1
|
van Baar MJB, van Bommel EJM, Smits MM, Touw DJ, Nieuwdorp M, Ten Kate RW, Joles JA, van Raalte DH. Whole-body insulin clearance in people with type 2 diabetes and normal kidney function: Relationship with glomerular filtration rate, renal plasma flow, and insulin sensitivity. J Diabetes Complications 2022; 36:108166. [PMID: 35221224 DOI: 10.1016/j.jdiacomp.2022.108166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/10/2022] [Accepted: 02/15/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Kidney insulin clearance, proposed to be the main route of extra-hepatic insulin clearance, occurs in tubular cells following glomerular filtration and peritubular uptake, a process that may be impaired in people with type 2 diabetes (T2D) and/or impaired kidney function. Human studies that investigated kidney insulin clearance are limited by the invasive nature of the measurement. Instead, we evaluated relationships between whole-body insulin clearance, and gold-standard measured kidney function and insulin sensitivity in adults with T2D and normal kidney function. RESEARCH DESIGN AND METHODS We determined insulin, inulin/iohexol and para-aminohippuric acid (PAH) clearances during a hyperinsulinemic-euglycemic clamp to measure whole-body insulin clearance and kidney function. Insulin sensitivity was expressed by glucose infusion rate (M value). Associations between whole-body insulin clearance, kidney function and insulin sensitivity were examined using univariable and multivariable linear regressions models. RESULTS We investigated 44 predominantly male (77%) T2D adults aged 63 ± 7, with fat mass 34.5 ± 9 kg, lean body mass 63.0 ± 11.8 kg, and HbA1c 7.4 ± 0.6%. Average whole-body insulin clearance was 1188 ± 358 mL/min. Mean GFR was 110 ± 22 mL/min, mean ERPF 565 ± 141 mL/min, and M value averaged 3.9 ± 2.3 mg/min. Whole-body insulin clearance was positively correlated with lean body mass, ERPF and insulin sensitivity, but not with GFR. ERPF explained 6% of the variance when entered in a nested multivariable linear regression model op top of lean body mass (25%) and insulin sensitivity (15%). CONCLUSIONS In adults with T2D and normal kidney function, whole-body insulin clearance was predicted best by lean body mass and insulin sensitivity, and to a lesser extent by ERPF. GFR was not associated with whole-body insulin clearance. In contrast to prior understanding, this suggests that in this population kidney insulin clearance may not play such a dominant role in whole-body insulin clearance.
Collapse
Affiliation(s)
- Michaël J B van Baar
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VUMC, Amsterdam, the Netherlands.
| | - Erik J M van Bommel
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VUMC, Amsterdam, the Netherlands
| | - Mark M Smits
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VUMC, Amsterdam, the Netherlands
| | - Daan J Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands
| | - Max Nieuwdorp
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VUMC, Amsterdam, the Netherlands
| | - Reinier W Ten Kate
- Department of Internal Medicine, Spaarne Gasthuis, Haarlem, the Netherlands
| | - Jaap A Joles
- Department of Nephrology and Hypertension, University Medical Center, Utrecht, the Netherlands
| | - Daniël H van Raalte
- Diabetes Center, Department of Internal Medicine, Amsterdam University Medical Centers, Location VUMC, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Engudar G, Rodríguez-Rodríguez C, Mishra NK, Bergamo M, Amouroux G, Jensen KJ, Saatchi K, Häfeli UO. Metal-ion coordinated self-assembly of human insulin directs kinetics of insulin release as determined by preclinical SPECT/CT imaging. J Control Release 2022; 343:347-360. [PMID: 35085699 DOI: 10.1016/j.jconrel.2022.01.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/23/2021] [Accepted: 01/19/2022] [Indexed: 12/25/2022]
Abstract
Human insulin (HI) has fascinating metal-facilitated self-assembly properties that are essential for its biological function. HI has a natural Zn2+ binding site and we have previously shown that covalently attached abiotic ligands (e.g., bipyridine, terpyridine) can lead to the formation of nanosized oligomeric structures through the coordination of metal ions. Here we studied the hypothesis that metal ions can be used to directly control the pharmacokinetics of insulin after covalent attachment of an abiotic ligand that binds metal ions. We evaluated the pharmacokinetics (PK) and biodistribution of HI self-assemblies directed by metal ion coordination (i.e., Fe2+/Zn2+, Eu3+/Zn2+, Fe2+/Co3+) using preclinical SPECT/CT imaging and ex vivo gamma counting. HI was site-specifically modified with terpyridine (Tpy) at the PheB1 or LysB29 position to create conjugates that bind either Fe2+ or Eu3+, while its natural binding site (e.g., HisB10) preferentially coordinates with either Zn2+ or Co3+. HI was also functionalized with trans-cyclooctene (TCO) opposite to Tpy at PheB1 or LysB29, respectively, to allow for tetrazine-TCO coupling via a tetrazine-modified DTPA followed by 111In-radiolabeling for SPECT/CT imaging. When the 111In-B29Tpy-HI conjugate was coordinated with Fe2+/Zn2+, its retention at the injection site 6 h after injection was ~8-fold higher than the control without the metal ions, while its kidney accumulation was lower. 111In-B1Tpy-HI showed comparable retention at the injection site 6 h after injection and slightly increased retention at 24 h. However, higher kidney accumulation and residence time of degraded 111In-B1Tpy-HI was observed compared to that of 111In-B29Tpy-HI. Quantitative PK analysis based on SPECT/CT images confirmed slower distribution from the injection site of the HI-metal ion assemblies compared to control HI conjugates. Our results show that the Tpy-binding site (i.e., PheB1 or LysB29) on HI and its coordination with the added metal ions (i.e., Fe2+/Zn2+ or Fe2+/Co3+) directed the distribution half-life of HI significantly. This clearly indicates that the PK of insulin can be controlled by complexation with different metal ions.
Collapse
Affiliation(s)
- Gokce Engudar
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Cristina Rodríguez-Rodríguez
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Department of Physics and Astronomy, Faculty of Science, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
| | - Narendra Kumar Mishra
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark
| | - Marta Bergamo
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Guillaume Amouroux
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Knud J Jensen
- Department of Chemistry, University of Copenhagen, Thorvaldsensvej 40, DK-1871 Frederiksberg C, Denmark.
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada.
| | - Urs O Häfeli
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| |
Collapse
|
3
|
Tokarz VL, MacDonald PE, Klip A. The cell biology of systemic insulin function. J Cell Biol 2018; 217:2273-2289. [PMID: 29622564 PMCID: PMC6028526 DOI: 10.1083/jcb.201802095] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 12/12/2022] Open
Abstract
Insulin is the paramount anabolic hormone, promoting carbon energy deposition in the body. Its synthesis, quality control, delivery, and action are exquisitely regulated by highly orchestrated intracellular mechanisms in different organs or "stations" of its bodily journey. In this Beyond the Cell review, we focus on these five stages of the journey of insulin through the body and the captivating cell biology that underlies the interaction of insulin with each organ. We first analyze insulin's biosynthesis in and export from the β-cells of the pancreas. Next, we focus on its first pass and partial clearance in the liver with its temporality and periodicity linked to secretion. Continuing the journey, we briefly describe insulin's action on the blood vasculature and its still-debated mechanisms of exit from the capillary beds. Once in the parenchymal interstitium of muscle and adipose tissue, insulin promotes glucose uptake into myofibers and adipocytes, and we elaborate on the intricate signaling and vesicle traffic mechanisms that underlie this fundamental function. Finally, we touch upon the renal degradation of insulin to end its action. Cellular discernment of insulin's availability and action should prove critical to understanding its pivotal physiological functions and how their failure leads to diabetes.
Collapse
Affiliation(s)
- Victoria L Tokarz
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Patrick E MacDonald
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Christensen EI, Wagner CA, Kaissling B. Uriniferous tubule: structural and functional organization. Compr Physiol 2013; 2:805-61. [PMID: 23961562 DOI: 10.1002/cphy.c100073] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The uriniferous tubule is divided into the proximal tubule, the intermediate (thin) tubule, the distal tubule and the collecting duct. The present chapter is based on the chapters by Maunsbach and Christensen on the proximal tubule, and by Kaissling and Kriz on the distal tubule and collecting duct in the 1992 edition of the Handbook of Physiology, Renal Physiology. It describes the fine structure (light and electron microscopy) of the entire mammalian uriniferous tubule, mainly in rats, mice, and rabbits. The structural data are complemented by recent data on the location of the major transport- and transport-regulating proteins, revealed by morphological means(immunohistochemistry, immunofluorescence, and/or mRNA in situ hybridization). The structural differences along the uriniferous tubule strictly coincide with the distribution of the major luminal and basolateral transport proteins and receptors and both together provide the basis for the subdivision of the uriniferous tubule into functional subunits. Data on structural adaptation to defined functional changes in vivo and to genetical alterations of specified proteins involved in transepithelial transport importantly deepen our comprehension of the correlation of structure and function in the kidney, of the role of each segment or cell type in the overall renal function,and our understanding of renal pathophysiology.
Collapse
|
5
|
|
6
|
Kolman P, Pica A, Carvou N, Boyde A, Cockcroft S, Loesch A, Pizzey A, Simeoni M, Capasso G, Unwin RJ. Insulin uptake across the luminal membrane of the rat proximal tubule in vivo and in vitro. Am J Physiol Renal Physiol 2009; 296:F1227-37. [PMID: 19261743 PMCID: PMC2681358 DOI: 10.1152/ajprenal.90351.2008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
We visualized insulin uptake in vivo across the apical membrane of the rat proximal tubule (PT) by confocal microscopy; we compared it with in vitro findings in a rat PT cell line (WKPT) using fluorescence microscopy and flow cytometry. Surface tubules were observed in vivo with a 633-nm single laser-illuminated real-time video-rate confocal scanning microscope in upright configuration for optical sectioning below the renal capsule. Fields were selected containing proximal and distal tubules; Cy5-labeled insulin was injected twice (the second time after ∼140 min) into the right jugular vein, and the fluorescence signal (at 650–670 nm) was recorded. Fluorescence was detected almost immediately at the brush-border membrane (BBM) of PT cells only, moving inside cells within 30–40 min. As a measure of insulin uptake, the ratio of the fluorescence signal after the second injection to the first doubled (ratio: 2.11 ± 0.26, mean ± SE, n = 10), indicating a “priming,” or stimulating, effect of insulin on its uptake mechanism at the BBM. This effect did not occur after pretreatment with intravenous lysine (ratio: 1.03 ± 0.07, n = 6; P < 0.01). Cy2- or Cy3-labeled insulin uptake in a PT cell line in vitro was monitored by 488-nm excitation fluorescence microscopy using an inverted microscope. Insulin localized toward the apical membrane of these cells. Semiquantitative analysis of insulin uptake by flow cytometry also demonstrated a priming effect (upregulation) on insulin internalization in the presence of increasing amounts of insulin, as was observed in vivo; moreover, this effect was not seen with, or affected by, the similarly endocytosed ligand β2-glycoprotein.
Collapse
Affiliation(s)
- Pavel Kolman
- Institute of Physical Engineering, Brno University of Technology, Brno, Czech Republic
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Ruiz OS, Qiu YY, Cardoso LR, Arruda JA. Regulation of the renal Na-HCO3 cotransporter: IX. Modulation by insulin, epidermal growth factor and carbachol. REGULATORY PEPTIDES 1998; 77:155-61. [PMID: 9809810 DOI: 10.1016/s0167-0115(98)00115-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
To examine the role of tyrosine kinase (TK) on basolateral membrane (BLM) transport, we looked for the presence of TK activity in these membranes and showed that the synthetic substrate for TK, poly [Glu80 Na, Tyr20] caused a three-fold increase in tyrosine phosphorylation. This effect was completely blocked by the TK inhibitors, 2-hydroxy-5(2,5-dihydroxybenzyl) aminobenzoic acid (HAC), 1 microM, and methyl 2,5-dihydroxycinnamate (DHC), 5 microM. We then examined the effect of agents that cause TK stimulation on tyrosine kinase immunocontent and on the Na-HCO3 cotransporter activity in BLM and in primary cultures of the proximal tubule. We utilized the cholinergic agent, carbachol (10(-4) M), epidermal growth factor (EGF 10(-8) M), and insulin (10(-8) M), well known activators of TK. Carbachol, insulin, and EGF caused a significant increase in TK immunoreactive protein content which was blocked by HAC and DHC. In BLM, carbachol significantly stimulated HCO3-dependent 22Na uptake and this effect was totally prevented by the monoclonal antibody against TK. In cultured proximal tubule cells, carbachol, EGF and insulin at physiologic concentration caused a significant stimulation of the cotransporter activity and this effect was completely blocked by the TK inhibitor, HAC. Increasing the dose of insulin 100-fold did not cause further stimulation of the cotransporter indicating that insulin plays a permissive role on the cotransporter. These results demonstrate the presence of TK in renal proximal tubule cells and show that activation of this kinase by dissimilar agents enhance the activity of the Na-HCO3 cotransporter.
Collapse
Affiliation(s)
- O S Ruiz
- Section of Nephrology, University of Illinois at Chicago, 60612-7315, USA
| | | | | | | |
Collapse
|
8
|
Sharma K, Considine RV, Michael B, Dunn SR, Weisberg LS, Kurnik BR, Kurnik PB, O'Connor J, Sinha M, Caro JF. Plasma leptin is partly cleared by the kidney and is elevated in hemodialysis patients. Kidney Int 1997; 51:1980-5. [PMID: 9186891 DOI: 10.1038/ki.1997.269] [Citation(s) in RCA: 184] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Leptin, the gene product of the ob gene, is important in the control of appetite in rodents and may have an important role in humans. The clearance of leptin from the circulation is unknown. As the leptin receptor is present in the kidney, we evaluated the role of the kidney in removing circulating leptin in humans. We measured leptin in aortic and renal vein plasma in 8 patients with intact renal function and 6 patients with impaired renal function who were undergoing elective cardiac catheterization. Renal blood flow was measured in all patients to calculate net mass balance across the kidney. In patients with intact renal function there is net renal uptake of 12% of circulating leptin, whereas in patients with renal insufficiency there is no renal uptake of leptin. In a separate cohort of 36 patients with end-stage renal failure on hemodialysis, peripheral leptin levels factored for body mass index was increased by > fourfold as compared to a group of healthy controls (N = 338). In addition, plasma leptin is not cleared by hemodialysis with a modified cellulose membrane. Additional studies are required to evaluate the role of leptin in mediating the anorexia of uremia.
Collapse
Affiliation(s)
- K Sharma
- Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Verbeke P, Perichon M, Schaeverbeke J, Bakala H. Effect of glycation of albumin on its binding to renal brush-border membrane vesicles: influence of aging in rats. BIOCHIMICA ET BIOPHYSICA ACTA 1996; 1282:93-100. [PMID: 8679665 DOI: 10.1016/0005-2736(96)00043-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Aging is associated with the loss of preferential urinary excretion of Amadori-product glycated albumin. We have measured the binding of 125I-labeled glycated albumin to the renal brush-border membrane vesicles from young and old rats to determine whether a specific receptor-mediated endocytosis system may be involved. 125I-Glycated albumin was specifically bound by renal brush-border membrane vesicles in a time- and temperature-dependent manner; the binding was concentration-dependent, saturable and reversible. Scatchard plots gave an apparent dissociation constant Km of 488 +/- 17 nM, and a number of binding sites N of 33.5 +/- 3.4 pmol/mg protein/min in membrane vesicles from young (3 months old) rats; the binding of native [125I]albumin, gave a Km of 1194 +/- 200 nM (P < 2%) and N of 82.4 +/- 16.3 pmol/mg protein/min (P < 3%). Vesicles from 10-month-old rats had a similar Km (619.6 +/- 135.3 nM) and N (21.91 +/- 2.98 pmol/mg protein/min), while those from older (30 months old) rats had significantly increased Km (1344 +/- 237 nM, P < 3%) and N (81.3 +/- 10.9 pmol/mg protein/min, P < 1%) for 125I-glycated albumin binding. 125I-Glycated HSA was not displaced by unlabeled native HSA in less than 100-fold excess and native [125I]HSA was only displaced by a 10-fold excess of unlabeled glycated HSA. The binding of native [125I]HSA was partly inhibited (85%) by unlabeled glycated HSA. Thus, there appear to be two different binding sites, one for glycated and the other for native albumin, lying close together; and the glycation site on albumin is the discriminatory recognition factor.
Collapse
Affiliation(s)
- P Verbeke
- Laboratoire de Biologie Cellulaire, Université Paris VII, France
| | | | | | | |
Collapse
|
10
|
|
11
|
Tremblay L, Gingras D, Boivin D, Béliveau R. Tyrosine protein kinase activity in renal brush-border membranes. BIOCHIMICA ET BIOPHYSICA ACTA 1992; 1108:183-9. [PMID: 1637843 DOI: 10.1016/0005-2736(92)90024-g] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Tyrosine protein kinase (TPK) activity was detected in rat renal brush-border membranes (BBM) using poly(Glu80Na,Tyr20) as a substrate. Maximal TPK activity required prior detergent dispersion of the membranes with 0.05% Triton X-100 and the presence of vanadate, a potent inhibitor of phosphotyrosine protein phosphatases, in the phosphorylation medium. Optimal conditions for measurement of TPK activity were 10 mM of MgCl2 and MnCl2, at 30 degrees C and pH 7.0. TPK activity was inhibited by genistein, with a IC50 value of 15 microM, while no inhibition was observed in the presence of 1-(5-isoquinolinesulfonyl)-2-methyl-piperazine dihydrochloride (H7), an inhibitor of serine-threonine kinases. TPK activity was enriched 4-fold in the BBM fraction relative to cortex homogenate. It was co-enriched with BBM enzyme markers, but not with those of the basolateral membrane (BLM). The endogenous substrates of TPK in brush-border and basolateral membranes were determined by Western blot analysis using an antiphosphotyrosine monoclonal antibody (PY20). Various phosphotyrosine-containing proteins were found in the BBM (31, 34, 46, 50, 53, 72, 90, 118 and 170 kDa) and in the BLM (37, 48, 50, 53, 72, 90, 130 and 170 kDa). Addition of exogenous insulin receptor to BBM and BLM increased the phosphorylation of most of the substrates. Solubilization of the TPK activity from BBM with 0.5% CHAPS and subsequent gel filtration on Superdex 75 yielded two peaks of tyrosine protein kinase activity with apparent molecular masses of 49 and 66 kDa. These results provide evidence for a non-receptor TPK activity associated with the renal tubular luminal membrane.
Collapse
Affiliation(s)
- L Tremblay
- Département de chimie-biochimie, Université du Québec à Montréal, Canada
| | | | | | | |
Collapse
|
12
|
Sekar MC, Yang M, Meezan E, Pillion DJ. Angiotensin II and bradykinin stimulate phosphoinositide breakdown in intact rat kidney glomeruli but not in proximal tubules: glomerular response modulated by phorbol ester. Biochem Biophys Res Commun 1990; 166:373-9. [PMID: 2154205 DOI: 10.1016/0006-291x(90)91955-r] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We have investigated the effect of angiotensin II, bradykinin, insulin and insulin-like growth factor I on phosphoinositide turnover in intact rat glomeruli and tubules. Angiotensin II produced a dose-dependent increase in inositol monophosphate formation with an IC50 of 10(-7)M, when added to isolated rat glomeruli. Angiotensin II-stimulated inositol phosphates formation was inhibited by the angiotensin receptor antagonist [Sar-Leu8]angiotensin II, indicating that the above response was mediated through activation of an angiotensin receptor in intact glomeruli. Besides angiotensin, in intact glomeruli, only bradykinin stimulated a phosphoinositide response, while in intact proximal tubules, none of the agonists tested produced an activation of the inositol phosphate formation. Angiotensin II- and bradykinin-stimulated inositol phosphate accumulation in intact glomeruli was inhibited by phorbol myristate acetate, an activator of protein kinase C.
Collapse
Affiliation(s)
- M C Sekar
- Department of Pharmacology, University of Alabama, Birmingham 35294
| | | | | | | |
Collapse
|
13
|
Bakala H, Perichon M, Sudey I, Schaeverbeke J. Binding of 125I-labelled albumin by isolated rat renal brush-border membrane vesicles. Evidence for uptake and internalization process. THE INTERNATIONAL JOURNAL OF BIOCHEMISTRY 1990; 22:1189-94. [PMID: 2289625 DOI: 10.1016/0020-711x(90)90120-r] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
1. In the kidney, filtered proteins are rapidly reabsorbed by the proximal tubule via adsorptive endocytosis. This process starts with the protein binding to the luminal brush-border membrane. 2. The binding of 125I-labelled albumin to rat renal brush-border membrane vesicles and the effect of a low molecular weight protein lysozyme on that binding was assessed by the filtration method. 3. The Scatchard plot revealed a one-component binding-type curve with a dissociation constant Kd of 430.9 nM and 39.6 pmol/mg membrane protein for the number of binding sites. 4. Albumin binding was saturable and reversible, time and temperature dependent and the initial rate enhanced by increasing amounts of lysozyme. 5. The fact that association of albumin with the brush-border membrane vesicles was dependent upon the intravesicular space suggested a double process, binding of the ligand to the membrane surface and its internalization. These data suggest that albumin has a different binding site than that of a low-molecular weight protein lysozyme, with a constant affinity value near physiological loads. That specificity may confer selectivity upon the endocytic uptake process.
Collapse
Affiliation(s)
- H Bakala
- Laboratoire de Biologie cellulaire, Université Paris 7, France
| | | | | | | |
Collapse
|
14
|
Nielsen S, Christensen EI. Insulin absorption in renal proximal tubules: a quantitative immunocytochemical study. JOURNAL OF ULTRASTRUCTURE AND MOLECULAR STRUCTURE RESEARCH 1989; 102:205-20. [PMID: 2699478 DOI: 10.1016/0889-1605(89)90015-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The purposes of the present study are mainly biological concerning proximal tubular handling of insulin: we will study the intracellular transport to subcellular compartments involved in insulin degradation, the specificity and saturability of the luminal endocytic absorption of insulin, the visualization of transtubular transport, and finally, if possible, the evaluation of the relative distribution (accumulation) of insulin in endocytic vacuoles and lysosomes. The second part is methodological: application of quantitative immunocytochemistry to endocytosis, quantitation of the effect of particle size and antigen density on labeling density on tissue sections, labeling at very low antigen densities, and effect of fish gelatin on background. Isolated renal proximal tubules were perfused with native insulin, 125I-insulin, or [leucineB-25]-insulin (2% receptor-binding ability and full immunoreactivity) or exposed to native insulin from the basolateral membranes. In conclusion, the luminal uptake of insulin is of low specificity, as native and [leucineB-25]-insulin were accumulated to the same extent. Endocytic uptake is of high capacity and the mechanism is saturable. Insulin accumulated in endocytic vacuoles and lysosomes, thus following the classical degradation pathway. No other subcellular compartment is associated with insulin degradation. It was not possible to detect the basolateral uptake, indicating loss of immunoreactivity after binding to its receptor. Absolute quantitative immunocytochemistry is applicable in studying endocytosis. The labeling density increases nonproportionally with antigen density probably caused by steric hindrances. Reduction of the particle size (16 to 6 nm) increased the labeling density 17.6 times.
Collapse
Affiliation(s)
- S Nielsen
- Department of Cell Biology, University of Aarhus, Denmark
| | | |
Collapse
|
15
|
Pillion DJ, Haskell JF, Atchison JA, Ganapathy V, Leibach FH. Receptors for IGF-I, but not for IGF-II, on proximal colon epithelial cell apical membranes. THE AMERICAN JOURNAL OF PHYSIOLOGY 1989; 257:E27-34. [PMID: 2546437 DOI: 10.1152/ajpendo.1989.257.1.e27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rabbit proximal colon epithelial cell apical membranes, which are known to contain receptors for insulin, were isolated by a Ca2+-precipitation technique. Binding assays with 125I-insulin-like growth factor I (IGF-I) revealed the presence of specific high-affinity binding sites, with 50% inhibition of binding observed at a concentration of 13.7 ng/ml IGF-I. In contrast, 50% inhibition of 125I-IGF-I binding was observed at an insulin concentration of 1.37 micrograms/ml, suggesting that 125I-IGF-I was not binding to insulin receptors present in this tissue. Cross-linking studies revealed an 125I-IGF-I binding subunit of relative molecular weight (Mr) of 130,000 under reducing conditions on docecyl sulfate-polyacrylamide gel electrophoresis that was similar to the IGF-I binding subunit in human placental membranes (Mr 140,000). Binding and cross-linking studies with 125I-insulin-like growth factor II (IGF-II), however, failed to reveal a specific receptor for this peptide in colon epithelial cell membranes. These results establish the coexistence of receptors for IGF-I and insulin, but not IGF-II, on rabbit proximal colon epithelial cell apical membranes and demonstrate that colon epithelial cells are capable of selective synthesis of various peptide hormone receptors.
Collapse
Affiliation(s)
- D J Pillion
- Department of Pharmacology, University of Alabama, Birmingham 35294
| | | | | | | | | |
Collapse
|