89
|
Animal Models of the Neuromuscular Junction, Vitally Informative for Understanding Function and the Molecular Mechanisms of Congenital Myasthenic Syndromes. Int J Mol Sci 2018; 19:ijms19051326. [PMID: 29710836 PMCID: PMC5983836 DOI: 10.3390/ijms19051326] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 04/24/2018] [Accepted: 04/25/2018] [Indexed: 01/16/2023] Open
Abstract
The neuromuscular junction is the point of contact between motor nerve and skeletal muscle, its vital role in muscle function is reliant on the precise location and function of many proteins. Congenital myasthenic syndromes (CMS) are a heterogeneous group of disorders of neuromuscular transmission with 30 or more implicated proteins. The use of animal models has been instrumental in determining the specific role of many CMS-related proteins. The mouse neuromuscular junction (NMJ) has been extensively studied in animal models of CMS due to its amenability for detailed electrophysiological and histological investigations and relative similarity to human NMJ. As well as their use to determine the precise molecular mechanisms of CMS variants, where an animal model accurately reflects the human phenotype they become useful tools for study of therapeutic interventions. Many of the animal models that have been important in deconvolving the complexities of neuromuscular transmission and revealing the molecular mechanisms of disease are highlighted.
Collapse
|
90
|
Yang JW, Dettmar AK, Kronbichler A, Gee HY, Saleem M, Kim SH, Shin JI. Recent advances of animal model of focal segmental glomerulosclerosis. Clin Exp Nephrol 2018; 22:752-763. [PMID: 29556761 DOI: 10.1007/s10157-018-1552-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 02/26/2018] [Indexed: 12/15/2022]
Abstract
In the last decade, great advances have been made in understanding the genetic basis for focal segmental glomerulosclerosis (FSGS). Animal models using specific gene disruption of the slit diaphragm and cytoskeleton of the foot process mirror the etiology of the human disease. Many animal models have been developed to understand the complex pathophysiology of FSGS. Therefore, we need to know the usefulness and exact methodology of creating animal models. Here, we review classic animal models and newly developed genetic animal models. Classic animal models of FSGS involve direct podocyte injury and indirect podocyte injury due to adaptive responses. However, the phenotype depends on the animal background. Renal ablation and direct podocyte toxin (PAN, adriamycin) models are leading animal models for FSGS, which have some limitations depending on mice background. A second group of animal models were developed using combinations of genetic mutation and toxin, such as NEP25, diphtheria toxin, and Thy1.1 models, which specifically injure podocytes. A third group of animal models involves genetic engineering techniques targeting podocyte expression molecules, such as podocin, CD2-associated protein, and TRPC6 channels. More detailed information about podocytopathy and FSGS can be expected in the coming decade. Different animal models should be used to study FSGS depending on the specific aim and sometimes should be used in combination.
Collapse
Affiliation(s)
- Jae Won Yang
- Department of Nephrology, Yonsei University Wonju College of Medicine, Wonju, Gangwon, Republic of Korea
| | - Anne Katrin Dettmar
- Pediatric Nephrology, Department of Pediatrics, Medical University Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Kronbichler
- Department of Internal Medicine IV (Nephrology and Hypertension), Universitätskliniken Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Moin Saleem
- Paediatric Renal Medicine, University of Bristol, Bristol, UK.,Children's Renal Unit, Bristol Royal Hospital for Children, Bristol, UK
| | - Seong Heon Kim
- Department of Pediatrics, Pusan National University Children's Hospital, Yangsan, Republic of Korea. .,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea.
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, 50 Yonsei-Ro, Seodaemun-gu, Seoul, 120-752, Republic of Korea.
| |
Collapse
|
91
|
Cil O, Perwad F. Monogenic Causes of Proteinuria in Children. Front Med (Lausanne) 2018; 5:55. [PMID: 29594119 PMCID: PMC5858124 DOI: 10.3389/fmed.2018.00055] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/15/2018] [Indexed: 01/02/2023] Open
Abstract
Glomerular disease is a common cause for proteinuria and chronic kidney disease leading to end-stage renal disease requiring dialysis or kidney transplantation in children. Nephrotic syndrome in children is diagnosed by the presence of a triad of proteinuria, hypoalbuminemia, and edema. Minimal change disease is the most common histopathological finding in children and adolescents with nephrotic syndrome. Focal segmental sclerosis is also found in children and is the most common pathological finding in patients with monogenic causes of nephrotic syndrome. Current classification system for nephrotic syndrome is based on response to steroid therapy as a majority of patients develop steroid sensitive nephrotic syndrome regardless of histopathological diagnosis or the presence of genetic mutations. Recent studies investigating the genetics of nephrotic syndrome have shed light on the pathophysiology and mechanisms of proteinuria in nephrotic syndrome. Gene mutations have been identified in several subcellular compartments of the glomerular podocyte and play a critical role in mitochondrial function, actin cytoskeleton dynamics, cell-matrix interactions, slit diaphragm, and podocyte integrity. A subset of genetic mutations are known to cause nephrotic syndrome that is responsive to immunosuppressive therapy but clinical data are limited with respect to renal prognosis and disease progression in a majority of patients. To date, more than 50 genes have been identified as causative factors in nephrotic syndrome in children and adults. As genetic testing becomes more prevalent and affordable, we expect rapid advances in our understanding of mechanisms of proteinuria and genetic diagnosis will help direct future therapy for individual patients.
Collapse
Affiliation(s)
- Onur Cil
- Department of Pediatrics, Division of Nephrology, University of California San Francisco, San Francisco, CA, United States
| | - Farzana Perwad
- Department of Pediatrics, Division of Nephrology, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
92
|
Ito M, Ohno K. Protein-anchoring therapy to target extracellular matrix proteins to their physiological destinations. Matrix Biol 2018; 68-69:628-636. [PMID: 29475025 DOI: 10.1016/j.matbio.2018.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/21/2022]
Abstract
Endplate acetylcholinesterase (AChE) deficiency is a form of congenital myasthenic syndrome (CMS) caused by mutations in COLQ, which encodes collagen Q (ColQ). ColQ is an extracellular matrix (ECM) protein that anchors AChE to the synaptic basal lamina. Biglycan, encoded by BGN, is another ECM protein that binds to the dystrophin-associated protein complex (DAPC) on skeletal muscle, which links the actin cytoskeleton and ECM proteins to stabilize the sarcolemma during repeated muscle contractions. Upregulation of biglycan stabilizes the DPAC. Gene therapy can potentially ameliorate any disease that can be recapitulated in cultured cells. However, the difficulty of tissue-specific and developmental stage-specific regulated expression of transgenes, as well as the difficulty of introducing a transgene into all cells in a specific tissue, prevents us from successfully applying gene therapy to many human diseases. In contrast to intracellular proteins, an ECM protein is anchored to the target tissue via its specific binding affinity for protein(s) expressed on the cell surface within the target tissue. Exploiting this unique feature of ECM proteins, we developed protein-anchoring therapy in which a transgene product expressed even in remote tissues can be delivered and anchored to a target tissue using specific binding signals. We demonstrate the application of protein-anchoring therapy to two disease models. First, intravenous administration of adeno-associated virus (AAV) serotype 8-COLQ to Colq-deficient mice, resulting in specific anchoring of ectopically expressed ColQ-AChE at the NMJ, markedly improved motor functions, synaptic transmission, and the ultrastructure of the neuromuscular junction (NMJ). In the second example, Mdx mice, a model for Duchenne muscular dystrophy, were intravenously injected with AAV8-BGN. The treatment ameliorated motor deficits, mitigated muscle histopathologies, decreased plasma creatine kinase activities, and upregulated expression of utrophin and DAPC component proteins. We propose that protein-anchoring therapy could be applied to hereditary/acquired defects in ECM and secreted proteins, as well as therapeutic overexpression of such factors.
Collapse
Affiliation(s)
- Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Japan.
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Japan
| |
Collapse
|
93
|
Generation of Functioning Nephrons by Implanting Human Pluripotent Stem Cell-Derived Kidney Progenitors. Stem Cell Reports 2018; 10:766-779. [PMID: 29429961 PMCID: PMC5918196 DOI: 10.1016/j.stemcr.2018.01.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/14/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) hold great promise for understanding kidney development and disease. We reproducibly differentiated three genetically distinct wild-type hPSC lines to kidney precursors that underwent rudimentary morphogenesis in vitro. They expressed nephron and collecting duct lineage marker genes, several of which are mutated in human kidney disease. Lentiviral-transduced hPSCs expressing reporter genes differentiated similarly to controls in vitro. Kidney progenitors were subcutaneously implanted into immunodeficient mice. By 12 weeks, they formed organ-like masses detectable by bioluminescence imaging. Implants included perfused glomeruli containing human capillaries, podocytes with regions of mature basement membrane, and mesangial cells. After intravenous injection of fluorescent low-molecular-weight dextran, signal was detected in tubules, demonstrating uptake from glomerular filtrate. Thus, we have developed methods to trace hPSC-derived kidney precursors that formed functioning nephrons in vivo. These advances beyond in vitro culture are critical steps toward using hPSCs to model and treat kidney diseases. Reproducible differentiation to kidney progenitors in 3 hESC lines After subcutaneous implantation, kidney-like tissues detectable by bioluminescence Implant nephrons contain glomeruli, proximal and distal tubules, and collecting ducts Vascularized glomeruli filter intravenously injected low-molecular-weight dextran
Collapse
|
94
|
Beaufils C, Farlay D, Machuca-Gayet I, Fassier A, Zenker M, Freychet C, Bonnelye E, Bertholet-Thomas A, Ranchin B, Bacchetta J. Skeletal impairment in Pierson syndrome: Is there a role for lamininβ2 in bone physiology? Bone 2018; 106:187-193. [PMID: 29051055 DOI: 10.1016/j.bone.2017.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/10/2017] [Accepted: 10/12/2017] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Pierson syndrome is caused by a mutation of LAMB2, encoding for laminin β2. Clinical phenotype is variable but usually associates congenital nephrotic syndrome (CNS) and ocular abnormalities. Neuromuscular impairment has also been described. METHODS We report on a 15-year old girl, suffering from Pierson Syndrome, who developed severe bone deformations during puberty. This patient initially displayed CNS and microcoria, leading to the clinical diagnosis of Pierson syndrome. Genetic analysis revealed a truncating mutation and a splice site mutation of LAMB2. The patient received a renal transplantation (R-Tx) at the age of 3. After R-Tx, renal evolution was simple, the patient receiving low-dose corticosteroids, tacrolimus and mycophenolate mofetil. At the age of 12, bone deformations progressively appeared. At the time of bone impairment, renal function was subnormal (glomerular filtration rate using iohexol clearance 50mL/min per 1.73m2), and parameters of calcium/phosphate metabolism were normal (calcium 2.45mmol/L, phosphorus 1.30mmol/L, PTH 81ng/L, ALP 334U/L, 25OH-D 73nmol/L). Radiographs showed major deformations such as scoliosis, genu varum and diffuse epiphyseal abnormalities. A high resolution scanner (HR-pQCT) was performed, demonstrating a bone of "normal low" quantity and quality; major radial and cubital deformations were observed. Stainings of laminin β2 were performed on bone and renal samples from the patient and healthy controls: as expected, laminin β2 was expressed in the control kidney but not in the patient's renal tissue, and a similar pattern was observed in bone. CONCLUSION This is the first case of skeletal impairment ever described in Pierson syndrome. Integrin α3β1, receptor for laminin β2, are found in podocytes and osteoblasts, and the observation of both the presence of laminin β2 staining in healthy bone and its absence in the patient's bone raises the question of a potential role of laminin β2 in bone physiology.
Collapse
Affiliation(s)
- Camille Beaufils
- Centre de Référence des Maladies Rénales Rares, Hôpital Femme Mère Enfant, Hospices, Civils de Lyon, 69677 Bron, France.
| | - Delphine Farlay
- INSERM, UMR 1033, Université Claude Bernard Lyon 1, Lyon, France
| | | | - Alice Fassier
- Service de Chirurgie Orthopédique Pédiatrique, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Martin Zenker
- Institute of Human Genetics, University Hospital Magdeburg, Germany
| | - Caroline Freychet
- Centre de Référence des Maladies Rénales Rares, Hôpital Femme Mère Enfant, Hospices, Civils de Lyon, 69677 Bron, France
| | - Edith Bonnelye
- INSERM, UMR 1033, Université Claude Bernard Lyon 1, Lyon, France
| | - Aurélia Bertholet-Thomas
- Centre de Référence des Maladies Rénales Rares, Hôpital Femme Mère Enfant, Hospices, Civils de Lyon, 69677 Bron, France
| | - Bruno Ranchin
- Centre de Référence des Maladies Rénales Rares, Hôpital Femme Mère Enfant, Hospices, Civils de Lyon, 69677 Bron, France
| | - Justine Bacchetta
- Centre de Référence des Maladies Rénales Rares, Hôpital Femme Mère Enfant, Hospices, Civils de Lyon, 69677 Bron, France; INSERM, UMR 1033, Université Claude Bernard Lyon 1, Lyon, France; Service de Chirurgie Orthopédique Pédiatrique, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France; Institute of Human Genetics, University Hospital Magdeburg, Germany; Faculté de Médecine Lyon Est, Université de Lyon, France, Lyon.
| |
Collapse
|
95
|
Abstract
The glomerular basement membrane (GBM) is a specialized structure with a significant role in maintaining the glomerular filtration barrier. This GBM is formed from the fusion of two basement membranes during development and its function in the filtration barrier is achieved by key extracellular matrix components including type IV collagen, laminins, nidogens, and heparan sulfate proteoglycans. The characteristics of specific matrix isoforms such as laminin-521 (α5β2γ1) and the α3α4α5 chain of type IV collagen are essential for the formation of a mature GBM and the restricted tissue distribution of these isoforms makes the GBM a unique structure. Detailed investigation of the GBM has been driven by the identification of inherited abnormalities in matrix proteins and the need to understand pathogenic mechanisms causing severe glomerular disease. A well-described hereditary GBM disease is Alport syndrome, associated with a progressive glomerular disease, hearing loss, and lens defects due to mutations in the genes COL4A3, COL4A4, or COL4A5. Other proteins associated with inherited diseases of the GBM include laminin β2 in Pierson syndrome and LMX1B in nail patella syndrome. The knowledge of these genetic mutations associated with GBM defects has enhanced our understanding of cell-matrix signaling pathways affected in glomerular disease. This review will address current knowledge of GBM-associated abnormalities and related signaling pathways, as well as discussing the advances toward disease-targeted therapies for patients with glomerular disease.
Collapse
Affiliation(s)
- Christine Chew
- Faculty of Biology Medicine and Health, Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Rachel Lennon
- Faculty of Biology Medicine and Health, Wellcome Trust Centre for Cell-Matrix Research, Division of Cell Matrix Biology, School of Biological Sciences, University of Manchester, Manchester, United Kingdom.,Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
96
|
Perry G, Xiao W, Welsh GI, Perriman AW, Lennon R. Engineered basement membranes: from in vivo considerations to cell-based assays. Integr Biol (Camb) 2018; 10:680-695. [DOI: 10.1039/c8ib00138c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Engineered basement membranes are required to mimic in vivo properties within cell-based assays.
Collapse
Affiliation(s)
- Guillaume Perry
- Sorbonne Université, Laboratoire d’Electronique et d’Electromagnétisme
- F-75005 Paris
- France
| | - Wenjin Xiao
- School of Cellular and Molecular Medicine, University of Bristol
- BS8 1TD Bristol
- UK
| | - Gavin I. Welsh
- Bristol Renal, Bristol Medical School, University of Bristol
- BS1 3NY Bristol
- UK
| | - Adam W. Perriman
- School of Cellular and Molecular Medicine, University of Bristol
- BS8 1TD Bristol
- UK
| | - Rachel Lennon
- Wellcome Trust Centre for Cell Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester
- M13 9PT Manchester
- UK
| |
Collapse
|
97
|
Funk SD, Bayer RH, Malone AF, McKee KK, Yurchenco PD, Miner JH. Pathogenicity of a Human Laminin β2 Mutation Revealed in Models of Alport Syndrome. J Am Soc Nephrol 2017; 29:949-960. [PMID: 29263159 DOI: 10.1681/asn.2017090997] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 11/19/2017] [Indexed: 01/15/2023] Open
Abstract
Pierson syndrome is a congenital nephrotic syndrome with eye and neurologic defects caused by mutations in laminin β2 (LAMB2), a major component of the glomerular basement membrane (GBM). Pathogenic missense mutations in human LAMB2 cluster in or near the laminin amino-terminal (LN) domain, a domain required for extracellular polymerization of laminin trimers and basement membrane scaffolding. Here, we investigated an LN domain missense mutation, LAMB2-S80R, which was discovered in a patient with Pierson syndrome and unusually late onset of proteinuria. Biochemical data indicated that this mutation impairs laminin polymerization, which we hypothesized to be the cause of the patient's nephrotic syndrome. Testing this hypothesis in genetically altered mice showed that the corresponding amino acid change (LAMB2-S83R) alone is not pathogenic. However, expression of LAMB2-S83R significantly increased the rate of progression to kidney failure in a Col4a3-/- mouse model of autosomal recessive Alport syndrome and increased proteinuria in Col4a5+/- females that exhibit a mild form of X-linked Alport syndrome due to mosaic deposition of collagen α3α4α5(IV) in the GBM. Collectively, these data show the pathogenicity of LAMB2-S80R and provide the first evidence of genetic modification of Alport phenotypes by variation in another GBM component. This finding could help explain the wide range of Alport syndrome onset and severity observed in patients with Alport syndrome, even for family members who share the same COL4 mutation. Our results also show the complexities of using model organisms to investigate genetic variants suspected of being pathogenic in humans.
Collapse
Affiliation(s)
- Steven D Funk
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri and
| | - Raymond H Bayer
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri and
| | - Andrew F Malone
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri and
| | - Karen K McKee
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | - Peter D Yurchenco
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey
| | - Jeffrey H Miner
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri and
| |
Collapse
|
98
|
Kang HG, Lee M, Lee KB, Hughes M, Kwon BS, Lee S, McNagny KM, Ahn YH, Ko JM, Ha IS, Choi M, Cheong HI. Loss of podocalyxin causes a novel syndromic type of congenital nephrotic syndrome. Exp Mol Med 2017; 49:e414. [PMID: 29244787 PMCID: PMC5750479 DOI: 10.1038/emm.2017.227] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/01/2017] [Accepted: 07/11/2017] [Indexed: 01/09/2023] Open
Abstract
Many cellular structures directly imply specific biological functions. For example, normal slit diaphragm structures that extend from podocyte foot processes ensure the filtering function of renal glomeruli. These slits are covered by a number of surface proteins, such as nephrin, podocin, podocalyxin and CD2AP. Here we report a human patient presenting with congenital nephrotic syndrome, omphalocele and microcoria due to two loss-of-function mutations in PODXL, which encodes podocalyxin, inherited from each parent. This set of symptoms strikingly mimics previously reported mouse Podxl−/− embryos, emphasizing the essential function of PODXL in mammalian kidney development and highlighting this patient as a human PODXL-null model. The results underscore the utility of current genomics approaches to provide insights into the genetic mechanisms of human disease traits through molecular diagnosis.
Collapse
Affiliation(s)
- Hee Gyung Kang
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Research Coordination Center for Rare Diseases, Seoul National University Hospital, Seoul, Republic of Korea
| | - Moses Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyoung Boon Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Michael Hughes
- The Biomedical Research Centre, The University of British Colombia, Vancouver, British Columbia, Canada
| | - Bo Sang Kwon
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sangmoon Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kelly M McNagny
- The Biomedical Research Centre, The University of British Colombia, Vancouver, British Columbia, Canada
| | - Yo Han Ahn
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Research Coordination Center for Rare Diseases, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jung Min Ko
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Research Coordination Center for Rare Diseases, Seoul National University Hospital, Seoul, Republic of Korea
| | - Il-Soo Ha
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Murim Choi
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hae Il Cheong
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea.,Research Coordination Center for Rare Diseases, Seoul National University Hospital, Seoul, Republic of Korea.,Kidney Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
99
|
Application of next-generation sequencing technology to diagnosis and treatment of focal segmental glomerulosclerosis. Clin Exp Nephrol 2017; 22:491-500. [PMID: 28752288 PMCID: PMC5956018 DOI: 10.1007/s10157-017-1449-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/06/2017] [Indexed: 12/15/2022]
Abstract
A broad range of genetic and non-genetic factors can lead to kidney injury that manifests as focal segmental glomerulosclerosis (FSGS), which can be classified into primary (idiopathic) and secondary forms. Previous genetic approaches to familial or sporadic cases of FSGS or steroid-resistant nephrotic syndrome identified causal mutations in a subset of genes. Recently, next-generation sequencing (NGS) approaches are becoming a part of a standard assessment in medical genetics. Current knowledge of the comprehensive genomic information is changing the way we think about FSGS and draws attention not only to identification of novel causal genes, but also to potential roles for combinations of mutations in multiple genes, mutations with complex inheritance, and susceptibility genes with variable penetrance carrying relatively minor but significant effects. This review provides an update on recent advances in the genetic analysis of FSGS and highlights the potential as well as the new challenges of NGS for diagnosis and mechanism-based treatment of FSGS.
Collapse
|
100
|
Kino J, Tsukaguchi H, Kimata T, Nguyen HT, Nakano Y, Miyake N, Matsumoto N, Kaneko K. Nephron development and extrarenal features in a child with congenital nephrotic syndrome caused by null LAMB2 mutations. BMC Nephrol 2017; 18:220. [PMID: 28683731 PMCID: PMC5501564 DOI: 10.1186/s12882-017-0632-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 06/22/2017] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Congenital nephrotic syndrome (CNS) is a rare disorder caused by various structural and developmental defects of glomeruli. It occurs typically as an isolated kidney disorder but associates sometimes with other systemic, extrarenal manifestations. CASE PRESENTATIONS An infant presented with severe CNS, which progressed rapidly to renal failure at age of 3 months and death at 27 months. The clinical phenotypes and genetic causes were studied, including the renal pathology at autopsy. Besides the CNS, the affected child had remarkable right-side predominant eye-ball hypoplasia with bilateral anterior chamber dysgenesis (microcoria). Brain MRI revealed grossly normal development in the cerebrum, cerebellum, and brain stem. Auditory brainstem responses were bilaterally blunted, suggesting a defective auditory system. At autopsy, both kidneys were mildly atrophied with persistent fetal lobulation. Microscopic examination showed a diffuse global sclerosis. However, despite of the smaller size of glomeruli, the nephron number remained similar to that of the age-matched control. Whole-exome sequencing revealed that the affected child was compound heterozygous for novel truncating LAMB2 mutations: a 4-bp insertion (p.Gly1693Alafs*8) and a splicing donor-site substitution (c.1225 + 1G > A), presumably deleting the coiled-coil domains that form the laminin 5-2-1 heterotrimer complex. CONCLUSIONS Our case represents a variation of Pierson syndrome that accompanies CNS with unilateral ocular hypoplasia. The average number but smaller glomeruli could reflect either mal-development or glomerulosclerosis. Heterogeneous clinical expression of LAMB2 defects may associate with the difference in fetal β1 subtype compensation among affected tissues. Further study is necessary to evaluate incidence and features of auditory defect under LAMB2 deficiency.
Collapse
Affiliation(s)
- Jiro Kino
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shimachi, Hirakata, Osaka, 573-1010, Japan
| | - Hiroyasu Tsukaguchi
- Second Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi Hirakata, Osaka, 573-1010, Japan.
| | - Takahisa Kimata
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shimachi, Hirakata, Osaka, 573-1010, Japan
| | - Huan Thanh Nguyen
- Second Department of Internal Medicine, Kansai Medical University, 2-5-1 Shinmachi Hirakata, Osaka, 573-1010, Japan
| | - Yorika Nakano
- Department of Pathology and Laboratory Medicine, Kansai Medical University, Osaka, Japan.,Present Address: Department of Histopathology and Cytology, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Kazunari Kaneko
- Department of Pediatrics, Kansai Medical University, 2-5-1 Shimachi, Hirakata, Osaka, 573-1010, Japan
| |
Collapse
|