1
|
Skoczynski K, Kraus A, Daniel C, Büttner-Herold M, Amann K, Schiffer M, Hermann K, Herrnberger-Eimer L, Tamm ER, Buchholz B. The extracellular matrix protein fibronectin promotes metanephric kidney development. Pflugers Arch 2024; 476:963-974. [PMID: 38563997 PMCID: PMC11139724 DOI: 10.1007/s00424-024-02954-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/07/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
Complex interactions of the branching ureteric bud (UB) and surrounding mesenchymal cells during metanephric kidney development determine the final number of nephrons. Impaired nephron endowment predisposes to arterial hypertension and chronic kidney disease. In the kidney, extracellular matrix (ECM) proteins are usually regarded as acellular scaffolds or as the common histological end-point of chronic kidney diseases. Since only little is known about their physiological role in kidney development, we aimed for analyzing the expression and role of fibronectin. In mouse, fibronectin was expressed during all stages of kidney development with significant changes over time. At embryonic day (E) 12.5 and E13.5, fibronectin lined the UB epithelium, which became less pronounced at E16.5 and then switched to a glomerular expression in the postnatal and adult kidneys. Similar results were obtained in human kidneys. Deletion of fibronectin at E13.5 in cultured metanephric mouse kidneys resulted in reduced kidney sizes and impaired glomerulogenesis following reduced cell proliferation and branching of the UB epithelium. Fibronectin colocalized with alpha 8 integrin and fibronectin loss caused a reduction in alpha 8 integrin expression, release of glial-derived neurotrophic factor and expression of Wnt11, both of which are promoters of UB branching. In conclusion, the ECM protein fibronectin acts as a regulator of kidney development and is a determinant of the final nephron number.
Collapse
Affiliation(s)
- Kathrin Skoczynski
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Andre Kraus
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Kerstin Amann
- Department of Nephropathology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Kristina Hermann
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | | | - Ernst R Tamm
- Institute of Human Anatomy and Embryology, University of Regensburg, Regensburg, Germany
| | - Bjoern Buchholz
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
2
|
Kanazawa Y, Takahashi T, Higuchi T, Miyachi R, Nagano M, Koinuma S, Shigeyoshi Y. Effects of stretching on the basement membrane structure in the soleus muscle of Wistar rats. Med Mol Morphol 2023; 56:11-19. [PMID: 36107241 DOI: 10.1007/s00795-022-00335-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 09/03/2022] [Indexed: 11/27/2022]
Abstract
The basement membrane (BM), mainly composed of collagen IV, plays an important role in the maintenance, protection, and recovery of muscle fibers. Collagen IV expression is maintained by the balance between synthetic and degradative factors, which changes depending on the level of muscle activity. For example, exercise increases collagen IV synthesis, whereas inactivity decreases collagen IV synthesis. However, the effects of stretching on the BM structure remain unclear. Therefore, to investigate the effects of stretching on the BM of the skeletal muscle, we continuously applied stretching to the rat soleus muscle and examined the altered expression of BM-related factors and structure using quantitative polymerase chain reaction (qPCR), western blotting, zymography, immunohistochemistry, and electron microscopy. The results show that stretching increased the matrix metalloproteinase 14 (MMP14) expression and MMP2 activity, and decreased the collagen IV expression and width of the lamina densa in the soleus muscle. These results suggest that stretching promotes BM degradation in the rat soleus muscle. The findings of this study indicate a new influence of stretching on skeletal muscles, and may contribute to the new use of stretching in rehabilitation and sports fields.
Collapse
Affiliation(s)
- Yuji Kanazawa
- Department of Medical Technology and Clinical Engineering, Hokuriku University, Ishikawa, Kanazawa, 920-1180, Japan.
| | - Tatsuo Takahashi
- Department of Clinical Pharmacology, Hokuriku University, Kanazawa, 920-1181, Japan
| | - Takashi Higuchi
- Department of Physical Therapy, Osaka University of Human Sciences, Shojyaku, Settsu, 566-8501, Japan
| | - Ryo Miyachi
- Department of Medical Technology and Clinical Engineering, Hokuriku University, Ishikawa, Kanazawa, 920-1180, Japan
| | - Mamoru Nagano
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kindai University, Ohnohigashi, Osakasayama, 589-8511, Japan
| | - Satoshi Koinuma
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kindai University, Ohnohigashi, Osakasayama, 589-8511, Japan
| | - Yasufumi Shigeyoshi
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kindai University, Ohnohigashi, Osakasayama, 589-8511, Japan
| |
Collapse
|
3
|
Kanazawa Y, Nagano M, Koinuma S, Sugiyo S, Shigeyoshi Y. Effects of Endurance Exercise on Basement Membrane in the Soleus Muscle of Aged Rats. Acta Histochem Cytochem 2021; 54:167-175. [PMID: 34764525 PMCID: PMC8569134 DOI: 10.1267/ahc.21-00057] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 09/17/2021] [Indexed: 01/22/2023] Open
Abstract
The basement membrane (BM)-related factors, including collagen IV, are important for the maintenance and recovery of skeletal muscles. Aging impairs the expression of BM-related factors during recovery after disuse atrophy. Muscle activity facilitates collagen synthesis that constitutes the BM. However, the effect of endurance exercise on the BM of aged muscles is unclear. Thus, to understand the effect of endurance exercise on the BM of the skeletal muscle in aged rats, we prescribed treadmill running in aged rats and compared the differences in the expression of BM-related factors between the aged rats with and without exercise habits. Aged rats were subjected to endurance exercise via treadmill running. Exercise increased the mRNA expression levels of the BM-related factors, the area and intensity of collagen IV-immunoreactivity and the width of lamina densa in the soleus muscle of aged rats. These finding suggests that endurance exercise promotes BM construction in aged rats.
Collapse
Affiliation(s)
- Yuji Kanazawa
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine
| | - Mamoru Nagano
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine
| | - Satoshi Koinuma
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine
| | - Shinichi Sugiyo
- Department of Physical Therapy, Osaka University of Human Sciences
| | - Yasufumi Shigeyoshi
- Department of Anatomy and Neurobiology, Kindai University Faculty of Medicine
| |
Collapse
|
4
|
Schuh MP, Alkhudairy L, Potter A, Potter SS, Chetal K, Thakkar K, Salomonis N, Kopan R. The Rhesus Macaque Serves As a Model for Human Lateral Branch Nephrogenesis. J Am Soc Nephrol 2021; 32:1097-1112. [PMID: 33789950 PMCID: PMC8259676 DOI: 10.1681/asn.2020101459] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/18/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Most nephrons are added in late gestation. Truncated extrauterine nephrogenesis in premature infants results in fewer nephrons and significantly increased risk for CKD in adulthood. To overcome the ethical and technical difficulties associated with studies of late-gestation human fetal kidney development, third-trimester rhesus macaques served as a model to understand lateral branch nephrogenesis (LBN) at the molecular level. METHODS Immunostaining and 3D rendering assessed morphology. Single-cell (sc) and single-nucleus (sn) RNA-Seq were performed on four cortically enriched fetal rhesus kidneys of 129-131 days gestational age (GA). An integrative bioinformatics strategy was applied across single-cell modalities, species, and time. RNAScope validation studies were performed on human archival tissue. RESULTS Third-trimester rhesus kidney undergoes human-like LBN. scRNA-Seq of 23,608 cells revealed 37 transcriptionally distinct cell populations, including naïve nephron progenitor cells (NPCs), with the prior noted marker genes CITED1, MEOX1, and EYA1 (c25). These same populations and markers were reflected in snRNA-Seq of 5972 nuclei. Late-gestation rhesus NPC markers resembled late-gestation murine NPC, whereas early second-trimester human NPC markers aligned to midgestation murine NPCs. New, age-specific rhesus NPCs (SHISA8) and ureteric buds (POU3F4 and TWIST) predicted markers were verified in late-gestation human archival samples. CONCLUSIONS Rhesus macaque is the first model of bona fide LBN, enabling molecular studies of late gestation, human-like nephrogenesis. These molecular findings support the hypothesis that aging nephron progenitors have a distinct molecular signature and align to their earlier human counterparts, with unique markers highlighting LBN-specific progenitor maturation.
Collapse
Affiliation(s)
- Meredith P. Schuh
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Lyan Alkhudairy
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Andrew Potter
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - S. Steven Potter
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Kashish Chetal
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Kairavee Thakkar
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Raphael Kopan
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
5
|
Nigro EA, Boletta A. Role of the polycystins as mechanosensors of extracellular stiffness. Am J Physiol Renal Physiol 2021; 320:F693-F705. [PMID: 33615892 DOI: 10.1152/ajprenal.00545.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Polycystin-1 (PC-1) is a transmembrane protein, encoded by the PKD1 gene, mutated in autosomal dominant polycystic kidney disease (ADPKD). This common genetic disorder, characterized by cyst formation in both kidneys, ultimately leading to renal failure, is still waiting for a definitive treatment. The overall function of PC-1 and the molecular mechanism responsible for cyst formation are slowly coming to light, but they are both still intensively studied. In particular, PC-1 has been proposed to act as a mechanosensor, although the precise signal that activates the mechanical properties of this protein has been long debated and questioned. In this review, we report studies and evidence of PC-1 function as a mechanosensor, starting from the peculiarity of its structure, through the long journey that progressively shed new light on the potential initiating events of cystogenesis, concluding with the description of PC-1 recently shown ability to sense the mechanical stimuli provided by the stiffness of the extracellular environment. These new findings have potentially important implications for the understanding of ADPKD pathophysiology and potentially for designing new therapies.NEW & NOTEWORTHY Polycystin-1 has recently emerged as a possible receptor able to sense extracellular stiffness and to negatively control the cellular actomyosin contraction machinery. Here, we revisit a large body of literature on autosomal dominant polycystic kidney disease providing a new possible mechanistic view on the topic.
Collapse
Affiliation(s)
- Elisa A Nigro
- Molecular Basis of Cystic Kidney Diseases, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Boletta
- Molecular Basis of Cystic Kidney Diseases, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
6
|
Hemker SL, Cerqueira DM, Bodnar AJ, Cargill KR, Clugston A, Anslow MJ, Sims-Lucas S, Kostka D, Ho J. Deletion of hypoxia-responsive microRNA-210 results in a sex-specific decrease in nephron number. FASEB J 2020; 34:5782-5799. [PMID: 32141129 DOI: 10.1096/fj.201902767r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/06/2020] [Accepted: 02/19/2020] [Indexed: 12/25/2022]
Abstract
Low nephron number results in an increased risk of developing hypertension and chronic kidney disease. Intrauterine growth restriction is associated with a nephron deficit in humans, and is commonly caused by placental insufficiency, which results in fetal hypoxia. The underlying mechanisms by which hypoxia impacts kidney development are poorly understood. microRNA-210 is the most consistently induced microRNA in hypoxia and is known to promote cell survival in a hypoxic environment. In this study, the role of microRNA-210 in kidney development was evaluated using a global microRNA-210 knockout mouse. A male-specific 35% nephron deficit in microRNA-210 knockout mice was observed. Wnt/β-catenin signaling, a pathway crucial for nephron differentiation, was misregulated in male kidneys with increased expression of the canonical Wnt target lymphoid enhancer binding factor 1. This coincided with increased expression of caspase-8-associated protein 2, a known microRNA-210 target and apoptosis signal transducer. Together, these data are consistent with a sex-specific requirement for microRNA-210 in kidney development.
Collapse
Affiliation(s)
- Shelby L Hemker
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Débora M Cerqueira
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew J Bodnar
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kasey R Cargill
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Andrew Clugston
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.,Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Melissa J Anslow
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Sunder Sims-Lucas
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Dennis Kostka
- Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.,Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jacqueline Ho
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Rangos Research Center, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
7
|
Genderen AM, Jansen J, Cheng C, Vermonden T, Masereeuw R. Renal Tubular- and Vascular Basement Membranes and their Mimicry in Engineering Vascularized Kidney Tubules. Adv Healthc Mater 2018; 7:e1800529. [PMID: 30091856 DOI: 10.1002/adhm.201800529] [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: 05/15/2018] [Revised: 07/18/2018] [Indexed: 01/09/2023]
Abstract
The high prevalence of chronic kidney disease leads to an increased need for renal replacement therapies. While there are simply not enough donor organs available for transplantation, there is a need to seek other therapeutic avenues as current dialysis modalities are insufficient. The field of regenerative medicine and whole organ engineering is emerging, and researchers are looking for innovative ways to create (part of) a functional new organ. To biofabricate a kidney or its functional units, it is necessary to understand and learn from physiology to be able to mimic the specific tissue properties. Herein is provided an overview of the knowledge on tubular and vascular basement membranes' biochemical components and biophysical properties, and the major differences between the two basement membranes are highlighted. Furthermore, an overview of current trends in membrane technology for developing renal replacement therapies and to stimulate kidney regeneration is provided.
Collapse
Affiliation(s)
- Anne Metje Genderen
- Division of PharmacologyUtrecht Institute for Pharmaceutical Sciences 3584 CG Utrecht The Netherlands
| | - Jitske Jansen
- Division of PharmacologyUtrecht Institute for Pharmaceutical Sciences 3584 CG Utrecht The Netherlands
| | - Caroline Cheng
- Regenerative Medicine Center UtrechtUniversity Medical Center Utrecht 3584 CT Utrecht The Netherlands
- Department of Nephrology and HypertensionUniversity Medical Center Utrecht 3508 GA Utrecht The Netherlands
- Department of Experimental CardiologyErasmus Medical Center 3015 GD Rotterdam The Netherlands
| | - Tina Vermonden
- Division of PharmaceuticsUtrecht Institute for Pharmaceutical Sciences 3584 CG Utrecht The Netherlands
| | - Rosalinde Masereeuw
- Division of PharmacologyUtrecht Institute for Pharmaceutical Sciences 3584 CG Utrecht The Netherlands
| |
Collapse
|
8
|
Mak KM, Mei R. Basement Membrane Type IV Collagen and Laminin: An Overview of Their Biology and Value as Fibrosis Biomarkers of Liver Disease. Anat Rec (Hoboken) 2017; 300:1371-1390. [PMID: 28187500 DOI: 10.1002/ar.23567] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/05/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022]
Abstract
Basement membranes provide structural support to epithelium, endothelium, muscles, fat cells, Schwann cells, and axons. Basement membranes are multifunctional: they modulate cellular behavior, regulate organogenesis, promote tissue repair, form a barrier to filtration and tumor metastasis, bind growth factors, and mediate angiogenesis. All basement membranes contain type IV collagen (Col IV), laminin, nidogen, and perlecan. Col IV and laminin self-assemble into two independent supramolecular networks that are linked to nidogen and perlecan to form a morphological discernable basement membrane/basal lamina. The triple helical region, 7S domain and NCI domain of Col IV, laminin and laminin fragment P1 have been evaluated as noninvasive fibrosis biomarkers of alcoholic liver disease, viral hepatitis, and nonalcoholic fatty liver disease. Elevated serum Col IV and laminin are related to degrees of fibrosis and severity of hepatitis, and may reflect hepatic basement membrane metabolism. But the serum assays have not been linked to disclosing the anatomical sites and lobular distribution of perisinusoidal basement membrane formation in the liver. Hepatic sinusoids normally lack a basement membrane, although Col IV is a normal matrix component of the space of Disse. In liver disease, laminin deposits in the space of Disse and codistributes with Col IV, forming a perisinusoidal basement membrane. Concomitantly, the sinusoidal endothelium loses its fenestrae and is transformed into vascular type endothelium. These changes lead to capillarization of hepatic sinusoids, a significant pathology that impairs hepatic function. Accordingly, codistribution of Col IV and laminin serves as histochemical marker of perisinusoidal basement membrane formation in liver disease. Anat Rec, 300:1371-1390, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ki M Mak
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Rena Mei
- Department of Medical Education and Center for Anatomy and Functional Morphology, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
9
|
Di Russo J, Hannocks MJ, Luik AL, Song J, Zhang X, Yousif L, Aspite G, Hallmann R, Sorokin L. Vascular laminins in physiology and pathology. Matrix Biol 2017; 57-58:140-148. [DOI: 10.1016/j.matbio.2016.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022]
|
10
|
Marciano DK. A holey pursuit: lumen formation in the developing kidney. Pediatr Nephrol 2017; 32:7-20. [PMID: 26902755 PMCID: PMC5495142 DOI: 10.1007/s00467-016-3326-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/23/2015] [Accepted: 01/12/2016] [Indexed: 02/07/2023]
Abstract
The formation of polarized epithelial tubules is a hallmark of kidney development. One of the fundamental principles in tubulogenesis is that epithelia coordinate the polarity of individual cells with the surrounding cells and matrix. A central feature in this process is the segregation of membranes into spatially and functionally distinct apical and basolateral domains, and the generation of a luminal space at the apical surface. This review examines our current understanding of the cellular and molecular mechanisms that underlie the establishment of apical-basal polarity and lumen formation in developing renal epithelia, including the roles of cell-cell and cell-matrix interactions and polarity complexes. We highlight growing evidence from animal models, and correlate these findings with models of tubulogenesis from other organ systems, and from in vitro studies.
Collapse
Affiliation(s)
- Denise K. Marciano
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. H5.102, Dallas, TX 75390-8856
| |
Collapse
|
11
|
Laurie G, Leblond C. What is Known of the Production of Basement Membrane Components. J Histochem Cytochem 2016; 31:159-163. [DOI: 10.1177/31.1a_suppl.6186721] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Immunohistochemistry was used to identify basement membrane components and examine their production by associated cells. Four substances were identified in a series of basement membranes in rats aged 20 days to 34 months, namely, type IV collagen, laminin, heparan sulfate proteoglycan, and fibronectin. They were then all localized to the basal lamina part of basement membranes and, presumably, are integrated within this layer. The production of type IV collagen was first examined in the embryonic endodermal cells associated with Reichert's membrane in the rat parietal yolk sac. The rough endoplasmic reticulum (rER), Golgi apparatus, and putative secretory granules of endodermal cells were immunostained, suggesting that these organelles participated in the biogenesis of type IV collagen. However, in rats aged 20 days or more, the cells associated with basement membranes were usually unstained. An exception was noted in the continually growing incisor tooth where the endothelial cells at the proliferating end usually showed immunostaining of rER and Golgi apparatus. It is, therefore, proposed that the formation of type IV collagen for basement membrane occurs at an early stage of development in the life of producer cells. Little is known of the formation of other basement membrane components during development, but there is immunohistochemical evidence that laminin and fibronectin are produced along the same secretory pathway as type IV collagen.
Collapse
Affiliation(s)
- G.W. Laurie
- Department of Anatomy, McGill University, Montreal, Quebec, Canada H3A 2B2 (OA 82-272S2)
| | - C.P. Leblond
- Department of Anatomy, McGill University, Montreal, Quebec, Canada H3A 2B2 (OA 82-272S2)
| |
Collapse
|
12
|
Halt KJ, Pärssinen HE, Junttila SM, Saarela U, Sims-Lucas S, Koivunen P, Myllyharju J, Quaggin S, Skovorodkin IN, Vainio SJ. CD146(+) cells are essential for kidney vasculature development. Kidney Int 2016; 90:311-324. [PMID: 27165833 DOI: 10.1016/j.kint.2016.02.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/06/2016] [Accepted: 02/11/2016] [Indexed: 01/14/2023]
Abstract
The kidney vasculature is critical for renal function, but its developmental assembly mechanisms remain poorly understood and models for studying its assembly dynamics are limited. Here, we tested whether the embryonic kidney contains endothelial cells (ECs) that are heterogeneous with respect to VEGFR2/Flk1/KDR, CD31/PECAM, and CD146/MCAM markers. Tie1Cre;R26R(YFP)-based fate mapping with a time-lapse in embryonic kidney organ culture successfully depicted the dynamics of kidney vasculature development and the correlation of the process with the CD31(+) EC network. Depletion of Tie1(+) or CD31(+) ECs from embryonic kidneys, with either Tie1Cre-induced diphtheria toxin susceptibility or cell surface marker-based sorting in a novel dissociation and reaggregation technology, illustrated substantial EC network regeneration. Depletion of the CD146(+) cells abolished this EC regeneration. Fate mapping of green fluorescent protein (GFP)-marked CD146(+)/CD31(-) cells indicated that they became CD31(+) cells, which took part in EC structures with CD31(+) wild-type ECs. EC network development depends on VEGF signaling, and VEGF and erythropoietin are expressed in the embryonic kidney even in the absence of any external hypoxic stimulus. Thus, the ex vivo embryonic kidney culture models adopted here provided novel ways for targeting renal EC development and demonstrated that CD146(+) cells are critical for kidney vasculature development.
Collapse
Affiliation(s)
- Kimmo J Halt
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland; Center of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
| | - Heikki E Pärssinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland; Center of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
| | - Sanna M Junttila
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland; Center of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
| | - Ulla Saarela
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland; Center of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
| | - Sunder Sims-Lucas
- Rangos Research Center, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Peppi Koivunen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland
| | - Johanna Myllyharju
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland; Center of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
| | - Susan Quaggin
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland; Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ilya N Skovorodkin
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland; Center of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland
| | - Seppo J Vainio
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, Oulu, Finland; Center of Excellence in Cell-Extracellular Matrix Research, Oulu, Finland; InfoTech Oulu, Oulu, Finland.
| |
Collapse
|
13
|
Hann BD, Baldelomar EJ, Charlton JR, Bennett KM. Measuring the intrarenal distribution of glomerular volumes from histological sections. Am J Physiol Renal Physiol 2016; 310:F1328-36. [PMID: 26984953 DOI: 10.1152/ajprenal.00382.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 03/11/2016] [Indexed: 11/22/2022] Open
Abstract
Glomerular volume is an important metric reflecting glomerular filtration surface area within the kidney. Glomerular hypertrophy, or increased glomerular volume, may be an important marker for renal stress. Current stereological techniques report the average glomerular volume (AVglom) within the kidney. These techniques cannot assess the spatial or regional heterogeneity common in developing renal pathology. Here, we report a novel "unfolding" technique to measure the actual distribution of individual glomerular volumes in a kidney from the two-dimensional glomerulus profiles observed by optical microscopy. The unfolding technique was first developed and tested for accuracy with simulations and then applied to measure the number of glomeruli (Nglom), AVglom, and intrarenal distribution of individual glomerular volume (IVglom) in the oligosyndactyl (Os/(+)) mouse model compared with wild-type (WT) controls. The Os/(+) mice had fewer and larger glomeruli than WT mice: Nglom was 12,126 ± 1,658 (glomeruli/kidney) in the WT mice and 5,516 ± 899 in the Os/(+) mice; AVglom was 2.01 ± 0.28 × 10(-4) mm(3) for the WT mice and 3.47 ± 0.35 × 10(-4) mm(3) for the Os/(+) mice. Comparing the glomerular volume distributions in Os/(+) and WT kidneys, we observed that the Os/(+) distribution peaked at a higher value of IVglom than the WT distribution peak, and glomeruli with a radius greater than 55 μm were more prevalent in the Os/(+) mice (3.4 ± 1.6% of total glomeruli vs. 0.6 ± 1.2% in WT). Finally, the largest profiles were more commonly found in the juxtamedullary region. Unfolding is a novel stereological technique that provides a new quantitative view of glomerular volume distribution in the individual kidney.
Collapse
Affiliation(s)
- Bradley D Hann
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, Hawaii
| | | | - Jennifer R Charlton
- Department of Pediatrics, Division of Nephrology, University of Virginia, Charlottesville, Virginia; and
| | - Kevin M Bennett
- Department of Biology, University of Hawaii at Manoa, Honolulu, Hawaii
| |
Collapse
|
14
|
González-Andrades M, Carriel V, Rivera-Izquierdo M, Garzón I, González-Andrades E, Medialdea S, Alaminos M, Campos A. Effects of Detergent-Based Protocols on Decellularization of Corneas With Sclerocorneal Limbus. Evaluation of Regional Differences. Transl Vis Sci Technol 2015; 4:13. [PMID: 25909036 DOI: 10.1167/tvst.4.2.13] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 01/21/2015] [Indexed: 11/24/2022] Open
Abstract
PURPOSE In this work, we decellularized whole porcine corneas including the sclerocorneal limbus (SCL) and we evaluated regional differences in order to identify an efficient method to decellularize whole corneas for future clinical use. METHODS We analyzed the efficiency of four decellularization protocols based on benzalkonium chloride (BAK), Igepal, sodium dodecyl sulfate (SDS), and Triton X-100 detergents on whole porcine corneas. RESULTS Results showed that the decellularization efficiency of most protocols was low, with the specific protocol resulting in more efficient levels of decellularization being 0.1% SDS for 48 hours, especially in the medium and posterior cornea regions. A significant correlation was found between the decellularization efficiency and the concentration of agent used (P = 0.0174; r = 0.1540), but not for the incubation time (P > 0.05). The analysis of cornea components preservation demonstrated that all protocols were able to preserve the integrity of the Bowman's layer and Descemet's membrane. Although the collagen structure was partially altered, the global decellularization groups showing highest preservation of the ECM collagen contents and orientation were Igepal and SDS, with differences among the three regions of the cornea. All global groups showed high levels of proteoglycan and glycoprotein preservation after decellularization, with the best results were found in the SDS group followed by the Igepal group. CONCLUSIONS These results suggest that very powerful protocols are necessary for whole-cornea decellularization. For the generation of lamelar corneas for clinical use, decellularization regional differences should be taken into account. TRANSLATIONAL RELEVANCE Decellularized whole corneas may be potential therapeutic agents for lamelar keratoplasty.
Collapse
Affiliation(s)
- Miguel González-Andrades
- Tissue Engineering Group, Department of Histology, University of Granada, Spain and Instituto de Investigación Biosanitaria ibs.GRANADA, Spain ; Division of Ophthalmology, University Hospital San Cecilio, Granada, Spain
| | - Victor Carriel
- Tissue Engineering Group, Department of Histology, University of Granada, Spain and Instituto de Investigación Biosanitaria ibs.GRANADA, Spain
| | - Mario Rivera-Izquierdo
- Tissue Engineering Group, Department of Histology, University of Granada, Spain and Instituto de Investigación Biosanitaria ibs.GRANADA, Spain
| | - Ingrid Garzón
- Tissue Engineering Group, Department of Histology, University of Granada, Spain and Instituto de Investigación Biosanitaria ibs.GRANADA, Spain
| | - Elena González-Andrades
- Tissue Engineering Group, Department of Histology, University of Granada, Spain and Instituto de Investigación Biosanitaria ibs.GRANADA, Spain
| | - Santiago Medialdea
- Division of Ophthalmology, University Hospital Virgen de las Nieves, Granada, Spain
| | - Miguel Alaminos
- Tissue Engineering Group, Department of Histology, University of Granada, Spain and Instituto de Investigación Biosanitaria ibs.GRANADA, Spain
| | - Antonio Campos
- Tissue Engineering Group, Department of Histology, University of Granada, Spain and Instituto de Investigación Biosanitaria ibs.GRANADA, Spain
| |
Collapse
|
15
|
Galarreta CI, Thornhill BA, Forbes MS, Simpkins LN, Kim DK, Chevalier RL. Transforming growth factor-β1 receptor inhibition preserves glomerulotubular integrity during ureteral obstruction in adults but worsens injury in neonatal mice. Am J Physiol Renal Physiol 2013; 304:F481-90. [PMID: 23303407 DOI: 10.1152/ajprenal.00496.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Unilateral ureteral obstruction (UUO), a widely used model of chronic kidney disease and congenital obstructive uropathy, causes proximal tubular injury and formation of atubular glomeruli. Because transforming growth factor-β1 (TGF-β1) is a central regulator of renal injury, neonatal and adult mice were subjected to complete UUO while under general anesthesia and treated with vehicle or ALK5 TGF-β1 receptor inhibitor (IN-1130, 30 mg·kg(-1)·day(-1)). After 14 days, glomerulotubular integrity and proximal tubular mass were determined by morphometry of Lotus tetragonolobus lectin distribution, and the fraction of atubular glomeruli was determined by serial section analysis of randomly selected individual glomeruli. Glomerular area, macrophage infiltration, fibronectin distribution, and interstitial collagen were measured by morphometry. Compared with placebo, inhibition of TGF-β1 by IN-1130 decreased apoptosis and formation of atubular glomeruli, prevented parenchymal loss, increased glomerular area and glomerulotubular integrity, and increased proximal tubule fraction of the adult obstructed kidney parenchyma from 17 to 30% (P < 0.05, respectively). IN-1130 decreased macrophage infiltration and fibronectin and collagen deposition in the adult obstructed kidney by ∼50% (P < 0.05, respectively). In contrast to these salutary effects in the adult, IN-1130 caused widespread necrosis in obstructed neonatal kidneys. We conclude that whereas IN-1130 reduces obstructive injury in adult kidneys through preservation of glomerulotubular integrity and proximal tubular mass, TGF-β1 inhibition aggravates obstructive injury in neonates. These results indicate that while caution is necessary in treating congenital uropathies, ALK5 inhibitors may prevent nephron loss due to adult kidney disease.
Collapse
Affiliation(s)
- Carolina I Galarreta
- Dept. of Pediatrics, Univ. of Virginia, Box 800386, Charlottesville, VA 22908, USA
| | | | | | | | | | | |
Collapse
|
16
|
Ultrastructure of the pronephric kidney of embryos and prolarvae of the sea lamprey, Petromyzon marinus. Tissue Cell 2012; 23:393-410. [PMID: 18621169 DOI: 10.1016/0040-8166(91)90057-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/1990] [Revised: 01/02/1991] [Indexed: 11/22/2022]
Abstract
Embryos of lampreys Petromyzon marinus were obtained through a technique of artificial fertilization. Samples of developmental intervals to the prolarval stage were prepared for transmission electron microscopy and the pronephros was examined. The pronephros was visible in the cardiac region of the coelom prior to the time of hatching of embryos and consisted of a renal corpuscle, nephrostomes, and proximal tubules connected to a pronephric duct. The renal corpuscle was comprised of poorly-defined vascular channels and a visceral epithelium of yolk-filled cells, the podocytes, with short major processes and pedicels resting on a basal lamina. The first proximal tubules possessed a delicate brush border of short microvilli but subsequent cellular differentiation yielded cells with all the components required for the process of endocytosis, a process which was demonstrated by uptake of the tracer, horseradish peroxidase. The distal tubules appeared later in development and were noted for abundant mitochondria and an extensive smooth tubular network. The timing of differentiation of various components of the nephron corresponds to that seen during morphogenesis of other vertebrate kidneys.
Collapse
|
17
|
Kao RM, Vasilyev A, Miyawaki A, Drummond IA, McMahon AP. Invasion of distal nephron precursors associates with tubular interconnection during nephrogenesis. J Am Soc Nephrol 2012; 23:1682-90. [PMID: 22904347 DOI: 10.1681/asn.2012030283] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Formation of a functional renal network requires the interconnection of two epithelial tubes: the nephron, which arises from kidney mesenchyme, and the collecting system, which originates from the branching ureteric epithelium. How this connection occurs, however, is incompletely understood. Here, we used high-resolution image analysis in conjunction with genetic labeling of epithelia to visualize and characterize this process. Although the focal absence of basal lamina from renal vesicle stages ensures that both epithelial networks are closely apposed, we found that a patent luminal interconnection is not established until S-shaped body stages. Precursor cells of the distal nephron in the interconnection zone exhibit a characteristic morphology consisting of ill-defined epithelial junctional complexes but without expression of mesenchymal markers such as vimentin and Snai2. Live-cell imaging revealed that before luminal interconnection, distal cells break into the lumen of the collecting duct epithelium, suggesting that an invasive behavior is a key step in the interconnection process. Furthermore, loss of distal cell identity, which we induced by activating the Notch pathway, prevented luminal interconnection. Taken together, these data support a model in which establishing the distal identity of nephron precursor cells closest to the nascent collecting duct epithelium leads to an active cell invasion, which in turn contributes to a patent tubular interconnection between the nephron and collecting duct epithelia.
Collapse
Affiliation(s)
- Robert M Kao
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, 1425 San Pablo Street, Los Angeles, CA 90089, USA
| | | | | | | | | |
Collapse
|
18
|
Xiao L, Li Z, Xu P, Li Z, Xu J, Yang Z. The expression of EPOR in renal cortex during postnatal development. PLoS One 2012; 7:e41993. [PMID: 22844537 PMCID: PMC3406037 DOI: 10.1371/journal.pone.0041993] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 06/28/2012] [Indexed: 12/03/2022] Open
Abstract
Erythropoietin (EPO), known for its role in erythroid differentiation, has been shown to be an important growth factor for brain and heart. EPO is synthesized by fibroblast-like cells in the renal cortex. Prompted by this anatomical relationship and its significant impact on the maturation process of brain and heart, we asked whether EPO could play a role during the development of renal cortex. The relationship between the development of renal cortex and the change of EPO receptor (EPOR), through which EPO could act as a renotropic cytokine, became interesting to us. In this study, the day of birth was recorded as postnatal day 0(P0). P7, P14, P21, P28, P35, P42 and mature mice (postnatal days>56) were used as the animal model of different developmental stages. Immunohistochemistry and Western blotting were used to detect the expression of EPOR in mouse renal cortex. Results showed that expression of EPOR decreased with the development of renal cortex and became stable when kidney became mature. The expression of EPOR was detected at the renal tubule of all developmental stages and a relatively higher expression was observed at P14. However, at the renal corpuscle the expression was only observed at P7 and quickly became undetectable after that. All these suggested that a translocation of EPOR from renal corpuscle to renal tubule may take place during the developmental process of renal cortex. Also, EPO may be an essential element for the maturation of renal cortex, and the requirement for EPO was changed during postnatal development process.
Collapse
Affiliation(s)
- Lu Xiao
- College of Medicine, Nankai University, Tianjin, China
| | - Zhanyong Li
- College of Life Sciences, Nankai University, Tianjin, China
| | - Pengjuan Xu
- College of Medicine, Nankai University, Tianjin, China
| | - Zhigui Li
- College of Medicine, Nankai University, Tianjin, China
| | - Jing Xu
- College of Medicine, Nankai University, Tianjin, China
| | - Zhuo Yang
- College of Medicine, Nankai University, Tianjin, China
- * E-mail:
| |
Collapse
|
19
|
Kuzma-Kuzniarska M, Rak-Raszewska A, Kenny S, Edgar D, Wilm B, Fuente Mora C, Davies JA, Murray P. Integration potential of mouse and human bone marrow-derived mesenchymal stem cells. Differentiation 2011; 83:128-37. [PMID: 22364880 DOI: 10.1016/j.diff.2011.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 10/17/2011] [Accepted: 11/08/2011] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are a multipotent cell population which has been described to exert renoprotective and regenerative effects in experimental models of kidney injury. Several lines of evidence indicate that MSCs also have the ability to contribute to nephrogenesis, suggesting that the cells can be employed in stem cell-based applications aimed at de novo renal tissue generation. In this study we re-evaluate the capacity of mouse and human bone marrow-derived MSCs to contribute to the development of renal tissue using a novel method of embryonic kidney culture. Although MSCs show expression of some genes involved in renal development, their contribution to nephrogenesis is very limited in comparison to other stem cell types tested. Furthermore, we found that both mouse and human MSCs have a detrimental effect on the ex vivo development of mouse embryonic kidney, this effect being mediated through a paracrine action. Stimulation with conditioned medium from a mouse renal progenitor population increases the ability of mouse MSCs to integrate into developing renal tissue and prevents the negative effects on kidney development, but does not appear to enhance their ability to undergo nephrogenesis.
Collapse
|
20
|
Yang DH, McKee KK, Chen ZL, Mernaugh G, Strickland S, Zent R, Yurchenco PD. Renal collecting system growth and function depend upon embryonic γ1 laminin expression. Development 2011; 138:4535-44. [PMID: 21903675 DOI: 10.1242/dev.071266] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In order to understand the functions of laminins in the renal collecting system, the Lamc1 gene was inactivated in the developing mouse ureteric bud (UB). Embryos bearing null alleles exhibited laminin deficiency prior to mesenchymal tubular induction and either failed to develop a UB with involution of the mesenchyme, or developed small kidneys with decreased proliferation and branching, delayed renal vesicle formation and postnatal emergence of a water transport deficit. Embryonic day 12.5 kidneys revealed an almost complete absence of basement membrane proteins and reduced levels of α6 integrin and FGF2. mRNA levels for fibroblast growth factor 2 (FGF2) and mediators of the GDNF/RET and WNT11 signaling pathway were also decreased. Furthermore, collecting duct cells derived from laminin-deficient kidneys and grown in collagen gels were found to proliferate and branch slowly. The laminin-deficient cells exhibited decreased activation of growth factor- and integrin-dependent pathways, whereas heparin lyase-treated and β1 integrin-null cells exhibited more selective decreases. Collectively, these data support a requirement of γ1 laminins for assembly of the collecting duct system basement membrane, in which immobilized ligands act as solid-phase agonists to promote branching morphogenesis, growth and water transport functions.
Collapse
Affiliation(s)
- Dong-Hua Yang
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Sarrab RM, Lennon R, Ni L, Wherlock MD, Welsh GI, Saleem MA. Establishment of conditionally immortalized human glomerular mesangial cells in culture, with unique migratory properties. Am J Physiol Renal Physiol 2011; 301:F1131-8. [PMID: 21653636 DOI: 10.1152/ajprenal.00589.2010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of this study was to establish an immortalized human mesangial cell line similar to mesangial cells in vivo for use as a tool for understanding glomerular cell function. Mesangial cells were isolated from glomerular outgrowths from a normal human kidney, then retrovirally transfected with a temperature-sensitive SV40T antigen+human telomerase (hTERT). Mesangial cells exhibited features of compact cells with small bodies in a confluent monolayer at 33°C, but the cell shape changed to flat and stellate after 5 days in growth-restrictive conditions (37°C). Western blot and immunofluorescence analysis showed that podocyte markers (nephrin, CD2AP, podocin, Wilms' tumor-1) and an endothelial-specific molecule (VE-cadherin) were not detectable in this cell line, whereas markers characteristic of mesangial cells (α-SMA, fibronectin, and PDGFβ-R) were strongly expressed. In migration assays, a significant reduction in wound surface was observed in podocyte and endothelial cells as soon as 12 h (75 and 62%, respectively) and complete wound closure after 24 h. In contrast, no significant change was observed in mesangial cells after 12 h, and even after 48 h the wounds were not completely closed. Until now, conditionally immortalized podocyte and endothelial cell lines derived from mice and humans have been described, and this has greatly boosted research on glomerular physiology and pathology. We have established the first conditionally immortalized human glomerular mesangial cell line, which will be an important adjunct in studies of representative glomerular cells, as well as in coculture studies. Unexpectedly, mesangial cells' ability to migrate seems to be slower than for other glomerular cells, suggesting this line will demonstrate functional properties distinct from previously available mesangial cell cultures. This conditionally immortalized human mesangial cell line represents a new tool for the study of human mesangial cell biology in vitro.
Collapse
Affiliation(s)
- Ramadan M Sarrab
- Children's Renal Unit and Academic Renal Unit, University of Bristol, Southmead Hospital, Bristol, United Kingdom
| | | | | | | | | | | |
Collapse
|
22
|
Kanwar YS, Venkatachalam MA. Ultrastructure of Glomerulus and Juxtaglomerular Apparatus. Compr Physiol 2011. [DOI: 10.1002/cphy.cp080101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Sarkar S, Schmued L. Kainic acid and 3-Nitropropionic acid induced expression of laminin in vascular elements of the rat brain. Brain Res 2010; 1352:239-47. [PMID: 20624377 DOI: 10.1016/j.brainres.2010.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 06/23/2010] [Accepted: 07/05/2010] [Indexed: 10/19/2022]
Abstract
Laminin is a glycoprotein component of the basement membrane and has been reported to be found in different areas of the nervous system including brain endothelial cells, Schwann cells and peripheral nerves. Although the in-vitro studies suggest that laminin plays an important role in growth and neurite extension of cultured neurons, localization of laminin in the brain has been controversial and inconsistent results have been reported. Recently, laminin immunoreactivity has been used as a marker for vascular elements in the brain. In this study, we have investigated the effect of two mechanistically different neurotoxins, kainic acid (KA), an NMDA agonist and 3-Nitropropionic acid (3-NPA), an inhibitor of mitochondrial respiration, on brain vascular elements revealed by laminin immunolabeling. We also explored whether administration of these two neurotoxic drugs correlate with the neuronal degeneration observed after neurotoxic insult by staining with Fluoro-Jade C dye. We have employed single immunolabeling to localize laminin in the brains. In KA treated rats, most of the laminin immunoreactivity is present in the piriform cortex, corpus callosum (myelinated tracts) amygdala, hippocampus, ventral thalamus and tenia tacta. In 3-NPA treated animals, laminin immunoreactivity was confined mostly to the striatum. In contrast, saline treated rats showed very little laminin immunolabeling around capillaries, arteries and in the meningeal membranes. To determine the effects of these neurotoxins on the integrity of the blood brain barrier (BBB), endothelial brain barrier antigen (EBA) immunolabeling was also performed. In addition, we performed CD11b immunolabeling to evaluate the effect of 3-NPA and KA on the activation of microglia in the brain. CD11b was dramatically increased in KA and 3-NPA treated animals. We have also combined laminin immunolabeling with Fluoro-Jade C labeling to evaluate the spatio-temporal association of degenerating neurons and the expression of laminin containing microvessels. Areas which showed intense laminin immunolabeling following KA or 3-NPA exposure correlated with those exhibiting the greatest number of degenerating neurons observed after Fluoro-Jade C staining. EBA-laminin double immunolabeling demonstrated that the expressions of laminin were predominantly localized in the areas (cortex, thalamus and hippocampus) where EBA has been either reduced or is absent. Our results from these experiments demonstrate that vascular laminin expression increases after treatment with KA or 3-NPA, suggesting the occurrence of neovascularization. Microglia may also contribute to the neurotoxic induced neovascularization and neurodegeneration.
Collapse
Affiliation(s)
- Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicological Research (NCTR), Jefferson, AR 72079, USA
| | | |
Collapse
|
24
|
Ren X, Zhang J, Gong X, Niu X, Zhang X, Chen P, Zhang X. Differentiation of murine embryonic stem cells toward renal lineages by conditioned medium from ureteric bud cells in vitro. Acta Biochim Biophys Sin (Shanghai) 2010; 42:464-71. [PMID: 20705585 DOI: 10.1093/abbs/gmq046] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The kidney is formed from two tissue populations derived from the intermediate mesoderm, the ureteric bud, and the metanephric mesenchyme. Metanephric mesenchyme is a pluripotent renal stem population, and conversion of renal mesenchyme into epithelia depends on the ureteric bud in vivo and in vitro. Embryonic stem (ES) cells have been induced to differentiate into a broad spectrum of specialized cell types in vitro, including hematopoietic, pancreatic, and neuronal cells. Such ES-derived cells can provide a valuable source of progenitor cells. However, whether ES cells can be stimulated by factors secreted from the fetal renal cells to differentiate into renal precursor cells in vitro has not been reported. In this study, we showed that murine ES cells can give rise to embryoid bodies in the absence of leukemia inhibitory factor. Culture conditions were optimized [6 days, 10 ng/ml activin and 10(-7) M retinoic acid (RA)] to generate maximal mesoderm populations specifically expressing Pax2 and brachyury. Results showed that 72% of the cells were brachyury positive by fluorescent activated cell sorter on Day 6 of EB cell differentiation. Conditioned medium collected from cultures of ureteric bud cells from renal cells of a 13-day-old fetus was added to the culture medium. Mesoderm cells were cultured for up to 10 days before showing expression of renal markers, initiation of nephrogenesis (WT-1 and Pax2), and terminally differentiated renal cell types (POD-1 and E-cadherin). This study suggests that ES cells pre-treated by RA and activin can interact with secreted molecules of the fetal renal cells to specifically differentiate into renal precursor cells. Our results provide an experimental basis for the development of in vitro assays to steer differentiation of ES cells toward renal lineages.
Collapse
Affiliation(s)
- Xiaohui Ren
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Sato Y, Uemura T, Morimitsu K, Sato-Nishiuchi R, Manabe RI, Takagi J, Yamada M, Sekiguchi K. Molecular basis of the recognition of nephronectin by integrin alpha8beta1. J Biol Chem 2009; 284:14524-36. [PMID: 19342381 DOI: 10.1074/jbc.m900200200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Integrin alpha8beta1 interacts with a variety of Arg-Gly-Asp (RGD)-containing ligands in the extracellular matrix. Here, we examined the binding activities of alpha8beta1 integrin toward a panel of RGD-containing ligands. Integrin alpha8beta1 bound specifically to nephronectin with an apparent dissociation constant of 0.28 +/- 0.01 nm, but showed only marginal affinities for fibronectin and other RGD-containing ligands. The high-affinity binding to alpha8beta1 integrin was fully reproduced with a recombinant nephronectin fragment derived from the RGD-containing central "linker" segment. A series of deletion mutants of the recombinant fragment identified the LFEIFEIER sequence on the C-terminal side of the RGD motif as an auxiliary site required for high-affinity binding to alpha8beta1 integrin. Alanine scanning mutagenesis within the LFEIFEIER sequence defined the EIE sequence as a critical motif ensuring the high-affinity integrin-ligand interaction. Although a synthetic LFEIFEIER peptide failed to inhibit the binding of alpha8beta1 integrin to nephronectin, a longer peptide containing both the RGD motif and the LFEIFEIER sequence was strongly inhibitory, and was approximately 2,000-fold more potent than a peptide containing only the RGD motif. Furthermore, trans-complementation assays using recombinant fragments containing either the RGD motif or LFEIFEIER sequence revealed a clear synergism in the binding to alpha8beta1 integrin. Taken together, these results indicate that the specific high-affinity binding of nephronectin to alpha8beta1 integrin is achieved by bipartite interaction of the integrin with the RGD motif and LFEIFEIER sequence, with the latter serving as a synergy site that greatly potentiates the RGD-driven integrin-ligand interaction but has only marginal activity to secure the interaction by itself.
Collapse
Affiliation(s)
- Yuya Sato
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Abrahamson DR. Development of kidney glomerular endothelial cells and their role in basement membrane assembly. Organogenesis 2009; 5:275-87. [PMID: 19568349 PMCID: PMC2659369 DOI: 10.4161/org.7577] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2008] [Accepted: 12/03/2008] [Indexed: 01/07/2023] Open
Abstract
Data showing that the embryonic day 12 (E12) mouse kidney contains its own pool of endothelial progenitor cells is presented. Mechanisms that regulate metanephric endothelial recruitment and differentiation, including the hypoxia-inducible transcription factors and vascular endothelial growth factor/vascular endothelial growth factor receptor signaling system, are also discussed. Finally, evidence that glomerular endothelial cells contribute importantly to assembly of the glomerular basement membrane (GBM), especially the laminin component, is reviewed. Together, this forum offers insights on blood vessel development in general, and formation of the glomerular capillary in particular, which inarguably is among the most unique vascular structures in the body.
Collapse
Affiliation(s)
- Dale R Abrahamson
- Department of Anatomy and Cell Biology; University of Kansas Medical Center; Kansas City, Kansas USA
| |
Collapse
|
27
|
Pitera JE, Scambler PJ, Woolf AS. Fras1, a basement membrane-associated protein mutated in Fraser syndrome, mediates both the initiation of the mammalian kidney and the integrity of renal glomeruli. Hum Mol Genet 2008; 17:3953-64. [PMID: 18787044 PMCID: PMC2638576 DOI: 10.1093/hmg/ddn297] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
FRAS1 is mutated in some individuals with Fraser syndrome (FS) and the encoded protein is expressed in embryonic epidermal cells, localizing in their basement membrane (BM). Syndactyly and cryptophthalmos in FS are sequelae of skin fragility but the bases for associated kidney malformations are unclear. We demonstrate that Fras1 is expressed in the branching ureteric bud (UB), and that renal agenesis occurs in homozygous Fras1 null mutant blebbed (bl) mice on a C57BL6J background. In vivo, the bl/bl bud fails to invade metanephric mesenchyme which undergoes involution, events replicated in organ culture. The expression of glial cell line-derived neurotrophic factor and growth-differentiation factor 11 was defective in bl/bl renal primordia in vivo, whereas, in culture, the addition of either growth factor restored bud invasion into the mesenchyme. Mutant primordia also showed deficient expression of Hoxd11 and Six2 transcription factors, whereas the activity of bone morphogenetic protein 4, an anti-branching molecule, was upregulated. In wild types, Fras1 was also expressed by nascent nephrons. Foetal glomerular podocytes expressed Fras1 transcripts and Fras1 immunolocalized in a glomerular BM-like pattern. On a mixed background, bl mutants, and also compound mutants for bl and my, another bleb strain, sometimes survive into adulthood. These mice have two kidneys, which contain subsets of glomeruli with perturbed nephrin, podocin, integrin α3 and fibronectin expression. Thus, Fras1 protein coats branching UB epithelia and is strikingly upregulated in the nephron lineage after mesenchymal/epithelial transition. Fras1 deficiency causes defective interactions between the bud and mesenchyme, correlating with disturbed expression of key nephrogenic molecules. Furthermore, Fras1 may also be required for the formation of normal glomeruli.
Collapse
Affiliation(s)
- Jolanta E Pitera
- Nephro-Urology Unit, UCL Institute of Child Health, London WC1 N 1EH, UK
| | | | | |
Collapse
|
28
|
Rahman A, Olivius P, Dirckx J, Von Unge M, Hultcrantz M. Stem cells and enhanced healing of chronic tympanic membrane perforation. Acta Otolaryngol 2008; 128:352-9. [PMID: 18368564 DOI: 10.1080/00016480701762508] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
CONCLUSIONS Important information about the basic reparative process of tympanic membrane (TM) healing is shown, which can be incorporated for further clinical understanding. This provides a basis for the exploration of stem cell treatment for TM perforations and holds promise for future improvements. OBJECTIVES This study aimed to analyse the healing of TM perforation by using stem cells and the stiffness of the membrane was tested in an acute and long-term study. MATERIALS AND METHODS Sprague-Dawley rats were used in a model of TM perforation. The perforation was performed with a laser system. Stem cells were applied and the healing time and morphological analysis were performed with light and transmission electron microscope. Stiffness was examined by moiré interferometry. RESULTS The stiffness of the perforated and healed TM was restored after just 2 weeks. In the chronic perforation model, mesenchymal stem cells enhanced the healing.
Collapse
|
29
|
A genome search for primary vesicoureteral reflux shows further evidence for genetic heterogeneity. Pediatr Nephrol 2008; 23:587-95. [PMID: 18197425 PMCID: PMC2259258 DOI: 10.1007/s00467-007-0675-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2007] [Revised: 09/24/2007] [Accepted: 10/16/2007] [Indexed: 11/28/2022]
Abstract
Vesicoureteral reflux (VUR) is the most common disease of the urinary tract in children. In order to identify gene(s) involved in this complex disorder, we performed a genome-wide search in a selected sample of 31 patients with primary VUR from eight families originating from southern Italy. Sixteen additional families with 41 patients were included in a second stage. Nonparametric, affected-only linkage analysis identified four genomic areas on chromosomes 1, 3, and 4 (p < 0.05); the best result corresponded to the D3S3681-D3S1569 interval on chromosome 3 (nonparametric linkage score, NPL = 2.75, p = 0.008). This region was then saturated with 26 additional markers, tested in the complete group of 72 patients from 24 families (NPL = 2.01, p = 0.01). We identified a genomic area on 3q22.2-23, where 26 patients from six multiplex families shared overlapping haplotypes. However, we did not find evidence for a common ancestral haplotype. The region on chromosome 1 was delimited to 1p36.2-34.3 (D1S228-D1S255, max. NPL = 1.70, p = 0.03), after additional fine typing. Furthermore, on chromosome 22q11.22-12.3, patients from a single family showed excess allele sharing (NPL = 3.35, p = 0.015). Only the chromosome 3q region has been previously reported in the single genome-wide screening available for primary VUR. Our results suggest the presence of several novel loci for primary VUR, giving further evidence for the genetic heterogeneity of this disorder.
Collapse
|
30
|
Abstract
Mohiuddin discusses the lessons learned from large animal xenograft models and why the immunological barrier is still the most important hurdle preventing clinical xenotransplantation of organs.
Collapse
Affiliation(s)
- Muhammad M Mohiuddin
- Cardiothoracic Surgery Research Program, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America.
| |
Collapse
|
31
|
Abstract
Interest in xenotransplantation has increased because conventional organ transplantation has been limited by a shortage of human organs. Although xenotransplantation could alleviate the existing and anticipated need for tissues and organs, the application is hindered by various biologic obstacles. This article reviews the basis for the demand for xenotransplantation, the obstacles to clinical application, and potential approaches to overcoming those obstacles.
Collapse
Affiliation(s)
- Zain Khalpey
- Transplantation Biology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
32
|
Menon S, Kakkar N, Radotra BD. Expression of laminin and fibronectin in renal dysplasia. Pediatr Dev Pathol 2004; 7:568-76. [PMID: 15630524 DOI: 10.1007/s10024-003-5057-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2003] [Accepted: 06/17/2004] [Indexed: 11/26/2022]
Abstract
The pathogenesis of renal dysplasia is a matter of debate. Recent theories have conceptualized the role of extracellular matrix proteins in the genesis of renal dysplasia. During normal nephrogenesis, collagen type I and III and fibronectins are lost and laminin and syndecan appear once proper induction has occurred. Any deviation from the normal pattern is said to lead to dysplasia. In this study, the expressions of adhesive glycoproteins, laminin, and fibronectin were studied immunohistochemically in 25 autopsy cases of renal dysplasia and normal age-matched control cases. These cases of renal dysplasia were categorized into 3 groups based on the period of gestation: 20 to 26 weeks, 27 to 33 weeks, and 34 to 40 weeks. The immunohistochemical findings were graded from 0 to 4+ based on the visual intensity. Chi-square analysis was used to calculate the difference in expressions of laminin and fibronectin in cases and controls as a whole and within and between age groups. Immunostaining for laminin in all age groups showed a significant difference in expression between dysplastic kidneys (less expression) and normal controls (greater expression). In the case of fibronectin expression, all but 1 group showed a significant difference, with dysplastic kidneys showing more and normal controls showing less expression. The inference derived is that laminin expression decreases and fibronectin expression increases in renal dysplasia compared with normal nephrogenesis.
Collapse
Affiliation(s)
- Santosh Menon
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Sector 12, Chandigarh, India, 160012.
| | | | | |
Collapse
|
33
|
Ye P, Habib SL, Ricono JM, Kim NH, Choudhury GG, Barnes JL, Abboud HE, Arar MY. Fibronectin induces ureteric bud cells branching and cellular cord and tubule formation. Kidney Int 2004; 66:1356-64. [PMID: 15458428 DOI: 10.1111/j.1523-1755.2004.00897.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND The extracellular matrix (ECM) protein fibronectin is involved in several stages of embryogenesis. Fibronectin exerts its effect through interaction with cellular integrin and nonintegrin receptors. METHODS We investigated the effect of fibronectin on branching and tubulogenesis of ureteric bud cells in a three-dimensional gel culture system. Primary ureteric bud cells from mouse embryos at gestation 11 days (E11) were isolated and established in culture. Fibronectin and integrin subunits were localized using immunoperoxidase staining. RESULTS In three-dimensional collagen type I gel culture of ureteric bud cell, fibronectin dose dependently induces cord and tubule formation. Both ureteric bud cells and ureteric bud branches in embryonic kidney express the same multiple integrin subunits that include beta(1), beta(3), alpha(3), alpha(4) and alpha(v). Embryonic kidneys examined at E12, E14, and E16 days of gestation express fibronectin in the undifferentiated mesenchyme especially next to ureteric bud branches and in the interstitium around glomerulotubular structures and blood vessels. Fibronectin expression was similar at the tips and stalks of branching ureteric bud. Fibronectin expression is maximum at E12 and decreases with advanced gestation. Cultured ureteric bud cells also express fibronectin. RGD peptides inhibit cord and tubular formation in the three-dimensional gel. Anti-alpha(3)beta(1) antibody partially inhibits fibronectin-induced cord and tubule formation. Hepatocyte growth factor (HGF), fibroblast growth factor (FGF), and glial cell line-derived neurotrophic factor (GDNF) induce ureteric bud cell cord formation in three-dimensional gel. The effects of growth factors are delayed and quantitatively less compared to the effect of fibronectin. CONCLUSION Fibronectin induces ureteric bud cells branching and tubulogenesis through interaction with multiple integrin receptors. Cultured ureteric bud cells express fibronectin and the origin of fibronectin at mesenchyme-ureteric bud interface is likely both the metanephric mesenchyme and ureteric bud epithelium. Addition of individual neutralizing antibodies to beta(1), beta(3), alpha(3), alpha(4,)alpha(6) and alpha(v) integrin subunits does not block the effect of fibronectin. Only an antibody to alpha(3)beta(1) integrin substantially blocks the effect of fibronectin. Other mechanisms, including unidentified integrins, are likely involved in fibronectin-induced cord and tubule formation.
Collapse
Affiliation(s)
- Peng Ye
- Department of Pediatrics and Department of Internal Medicine, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Despite improvements in pharmacological therapies, the outlook for patients with severe cardiac disease remains poor. At present, only transplantation can 'cure' end-stage cardiac failure. However, fewer than 5% of those who need a cardiac transplant receive one in the United States each year. To address this problem, some propose using animals as a source of organs for transplantation, that is, xenotransplantation. Here, we discuss the rationale for xenotransplantation beyond overcoming the shortage of human organs, and we weigh xenotransplantation against other new technologies that might be used for the treatment of cardiac failure.
Collapse
Affiliation(s)
- Kiyoshi Ogata
- Transplantation Biology, Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
35
|
Chi L, Zhang S, Lin Y, Prunskaite-Hyyryläinen R, Vuolteenaho R, Itäranta P, Vainio S. Sprouty proteins regulate ureteric branching by coordinating reciprocal epithelialWnt11, mesenchymalGdnfand stromalFgf7signalling during kidney development. Development 2004; 131:3345-56. [PMID: 15201220 DOI: 10.1242/dev.01200] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The kidney is a classic model for studying mechanisms of inductive tissue interactions associated with the epithelial branching common to many embryonic organs, but the molecular mechanisms are still poorly known. Sprouty proteins antagonize tyrosine kinases in the Egf and Fgf receptors and are candidate components of inductive signalling in the kidney as well. We have addressed the function of sprouty proteins in vivo by targeted expression of human sprouty 2 (SPRY2) in the ureteric bud, which normally expresses inductive signals and mouse sprouty 2 (Spry2). Ectopic SPRY2 expression led to postnatal death resulting from kidney failure, manifested as unilateral agenesis, lobularization of the organ or reduction in organ size because of inhibition of ureteric branching. The experimentally induced dysmorphology associated with deregulated expression of Wnt11, Gdnf and Fgf7 genes in the early stages of organogenesis indicated a crucial role for sprouty function in coordination of epithelial-mesenchymal and stromal signalling, the sites of expression of these genes. Moreover, Fgf7 induced Spry2 gene expression in vitro and led with Gdnf to a partial rescue of the SPRY2-mediated defect in ureteric branching. Remarkably, it also led to supernumerary epithelial bud formation from the Wolffian duct. Together, these data suggest that Spry genes contribute to reciprocal epithelial-mesenchymal and stromal signalling controlling ureteric branching, which involves the coordination of Ffg/Wnt11/Gdnf pathways.
Collapse
Affiliation(s)
- Lijun Chi
- Biocenter Oulu and Department of Biochemistry, Faculties of Science and Medicine, University of Oulu, PO Box 3000, FIN-90014 Oulu, Finland
| | | | | | | | | | | | | |
Collapse
|
36
|
Ogata K, Platt JL. Potential applications and prospects for cardiac xenotransplantation. J Heart Lung Transplant 2004; 23:515-26. [PMID: 15135365 DOI: 10.1016/j.healun.2003.07.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2003] [Revised: 07/21/2003] [Accepted: 07/26/2003] [Indexed: 11/25/2022] Open
Abstract
Despite improvements in pharmacologic therapies, the outlook for patients with severe cardiac disease remains poor. At present, the only "cure" for end-stage heart failure is transplantation. However, fewer than 5% of those who need a cardiac transplant receive one in the United States each year. As an alternative, some propose using animals as a source of organs for transplantation (i.e., xenotransplantation). In this article we review the potential applications of xenotransplantation for the treatment of cardiac disease, and weigh xenotransplantation against other new technologies that might be used. We also consider the current status of addressing the hurdles to application of xenotransplantation.
Collapse
Affiliation(s)
- Kiyoshi Ogata
- Transplantation Biology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
37
|
Discenza MT, Vaz D, Hassell JA, Pelletier J. Activation of the WT1 tumor suppressor gene promoter by Pea3. FEBS Lett 2004; 560:183-91. [PMID: 14988020 DOI: 10.1016/s0014-5793(04)00104-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2003] [Revised: 01/14/2004] [Accepted: 01/21/2004] [Indexed: 01/08/2023]
Abstract
Gene array profiling of RNA from cells engineered to express a dominant-negative version of the ETS family member transcription factor Pea3 (polyomavirus enhancer activator 3) identified WT1 as a candidate downstream gene. Given the co-expression of WT1 and Pea3 in developing kidney and breast tissue undergoing mesenchymal to epithelial transitions, we further characterized this potential gene hierarchy. Analysis of the human WT1 promoter revealed several potential binding sites for Pea3. Pea3 transactivated the WT1 promoter in transient transfection assays and bound to specific sites within the WT1 promoter in vitro. Our results position Pea3 upstream of WT1 and define a gene hierarchy important for mesenchymal-epithelial transitions.
Collapse
Affiliation(s)
- Maria Teresa Discenza
- Department of Biochemistry, McIntyre Medical Sciences Building, McGill University, Montreal, QC, Canada H3G 1Y6
| | | | | | | |
Collapse
|
38
|
Kanwar YS, Wada J, Lin S, Danesh FR, Chugh SS, Yang Q, Banerjee T, Lomasney JW. Update of extracellular matrix, its receptors, and cell adhesion molecules in mammalian nephrogenesis. Am J Physiol Renal Physiol 2004; 286:F202-15. [PMID: 14707006 DOI: 10.1152/ajprenal.00157.2003] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
One of the hallmarks of mammalian nephrogenesis includes a mesenchymal-epithelial transition that is accomplished by intercalation of the ureteric bud, an epithelium-lined tubelike structure, into an undifferentiated mesenchyme, and the latter then undergoes an inductive transformation and differentiates into an epithelial phenotype. At the same time, the differentiating mesenchyme reciprocates by inducing branching morphogenesis of the ureteric bud, which forms a treelike structure with dichotomous iterations. These reciprocal inductive interactions lead to the development of a functioning nephron unit made up of a glomerulus and proximal and distal tubules. The inductive interactions and differentiation events are modulated by a number of transcription factors, protooncogenes, and growth factors and their receptors, which regulate the expression of target morphogenetic modulators including the ECM, integrin receptors, and cell adhesion molecules. These target macromolecules exhibit spatiotemporal and stage-specific developmental regulation in the metanephros. The ECM molecules expressed at the epithelial-mesenchymal interface are perhaps the most relevant and conducive to the paracrine-juxtacrine interactions in a scenario where the ligand is expressed in the mesenchyme while the receptor is located in the ureteric bud epithelium or vice versa. In addition, expression of the target ECM macromolecules is regulated by matrix metalloproteinases and their inhibitors to generate a concentration gradient at the interface to further propel epithelial-mesenchymal interactions so that nephrogenesis can proceed seamlessly. In this review, we discuss and update our current understanding of the role of the ECM and related macromolecules with respect to metanephric development.
Collapse
Affiliation(s)
- Yashpal S Kanwar
- Department of Pathology, Northwestern Univ. Medical School, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Mascarenhas JB, Rüegg MA, Winzen U, Halfter W, Engel J, Stetefeld J. Mapping of the laminin-binding site of the N-terminal agrin domain (NtA). EMBO J 2003; 22:529-36. [PMID: 12554653 PMCID: PMC140726 DOI: 10.1093/emboj/cdg041] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Agrin is a key organizer of acetylcholine receptor (AChR) clustering at the neuromuscular junction. The binding of agrin to laminin is required for its localization to synaptic basal lamina and other basement membranes. The high-affinity interaction with the coiled-coil domain of laminin is mediated by the N-terminal domain of agrin. We have adopted a structurally guided site-directed mutagenesis approach to map the laminin-binding site of NtA. Mutations of L117 and V124 in the C-terminal helix 3 showed that they are crucial for binding. Both residues are located in helix 3 and face the groove between the beta-barrel and the C-terminal helical segment of NtA. Remarkably, the distance between both residues matches a heptad repeat distance of two aliphatic residues which are solvent exposed in the coiled-coil domain of laminin. A lower but significant contribution originates from R43 and a charged cluster (E23, E24 and R40) at the open face of the beta-barrel structure. We propose that surface-exposed, conserved residues of the laminin gamma1 chain interact with NtA via hydrophobic and ionic interactions.
Collapse
Affiliation(s)
| | - Markus A. Rüegg
- Departments of Biophysical Chemistry and
Neurobiology/Pharmacology, Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland and Department of Neurobiology, University of Pittsburgh, Anatomy and Cell Science, Pittsburgh, PA, USA Corresponding author e-mail:
| | - Uwe Winzen
- Departments of Biophysical Chemistry and
Neurobiology/Pharmacology, Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland and Department of Neurobiology, University of Pittsburgh, Anatomy and Cell Science, Pittsburgh, PA, USA Corresponding author e-mail:
| | - Willi Halfter
- Departments of Biophysical Chemistry and
Neurobiology/Pharmacology, Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland and Department of Neurobiology, University of Pittsburgh, Anatomy and Cell Science, Pittsburgh, PA, USA Corresponding author e-mail:
| | | | - Jörg Stetefeld
- Departments of Biophysical Chemistry and
Neurobiology/Pharmacology, Biozentrum, University of Basel, Klingelbergstrasse 70, CH-4056 Basel, Switzerland and Department of Neurobiology, University of Pittsburgh, Anatomy and Cell Science, Pittsburgh, PA, USA Corresponding author e-mail:
| |
Collapse
|
40
|
Abstract
For nearly a century, xenotransplantation has been seen as a potential approach to replacing organs and tissues damaged by disease. Until recently, however, the application of xenotransplantation has seemed only a remote possibility. What has changed this perspective is the advent of genetic engineering of large animals; that is, the ability to add genes to and remove genes from lines of animals that could provide an enduring source of tissues and organs for clinical application. Genetic engineering could address the immunologic, physiologic and infectious barriers to xenotransplantation, and could allow xenotransplantation to provide a source of cells with defined and even controlled expression of exogenous genes. This communication will consider one perspective on the application of genetic engineering in xenotransplantation.
Collapse
Affiliation(s)
- J L Platt
- Transplantation Biology, Department of Surgery, Medical Sciences Building 2-66, Mayo Clinic, 200 1st Street S.W., Rochester, MN 55905, USA.
| |
Collapse
|
41
|
Leimeister C, Steidl C, Schumacher N, Erhard S, Gessler M. Developmental expression and biochemical characterization of Emu family members. Dev Biol 2002; 249:204-18. [PMID: 12221002 DOI: 10.1006/dbio.2002.0764] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Kidney development has often served as a model for epithelial-mesenchymal cell interaction where the branching epithelium of the ureteric bud induces the metanephrogenic mesenchyme to form epithelial nephrons. In a screen for genes differentially expressed during kidney development, we have identified a novel gene that is dynamically expressed in the branching ureter and the developing nephrons. It was designated Emu1 since it shares an N-terminal cysteine-rich domain with Emilin1/2 and Multimerin. This highly conserved EMI domain is also found in another novel protein (Emu2) of similar protein structure: an N-terminal signal peptide followed by the EMI domain, an interrupted collagen stretch, and a conserved C-terminal domain of unknown function. We identified two further secreted EMI domain proteins, prompting us to compare their gene and protein structures, the EMI domain phylogeny, as well as the embryonic expression pattern of known (Emilin1/2, Multimerin) and novel (Emu1/2, Emilin3, Multimerin2) Emu gene family members. Emu1 and Emu2 not only show a similar structural organization, but furthermore a striking complementary expression in organs developing through epithelial-mesenchymal interactions. In these tissues, Emu1 is restricted to epithelial and Emu2 to mesenchymal cells. Preliminary biochemical analysis of Emu1/2 confirmed that they are secreted glycoproteins which are attached to the extracellular matrix and capable of forming homo- and heteromers via disulfide bonding. The widespread, but individually distinct expression patterns of all Emu gene family members suggest multiple functions during mouse embryogenesis. Their multidomain protein structure may indicate that Emu proteins interact with several different extracellular matrix components and serve to connect and integrate the function of multiple partner molecules.
Collapse
Affiliation(s)
- Cornelia Leimeister
- Theodor-Boveri-Institute, Physiological Chemistry I, University of Wuerzburg, 97074, Wuerzburg, Germany
| | | | | | | | | |
Collapse
|
42
|
Petäjäniemi N, Korhonen M, Kortesmaa J, Tryggvason K, Sekiguchi K, Fujiwara H, Sorokin L, Thornell LE, Wondimu Z, Assefa D, Patarroyo M, Virtanen I. Localization of laminin alpha4-chain in developing and adult human tissues. J Histochem Cytochem 2002; 50:1113-30. [PMID: 12133914 DOI: 10.1177/002215540205000813] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Recent studies suggest important functions for laminin-8 (Ln-8; alpha4beta1gamma1) in vascular and blood cell biology, but its distribution in human tissues has remained elusive. We have raised a monoclonal antibody (MAb) FC10, and by enzyme-linked immunoassay (EIA) and Western blotting techniques we show that it recognizes the human Ln alpha4-chain. Immunoreactivity for the Ln alpha4-chain was localized in tissues of mesodermal origin, such as basement membranes (BMs) of endothelia, adipocytes, and skeletal, smooth, and cardiac muscle cells. In addition, the Ln alpha4-chain was found in regions of some epithelial BMs, including epidermis, salivary glands, pancreas, esophageal and gastric glands, intestinal crypts, and some renal medullary tubules. Developmental differences in the distribution of Ln alpha4-chain were detected in skeletal muscle, walls of vessels, and intestinal crypts. Ln alpha4- and Ln alpha2-chains co-localized in BMs of fetal skeletal muscle cells and in some epithelial BMs, e.g., in gastric glands and acini of pancreas. Cultured human pulmonary artery endothelial (HPAE) cells produced Ln alpha4-chain as M(r) 180,000 and 200,000 doublet and rapidly deposited it to the growth substratum. In cell-free extracellular matrices of human kidney and lung, Ln alpha4-chain was found as M(r) 180,000 protein.
Collapse
Affiliation(s)
- Noora Petäjäniemi
- Institute of Biomedicine/Anatomy, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Malhas AN, Abuknesha RA, Price RG. Interaction of the leucine-rich repeats of polycystin-1 with extracellular matrix proteins: possible role in cell proliferation. J Am Soc Nephrol 2002; 13:19-26. [PMID: 11752017 DOI: 10.1681/asn.v13119] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Polycystin-1, the product of the PKD1 gene, is a membrane-bound multidomain protein with a unique structure and a molecular weight of approximately 460 kD. The purpose of this study is to investigate the binding of the cystein-flanked leucine-rich repeats (LRR) of polycystin-1 to extracellular matrix (ECM) components. These interactions may play a role in normal renal development as well as the pathogenesis of autosomal-dominant polycystic kidney disease (ADPKD). In vitro assays were used to assess the binding of a fusion protein containing the LRR of polycystin-1 and that of affinity purified polycystin-1 to a number of ECM components. The results showed that the LRR modulate the binding of polycystin-1 to collagen I, fibronectin, laminin, and cyst fluid-derived laminin fragments. The addition of the LRR fusion protein to cells in culture resulted in a significant dose-dependent reduction in the rate of proliferation. Cyst fluid-derived laminin fragments had a stimulatory effect on cell proliferation, which was reversed by the LRR fusion protein. These results suggest that the LRR of polycystin-1 act as mediators of the polycystin-1 interaction with the ECM. The observed suppression effect of the LRR on cell proliferation suggests a functional role of the LRR-mediated polycystin-1 involvement in cell-matrix and cell-cell interactions. These interactions may result in the enhanced cell proliferation that is a characteristic feature of ADPKD.
Collapse
Affiliation(s)
- Ashraf N Malhas
- Division of Life Sciences, King's College London, London, United Kingdom
| | | | - Robert G Price
- Division of Life Sciences, King's College London, London, United Kingdom
| |
Collapse
|
45
|
Abstract
Exciting new technologies, such as cellular transplantation, organogenesis and xenotransplantation, are thought to be promising approaches for the treatment of human disease. The feasibility of applying these technologies, however, might be limited by biological and immunological hurdles. Here, we consider whether, and how, xenotransplantation and various other technologies might be applied in future efforts to replace or supplement the function of human organs and tissues.
Collapse
Affiliation(s)
- M Cascalho
- Department of Surgery and Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
46
|
|
47
|
|
48
|
Lozanoff S, Johnston J, Ma W, Jourdan-Le Saux C. Immunohistochemical localization of Pax2 and associated proteins in the developing kidney of mice with renal hypoplasia. J Histochem Cytochem 2001; 49:1081-97. [PMID: 11511678 DOI: 10.1177/002215540104900903] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Pax2 has been identified as a key regulatory protein associated with renal developmental malformations. The purpose of this study was to determine whether Pax2 protein expression, and that of other proteins important for normal renal development, is abnormally distributed in the prenatal kidney of the Brachyrrhine (Br) mouse that displays heritable renal hypoplasia. Embryonic 3H1 +/+ and Br/Br mice were collected between E11.0 and E18.0. Routine light microscopy and immunohistochemical analysis using antibodies to Pax2, E-cadherin, fibronectin, laminin, and Type IV collagen were applied to sequential tissue sections. E-cadherin stained consistently in the renal tubules of both normal and mutant animals. Whereas the initial expression of Pax2 corresponded between normal and mutant kidneys, it became progressively limited to the nephrogenic zone in +/+ animals, while distributing erratically in the Br/Br kidney. Fibronectin was not expressed in the normal nephrogenic zone but remained abundantly distributed throughout the Br/Br kidney. Luminin and Type IV collagen staining revealed a deficiency in renal vasculature formation in Br/Br kidneys. Results suggest that initial morphological differentiation occurs normally in the Br kidney but that subsequent nephric formation is associated with abnormal distribution of Pax2 and ECM proteins. (J Histochem Cytochem 49:1081-1097, 2001)
Collapse
Affiliation(s)
- S Lozanoff
- Department of Anatomy and Reproductive Biology, University of Hawai'i School of Medicine, 1960 East-West Road, Honolulu, HI 96822, USA.
| | | | | | | |
Collapse
|
49
|
Brandenberger R, Schmidt A, Linton J, Wang D, Backus C, Denda S, Müller U, Reichardt LF. Identification and characterization of a novel extracellular matrix protein nephronectin that is associated with integrin alpha8beta1 in the embryonic kidney. J Cell Biol 2001; 154:447-58. [PMID: 11470831 PMCID: PMC2150762 DOI: 10.1083/jcb.200103069] [Citation(s) in RCA: 189] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2001] [Revised: 05/22/2001] [Accepted: 05/23/2001] [Indexed: 12/02/2022] Open
Abstract
The epithelial-mesenchymal interactions required for kidney organogenesis are disrupted in mice lacking the integrin alpha8beta1. None of this integrin's known ligands, however, appears to account for this phenotype. To identify a more relevant ligand, a soluble integrin alpha8beta1 heterodimer fused to alkaline phosphatase (AP) has been used to probe blots and cDNA libraries. In newborn mouse kidney extracts, alpha8beta1-AP detects a novel ligand of 70-90 kD. This protein, named nephronectin, is an extracellular matrix protein with five EGF-like repeats, a mucin region containing a RGD sequence, and a COOH-terminal MAM domain. Integrin alpha8beta1 and several additional RGD-binding integrins bind nephronectin. Nephronectin mRNA is expressed in the ureteric bud epithelium, whereas alpha8beta1 is expressed in the metanephric mesenchyme. Nephronectin is localized in the extracellular matrix in the same distribution as the ligand detected by alpha8beta1-AP and forms a complex with alpha8beta1 in vivo. Thus, these results strongly suggest that nephronectin is a relevant ligand mediating alpha8beta1 function in the kidney. Nephronectin is expressed at numerous sites outside the kidney, so it may also have wider roles in development. The approaches used here should be generally useful for characterizing the interactions of novel extracellular matrix proteins identified through genomic sequencing projects.
Collapse
Affiliation(s)
- R Brandenberger
- Howard Hughes Medical Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Lehtonen S, Lehtonen E. HMG-17 is an early marker of inductive interactions in the developing mouse kidney. Differentiation 2001; 67:154-63. [PMID: 11683498 DOI: 10.1046/j.1432-0436.2001.670407.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We studied the relationship between proliferation, differentiation, and the expression of high-mobility-group protein 17 (HMG-17) during metanephric kidney development. Proliferating cells were found homogenously throughout the early kidney rudiment. The expression pattern of HMG-17 mRNA, on the other hand, was distinctly uneven: In the undifferentiated mesenchyme, the cells in the cranial "tail" part of the mesenchyme showed only a weak signal, whereas a group of cells lying close to the tip of the ureteric bud showed strong HMG-17 expression. The region distinctly positive for HMG-17 is known to contain the first cells to undergo mesenchyme-to-epithelium transition. Using the transfilter model system, strong expression of HMG-17 mRNA, followed by mesenchyme-to-epithelium transition, could be induced also in the "tail" part of the mesenchyme. The upregulation of HMG-17 in the metanephrogenic mesenchyme thus results from interaction with an inductor tissue. Throughout the renal development, the HMG-17 mRNA was also abundant in those epithelial and mesenchymal cells that were undergoing active cell differentiation, and the transcript was downregulated in mature cells. HMG-17 expression thus correlated with the processes of induction and differentiation rather than with proliferation. The present results suggest that HMG-17 could have a role in the activation of the genes regulating kidney organogenesis.
Collapse
Affiliation(s)
- S Lehtonen
- Department of Pathology, Haartman Institute University of Helsinki, Finland
| | | |
Collapse
|