Academic Appointments


Honors & Awards


  • Glaxo-Allenbury Award for Research in Endocrinology and Metabolism, Glaxo-Allenbury Award (1969)
  • Kidney Research Award, 1974 Charlotte Roberts Trust Fund (1974)
  • Fellow, Royal College of Physicians and Surgeons, Royal College of Physicians and Surgeons, Glasgow (1982)
  • Member, American Society of Clinical Investigators, American Society of Clinical Investigators (1982)
  • Training Award, Juvenile Diabetes Foundation International (1989)
  • Fellow, American Heart Association (2003)

Professional Education


  • MB. MBCHB, University Cape Town, Medicine (1960)
  • MD, University Witwatersrand, Medicine (1975)

Current Research and Scholarly Interests


I am an Active Emeritus Professor Medicine/ Nephrology at Stanford University. I began my long career in investigative medicine over 40 years ago. My first major contribution was one of the earliest randomized clinical trials evaluating beta-blockade for angina (Rabkin R et al. The prophylactic value of propranolol in angina pectoris. Am J Cardiol. 1966;18:370-83.). Moving from clinical investigation, (e.g. The effect of renal disease on the renal uptake and excretion of insulin in man. Rabkin R et al, NEJM, 1970, 282:182-187), my research became more basic orientated and focused on the renal metabolism of peptide hormones (Factors influencing the handling of insulin by the isolated rat kidney. Rabkin, R. and A.E. Kitabchi. J. Clin. Invest. 1978, 161:169-175. The handling of immunoreactive vasopressin by the isolated perfused rat kidney. Rabkin, R et al. J. Clin. Invest. 1979. 63:6-13). More recently I have focused on the mechanisms accounting for the resistance to growth hormone, insulin-like growth factor-1 and amino acids in uremia to explain their role in the impaired body growth and muscle wasting that is common in chronic kidney disease (CKD) (Schaefer F, Chen Y, Tsao T, Nouri P, and Rabkin R. (2001). Impaired JAK-STAT signal transduction contributes to growth hormone resistance in chronic uremia. J Clin Invest 108: 467-475) Chen Y, Sood S, McIntire K, Roth R, Rabkin R. Leucine stimulated mTOR signaling is partly attenuated in skeletal muscle of chronically uremic rats especially when work overloaded. American Journal of Physiology. Endocrinology and metabolism. (2011) 301, E873-871. Intrigued by my findings in rats with CKD, that exercise can correct some of the skeletal muscle abnormalities in signal transduction and IGF-1 and myostatin gene expression, I have been motivated to go from the bench to the bedside, and have brought together an outstanding group of clinical investigators with expertise in exercise rehabilitation as well as clinical trials in CKD patients and together we are embarking on a 4 year VA Merit Review Funded study “Exercise to Prevent Muscle Mass and Functional Loss in Elderly Dialysis Patients”.

Clinical Trials


  • Exercise to Prevent Muscle Mass and Functional Loss in Elderly Dialysis Patients Recruiting

    The majority of individuals with advanced ESRD have reduced exercise capacity in part due to decreased muscle mass. This leads to a reduced ability to perform daily activities, a greater incidence of falls, and a reduced quality of life. The mechanisms responsible for the loss of muscle mass in ESRD are not understood very well. This study is designed to determine the effectiveness of an exercise program on improving muscle mass, exercise capacity and quality of life in persons with ESRD. In addition, the study will attempt to better understand why muscle loss occurs in people with ESRD, the influence exercise has on these mechanisms, and whether the response to exercise can be enhanced with nutrient supplementation.

    View full details

  • Testosterone Replacement Therapy in Advanced Chronic Kidney Disease Not Recruiting

    Muscle wasting is common in advanced chronic kidney disease (CKD) and adversely affects morbidity and mortality. In 2/3 of males with advanced CKD serum testosterone (TT) levels are reduced, and likely contributes to the wasting. As TT in relatively safe physiologic replacement doses, increases muscle mass in otherwise normal TT deficient subjects, we hypothesize that physiologic TT replacement will be effective in preventing and treating the loss of muscle mass and function in CKD patients, will improve quality of life and may reduce some cardiovascular disease (CVD) risk factors.

    Stanford is currently not accepting patients for this trial.

    View full details

Graduate and Fellowship Programs


All Publications


  • SOCS2 Silencing Improves Somatic Growth without Worsening Kidney Function in CKD. American journal of nephrology Landau, D. n., Assadi, M. H., Abu Hilal, R. n., Chen, Y. n., Rabkin, R. n., Segev, Y. n. 2020: 1–7

    Abstract

    Growth hormone (GH) resistance in CKD is partly due to increased expression of SOCS2, a GH signaling negative regulator. In SOCS2 absence, body growth is exaggerated. However, GH overexpression in mice causes glomerulosclerosis. Accordingly, we tested whether lack of SOCS2 improves body growth, but accelerates kidney damage in CKD.Eight-week-old mutant SOCS2-deficient high growth (HG) and normal wild-type mice (N) underwent 5/6 nephrectomy (CKD) or sham operation (C) and were sacrificed after 12 weeks, generating 4 groups: C-N, C-HG, CKD-N, CKD-HG.Somatic growth, inhibited in CKD-N, increased significantly in CKD-HG. Liver p-STAT5, a key intracellular signal of GH receptor (GHR) activation, was decreased in CKD-N but not in CKD-HG. Serum Cr as well as histopathological scores of renal fibrosis were similar in both CKD groups. Kidney fibrogenic (TGF-β and collagen type IV mRNA) and inflammatory precursors (IL6, STAT3, and SOCS3 mRNA) were similarly increased in C-HG, CKD-HG, and CKD-N versus C-N. Renal GHR mRNA was decreased in C-HG, CKD-HG, and CKD-N versus C-N. Kidney p-STAT5 was decreased in CKD-N but not elevated in CKD-HG.CKD-related growth retardation is overcome by SOCS2 silencing, in association with increased hepatic STAT5 phosphorylation. Renal insufficiency is not worsened by SOCS2 absence, as kidney GHR and STAT5 are not upregulated. This may be due to elevated kidney proinflammatory cytokines and their mediators, phospho-STAT3 and SOCS3, which may counteract for the absence in SOCS2 and explain the renal safety of prolonged GH therapy in CKD.

    View details for DOI 10.1159/000508224

    View details for PubMedID 32541140

  • A randomized controlled trial of exercise to prevent muscle mass and functional loss in elderly hemodialysis patients: Rationale, study design, and baseline sample. Contemporary clinical trials communications Chan, K. N., Chen, Y. n., Lit, Y. n., Massaband, P. n., Kiratli, J. n., Rabkin, R. n., Myers, J. N. 2019; 15: 100365

    Abstract

    Elderly maintenance hemodialysis (MHD) patients exhibit muscle wasting and impaired physical function. This trial determines whether MHD patients benefit from a 12-week home-based exercise program, protein supplementation, or both.and Methods: This is a randomized, blinded controlled trial involving 60 elderly MHD patients with impaired exercise capacity and function. Patients are randomized into either a homebased exercise program or normal care over a 12-week period. Measures at baseline include peak VO2, strength and body composition as well as cognitive and disease-specific questionnaires. Muscle biopsies are obtained and analyzed for protein signaling, expression of IGF-1, androgen receptors, and myostatin.At baseline, patient characteristics in the exercise and normal care groups were similar by age, gender and anthropomorphic measures. Peak VO2 was impaired (14.7 ± 3.3 ml/kg/min), representing 55 ± 14% of the age-predicted value. Six-minute walk distance was 322 ± 71 m, and the mean 1-min sit to stand test was 18 ± 8 repetitions, representing 69 ± 16% and 55 ± 22% of the age-predicted values, respectively. Indices of muscle function, including upper and lower body and hand grip strength all indicate marked impairment. Quality of life (QoL) using the SF36, the Beeson cognitive test, and KDQOL all suggest marked impairments compared to age-expected reference values for non-MHD patients.Patients undergoing MHD exhibit markedly reduced physical function and QoL. Thus, there are potentially significant gains to be made through a program of aerobic and resistance exercise. We anticipate this trial will demonstrate that home-based exercise improves cardiopulmonary function, protein signaling and QoL, and increases muscle mass, strength, and body composition.

    View details for DOI 10.1016/j.conctc.2019.100365

    View details for PubMedID 31193611

    View details for PubMedCentralID PMC6536673

  • Prehabilitation for Kidney Transplant Candidates: Is it Time? Clinical transplantation Cheng, X. S., Myers, J. N., Chertow, G. M., Rabkin, R., Chan, K. N., Chen, Y., Tan, J. C. 2017

    Abstract

    Many patients become frail with diminished cardiorespiratory fitness while awaiting kidney transplantation. Frailty and poor fitness powerfully predict mortality, transplant graft survival, and health care utilization after kidney transplantation. Efforts to intervene with post-transplant physical therapy have been met with limited success, in large part due to high study drop-out. We reviewed the literature on chronic kidney disease and exercise to propose a clinical framework for physical therapy interventions to improve fitness, scheduled for before the transplant. This framework may lead to better patient retention and compliance, and thus demonstrate better efficacy in mitigating the effects of frailty and poor fitness after kidney transplantation. This article is protected by copyright. All rights reserved.

    View details for DOI 10.1111/ctr.13020

    View details for PubMedID 28564126

  • Endurance exercise and growth hormone improve bone formation in young and growth-retarded chronic kidney disease rats. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association Troib, A., Guterman, M., Rabkin, R., Landau, D., Segev, Y. 2016; 31 (8): 1270-1279

    Abstract

    Childhood chronic kidney disease (CKD) is associated with both short stature and abnormal bone mineralization. Normal longitudinal growth depends on proper maturation of epiphyseal growth plate (EGP) chondrocytes, leading to the formation of trabecular bone in the primary ossification centre. We have recently shown that linear growth impairment in CKD is associated with impaired EGP growth hormone (GH) receptor signalling and that exercise improved insulin-like growth factor I (IGF-I) signalling in CKD-related muscle atrophy.In this study, 20-day-old rats underwent 5/6 nephrectomy (CKD) or sham surgery (C) and were exercised with treadmill, with or without GH supplementation.CKD-related growth retardation was associated with a widened EGP hypertrophic zone. This was not fully corrected by exercise (except for tibial length). Exercise in CKD improved the expression of EGP key factors of endochondral ossification such as IGF-I, vascular endothelial growth factor (VEGF), receptor activator of nuclear factor kappa-B ligand (RANKL) and osteocalcin. Combining GH treatment with treadmill exercise for 2 weeks improved the decreased trabecular bone volume in CKD, as well as the expression of growth plate runt-related transcription factor 2, RANKL, metalloproteinase 13 and VEGF, while GH treatment alone could not do that.Treadmill exercise improves tibial bone linear growth, as well as growth plate local IGF-I. When combined with GH treatment, running exercise shows beneficial effects on trabecular bone formation, suggesting the potential benefit of this combination for CKD-related short stature and bone disease.

    View details for DOI 10.1093/ndt/gfv373

    View details for PubMedID 26560811

  • Impaired renal growth hormone JAK/STAT5 signaling in chronic kidney disease. Nephrology, dialysis, transplantation : official publication of the European Dialysis and Transplant Association - European Renal Association Wiezel, D., Assadi, M. H., Landau, D., Troib, A., Kachko, L., Rabkin, R., Segev, Y. 2014; 29 (4): 791-799

    Abstract

    Treatment with recombinant human growth hormone (GH) is the standard therapy for short stature in children with chronic kidney disease (CKD). However, concerns have been raised on the potential renal fibrogenic effects of GH. There is no information regarding the renal GH receptor (GHR)-JAK-STAT signaling pathway in CKD.Subtotal nephrectomized (CKD) and pair-fed sham-operated control (C) juvenile rats were treated with subcutaneous GH or saline for 2 weeks. A single intravenous GH bolus or vehicle was provided prior to euthanasia.Reduced body weight in CKD was improved with GH therapy. The remnant kidney showed glomerular hypertrophy and early interstitial fibrosis without inflammatory infiltration. Treatment of CKD rats with GH did not worsen renal function or fibrosis. Kidney GHR mRNA and protein levels were reduced and basal phosphorylation of JAK2 and STAT5 was significantly impaired. However, intravenous GH administration prior to sacrifice normalized STAT5 phosphorylation. Basal renal IL6 mRNA and phosphorylation of its downstream signaling molecule STAT3 were increased as was the product of its action, the suppressor of cytokine signaling 3 (SOCS3) mRNA.Despite known unaltered circulating GH levels, remnant kidneys of uremic growth retarded juvenile rats show impaired basal signaling along the GH-activated JAK2/STAT5 signaling pathway. This may well be a consequence of the reduced GHR level and the inhibitory effect of the increase in IL-6-mediated SOCS3 expression. This renal GH insensitivity, if present in humans, may protect against the potential adverse renal effects of GH administration in CKD patients.

    View details for DOI 10.1093/ndt/gfu003

    View details for PubMedID 24463190

  • Acute uremia suppresses leucine-induced signal transduction in skeletal muscle. Kidney international McIntire, K. L., Chen, Y., Sood, S., Rabkin, R. 2014; 85 (2): 374-382

    Abstract

    Adequate nutrient intake in acute uremia is a key part of patient management especially as food utilization is usually impaired. Leucine is important as it comprises about one-fifth of essential amino acid needs and, apart from serving as a substrate, it directly activates the mTOR signaling pathway stimulating protein synthesis and inhibiting autophagy. Here we tested whether leucine activation of the mTOR signaling pathway in muscle is severely disrupted in acute uremia. Several abnormalities were identified in bilateral ureteral ligated (model of acute uremia) compared to sham-operated pair-fed control rats. Levels of several signaling proteins increased significantly while leucine-induced phosphorylation of mTOR and downstream proteins, 4e-BP1 and S6K1, was completely suppressed. Levels of LC3B-II, a specific autophagosomal membrane-associated protein used as a marker of autophagy, increased threefold in uremia. Furthermore, while leucine suppressed LC3B-II levels in controls, it failed to do so in uremic rats. Muscle IL-6 mRNA levels increased, while IGF-1 mRNA levels decreased in uremia. These findings establish that, in acute uremia, severe resistance to leucine-induced activation of the mTOR anabolic signaling pathway develops. Thus, leucine resistance, together with the reduction in IGF-1 and increase in IL-6 expression, may explain why the anabolic effect of nutritional therapy is diminished in acute uremic patients.

    View details for DOI 10.1038/ki.2013.216

    View details for PubMedID 23783244

  • Acute Acidosis Attenuates Leucine Stimulated Signal Transduction and Protein Synthesis in Rat Skeletal Muscle AMERICAN JOURNAL OF NEPHROLOGY Sood, S., Chen, Y., McIntire, K., Rabkin, R. 2014; 40 (4): 362-370

    View details for DOI 10.1159/000366524

    View details for Web of Science ID 000345194900008

  • Acute acidosis attenuates leucine stimulated signal transduction and protein synthesis in rat skeletal muscle. American journal of nephrology Sood, S., Chen, Y., McIntire, K., Rabkin, R. 2014; 40 (4): 362-370

    Abstract

    Critical illnesses are often complicated by acute metabolic acidosis, which if persistent, adversely affects outcome. Among the harmful effects that it might cause are impaired utilization of nutrients, increased proteolysis and depressed protein synthesis, leading to muscle wasting. As the amino acid leucine stimulates protein synthesis by activating mTOR signaling, we explored whether in acidosis, impaired leucine-stimulated signaling might be a contributor to the depressed protein synthesis.Male pair-fed rats were gavaged with NH4Cl (acidosis) or NaCl (control) for 2 days and then gavaged once with leucine and sacrificed 45 min later. Extensor digitorum longus muscles were isolated, incubated with or without leucine and protein synthesis measured. The anterior tibial muscle signaling was analysed by Western immunobloting.Despite pair-feeding, acidotic rats lost body and muscle weight vs. controls. Moreover, leucine-induced protein synthesis in isolated muscle from acidotic rats was impaired. In-vivo, 45 min after an oral leucine load, anterior tibial muscle mTOR and 4E-BP1 phosphorylation increased significantly and comparably in control and acidotic rats. In contrast, leucine-stimulated phosphorylation of S6K1, a regulator of translation initiation and protein synthesis, was attenuated to approximately 56% of the control value (p < 0.05).This study reveals that an acute metabolic acidosis impairs leucine-stimulated protein synthesis and activation of signaling downstream of mTOR at the level of S6K1. We propose that this S6K1 abnormality may account in part, for the resistance to leucine-stimulated muscle protein synthesis, and may thereby contribute to the impaired nutrient utilization and ultimately the muscle wasting that develops in acidosis.

    View details for DOI 10.1159/000366524

    View details for PubMedID 25358492

  • Epiphyseal growth plate growth hormone receptor signaling is decreased in chronic kidney disease-related growth retardation. Kidney international Troib, A., Landau, D., Kachko, L., Rabkin, R., Segev, Y. 2013; 84 (5): 940-949

    Abstract

    Linear growth retardation in children with chronic kidney disease (CKD) has been ascribed to insensitivity to growth hormone. This resistance state has been attributed to impaired growth hormone signaling through the JAK2/STAT5 pathway in liver and skeletal muscle leading to reduced insulin-like growth factor-I (IGF-I). Here we determine whether systemic and growth plate alterations in growth hormone signaling contribute to CKD-induced linear growth retardation using partially nephrectomized and pair-fed control 20-day-old rats. Serum growth hormone did not change in rats with CKD, yet serum IGF-I levels were decreased and growth retarded. The tibial growth plate hypertrophic zone was wider and vascularization at the primary ossification center was reduced in CKD. This was associated with a decrease in growth plate vascular endothelial growth factor (VEGF) mRNA and immunostainable VEGF and IGF-I levels. Growth plate growth hormone receptor and STAT5 protein levels were unchanged, while JAK2 was reduced. Despite comparable growth hormone and growth hormone receptor levels in CKD and control rats, relative STAT5 phosphorylation was significantly depressed in CKD. Of note, the mRNA of SOCS2, an inhibitor of growth hormone signaling, was increased. Thus, linear growth impairment in CKD can in part be explained by impaired long bone growth plate growth hormone receptor signaling through the JAK2/STAT5 pathway, an abnormality that may be caused by an increase in SOCS2 expression.Kidney International advance online publication, 29 May 2013; doi:10.1038/ki.2013.196.

    View details for DOI 10.1038/ki.2013.196

    View details for PubMedID 23715123

  • Leucine-stimulated mTOR signaling is partly attenuated in skeletal muscle of chronically uremic rats AMERICAN JOURNAL OF PHYSIOLOGY-ENDOCRINOLOGY AND METABOLISM Chen, Y., Sood, S., McIntire, K., Roth, R., Rabkin, R. 2011; 301 (5): E873-E881

    Abstract

    The branched-chain amino acid leucine stimulates muscle protein synthesis in part by directly activating the mTOR signaling pathway. Furthermore, leucine, if given in conjunction with resistance exercise, enhances the exercise-induced mTOR signaling and protein synthesis. Here we tested whether leucine can activate the mTOR anabolic signaling pathway in uremia and whether it can enhance work overload (WO)-induced signaling through this pathway. Chronic kidney disease (CKD) and control rats were studied after 7 days of surgically induced unilateral plantaris muscle WO and a single leucine or saline load. In the basal state, 4E-BP1 phosphorylation was modestly depressed in non-WO muscle of CKD rats, whereas rpS6 phosphorylation was nearly completely suppressed. After oral leucine mTOR, S6K1 and rpS6 phosphorylation increased similarly in both groups, whereas the phospho-4E-BP1 response was modestly attenuated in CKD. WO alone activated the mTOR signaling pathway in control and CKD rats. In WO CKD, muscle leucine augmented mTOR and 4E-BP1 phosphorylation, but its effect on S6K1 phosphorylation was attenuated. Taken together, this study has established that the chronic uremic state impairs basal signaling through the mTOR anabolic pathway, an abnormality that may contribute to muscle wasting. However, despite this abnormality, leucine can stimulate this signaling pathway in CKD, although its effectiveness is partially attenuated, including in skeletal muscle undergoing sustained WO. Thus, although there is some resistance to leucine in CKD, the data suggest a potential role for leucine-rich supplements in the management of uremic muscle wasting.

    View details for DOI 10.1152/ajpendo.00068.2011

    View details for Web of Science ID 000296585400015

    View details for PubMedID 21791619

  • The effects of type 1 IGF receptor inhibition in a mouse model of diabetic kidney disease GROWTH HORMONE & IGF RESEARCH Troib, A., Landau, D., Youngren, J. F., Kachko, L., Rabkin, R., Segev, Y. 2011; 21 (5): 285-291

    Abstract

    We have recently shown increased sensitivity to IGF-I induced signal transduction in kidneys of diabetic mice. Accordingly we investigated the effects of PQ401, a novel diarylurea compound that inhibits IGF1R autophosphorylation in type I diabetes.Control (C) and Diabetic (D) mice were administered PQ401 (CP, DP) or vehicle (C, D) for 3weeks.CP animals showed a decrease in renal phosphorylated (p-)AKT and p-IGF1R. However, PQ401 had no effect on diabetic state (hyperglycemia, weight loss) or renal disease parameters (hypertrophy, hyperfiltration and albuminuria). Type IV collagen as well as TGF-β mRNA increased in DP and D compared to C. In the CP group renal hypertrophy with fat accumulation in proximal tubuli and increased renal IGF-I, collagen IV and TGF-β mRNA were seen.IGF1R inhibition by PQ401 exerted no significant effects on diabetic kidney disease parameters, arguing against a role for IGF-I in the pathogenesis of diabetic kidney disease. However, PQ401 affects normal kidneys, inducing renal hypertrophy as well as collagen and fat accumulation, with increased renal IGF-I mRNA, suggestive of a damage-regeneration process. Therefore, this diarylurea compound is not beneficial in early diabetic kidney disease. Its potential deleterious effects on kidney tissue need to be further investigated.

    View details for DOI 10.1016/j.ghir.2011.07.007

    View details for Web of Science ID 000295444300008

    View details for PubMedID 21865067

  • A marked deficiency in circulating and renal IGF-I peptide does not inhibit compensatory renal enlargement in uninephrectomized mice GROWTH HORMONE & IGF RESEARCH Landau, D., Biada, J., Chen, Y., Sood, S., Yakar, S., LeRoith, D., Segev, Y., Rabkin, R. 2011; 21 (5): 279-284

    Abstract

    Increase in kidney IGF-I levels due to its increased trapping from the circulation was hypothesized to be a key mediator of compensatory renal enlargement. We tested this hypothesis using genetically engineered mice with extremely low circulating IGF-I levels.Both IGF-I deficient (ID) and normal (N) mice underwent a uninephrectomy (UNx) and sacrificed 2 or 9days later.Initial body weight (BW) and kidney weight (KW) were significantly reduced in ID vs. N mice, while KW/BW ratios were similar. KW increased post-UNx to a comparable extent in ID and N mice (125±4 and 118±6% of pre-UNx KW, p<0.05 vs. C). Kidney IGF-I mRNA levels were similar in the ID and N mice and did not change post-UNx. Kidney IGF-I peptide levels pre-UNx were significantly lower in ID vs. N mice (25±5 vs. 305±39ng/g) and increased in both groups after UNx, remaining low in ID mice (45±4 in ID vs 561±64ng/g in N). IGF type 1 receptor phosphorylation was unchanged.While a severe deficiency of circulating IGF-I impairs body growth, UNx induces a significant and proportional increase in renal mass in ID mice despite markedly decreased kidney IGF-I levels (>90% reduction) and no significant change in receptor phosphorylation. This all suggests that factors other than circulating and locally produced IGF-I are responsible for compensatory renal enlargement.

    View details for DOI 10.1016/j.ghir.2011.07.008

    View details for Web of Science ID 000295444300007

    View details for PubMedID 21862442

  • Uremia attenuates growth hormone-stimulated insulin-like growth factor-1 expression, a process worsened by inflammation KIDNEY INTERNATIONAL Chen, Y., Biada, J., Sood, S., Rabkin, R. 2010; 78 (1): 89-95

    Abstract

    Growth hormone (GH) resistance is common in uremia and together with resistance to insulin-like growth factor-1 (IGF-1) contributes to uremic growth retardation and muscle wasting. Previously, we found decreased GH-stimulated janus-kinase 2-signal transducers and activators of transcription 5 (STAT5) phosphorylation and nuclear translocation in uremia; however, it is unclear whether there are more distal defects. Therefore, we tested whether the binding of phosphorylated STAT5b to DNA is intact in uremia. Using uremic rats we found that in addition to impaired hepatic STAT5b phosphorylation, the binding of available phospho-STAT5b to DNA is decreased thus contributing to impaired IGF-1 gene expression. As sepsis-induced inflammation causes a loss of body protein and as Gram-negative infections are relatively common in uremia, we also characterized mechanisms in which acute inflammation might contribute to GH resistance in uremia. Endotoxin-induced inflammation markedly increased the resistance to GH-mediated STAT5b signaling, and further decreased STAT5b binding to DNA and IGF-1 gene expression. These perturbations appear to be related to increased cytokine expression. Thus, our findings indicate that hepatic resistance to GH-induced IGF-1 expression in uremia arises due to defects in STAT5b phosphorylation and its impaired binding to DNA, processes further aggravated by inflammation.

    View details for DOI 10.1038/ki.2010.85

    View details for Web of Science ID 000278729200014

    View details for PubMedID 20375991

  • Endotoxin-Induced Growth Hormone Resistance in Skeletal Muscle ENDOCRINOLOGY Chen, Y., Sood, S., Krishnamurthy, V. M., Rotwein, P., Rabkin, R. 2009; 150 (8): 3620-3626

    Abstract

    Inflammation-induced skeletal muscle wasting is a serious clinical problem and arises in part because of resistance to GH-stimulated IGF-I expression. Although it is established that in the liver, resistance develops because of impaired signaling through the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 5 (STAT5) transduction pathway, together with a more distal defect in STAT5 DNA-binding activity, the situation in skeletal muscle is unclear. Accordingly, we set out to characterize the mechanisms behind the skeletal muscle resistance to GH in rats with acute inflammation induced by endotoxin. Endotoxin caused significant declines in GH-stimulated STAT5a/b phosphorylation and IGF-I gene expression, and this occurred despite a lack of change in signaling protein levels or phosphorylation of JAK2. In whole muscle, GH-stimulated phospho-STAT5a/b levels were reduced by half, and in the nucleus, phospho-STAT5b levels were similarly reduced. Furthermore, the binding of phosphorylated STAT5b to DNA was reduced and to a similar extent to the reduction in nuclear phosphorylated STAT5b. Interestingly, GH-induced androgen receptor gene expression was also suppressed. Thus, it appears that skeletal muscle resistance to GH-stimulated IGF-I expression in acute endotoxemia arises from a defect in STAT5b signaling, with a proportionate reduction in STAT5b DNA binding. Finally, it appears that resistance to GH-induced androgen receptor expression also develops and, together with the attenuated GH-induced IGF-I expression, likely plays an important role in the muscle wasting that arises in endotoxin-induced inflammation.

    View details for DOI 10.1210/en.2008-1703

    View details for Web of Science ID 000268158400024

    View details for PubMedID 19443577

    View details for PubMedCentralID PMC2717874

  • Increased renal Akt/mTOR and MAPK signaling in type I diabetes in the absence of IGF type 1 receptor activation ENDOCRINE Landau, D., Eshet, R., Troib, A., Gurman, Y., Chen, Y., Rabkin, R., Segev, Y. 2009; 36 (1): 126-134

    Abstract

    Growth hormone (GH) and IGF-I have been implicated in the pathogenesis of type I diabetic (DM) nephropathy. We investigated renal GH receptor (GHR) and IGF-type 1 receptor (IGF1R) signaling in an animal model of type I DM. Kidney tissue was examined for GHR and IGF1R key signaling molecules. GHR levels were unchanged and IGF-I mRNA levels were decreased in the diabetic group (D). Basal and GH stimulated phosphorylated (p-) JAK2 and STAT5 levels were similar in controls (C) and D. The levels of p-IGF1R were similar in the two groups at baseline, while pAkt, pGSK3, p-mTOR, p-rpS6, p-erk1/2 (Mapk), and pSTAT-3 were increased in D. Following IGF-I administration p-Akt, p-rpS6, p-Mapk, and p-GSK levels increased more pronouncedly in D versus C. In conclusion, the lack of JAK2-STAT5 activation and the decrease in kidney IGF-I mRNA levels in D argue against a role for the GH activated JAK2-STAT5 pathway in the pathogenesis of diabetic nephropathy. On the other hand while IGF1R phosphorylation was unchanged, Akt/mTOR and MAPK signaling were hyperactivate in DM, suggesting their involvement. The increase in baseline activated Akt, mTOR, rpS6, and MAPK cannot be explained by activation of the IGF1R, but may be triggered by other growth factors and nutrients.

    View details for DOI 10.1007/s12020-009-9190-2

    View details for Web of Science ID 000267781000020

    View details for PubMedID 19387875

  • Low-dose growth hormone is cardioprotective in uremia JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Rabkin, R., Awwad, I., Chen, Y., Ashley, E. A., Sun, D., Sood, S., Clusin, W., Heidenreich, P., Piecha, G., Gross, M. 2008; 19 (9): 1774-1783

    Abstract

    Growth hormone (GH) is required to maintain normal cardiac structure and function and has a positive effect on cardiac remodeling in experimental and possibly human disease. Cardiac resistance to GH develops in the uremic state, perhaps predisposing to the characteristic cardiomyopathy associated with uremia. It was hypothesized that administration of low-dosage GH may have a salutary effect on the cardiac remodeling process in uremia, but because high levels of GH have adverse cardiac effects, administration of high-dosage GH may worsen uremic cardiomyopathy. In rats with chronic renal failure, quantitative cardiac morphology revealed a decrease in total capillary length and capillary length density and an increase in mean intercapillary distance and fibroblast volume density evident. Low-dosage GH prevented these changes. Collagen and TGF-beta immunostaining, increased in chronic renal failure, were also reduced by GH, suggesting a mechanism for its salutary action. Low-dosage GH also prevented thickening of the carotid artery but did not affect aortic pathology. In contrast, high-dosage GH worsened several of these variables. These results suggest that low-dosage GH may benefit the heart and possibly the carotid arteries in chronic renal failure.

    View details for DOI 10.1681/ASN.2007121386

    View details for Web of Science ID 000259167000021

    View details for PubMedID 18650479

    View details for PubMedCentralID PMC2518445

  • Increased workload fully activates the blunted IRS-1/PI3-kinase/Akt signaling pathway in atrophied uremic muscle KIDNEY INTERNATIONAL Chen, Y., Sood, S., Biada, J., Roth, R., Rabkin, R. 2008; 73 (7): 848-855

    Abstract

    Muscle wasting in chronic renal failure is associated with increased morbidity and mortality; however, resistance exercise is effective at increasing muscle mass while improving muscle strength and function. To study the mechanism by which this occurs, we compared uremic and control rats where work overload was surgically induced unilaterally in the plantaris muscle. We found that work overload increases muscle insulin-like growth factor-1 and mechano-growth factor expression. This in addition to direct mechanical activation of signaling was likely the cause of the observed increased in the protein levels and phosphorylation of the mediators of these growth factors, the insulin receptor substrate-1/phosphoinositide 3-kinase/Akt pathway. The mechanical enhancement of signal transduction appeared to be mediated in part by increased signal protein levels and decreased SOCS2 mRNA expression (suppressor of cytokine signaling-2). Despite impaired basal signaling, the work-induced signaling response was similar to that observed in nonuremic rats and produced changes consistent with decreased muscle protein degradation, increased protein synthetic capacity, and an increased number of multinucleated muscle cells. Our studies suggest that these work-induced changes account for the improved uremic muscle mass reaching levels comparable to those seen in normal rats.

    View details for DOI 10.1038/sj.ki.5002801

    View details for Web of Science ID 000254085100011

    View details for PubMedID 18216779

  • Growth hormone treatment prevents osteoporosis in uremic rats HISTOLOGY AND HISTOPATHOLOGY Berger, I., Piecha, G., Rabkin, R., Kaya, N., Geldyyev, A., Sun, D., Chen, Y., Koleganova, N., Gross, M. 2007; 22 (11): 1231-1239

    Abstract

    Growth hormone (GH) is responsible for longitudinal bone growth. GH-receptor in the growth plate was found to be decreased in chronic renal insufficiency. A therapeutic use of GH in chronic renal insufficiency is not established. The current study aims to clarify the effects of GH treatment on bone metabolism in a uremic rat model.Sprague Dawley rats were subjected to subtotal surgical renal ablation (SNX) or sham operation. SNX rats were randomized into 4 groups: treated with different doses of GH (1.5, 4.0, or 10.0 mg/kg) or vehicle after 10 weeks of uremia and treated for 6 weeks. Bone and renal morphology was evaluated: bone density, thickness of spongiosa, osteoblast surface, osteoid volume, osteoclast quantity, and resorptive volume.GH treatment resulted in a decrease of resorption area and lower number of osteoclasts. Osteoid volume, number of osteoblasts, percentage of active osteoblasts, thickness of the growth plate and mean cortical width increased. GH receptor (GHR) protein expression increased in GH treated rats. IGF-1 expression was decreased in osteoblasts and chondroblasts of SNX-V rats and increased following GH treatment. The TGF-beta expression was down regulated in SNX+V group in osteocytes and chondroblasts as compared to sham operated animals. The down regulation was prevented in treated animals irrespective of the dose given.Treatment with GH in uremic animals increased bone density to the levels of non-uremic controls. Thus GH seems to have a potential of preventing renal osteodystrophy.

    View details for Web of Science ID 000248748000007

    View details for PubMedID 17647196

  • Endotoxin attenuates growth hormone-induced hepatic insulin-like growth factor I expression by inhibiting JAK2/STAT5 signal transduction and STAT5b DNA binding AMERICAN JOURNAL OF PHYSIOLOGY-ENDOCRINOLOGY AND METABOLISM Chen, Y., Sun, D., Krishnamurthy, V. M., Rabkin, R. 2007; 292 (6): E1856-E1862

    Abstract

    Gram-negative sepsis with release of endotoxin is a frequent cause of cachexia that develops partly because of resistance to growth hormone (GH) with reduced insulin-like growth factor-I (IGF-I) expression. We set out to more fully characterize the mechanisms for the resistance and to determine whether in addition to a defect in the janus kinase 2 (JAK2)-signal transducer and activator of transcription (STAT) 5b pathway, required for GH-induced IGF-I expression, there might also be a more distal defect. Conscious rats were given endotoxin and studied 4 h later. In liver of these animals, GH-induced JAK2 and STAT5 phosphorylation was impaired and appeared to be caused, at least in part, by a marked increase in hepatic tumor necrosis factor-alpha and interleukin-6 mRNA expression accompanied by elevated levels of inhibitors of GH signaling, namely cytokine-inducible suppressors of cytokine signaling-1 and -3 and cytokine-inducible SH2 protein (CIS). Nuclear phosphorylated STAT5b levels were significantly depressed to 61% of the control values and represent a potential cause of the reduced GH-induced IGF-I expression. In addition, binding of phosphorylated STAT5b to DNA was reduced to an even greater extent and averaged 17% of the normal control value. This provides a further explanation for the impaired IGF-I gene transcription. Interestingly, when endotoxin-treated rats were treated with GH, there was a marked increase in proinflammatory cytokine gene expression in the liver. If such a response were to occur in humans, this might provide a partial explanation for the adverse effect of GH treatment reported in critically ill patients.

    View details for DOI 10.1152/ajpendo.00581.2006

    View details for Web of Science ID 000247939100042

    View details for PubMedID 17327369

  • Protective effect of low dose growth hormone on uremic cardiomyopathy Rabkin, R., Awwad, I., Sun, D., Chen, Y., Berger, I., Gross, M. ELSEVIER GMBH, URBAN & FISCHER VERLAG. 2007: 334–35
  • Work-induced changes in skeletal muscle IGF-1 and myostatin gene expression in uremia KIDNEY INTERNATIONAL Sun, D. F., Chen, Y., Rabkin, R. 2006; 70 (3): 453-459

    Abstract

    Resistance to growth hormone (GH)-induced insulin-like growth factor-1 (IGF-1) gene expression contributes to uremic muscle wasting. Since exercise stimulates muscle IGF-1 expression independent of GH, we tested whether work overload (WO) could increase skeletal muscle IGF-1 expression in uremia and thus bypass the defective GH action. Furthermore, to provide insight into the mechanism of uremic wasting and the response to exercise we examined myostatin expression. Unilateral plantaris muscle WO was initiated in uremic and pairfed (PF) normal rats by ablation of a gastrocnemius tendon and adjoining part of this muscle with the contralateral plantaris as a control. Some rats were GH treated for 7 days. WO led to similar gains in plantaris weight in both groups and corrected the uremic muscle atrophy. GH increased plantaris IGF-1 mRNA >twofold in PF rats but the response in uremia was severely attenuated. WO increased the IGF-1 mRNA levels significantly in both uremic and PF groups, albeit less brisk in uremia; however, after 7 days IGF-1 mRNA levels were elevated similarly, >2-fold, in both groups. In the atrophied uremic plantaris muscle basal myostatin mRNA levels were increased significantly and normalized after an increase in WO suggesting a myostatin role in the wasting process. In the hypertrophied uremic left ventricle the basal myostatin mRNA levels were reduced and likely favor the cardiac hypertrophy. Together the findings provide insight into the mechanisms of skeletal muscle wasting in uremia and the hypertrophic response to exercise, and suggest that alterations in the balance between IGF-1 and myostatin play an important role in these processes.

    View details for DOI 10.1038/sj.ki.5001532

    View details for Web of Science ID 000239390200009

    View details for PubMedID 16871256

  • Transcriptional profiling of aging in human muscle reveals a common aging signature PLOS GENETICS Zahn, J. M., Sonu, R., Vogel, H., Crane, E., Mazan-Mamczarz, K., Rabkin, R., Davis, R. W., Becker, K. G., Owen, A. B., Kim, S. K. 2006; 2 (7): 1058-1069

    Abstract

    We analyzed expression of 81 normal muscle samples from humans of varying ages, and have identified a molecular profile for aging consisting of 250 age-regulated genes. This molecular profile correlates not only with chronological age but also with a measure of physiological age. We compared the transcriptional profile of muscle aging to previous transcriptional profiles of aging in the kidney and the brain, and found a common signature for aging in these diverse human tissues. The common aging signature consists of six genetic pathways; four pathways increase expression with age (genes in the extracellular matrix, genes involved in cell growth, genes encoding factors involved in complement activation, and genes encoding components of the cytosolic ribosome), while two pathways decrease expression with age (genes involved in chloride transport and genes encoding subunits of the mitochondrial electron transport chain). We also compared transcriptional profiles of aging in humans to those of the mouse and fly, and found that the electron transport chain pathway decreases expression with age in all three organisms, suggesting that this may be a public marker for aging across species.

    View details for DOI 10.1371/journal.pgen.0020115

    View details for PubMedID 16789832

  • Circulating growth hormone binding protein levels and mononuclear cell growth hormone receptor expression in uremia 7th Symposium on Growth and Development in Children with Chronic Kidney Disease Greenstein, J., Guest, S., Tan, J. C., Tummala, P., Busque, S., Rabkin, R. W B SAUNDERS CO-ELSEVIER INC. 2006: 141–49

    Abstract

    Resistance to growth hormone (GH) in end-stage renal disease (ESRD) causes growth retardation and muscle wasting. In humans, circulating GH binding protein (GHBP), the extracellular domain of the GH receptor that is shed into the circulation and is believed to reflect tissue GH receptor levels, is reduced in uremia and suggests that cellular GH receptor levels are correspondingly reduced. If true, this could be a cause of GH resistance. We set out to establish whether serum GHBP levels reflect cellular GH receptor levels and whether changes in serum GHBP levels are related to nutritional or inflammatory status.GH receptor protein expression in peripheral blood mononuclear cells (PBMC) from 21 ESRD and 14 normal subjects were analyzed by fluorochrome flow cytometry.The GH receptor density and percent total PBMCs expressing the GH receptor were similar in the 2 groups, and there was no difference in percent GH receptor positive T or B cells or monocytes. In contrast, serum GHBP levels were 80% lower in ESRD. GHBP levels did not correlate with serum albumin, body mass index, or muscle mass but seemed to be partly related to the log serum C-reactive protein levels.Serum GHBP levels are markedly reduced in ESRD; this seems to occur independent of nutritional status and may in part be caused by inflammation. Because GH receptor expression on PBMC of ESRD and control subjects was similar, our findings argue against a reduction in GH receptor as a cause of GH resistance and the use of serum GHBP levels as a reliable marker of specific tissue GH receptor levels.

    View details for DOI 10.1053/j.jrn.2006.01.007

    View details for Web of Science ID 000236735600007

    View details for PubMedID 16567271

  • Suppressors of cytokine signaling in health and disease PEDIATRIC NEPHROLOGY Tan, J. C., Rabkin, R. 2005; 20 (5): 567-575

    Abstract

    Cytokines consist of a large family of secreted proteins, including pro-inflammatory agents, growth hormone and erythropoietin, that utilize the Janus kinase (JAK) signal transducer and activator of transcription (STAT) signal transduction pathway to mediate many of their key physiologic and pathologic actions. These actions include cytokine-mediated inflammation, immunoregulation, hematopoiesis and growth. The JAK-STAT pathway is regulated by several processes, among which negative feedback regulation by the suppressors of cytokine signaling (SOCS), members of a family of eight proteins, is particularly important. Each cytokine induces one or more specific SOCS proteins that in turn down-regulate the signal initiated by the cytokine. Through their impact on the cytokine-activated JAK-STAT pathway, the SOCS proteins are involved in many diseases that come to the attention of the pediatric nephrologist. For example, an increase in the expression of SOCS-2 and -3 may be a cause of growth hormone resistance and thus may contribute to the growth retardation that affects children with chronic renal failure. Because of their obvious biologic importance, the SOCS proteins have been the subject of intense research that includes the development of strategies to utilize these proteins to control cytokine-induced JAK/STAT signal transduction for therapeutic purposes.

    View details for DOI 10.1007/s00467-004-1766-8

    View details for Web of Science ID 000228535900003

    View details for PubMedID 15723195

  • Growth hormone resistance in uremia, a role for impaired JAK/STAT signaling 7th Symposium on Growth and Development in Children with Chronic Kidney Disease Rabkin, R., Sun, D. F., Chen, Y., Tan, J., Schaefer, F. SPRINGER. 2005: 313–18

    Abstract

    Resistance to growth hormone (GH) is a significant complication of advanced chronic renal failure. Thus while the circulating GH levels are normal or even elevated in uremia, resistance to the hormone leads to stunting of body growth in children and contributes to muscle wasting in adults. Insensitivity to GH is the consequence of multiple defects in the GH/insulin-like growth factor-1 (IGF-1) system. Expression of the GH receptor may be reduced, although this is not a consistent finding, GH activation of the Janus kinase 2-signal transducer (JAK2) and activator of transcription (STAT) signal transduction pathway is depressed and this leads to reduced IGF-1 expression, and finally there is resistance to IGF-1, a major mediator of GH action. We review these various defects with an emphasis on the GH-activated JAK2-STAT5 pathway, since this pathway is essential for normal body growth and there has been recent progress in our understanding of the perturbations that occur in uremia.

    View details for DOI 10.1007/s00467-004-1713-8

    View details for Web of Science ID 000227713600011

    View details for PubMedID 15692835

  • Cardiac resistance to growth hormone in uremia KIDNEY INTERNATIONAL Zheng, Z. L., Sun, D. F., Tummala, P., Rabkin, R. 2005; 67 (3): 858-866

    Abstract

    Cardiovascular disease is a major cause of death in end-stage renal disease (ESRD). Since growth hormone is required for maintaining normal cardiac structure and function and as growth hormone has a salutary effect on cardiac remodeling in disease, we postulated that if cardiac resistance to growth hormone develops in chronic renal failure (CRF) this may predispose to the cardiomyopathy of uremia. We set out to test whether in CRF there is resistance to the cardiac action of growth hormone and whether this defect might be caused by altered growth hormone signaling.Growth hormone-deficient (dw/dw) rats and growth hormone-intact Sprague-Dawley rats underwent a subtotal nephrectomy or sham operation and pair feeding.In dw/dw rats treated with growth hormone for 8 days there was a significant increase in insulin-like growth factor-1 (IGF-1) mRNA levels in controls but this response was attenuated in CRF. Next, growth hormone-stimulated Janus kinase-signal transducers and activators of transcription (JAK2-STAT5) signaling was studied 15 minutes after intravenous growth hormone in dw/dw and Sprague-Dawley rats. Growth hormone receptor, JAK2, STAT5a, and STAT5b protein levels were unaltered in CRF. Growth hormone-induced JAK2, growth hormone receptor (GHR), and STAT5 tyrosine phosphorylation was significantly depressed in CRF as was nuclear translocation of phosphorylated STAT5. When rats were treated with pharmacologic dose growth hormone, STAT5 phosphorylation increased similarly in CRF and control rats.Uremic rats develop cardiac resistance to growth hormone caused at least, in part, by a postreceptor defect in growth hormone-induced signaling that is characterized by impaired phosphorylation and nuclear translocation of STAT5. These findings raise the question whether growth hormone resistance contributes to the cardiac changes of uremia.

    View details for Web of Science ID 000227013500006

    View details for PubMedID 15698425

  • Chronic uremia attenuates growth hormone-induced signal transduction in skeletal muscle JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Sun, D. F., Zheng, Z. L., Tummala, P., Oh, J., Schaefer, F., Rabkin, R. 2004; 15 (10): 2630-?

    Abstract

    Malnutrition and muscle wasting are common in chronic renal failure (CRF) and adversely affect morbidity and mortality. Contributing to the muscle wasting is resistance to growth hormone (GH). For testing whether impaired GH signaling is a cause of the skeletal muscle GH resistance and for elucidating its mechanisms, muscle GH signaling and action were studied in GH-deficient rats with surgically induced CRF and sham-operated pairfed control rats. GH treatment increased gastrocnemius muscle IGF-1 mRNA levels significantly in control but not in CRF rats. GH-activated Janus-associated kinase 2 (JAK2)-signal transducers and activators of transcription 5 (STAT5) signaling was impaired in CRF rats, despite normal GH receptor (GHR), JAK2, and STAT5 protein levels. Phosphorylation of the GHR, JAK2, and STAT5 in response to GH was depressed by nearly half in CRF (P < 0.05), and nuclear phospho-STAT5 levels were depressed by approximately one third (P < 0.01). GH-stimulated suppressors of cytokine signaling 2 mRNA levels were significantly higher in CRF. This may be related to inflammatory cytokine activity because C-reactive protein levels were elevated. Muscle protein-tyrosine phosphatase activity was also increased significantly by twofold. In conclusion, rats with CRF acquire skeletal muscle resistance to GH that is caused at least in part by impaired JAK2-GHR-STAT5 phosphorylation and nuclear STAT5 translocation. Furthermore, it seems that the attenuated JAK2-STAT5 phosphorylation may be caused by at least two different processes. One involves depressed phosphorylation of the signaling proteins because of increased suppressors of cytokine signaling 2 expression that may be linked to low-grade inflammation. The other may involve increased signaling protein dephosphorylation because of heightened protein-tyrosine phosphatase activity.

    View details for DOI 10.1097/01.ASN.00001139492.36400.6C

    View details for Web of Science ID 000224138300014

    View details for PubMedID 15466267

  • Growth hormone-mediated janus associated kinase-signal transducers and activators of transcription signaling in the growth hormone-resistant potassium-deficient rat JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Schaefer, F., Yoon, S. A., Nouri, P., Tsao, T., Tummala, P., Deng, E., Rabkin, R. 2004; 15 (9): 2299-2306

    Abstract

    Potassium deficiency (KD) is associated with severe growth failure, in part caused by growth hormone (GH) resistance. This study set out to determine whether the resistance could be caused by a defect in GH-mediated janus associated kinase-signal transducers and activators of transcription (STAT) signaling as occurs in uremia. To this end, rats were fed a K-deficient diet for 8 d and pair-fed controls received a K-replete diet. Animals from each group received GH or vehicle, and during this period, KD rats were GH resistant; GH induced body and liver weight gain and linear body growth were severely attenuated in these rats. In addition, signal transduction was studied in the liver of rats that were killed 10 or 15 min after an intravenous GH bolus or vehicle. When the rats were killed, GH receptor mRNA and protein levels were similar in the two groups. The abundance of STAT5, STAT3, and STAT1, proteins that mediate GH signaling, was significantly increased by 40 to 130% in KD. Furthermore, GH induced a far greater increase in STAT5 and STAT3 phosphorylation in this group. STAT5 phosphorylation was enhanced fourfold even when normalized for total STAT5 content. Phosphorylated STAT5 and STAT3 proteins were also increased in nuclear extracts, suggesting normal nuclear translocation of the activated signaling proteins. DNA binding of nuclear STAT5 was unaltered. Thus, in KD, there is resistance to the growth-promoting action of GH despite hyperactivation of the janus associated kinase-STAT signaling pathway. This suggests the presence of a defect distal to the nuclear binding of STAT or, alternatively, a defect in a STAT-independent GH-activated signaling pathway.

    View details for DOI 10.1097/01.ANS.0000137885.63580.92

    View details for Web of Science ID 000223668200008

    View details for PubMedID 15339979

  • New concepts: growth hormone, insulin-like growth factor-I and the kidney GROWTH HORMONE & IGF RESEARCH Rabkin, R., Schaefer, F. 2004; 14 (4): 270-276

    Abstract

    Both growth hormone (GH) and IGF-1 have major effects on normal kidney growth, structure and function and participate in the pathogenesis of certain kidney diseases. Furthermore when the kidneys fail there are profound changes in the circulating GH-IGF-1 system and the renal and systemic responses to these hormones. In this brief review we address the advances that have been made in our understanding of the relationship between growth hormone GH and IGF-1 and the kidney in health and the systemic and local perturbations that occur in kidney disease and identify key unanswered questions.

    View details for DOI 10.1016/j.ghir.2004.02.001

    View details for Web of Science ID 000222744900002

    View details for PubMedID 15231295

  • Albumin stimulates the accumulation of extracellular matrix in renal tubular epithelial cells AMERICAN JOURNAL OF NEPHROLOGY Stephan, J. P., Mao, W. G., Filvaroff, E., Cai, L. P., Rabkin, R., Pan, G. H. 2004; 24 (1): 14-19

    Abstract

    The accumulation of a large amount of plasma proteins in the urine, previously regarded as a marker of glomerular damage, is now recognized as a mediator of tubulointerstitial damage. Using an in vitro approach, several key extracellular matrix (ECM) proteins were analyzed after treatment of primary human renal proximal tubular epithelial cells with fatty acid free human albumin. We demonstrate that human albumin stimulates the accumulation of ECM proteins by proximal tubular epithelial cells through a post-transcriptional mechanism. Albumin induced a significant increase in tissue inhibitor of metalloproteinases (TIMP)-1 and TIMP-2. Taken together, our data suggest that ECM protein accumulation in response to albumin resulted partly from inhibition of ECM degradation. Addition of transforming growth factor beta (TGF-beta)-specific neutralizing antibody failed to alter ECM protein levels after albumin treatment, indicating that the albumin-induced increase in ECM is TGF-beta independent. In conclusion, we have shown that exposure of cultured human proximal tubular cell to albumin leads to the TGF-beta-independent accumulation of ECM proteins, suggesting that albumin may be a contributing factor to the progression of kidney fibrosis in proteinuric states.

    View details for DOI 10.1159/000075347

    View details for Web of Science ID 000188956900002

    View details for PubMedID 14654729

  • Growth hormone (GH) receptor expression in uremia. 36th Annual Meeting of the American-Society-of-Nephrology Greenstein, J., Guest, S., Tummala, P., Tan, J., Rabkin, R. AMER SOC NEPHROLOGY. 2003: 537A–538A
  • Growth hormone (GH) receptors in uremia. Western Regional Meeting of the American-Federation-for-Medical-Research Greenstein, J., Guest, S., Tummala, P., Rabkin, R., Tan, J. LIPPINCOTT WILLIAMS & WILKINS. 2003: S152–S152
  • Diabetic nephropathy. Clinical cornerstone Rabkin, R. 2003; 5 (2): 1-11

    Abstract

    As the epidemic of diabetes spreads so does the number of patients at risk for developing diabetic nephropathy, which occurs in 20% to 40% of all diabetic patients. Indeed, diabetes is the most common cause of end-stage renal disease (ESRD) in the United States, accounting for > 40% of patients starting renal replacement therapy each year. Unfortunately, the outcome for diabetic patients with ESRD is worse than that for nondiabetic patients because of comorbid conditions in the diabetic population. However, with early and intensive blood glucose and blood pressure control--including the use of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers--the development and progression of diabetic kidney disease can be slowed.

    View details for PubMedID 12800476

  • The role of growth factors and ammonia in the genesis of hypokalemic nephropathy JOURNAL OF RENAL NUTRITION Fervenza, F. C., Rabkin, R. 2002; 12 (3): 151-159

    Abstract

    Hypokalemia is a common electrolyte abnormality encountered in clinical practice. It can be identified in an asymptomatic patient undergoing routine electrolyte screening or can manifest itself as part of a number of functional abnormalities in a variety of organs and systems. Among the most commonly recognized complications are profound effects on the cardiovascular and neuromuscular systems, together with abnormalities in acid-base regulation. In humans, hypokalemia contributes to the development of hypertension and predisposes patients to a variety of ventricular arrhythmias, including ventricular fibrillation. Commonly recognized neuromuscular complications include weakness, cramping, and myalgia. Hypokalemia also affects systemic acid-base homeostasis by interfering with multiple components of the renal acid-base regulation and is a frequent cause of metabolic alkalosis. Less known, however, is the role of potassium deficiency in causing progressive renal failure. In animals, potassium deficiency stimulates renal enlargement because of cellular hypertrophy and hyperplasia. If potassium deficiency persists, interstitial infiltrates appear in the renal interstitial compartment, and eventually tubulointerstitial fibrosis develops. In humans, longstanding hypokalemia is associated with the development of renal cysts, chronic interstitial nephritis, and progressive loss of renal function, the so-called hypokalemic nephropathy. This review focuses on the potential mechanisms involved in the development of the hypokalemic nephropathy with emphasis on the role of ammonia and growth factors in its pathogenesis.

    View details for DOI 10.1053/jren.2002.33511

    View details for Web of Science ID 000176676800003

    View details for PubMedID 12105812

  • Effect of prolonged uremia on insulin-like growth factor-I receptor autophosphorylation and tyrosine kinase activity in kidney and muscle EXPERIMENTAL NEPHROLOGY Tsao, T., Fervenza, F., Friedlaender, M., Chen, Y., Rabkin, R. 2002; 10 (4): 285-292

    Abstract

    Recently, based on a study in rats with chronic renal failure (CRF), it has been suggested that IGF-I resistance in uremia may be caused in part by defective IGF-I receptor autophosphorylation and tyrosine kinase activity. Thus if such a defect were to develop in prolonged acute renal failure (ARF), this may explain why IGF-I therapy, effective in rats, has failed to promote recovery from ARF in patients. Accordingly, we examined IGF-I receptor function in rats with uremia of increasing duration and in pair-fed sham-operated controls. After 6 days of prolonged ARF, kidney IGF-I receptor binding increased twofold, while IGF-I stimulated receptor phosphorylation and tyrosine kinase activity were unchanged. Muscle receptor binding, autophosphorylation and tyrosin kinase activity were similar to control values after 6 or even 21 days of uremia. Taking all these findings together it appears that IGF-I resistance in uremia cannot be attributed to a receptor defect. This in turn argues against altered receptor function as a cause of the failure of IGF-I to modify clinical ARF.

    View details for Web of Science ID 000176984800007

    View details for PubMedID 12097832

  • Impaired JAK-STAT signal transduction contributes to growth hormone resistance in chronic uremia JOURNAL OF CLINICAL INVESTIGATION Schaefer, F., Chen, Y., Tsao, T., Nouri, P., Rabkin, R. 2001; 108 (3): 467-475

    Abstract

    Chronic renal failure (CRF) is associated with resistance to the growth-promoting and anabolic actions of growth hormone (GH). In rats with CRF induced by partial renal ablation, 7 days of GH treatment had a diminished effect on weight gain and hepatic IGF-1 and IGFBP-1 mRNA levels, compared with sham-operated pair-fed controls. To assess whether GH resistance might be due to altered signal transduction, activation of the JAK-STAT pathway was studied 10 or 15 minutes after intravenous injection of 5 mg/kg GH or vehicle. Hepatic GH receptor (GHR) mRNA levels were significantly decreased in CRF, but GHR protein abundance and GH binding to microsomal and plasma membranes was unaltered. JAK2, STAT1, STAT3, and STAT5 protein abundance was also unchanged. However, GH-induced tyrosine phosphorylation of JAK2, STAT5, and STAT3 was 75% lower in the CRF animals. Phosphorylated STAT5 and STAT3 were also diminished in nuclear extracts. The expression of the suppressor of cytokine signaling-2 (SOCS-2) was increased twofold in GH-treated CRF animals, and SOCS-3 mRNA levels were elevated by 60% in CRF, independent of GH treatment. In conclusion, CRF causes a postreceptor defect in GH signal transduction characterized by impaired phosphorylation and nuclear translocation of GH-activated STAT proteins, which is possibly mediated, at least in part, by overexpression of SOCS proteins.

    View details for Web of Science ID 000170318300018

    View details for PubMedID 11489940

  • Growth factors in the treatment of wasting in kidney failure JOURNAL OF RENAL NUTRITION Chen, Y., Fervenza, F. C., Rabkin, R. 2001; 11 (2): 62-66

    Abstract

    Protein-energy malnutrition is relatively common in patients with advanced renal failure and is associated with an increase in morbidity and mortality. Accordingly, there is a need to develop more effective treatments to enhance the nutritional status of these patients. One such approach is the use of recombinant growth factors and anabolic steroids for malnourished patients refractory to standard nutritional therapy. In this article, we review the current state of our knowledge regarding the potential use of these agents in malnourished renal failure patients and conclude that although the studies performed to date are indeed encouraging, further and more extensive clinical trials are required to firmly establish their efficacy and safety profile and the indications for clinical use.

    View details for DOI 10.1053/jren.2001.23468

    View details for Web of Science ID 000173725200002

    View details for PubMedID 11295025

  • Hepatocyte growth factor receptor in acute tubular necrosis JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Rabkin, R., Fervenza, F., Tsao, T., Sibley, R., Friedlaender, M., Hsu, F., Lassman, C., Hausmann, M., Huie, P., Schwall, R. H. 2001; 12 (3): 531-540

    Abstract

    In acute tubular necrosis, there are early transient increases in circulating and local bioactive hepatocyte growth factor (HGF) levels and renal HGF receptor (c-MET) gene expression. It has therefore been suggested that endogenous HGF may play a role in initiating renal repair. To test this hypothesis, changes in the levels, activity, and anatomic distribution of c-MET protein were characterized in relation to the onset and localization of DNA synthesis in kidneys of rats with ischemia-induced acute tubular necrosis. Whole-kidney c-MET protein levels were significantly increased in the injured kidneys 12 h after injury and rose to a maximum after 1 d, exceeding the control values by sevenfold. Eight days after injury, c-MET levels, although decreasing, were still elevated above control values. An increase in the levels of activated c-MET, i.e., tyrosine-phosphorylated c-MET, was also evident as early as 12 h after injury. Histologic analyses demonstrated that the increase in c-MET immunoreactivity was most marked in the most severely damaged nephron segments in the outer medulla. In injured proximal tubules, the receptor was redistributed from an apical location to an intracellular location. DNA synthesis was increased in the injured kidneys, especially in the outer medulla, where the increase in c-MET protein levels was most prominent. The increase in DNA synthesis was first detected 12 h after the initial increase in activated c-MET levels. It is concluded that the early increases in the levels of c-MET protein and activated receptor support the hypothesis that HGF participates in the initiation of renal regeneration. In addition, the persistent elevation of c-Met protein levels suggests that prolonged and even late treatment with HGF may be of therapeutic value

    View details for Web of Science ID 000167221400014

    View details for PubMedID 11181801

  • Expression of insulin-like growth factor-I and transforming growth factor-beta in hypokalemic nephropathy in the rat KIDNEY INTERNATIONAL Tsao, T., Fawcett, J., Fervenza, F. C., Hsu, F. W., Huie, P., Sibley, R. K., Rabkin, R. 2001; 59 (1): 96-105

    Abstract

    Potassium deficiency (KD) in the rat retards body growth but stimulates renal enlargement caused by cellular hypertrophy and hyperplasia, which is most marked in the outer medulla. If hypokalemia persists, interstitial infiltrates appear and eventually fibrosis. Since early in KD insulin-like growth factor-I (IGF-I) levels in the kidney are elevated, suggesting that it may be an early mediator of the exaggerated renal growth, and as transforming growth factor-beta (TGF-beta) promotes cellular hypertrophy and fibrosis, we examined the renal expression of these growth factors in prolonged KD.Rats were given a K-deficient diet or were pair fed or ad libitum fed a K-replete diet for 21 days. Growth factor mRNA levels were measured in whole kidney and protein expression localized by immunohistochemistry.KD rats weighed less than pair-fed controls, while the kidneys were 49% larger. Their serum IGF-I and kidney IGF-I protein levels were depressed, as were their IGF-I mRNA levels in liver, kidney, and muscle. These changes can largely be attributed to decreased food intake. In contrast, kidney IGF binding protein-1 (IGFBP-1) mRNA and TGF-beta mRNA levels were increased significantly. Histology of outer medulla revealed marked hypertrophy and adenomatous hyperplasia of the collecting ducts and hypertrophy of the thick ascending limbs of Henle with cellular infiltrates in the interstitium. Both nephron segments immunostained strongly for IGF-I and IGFBP-1, but only the nonhyperplastic enlarged thick ascending Henle limb cells immunostained for TGF-beta, which was strongly positive. Prominent interstitial infiltrates with ED1 immunostained monocytes/macrophages were present.These findings are consistent with a sustained role for IGF-I in promoting the exaggerated renal growth of KD and appear to be mediated through local trapping of IGF-I by the overexpressed IGFBP-1, which together with IGF-I can promote renal growth. The selective localization of TGF-beta to hypertrophied nonhyperplastic nephron segments containing IGF-I raises the possibility that TGF-beta may be serving to convert the mitogenic action of IGF-I into a hypertrophic response in these segments. It is also conceivable that TGF-beta may be a cause of the tubulointerstitial infiltrate. Finally, the low circulating IGF-I levels likely contribute to the impaired body growth.

    View details for Web of Science ID 000166294700012

    View details for PubMedID 11135062

  • Growth factor insensitivity in renal failure Ninth International Symposium of Nephrology at Montecatini Italy: Kidney Proteins and Growth Factors Rabkin, R. INFORMA HEALTHCARE. 2001: 291–300

    Abstract

    Advanced chronic renal failure is associated with multiple endocrine and metabolic abnormalities that result from changes in the secretion and metabolism of hormones and growth factors and the target organ sensitivity to their physiological actions. As a consequence, growth retardation, bone disease, pertubations in lipid, carbohydrate and protein metabolism are commonly seen in patients with chronic renal failure. The recent availability of recombinant growth factors has provided new therapeutic opportunities for correcting these abnormalities. However because of the presence of end-organ resistance relatively high dose therapy is required and this carries an increased risk of side effects. One logical approach to this problem would be to prevent or treat the underlying resistance and thus restore sensitivity to endogenous GH or low doses of the recombinant molecule. To achieve this goal, a better understanding of the mechanism of growth factor resistance is required. In this lecture, in honor of the memory of Frank Carone. I review our current state of knowledge of the impact of advanced renal failure on the tissue sensitivity to insulin, growth hormone and insulin-like-growth factor I.

    View details for Web of Science ID 000170256000001

    View details for PubMedID 11499545

  • Paradoxical body and kidney growth in potassium deficiency Ninth International Symposium of Nephrology at Montecatini Italy: Kidney Proteins and Growth Factors Fervenza, F., Tsao, T., Rabkin, R. INFORMA HEALTHCARE. 2001: 339–46

    Abstract

    In the growing animal, K deficiency (KD) retards body growth, but paradoxically stimulates renal growth. If KD persists, interstitial infiltrates appear and eventually tubulointerstitial fibrosis develops. In patients with chronic KD, renal cysts may form and with time tubulointerstitial disease with renal failure develops. Since early in KD, kidney IGF-I levels increase and may be a cause of the renal hypertrophy, and as TGF-beta promotes hypertrophy and fibrosis, we examined the expression of these growth factors in chronic KD. Rats were given a KD diet or pair or ad-lib fed a normal K diet. After 21 days, KD rats weighed less than pair fed controls, while the kidneys were 49% larger Serum IGF-I and kidney IGF-I protein levels were depressed, as were IGF-I mRNA levels, and is largely attributable to decreased food intake. Kidney IGFBP-1 and TGF-beta mRNA levels were increased (p < 0.05). There was marked hypertrophy and adenomatous hyperplasia of outer medullary collecting ducts, hypertrophy of thick ascending limbs of Henle (TALH) and interstitial infiltrates. Both nephron segments stained strongly for IGF-I and IGFBP-1. Only the non-hyperplastic TALH was strongly TGF-beta positive. Interstitial infiltrates containing monocytes/macrophages were prominent. These findings are consistent with a sustained role for IGF-I in promoting the renal hypertrophy of KD and appear to be caused by local trapping of IGF-I by the over-expressed IGFBP-1. Localization of TGF-beta to the hypertrophied non-hypoplastic tubules containing IGF-I, suggests that TGF-beta may be acting to convert the proliferative action of IGF-I into a hypertrophic response. TGF-beta may also contribute to the genesis of the tubulointerstitial infiltrate. Finally, the reduced levels of serum IGF-1 levels may be a cause of the blunted body growth.

    View details for Web of Science ID 000170256000006

    View details for PubMedID 11499550

  • Effect of metabolic acidosis on the insulin-like growth factor-I system and cathepsins B and L gene expression in the kidney JOURNAL OF LABORATORY AND CLINICAL MEDICINE Fawcett, J., Hsu, F. W., Tsao, T., Rabkin, R. 2000; 136 (6): 468-475

    Abstract

    Prolonged acidemia causes growth retardation and muscle wasting, in part because of reduced food intake, depressed growth hormone secretion, and low serum insulin-like growth factor-I (IGF-I) levels. Paradoxically, in the rat kidney, protein synthesis increases, cathepsin B and L activities decline, protein degradation falls, and the kidneys enlarge. Because IGF-I has been implicated as a cause of renal hypertrophy in a variety of conditions, we examined whether IGF-I could be playing a role in the renal hypertrophy of acidosis. Rats were gavaged with NH4Cl or water for 4 days. Water-gavaged rats either were pair-fed with the NH4Cl-loaded rats (pH 7.15) or were given free access to food and served as controls. After 2 days, kidney weight and IGF-I mRNA levels did not differ between the groups, but kidney IGF-I protein levels were significantly higher in the acidotic rats. After 4 days the kidneys of the acidotic rats were significantly larger than the kidneys in both control groups but the renal IGF-I levels did not differ between the groups. It is notable that renal cathepsin B and L mRNA levels were reduced by 30% to 50% at both times. Thus the transient increase in renal IGF-I protein levels in acidosis, before the onset of hypertrophy, suggests that IGF-I may play a role in initiating kidney growth. Furthermore, it appears that reduced cathepsin B and L gene expression is a cause of the low renal cathepsin activity seen in acidosis. This likely contributes to the depressed renal proteolysis caused by acidosis.

    View details for DOI 10.1067/mlc.2000.110606

    View details for Web of Science ID 000165697900005

    View details for PubMedID 11128748

  • Response to growth hormone therapy in experimental ischemic acute renal failure JOURNAL OF LABORATORY AND CLINICAL MEDICINE Fervenza, F. C., Hsu, F. W., Tsao, T., Friedlaender, M. M., Rabkin, R. 1999; 133 (5): 434-439

    Abstract

    In acute renal failure (ARF), the gene and peptide expression of insulin-like growth factor-I (IGF-I) falls. Because IGF-I is regulated by growth hormone (GH) and because kidney GH receptor expression is also attenuated in ARF, the impaired IGF-I expression may partly reflect local GH resistance. Because IGF-I treatment accelerates recovery from ARF, we determined whether high-dose GH therapy could overcome this putative GH resistance, stimulate IGF-I production, and enhance recovery. Rats with ARF were given 2.5 mg GH or vehicle (V) over 2 days, beginning 24 hours before the onset of ARF. GH prevented weight loss but did not modify the course of ARF. Next we determined whether the failure of GH to modify kidney recovery could reflect a failure to stimulate renal IGF-I gene expression. Rats were treated with GH or V over an 18-hour period beginning 1 day after the induction of ARF. Hepatic IGF-I mRNA and serum IGF-I peptide levels rose significantly with GH treatment, but the low kidney IGF-I mRNA levels did not respond. We conclude that the failure of GH to enhance recovery from ARF is caused by impaired GH-stimulated renal IGF-I production, while the maintenance of body weight likely reflects the systemic effects of the increase in hepatic IGF-I production.

    View details for Web of Science ID 000080120000005

    View details for PubMedID 10235126

  • Intrarenal insulin-like growth factor-1 axis after unilateral nephrectomy in rat JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Fervenza, F. C., Tsao, T., Hsu, F., Rabkin, R. 1999; 10 (1): 43-50

    Abstract

    It has been suggested that insulin-like growth factor-1 (IGF-1) may play a role in early compensatory renal growth. Since IGF-1 action is influenced by IGF binding proteins (IGFBP), this study was conducted to characterize the changes in gene expression not only of IGF-1 and its receptor, but also of IGFBP in the hypertrophying kidney of adult and weanling rats 1 wk after removal of the other kidney. At this time, there were distinct age-dependent changes in the renal IGF-1 axis. In the mature kidney, IGF-1 mRNA levels fell without a change in kidney IGF-1 peptide content. Likewise, although IGFBP-2, -3, and -5 mRNA levels fell, membrane-associated IGFBP did not change. IGF-1 receptor mRNA levels and IGF-1 receptor number both fell. In the weanling kidneys, IGF-1 mRNA and peptide levels and IGF-1 receptor binding were unaltered. However, IGFBP-3, -4, and -5 mRNA levels were increased, as were plasma membrane-associated IGFBP. Although these changes in the intrarenal IGF-1 axis were distinct, it is difficult to conceive how in either the mature or immature rat they could contribute to the ongoing compensatory renal growth that occurs 1 wk after loss of kidney mass unless IGF-1 were acting in a synergistic manner with other growth promoters.

    View details for Web of Science ID 000077878300006

    View details for PubMedID 9890308

  • Role of insulinlike growth factor I in renal transplantation: Evaluation of intra- and postoperative effects Congress on New Dimensions in Transplantation - Weaving the Future Maestri, M., Adams, G., Gaspari, A., Passalia, L., Innocente, F., Rademacher, J., Luzzana, F., Polese, M., Bolla, E., Noli, S., Scotti-Foglieni, A., Scotti-Foglieni, C., Dafoe, D., Dionigi, P., Zonta, A., Rabkin, R. ELSEVIER SCIENCE INC. 1998: 2019–23

    View details for Web of Science ID 000075412900158

    View details for PubMedID 9723374

  • Insulin-like growth factor-I treatment to enhance renal function in advanced chronic renal failure 8th International Symposium of Nephrology Fervenza, F. C., Friedlaender, M. M., Ike, J. O., Rabkin, R. TAYLOR & FRANCIS INC. 1998: 349–56

    Abstract

    Because of the potential for IGF-I to enhance renal function in advanced chronic renal failure (CRF) we set out to determine whether IGF-I can induce a sustained increase in renal function in patients with near end-stage renal failure. To this end we first examined the impact of CRF on the pharmacokinetics of IGF-I and then we examined the effect of prolonged IGF-I treatment on the renal function of patients with an average GFR of 17 mL/min/1.73 m2. Interestingly the metabolic clearance rate of IGF-I in CRF subjects was similar to that in normal subjects even though the total serum IGF-I levels rose to higher maximum levels. This increase was due to a reduced volume of IGF-I distribution, a consequence of the elevated serum IGF binding proteins in CRF subjects. Treatment with IGF-I (60 mg/kg twice daily sc) for 31 days resulted in a 14% and 18% increase in the inulin and PAH clearances respectively (n = 6 patients). These parameters returned to basal levels on stopping treatment. Serum immunoreactive IGFBP-3 levels fell and IGFBP-2 and -3 levels rose during IGF-I therapy. Adverse effects were mild, of short duration and easily manageable. Thus IGF-I pharmacokinetics are largely unchanged in CRF and the administration of IGF-I produces a modest improvement in the GFR. These results appear to justify more extensive examination of the therapeutic role of IGF-I in the treatment of CRF.

    View details for Web of Science ID 000073058400021

    View details for PubMedID 9574462

  • The insulin-like growth factor-I axis in acute renal failure 8th International Symposium of Nephrology Friedlaender, M. M., Fervenza, F. C., Tsao, T., Hsu, F., Rabkin, R. TAYLOR & FRANCIS INC. 1998: 343–48

    Abstract

    We have examined the response of the renal insulin-like growth factor (IGF-I) axis to acute ischemic injury in the rat Key findings included a decrease in IGF-I mRNA and peptide levels, a decrease in GH receptor gene plus protein expression and a decrease in the IGF binding proteins except for IGF binding protein I. Administration of GH to compensate for the reduced GH receptor binding corrected the IGF-I mRNA levels suggesting a relative GH deficiency. Interestingly, IGF-I receptor mRNA levels were unchanged while plasma membrane IGF-I receptor number increased two fold. This appeared to be due to a redistribution of receptors to a membrane location. IGF-I receptor autophosphorylation and tyrosine kinase activity were intact despite severe uremia for up to 6 days. We propose that this increase of functional IGF-I receptors following acute tubular necrosis will sensitize the kidney to the administration of exogenous IGF-I.

    View details for Web of Science ID 000073058400020

    View details for PubMedID 9574461

  • Effect of insulin-like growth factor binding proteins on the response of proximal tubular cells to insulin-like growth factor-I KIDNEY INTERNATIONAL Yap, J., Tsao, T., Fawcett, J., Fielder, P. J., Keller, G. A., Rabkin, R. 1997; 52 (5): 1216-1223

    Abstract

    The insulin-like growth factor binding proteins (IGFBP) are major modulators of insulin-like growth factor-I (IGF-I) action, but relatively little is known about their production by kidney tubular cells or about their modulating effects on the action of IGF-I on these cells. In this study we demonstrated that rabbit proximal tubular cells express the genes for IGFBP-2, -4 and -5 and secrete 24 and 32 kDa size binding proteins. The rate of IGFBP production by these cells was regulated by several growth factors including hydrocortisone, which was potently stimulatory, and EGF, which was inhibitory. The overall effect of these kidney cell-secreted IGFBPs was to inhibit the mitogenic activity of IGF-I. Similarly, recombinant IGFBP-3, the major circulating IGFBP that in kidney is produced close to the proximal tubules, also inhibited IGF-I stimulated DNA synthesis in cultured rabbit proximal tubular cells and in cultured opossum kidney (OK) cells. IGFBP-3 also inhibited basal DNA synthesis in OK cells in the absence of added IGF-I, suggesting that this IGFBP may have an IGF-I independent action. These findings highlight the important effect that IGFBPs have on the action of IGF-I on kidney cells and support the notion that the changes in IGFBPs observed in various renal diseases may contribute to the pathophysiology of these diseases.

    View details for Web of Science ID A1997YC33900005

    View details for PubMedID 9350644

  • The IGF-I axis in kidney and skeletal muscle of potassium deficient rats KIDNEY INTERNATIONAL Hsu, F. W., Tsao, T., Rabkin, R. 1997; 52 (2): 363-370

    Abstract

    Potassium deficiency in the rat results in growth retardation, muscle wasting and renal hypertrophy. This study tests the thesis that K deficiency leads to tissue distinct changes in the local IGF-I system and cell sensitivity to IGF-I that favors renal enlargement on the one hand and impaired muscle growth on the other. In rats after eight days of K deficiency, compared to pair-fed control rats, food utilization and muscle and body wt gain were attenuated while the kidneys enlarged. In muscle GH receptor and IGF-I gene expression, IGF-I peptide and IGF binding protein-5 (IGFBP) levels were decreased. Together with reduced food utilization, these changes may contribute to the attenuated muscle growth. In the enlarged kidneys despite a fall in IGF-I mRNA level, IGF-I peptide concentration was increased more than twofold. This increase in IGF-I could be caused by the increase in kidney IGFBP-1 gene and protein expression and the decrease in kidney IGF-I degrading activity noted in K deficiency. Treatment with IGF-I failed to induce body or muscle growth, but induced a further increase in kidney size and enlargement of the spleen. Thus, in K deficiency the spontaneous increase in IGF-I levels in the kidney that is IGF-I sensitive may well be a cause of the renal hypertrophy.

    View details for Web of Science ID A1997XP20500009

    View details for PubMedID 9263991

  • Insulin-like growth factor-I ameliorates delayed kidney graft function and the acute nephrotoxic effects of cyclosporine TRANSPLANTATION Maestri, M., Dafoe, D. C., Adams, G. A., Gaspari, A., Luzzana, F., Innocente, F., Rademacher, J., Dionigi, P., Barbieri, A., Zonta, F., Zonta, A., Rabkin, R. 1997; 64 (2): 185-190

    Abstract

    Delayed graft function (DGF) is a relatively common complication after cadaveric renal transplantation. The adverse effect of DGF on long-term graft survival has lead to intensive efforts to reduce ischemic graft injury. In this study we examined the effects of a new protective treatment based on insulin growth factor (IGF)-I. We evaluated the impact of the treatment on renal recovery and on the nephrotoxicity that is a common side effect of mainstream immunosuppressants. Because therapy with IGF-I or the analog des(1-3)IGF-I is effective in treating experimental ischemic renal failure, these peptides may be useful as perspective clinical treatments.We have addressed three areas relating to the potential use of IGF-I and its analog des(1-3)IGF-I. First, because of the immunogenic properties of IGF-I, we assessed the effect of des(1-3)IGF-I on the rejection of skin allografts in Lewis rats. Next we determined whether treatment with des(1-3)IGF-I influences the early function of transplanted kidneys in a model of DGF induced by a combination of warm and cold ischemia. Finally we tested whether IGF-I protects against acute cyclosporine nephrotoxicity.Des(1-3)IGF-I did not accelerate the rejection of the skin grafts (P=0.57). The administration of this peptide in a model of syngenic renal transplant improved the early function of the graft. Postoperative values of creatinine and blood urea nitrogen were significantly better (P<0.05) in treated animals. IGF-I also ameliorated the nephrotoxicity of cyclosporine, with better values of creatinine and blood urea nitrogen (P<0.05).In evaluating this study it should be recognized that the animal models studied, although widely used, differ from the human condition. However, IGF-I and des(1-3)IGF-I exhibit properties that strongly suggest their value in preventing clinical DGF, and they deserve further studies.

    View details for Web of Science ID A1997XN86900001

    View details for PubMedID 9256171

  • The use of growth factors to increase glomerular filtration rate in chronic renal failure patients CURRENT OPINION IN NEPHROLOGY AND HYPERTENSION Miller, S. B., Rabkin, R. 1997; 6 (4): 401-404

    Abstract

    Insulin-like growth factor-1 has been safely administered to humans with chronic renal failure in an attempt to increase glomerular filtration rate. The results of short-term studies have been encouraging. Further studies will be required to better define the role of this or other growth factors in increasing glomerular filtration rate in patients with chronic renal failure.

    View details for Web of Science ID A1997XK26600016

    View details for PubMedID 9263692

  • Nutrient regulation of insulin-like growth factor-1 8th International Congress on Renal Nutrition and Metabolism Rabkin, R. KARGER. 1997: 157–60

    Abstract

    Insulin-like growth factor-I (IGF-I) is a nutrient-regulated growth factor. In malnutrition, serum IGF-I levels fall despite normal or elevated growth hormone (GH) levels and this is caused by resistance to GH, defects in IGF-I gene transcription and translation and mRNA instability. Because IGF-I production is sensitive to protein and energy deficiencies serum IGF-I levels may be valuable in the assessment of the nutritional status of patients with wasting diseases, including advanced renal failure. It should be recognized, however, that serum or tissue IGF-I peptide levels do not always reflect IGF-I gene expression. For example, in the K-deficient rat, kidney IGF-I levels are increased even though IGF-I mRNA levels are low. This appears to be due in part to increased sequestration by IGF-binding proteins and by decreased kidney IGF-I degradation. The increase in kidney IGF-I may contribute to the exaggerated renal growth that occurs in hypokalemia.

    View details for Web of Science ID A1997YE34400007

    View details for PubMedID 9387107

  • Nuclear transport of insulin-like growth factor-I and insulin-like growth factor binding protein-3 in opossum kidney cells. ENDOCRINOLOGY Li, W. L., Fawcett, J., Widmer, H. R., Fielder, P. J., Rabkin, R., Keller, G. A. 1997; 138 (4): 1763-1766

    Abstract

    When added to cultured opossum kidney cells, IGF-I is internalized and transported to distinct intracellular compartments that depend on the cell location within the monolayer. In resting cells away from the periphery of the monolayer, IGF-I is internalized by a clathrin coated pit pathway and delivered to the endosomal compartment. In contrast, cells growing at the edges of a monolayer or an experimental wound internalize IGF-I by an alternative route which rapidly delivers IGF-I to the nucleus. Similarly to IGF-I, IGFBP-3 is also internalized and accumulates in the endosomal compartment in resting cells whereas it is targeted to the nucleus in proliferating cells. IGFBP-3, which contains a putative nuclear targeting signal, may act as a carrier for IGF-I nuclear transport. The transport of IGF-I and IGFBP-3 to two different compartments may influence their biological activity.

    View details for Web of Science ID A1997WP33500053

    View details for PubMedID 9075741

  • Early experience with extended use of insulin-like growth factor-1 in advanced chronic renal failure KIDNEY INTERNATIONAL Ike, J. O., Fervenza, F. C., Hoffman, A. R., Yeh, I., Hintz, R. L., Liu, F., Rabkin, R. 1997; 51 (3): 840-849

    Abstract

    Short-term high-dose insulin-like growth factor-1 (IGF-1) therapy has been shown to enhance glomerular filtration rate (GFR) in end-stage chronic renal failure (CRF), but the efficacy and safety of prolonged therapy is unproven. To determine if prolonged therapy with IGF-1 can enhance renal function in advanced CRF, eight patients were entered into a study to receive one month of IGF-1 treatment, 60 micrograms/kg subcutaneously b.i.d. Six patients completed the study and two dropped out for reasons considered to be unrelated to the IGF-I treatment. Baseline inulin and PAH clearances averaged 17 +/- 3 and 66 +/- 14 ml/min/1.73 m2, respectively, in the subjects who completed the study. With treatment there was a modest 14% increase in the average GFR which approached statistical significance (P = 0.051). After stopping treatment the clearance values returned to basal values. The PAH clearance showed a similar trend. There were no significant changes in BUN, serum creatinine or electrolyte levels. On the other hand there were marked changes in the serum IGF binding protein (IGFBP) profile. Serum IGFBP-3 levels fell while IGFBP-1 and -2 levels rose during treatment, changes that likely affect the bioavailability of IGF-I. Thus, in this small series of patients IGF-1 treatment produced significant changes in the serum IGFBP profile and a modest upward trend in the GFR.

    View details for Web of Science ID A1997WL44800030

    View details for PubMedID 9067919

  • IGF-I receptor binding, autophosphorylation, and kinase activity in kidney and muscle of acutely uremic rats AMERICAN JOURNAL OF PHYSIOLOGY-RENAL PHYSIOLOGY Tsao, T., Hsu, F. W., Rabkin, R. 1997; 272 (3): F325-F332

    Abstract

    Following acute tubular necrosis (ATN), kidney plasma membrane insulin-like growth factor-I (IGF-I) receptor number increases markedly, although IGF-I receptor mRNA levels do not change. To determine whether this increase could represent a redistribution of intracellular receptors and whether receptor function is intact in acute uremia, rats with ATN of 2 days duration and pair-fed controls were studied. Skeletal muscle receptor binding was unchanged. In contrast, binding to receptors in solubilized cortex and isolated cortical plasma membranes increased significantly due to an increase in receptor number. However, the increase in membrane binding was threefold greater than the increase in solubilized cortex binding. This indicates that the increase in total cellular IGF-I receptors can only account for a minor portion of the increase in abundance of plasma membrane receptors number and is consistent with a redistribution of receptors from an intracellular to a membrane location as the major mechanism. Autophosphorylation and receptor kinase activity were unaffected by the uremia (blood urea nitrogen of approximately 198 mg/dl). Since these early steps of IGF-I receptor signaling are intact early in acute uremia, it is likely that at this time in the course of the disease the increase in receptor number will heighten the sensitivity to IGF-I and may thus favor its participation in renal repair.

    View details for Web of Science ID A1997WP85000007

    View details for PubMedID 9087675

  • Regional changes in the intrarenal insulin-like growth factor-I axis in diabetes KIDNEY INTERNATIONAL Fervenza, F. C., Tsao, T., Hoffman, A. R., Rabkin, R. 1997; 51 (3): 811-818

    Abstract

    Since insulin-like growth factor-I (IGF-I) has been shown to promote renal growth and as kidney IGF-I content increases during the early days after the onset of diabetes, it is likely that this growth factor contributes to initial diabetic renal hypertrophy. However, it is unclear whether IGF-I contributes to the continued renal growth that occurs in diabetes. Since IGF-I action is mediated through its receptor and as its bioavailability is regulated by IGF binding proteins (IGFBP), we postulated that changes in IGF-I receptor binding or IGFBP production may favor a role for IGF-I in diabetic renal growth when kidney IGF-I levels have returned to normal. To test this thesis, we studied kidneys of rats after seven days of streptozotocin diabetes. In diabetic cortex and medulla, growth hormone receptor mRNA levels and IGF-I and IGF-I receptor mRNA and protein product levels were unchanged. In cortex IGFBP-1 mRNA levels were increased while IGFBP-2 and -4 mRNA levels decreased. In medulla the only change was a fall in IGFBP-1 mRNA levels. Using Western ligand blot we observed an increase in a 32 kDa plasma membrane-associated IGFBP. Insulin therapy reversed all changes except the elevated cortical IGFBP-1 mRNA levels, indicating the presence of regional heterogeneity in the IGFBP response to diabetes in the kidney. However, the lack of change in IGF-I, IGF-I receptor and growth hormone receptor gene expression and protein products after one week of diabetes argues against a role for IGF-I in sustaining diabetic renal growth beyond the initial growth phase.

    View details for Web of Science ID A1997WL44800026

    View details for PubMedID 9067915

  • Renal hypertrophy and kidney disease in diabetes DIABETES-METABOLISM REVIEWS Rabkin, R., Fervenza, F. C. 1996; 12 (3): 217-241

    View details for Web of Science ID A1996VW07300004

    View details for PubMedID 8959386

  • Processing of I-125-insulin by polarized cultured kidney cells EXPERIMENTAL CELL RESEARCH Rabkin, R., Hamik, A., Yagil, C., Hamel, F. G., Duckworth, W. C., Fawcett, J. 1996; 224 (1): 136-142

    Abstract

    Renal clearance of insulin is achieved by glomerular filtration and by passage from the postglomerular peritubular circulation into the renal interstitium. In the proximal tubule, filtered insulin binds to the apical membrane and is internalized and degraded while insulin in the interstitium is taken up by receptor-mediated endocytosis and degraded. To study these processes we have utilized cultured opossum kidney cells. These cells have proximal-like features and process insulin in a manner consistent with that described in vivo. To study apical and basolateral uptake and metabolism of insulin independently, cells were grown on filters suspended in culture wells. insulin was degraded to large insulin-size intermediates and low-molecular-weight products. This occurred whether the protein was internalized from the apical or basolateral pole of the cells. Analysis of the intermediate products by reverse-phase high-performance liquid chromatography revealed that products formed after apical or basolateral internalization were similar. Since products were preferentially released from the side of uptake, it is likely that apically and basolaterally internalized insulin is degraded in comparable organelles located in different regions of the cell. Most of the internalized insulin traversed the degradative pathway but some insulin followed a retroendocytic or minor transcytotic pathway. Degradation was inhibited by chloroquine, which also selectively increased the release of internalized insulin from the apical pole irrespective of the side of uptake. Thus while the polar degradative processes appear to be similar in nature, the polar exocytotic processes appear to be different.

    View details for Web of Science ID A1996UH25100017

    View details for PubMedID 8612678

  • Pharmacokinetics of insulin-like growth factor-1 in advanced chronic renal failure KIDNEY INTERNATIONAL Rabkin, R., Fervenza, F. C., Maidment, H., Ike, J., Hintz, R., Liu, F., Bloedow, D. C., Hoffman, A. R., Gesundheit, N. 1996; 49 (4): 1134-1140

    Abstract

    Information regarding the impact of chronic renal failure (CRF) on IGF-1 serum clearance is limited. Thus we evaluated the pharmacokinetics of insulin-like growth factor-1 (IGF-1) in six normal adults and six adults with advanced CRF (serum creatinine 7 +/- 0.8 mg/dl). All subjects were given 80 micrograms/kg recombinant human IGF-1 s.c. and blood was sampled over 48 hours. Baseline total serum IGF-1 levels were similar in both groups, but peak levels were elevated significantly in CRF; this was apparently related to the reduced distribution volume in CRF subjects. CRF did not affect the metabolic clearance rate (MCR) of total serum IGF-1. Immunoreactive IGF binding protein-3 (IGFBP-3) levels were greater in CRF. Western immunoblots revealed that the apparent increase in IGFBP-3 was largely due to an increase in immunoreactive fragments. IGFBP-3 protease activity was not increased. Thus IGFBP fragment accumulation likely reflects reduced fragment clearance. Western ligand blots revealed elevated 30 and 34 kDa IGFBP levels and IGFBP products in CRF serum. Serum acid labile subunit levels were unchanged in CRF. Peak free IGF-1 levels and the MCR of free IGF-1 did not differ between groups. In both groups the MCR of free IGF-1 exceeded the MCR of total IGF-1 by approximately 30-fold. These data suggest that in CRF patients receiving s.c. IGF-1: (a) total serum IGF-1 levels are increased as a result of elevated circulating IGFBPs that may restrict the distribution of IGF-1 beyond plasma; (b) serum free IGF-1 levels are not altered; and (c) the IGF-1 MCR is unchanged in CRF. Thus, in advanced CRF, apart from a reduction in the total IGF-1 volume of distribution the pharmacokinetics of IGF-1 are largely unaltered.

    View details for Web of Science ID A1996UB11200035

    View details for PubMedID 8691735

  • Response of the intrarenal insulin-like growth factor-I axis to acute ischemic injury and treatment with growth hormone and epidermal growth factor KIDNEY INTERNATIONAL Fervenza, F. C., Tsao, T., Rabkin, R. 1996; 49 (2): 344-354

    Abstract

    We previously reported that following bilateral acute tubular necrosis (ATN) profound changes in the intrarenal insulin-like growth factor-I axis occurs which are unrelated to altered nutritional intake. In this current report we studied rats with unilateral ATN to assess whether these changes reflect a response to acute injury or the accompanying uremia. Compared to the contralateral kidney, the injured kidney showed an increase in IGF-I receptor number without a change in IGF-I receptor mRNA levels, a decrease in IGF-I mRNA and IGF-I protein levels, a decrease in growth hormone (GH) receptor mRNA abundance and receptor binding. There was also a decrease in IGF binding protein-2, -3 and -5 mRNA levels together with a fall in protein products. Since this unilateral ATN model excludes the influence of uremia and reduced nutritional intake, we surmised that these changes reflect a direct response to injury. Next, because of the reduced GH receptor binding noted above and the reported decrease in epidermal growth factor (EGF) expression in ATN, we tested the thesis that the low kidney IGF-I mRNA levels in ATN are partly due to a relative or absolute deficiency of these hormones. Administration of EGF or GH promptly increased ATN kidney IGF-I mRNA levels to control kidney values, lending support to the thesis. The response to EGF also suggests that the salutary effect of EGF treatment in ATN may partly be mediated by stimulating IGF-I production.

    View details for Web of Science ID A1996TR10300006

    View details for PubMedID 8821816

  • Protein turnover in the hypertrophying kidney 7th International Congress on Renal Nutrition and Metabolism Rabkin, R., Shechter, P., Shi, J. D., Boner, G. KARGER. 1996: 153–56

    Abstract

    To establish whether altered proteolysis contributes to the increase in protein content in hypertrophying kidneys, we studied protein turnover in proximal renal tubules isolated from rats with three forms of renal hypertrophy, diabetes mellitus (DM), ammonium chloride-induced acidosis and compensatory renal growth (CRG). We found that in DM and in chronic acidosis the normal balance in protein turnover is altered due to attenuated proteolysis and accelerated protein synthesis. Together this favors an increase in kidney protein content. In contrast, in CRG, the increase in protein content is entirely due to increased protein synthesis. Thus, the changes in protein turnover leading to the net gain in kidney protein content in renal hypertrophy depends on the cause of hypertrophy.

    View details for Web of Science ID A1996TG87800034

    View details for PubMedID 8676809

  • EXPRESSION OF THE GENES ENCODING THE RAT RENAL INSULIN-LIKE GROWTH-FACTOR-I SYSTEM JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Rabkin, R., Brody, M., LU, L. H., Chan, C., Shaheen, A. M., Gillet, N. 1995; 6 (5): 1511-1518

    Abstract

    Insulin-like growth factor-I (IGF-I) modulates renal function, growth, and repair. IGF-I produced in the kidney is one component of the intrarenal IGF-I system comprising the IGF-I receptor (IGF-IR) and six IGF-binding proteins (IGFBP). Because of the physiologic importance of IGF-I and its potential therapeutic properties, the renal sites of mRNA synthesis for IGF-I, IGF-IR, and IGFBP-I through IGFBP-5 were characterized in rat kidney by in situ hybridization. Anatomical heterogeneity was prominent. IGF-I mRNA was present in the thick ascending limb of Henle in the outer medulla, whereas IGF-IR mRNA was diffusely present at low levels throughout the kidney. IGFBP-I mRNA was localized to cells within the distal convoluted tubules as well as the thick ascending limb of Henle. IGFBP-2 mRNA was expressed in glomeruli, medullary ray collecting ducts, pelvic smooth muscle and uroepithelium, and the papilla tip; IGFBP-3 mRNA was localized to the cortical interstitium, whereas IGFBP-4 mRNA was expressed in proximal tubules, medullary ray collecting ducts, and glomeruli. IGFBP-5 was strongly positive throughout the medulla with lesser expression in the distal convoluted tubules and glomeruli. This study highlights the complexity of the intrarenal IGF-I system. The striking heterogeneity of IGFBP gene expression suggests that the various IGFBP may have diverse modulatory effects on the action of IGF-I or discrete effects of their own.

    View details for Web of Science ID A1995TF75200027

    View details for PubMedID 8589331

  • RENAL GROWTH HORMONE-INSULIN-LIKE GROWTH-FACTOR-I SYSTEM IN ACUTE-RENAL-FAILURE KIDNEY INTERNATIONAL Tsao, T., Wang, J., Fervenza, F. C., Vu, T. H., JIN, I. H., Hoffman, A. R., Rabkin, R. 1995; 47 (6): 1658-1668

    Abstract

    The renal growth hormone--insulin-like growth factor-I system in acute ischemic renal failure. Recovery from acute tubular necrosis (ATN) is accelerated by IGF-I therapy. Furthermore, the local renal growth hormone-IGF-I system may participate in the natural repair. We examined the IGF-I system in rat kidneys subjected to 60 minute ischemia compared to sham operated controls. Two days after injury, growth hormone receptor mRNA and IGF-I mRNA levels fell approximately 9 to 33% of control values. This was associated with a reduction in kidney immunoreactive IGF-I levels. In contrast, IGF-I receptor mRNA abundance was unchanged. However, plasma membrane IGF-I receptor binding on day 2 and day 7 was near double the control values (P < 0.01). Scatchard analysis revealed a near twofold increase in receptor number. Since receptor mRNA levels were unchanged, this implies receptor protein up-regulation. In contrast to unchanged IGF-I receptor mRNA levels, the abundance of mRNA levels of insulin-like growth factor binding proteins (IGFBP) -2, -3, -4 and -5 fell approximately 14 to 62% of control levels day 2 after injury (P < 0.05), suggesting reduced IGFBP production. Thus, the renal response to ischemic ATN, namely, low IGFBP mRNA levels and high IGF-I receptor number, may function to increase IGF-I bioavailability and thereby enhance the reparative actions of local and circulating IGF-I in ischemic ATN.

    View details for Web of Science ID A1995QZ86800020

    View details for PubMedID 7543960

  • INSULIN-LIKE GROWTH-FACTOR-I TREATMENT OF ACUTE-RENAL-FAILURE JOURNAL OF LABORATORY AND CLINICAL MEDICINE Rabkin, R. 1995; 125 (6): 684-685

    View details for Web of Science ID A1995RB44500005

    View details for PubMedID 7769361

  • THE PROCESSING OF INSULIN-LIKE GROWTH-FACTOR-I (IGF-I) BY A CULTURED KIDNEY-CELL LINE IS ALTERED BY IGF-BINDING PROTEIN-3 ENDOCRINOLOGY Fawcett, J., Rabkin, R. 1995; 136 (4): 1340-1347

    Abstract

    The proximal renal tubule is a common site of peptide hormone metabolism, including that of insulin-like growth factor-I (IGF-I). To further explore the renal uptake and processing of IGF-I, a study was carried out with the proximal-like cultured opossum kidney (OK) cell line. [125I]IGF-I associated with these cells in a specific manner. Association was competitively inhibited by IGF-I. Des(1-3)-IGF-I was equally effective, insulin had only a small effect, and the unrelated peptides, glucagon and GH, were without effect. Degradation was inhibited in similar manner. Comparisons of [125I]IGF-I with [125I]insulin revealed comparable cell association, but degradation of internalized IGF-I was several-fold slower. Furthermore, IGF-I degradation was less sensitive, by half, to the inhibitory effect of chloroquine. When OK cells were exposed to [125I]IGF-I in the presence of IGF-binding protein-3 (IGFBP-3) cell association (binding and internalization) was reduced significantly. Of note, total cell degradation was reduced (P < 0.01), but the IGF-I that was internalized was degraded more rapidly than in control cells. Gel filtration and reverse phase HPLC revealed that the products of IGF-I degradation included large IGF-I-size intermediates in addition to trichloroacetic acid-soluble material. This product profile was not altered by IGFBP-3. Thus, as previously described for insulin, cultured OK cells possess specific IGF-I receptors and degrade internalized IGF-I. However, IGF-I processing differs from that of insulin, in that degradation is slower and relatively insensitive to competition by insulin. This study also shows that IGFBP-3 inhibits the binding and uptake of [125I]IGF-I by these kidney cells. However, once IGF-I is internalized, IGFBP-3 enhances degradation. Although the mechanism of this paradoxical action requires further study, analysis of the products of degradation suggests that the same enzymes are involved in IGF-I degradation regardless of whether IGFBP-3 is present.

    View details for Web of Science ID A1995QP11700003

    View details for PubMedID 7534695

  • SEQUENTIAL PROCESSING OF INSULIN BY CULTURED KIDNEY-CELLS ENDOCRINOLOGY Fawcett, J., Rabkin, R. 1995; 136 (1): 39-45

    Abstract

    It is now well established that in kidney, as in liver, endosomes participate in the degradation of insulin. Degradation in this compartment involves the action of the insulin-degrading enzyme or a similar enzyme with the formation of large intermediate products. The role of lysosomes is less clear, for although earlier studies suggested that they are the major or sole site of degradation, this has been increasing questioned. More recently, it has been concluded that endosomes, at least in liver, are the major site of insulin degradation and that classic lysosomes are only involved in the latter stages of degradation if at all. As intracellular insulin processing varies among cell types, we set out to examine directly the processing of insulin within cultured proximal-like opossum kidney cells. By means of analytical subcellular fractionation and reverse phase HPLC analysis of products, we established the following sequence of events. After internalization, [125I]A14-insulin is partially degraded in endosomes. The formed products together with intact insulin, which accounts for most of the radioactive material in the endosomes, are then directed to the lysosomal compartment, where degradation proceeds rapidly to completion. Bacitracin inhibited degradation in both compartments and, although not eliminating insulin trafficking, may impair the transfer of insulin from endosomes to lysosomes. This study establishes a major role for lysosomes in kidney cell insulin degradation.

    View details for Web of Science ID A1995QD58400007

    View details for PubMedID 7828556

  • RENAL TUBULAR CELL PROTEIN BREAKDOWN IN UNINEPHRECTOMIZED AND AMMONIUM CHLORIDE-LOADED RATS JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Shechter, P., Shi, J. D., Rabkin, R. 1994; 5 (5): 1201-1207

    Abstract

    Kidney enlargement after unilateral nephrectomy or the induction of a systemic acidosis with ammonium chloride is associated with an increase in kidney protein content. This reflects an imbalance between protein breakdown and protein synthesis. Because it has been shown in diabetic nephromegaly that depressed protein breakdown contributes to the increase in kidney protein content, this study examined whether altered protein breakdown is common to all forms of renal hypertrophy. Accordingly, protein turnover was measured in isolated proximal tubules from kidney in rats undergoing renal enlargement after uninephrectomy or chronic ammonium chloride-induced acidosis. In both conditions, kidney protein content and protein synthesis ([14C]valine incorporation) increased significantly. Fractional protein degradation was depressed in renal tubules isolated from the acidotic rats and was accompanied by a decrease in proximal tubule cathepsin B and combined B and L activities. These changes are comparable to earlier observations with the diabetic kidney. In contrast, after unilateral nephrectomy, protein breakdown is not reduced, and it can reasonably be concluded that, in this condition, protein gain reflects increased protein synthesis alone. It was concluded that the pattern of protein turnover leading to protein accretion in renal hypertrophy varies according to the initial stimulus for renal growth.

    View details for Web of Science ID A1994PU62400006

    View details for PubMedID 7873730

  • THE INTRARENAL IGF-I AXIS IN COMPENSATORY RENAL GROWTH Fervenza, F., Tsao, T., Hsu, F., Rabkin, R. AMER SOC NEPHROLOGY. 1994: 691–91
  • HETEROGENEITY OF IGF-I, IGF-I RECEPTOR AND IGF BINDING-PROTEIN GENE-EXPRESSION IN KIDNEY Rabkin, R., Brody, M., Lu, L., Chan, C., Gillett, N. A. AMER SOC NEPHROLOGY. 1994: 793–93
  • INSULIN-LIKE GROWTH-FACTOR-I GENE-EXPRESSION IN ACUTE TUBULAR-NECROSIS (ATN) Fervenza, F., Tsao, T., Hsu, F., Rabkin, R. AMER SOC NEPHROLOGY. 1994: 897–97
  • RECOVERY FROM ACUTE ISCHEMIC RENAL-FAILURE IS ACCELERATED BY DES(1-3)-INSULIN-LIKE GROWTH-FACTOR-I CLINICAL SCIENCE Clark, R., Mortensen, D., Rabkin, R. 1994; 86 (6): 709-714

    Abstract

    1. Acute renal failure carries a high risk of morbidity and mortality, so there is a need for agents that minimize renal injury after an insult and that hasten repair. Insulin-like growth factor-1 is mitogenic for renal tubular cells; in normal kidneys it has haemodynamic effects and it is potently anabolic. We tested the theory that insulin-like growth factor-1 may be of use in the treatment of acute renal failure by administering recombinant des-(1-3)-insulin-like growth factor-1, a truncated form of insulin-like growth factor-1, which occurs naturally. Ischaemic renal failure was induced in normal rats by occluding both renal pedicles for 60 min. Then des-(1-3)-insulin-like growth factor-1 (0.8 mg day-1 kg-1) or vehicle was given by subcutaneous minipump for 7 days. The rats were weighed and bled daily and in one experiment were housed in metabolic cages and urine was collected. 2. Des-(1-3)-insulin-like growth factor-1 caused a lower and earlier peak in both serum creatinine and blood urea-nitrogen levels, and a more rapid and complete return toward basal values than in untreated animals. Also des-(1-3)-insulin-like growth factor-1 significantly increased creatinine clearance and reduced fractional excretion of filtered sodium. Besides these beneficial effects on kidney function, des-(1-3)-insulin-like growth factor-1 was anabolic as treated rats gained weight while control rats lost weight. The mortality in control rats was 28% compared with 6% in treated rats.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1994NR97700009

    View details for PubMedID 8062506

  • EFFECTS OF FLUOXETINE ON MOOD AND IMMUNE STATUS IN DEPRESSED-PATIENTS WITH HIV ILLNESS JOURNAL OF CLINICAL PSYCHIATRY Rabkin, J. G., Rabkin, R., Wagner, G. 1994; 55 (3): 92-97

    Abstract

    When present, depression in the context of human immunodeficiency virus (HIV) illness not only detracts from quality of remaining life but may interfere with motivation to obtain good medical care as well, thus directly influencing life's duration. This study was undertaken to assess the efficacy of fluoxetine in treating depression occurring in the context of HIV illness and to assess effects, if any, of fluoxetine on immune status (T-cell subsets).Patients had originally participated in a double-blind, placebo-controlled, 6-week study of imipramine. Imipramine nonresponders or relapsers and side effect dropouts were offered open treatment with fluoxetine for 12 weeks. Adjunctive dextroamphetamine was prescribed if the treating psychiatrist considered it clinically indicated. Eligibility criteria for the original study included a DSM-III-R diagnosis of major depression, dysthymia, or both. Concurrent HIV medications were permitted.Measures included the clinician-rated Hamilton Rating Scale for Depression and Clinical Global Impressions Scale, and patient-rated Brief Symptom Inventory and Beck Hopelessness Scale. Of the 23 patients receiving only fluoxetine, 83% (N = 19) were classified as responders. Of the 7 (30%) who also received adjunctive dextroamphetamine, all responded. Patients with CD4 cell counts under 200/cu mm did as well as others. CD4 cell count was not influenced by duration of treatment with fluoxetine; the average decline was that expected due to the passage of time alone. Side effects were mild and relatively infrequent.In this open treatment study, fluoxetine alone and fluoxetine plus dextroamphetamine were found to be effective treatments for patients with HIV illness and Axis I depression, regardless of the initial level of immune deficiency or number or type of HIV medications used concurrently. No negative effects on immune status were observed.

    View details for Web of Science ID A1994NF70500003

    View details for PubMedID 7915270

  • TUBULAR CELL PROTEIN-DEGRADATION IN EARLY DIABETIC RENAL HYPERTROPHY JOURNAL OF THE AMERICAN SOCIETY OF NEPHROLOGY Shechter, P., Boner, G., Rabkin, R. 1994; 4 (8): 1582-1587

    Abstract

    Renal hypertrophy in diabetes is accompanied by an increase in kidney protein content, which reflects an imbalance between protein synthesis and degradation. This study determines whether altered cellular protein degradation contributes to the imbalance. Diabetes was induced in rats with streptozotocin (55 mg/kg/ip). After 2 or 4 days of diabetes, kidney weight and protein content were measured. Over the 4 days, despite a loss in body weight, kidney wet weight increased by 35% and protein content by 37% in the diabetic rats. Treatment with insulin prevented this increase. Long-lived protein degradation was measured in isolated proximal tubules prelabeled with (14C)valine in vivo. Two days after streptozotocin, protein degradation was depressed by 19% (P < 0.05) and by the fourth day by 27% compared with that in nondiabetic controls (2.6% +/- 0.2 versus 1.9 +/- 0.1% degraded/h; P < 0.01). This was accompanied by a similar diabetes-induced decrease in proximal tubule cathepsin B and L activity. Accordingly, this study provides direct evidence that, in diabetes, tubular cell protein breakdown is depressed and suggests that altered lysosomal cathepsin activity may contribute to this effect. Depressed proteolysis likely contributes to the increase in kidney protein content and hence to diabetic renal hypertrophy.

    View details for Web of Science ID A1994MZ64000010

    View details for PubMedID 8025232

  • DEGRADATION OF INSULIN BY ISOLATED RAT RENAL CORTICAL ENDOSOMES ENDOCRINOLOGY Fawcett, J., Rabkin, R. 1993; 133 (4): 1539-1547

    Abstract

    It has been widely accepted that in kidney, degradation of insulin occurs in lysosomes. It is thought that after internalization into the cell, insulin dissociates from its receptor, which then recycles to the plasma membrane, while the hormone is transported in endosomes to the lysosomes, where it is degraded. However, earlier studies from this laboratory have suggested that insulin may also be degraded in an extralysosomal site, most likely endosomes. Indeed, studies in other tissues, most notably liver, have shown that insulin degradation does take place in endosomes. Since the intracellular processing of insulin differs between different tissues and cell types, and as the kidney is a major site of insulin degradation, we set out to determine directly whether endosomes degrade internalized insulin in the kidney. Rats were injected with [125I]monoiodoinsulin, labeled at either the A14 or B26 tyrosine. After killing, the kidney cortex was excised, and heavy endosomes were prepared by differential and isopycnic centrifugation. The isolated [125I]insulin-loaded endosomes were incubated for up to 60 min in intracellular medium, and degradation of [125I] insulin was estimated by means of precipitation in trichloroacetic acid. In the presence of ATP (10 mM), the percent degraded was increased over the control value (no ATP present), but under these circumstances, degradation was greater when the endosomes contained internalized 125I-labeled [B26]insulin than with A14-labeled [125I]insulin (26% vs. 13% degraded/h). In the absence of ATP, the percent degraded increased when the pH of the incubation medium was lowered. Radiolabeled material was extracted from endosomes, and Sephadex G-50 analysis revealed the presence of high mol wt, insulin-size, and low mol wt material. Reverse phase HPLC analysis of the insulin-size material revealed the presence of intact insulin and a number of degradation products. The elution profiles of some of these products were consistent with that reported to arise from the action of the insulin-degrading enzyme. Western blot analysis with the antiinsulin-degrading enzyme monoclonal antibody 9B12 confirmed the presence of the enzyme in endosomal preparations. We conclude that degradation of insulin does occur in kidney cortical endosomes, probably involves the insulin-degrading enzyme, and results in the formation of relatively large intermediate products as well as low mol wt products.

    View details for Web of Science ID A1993MA41100008

    View details for PubMedID 8404592

  • INSULIN UPTAKE AND PROCESSING BY CULTURED MOUSE GLOMERULAR ENDOTHELIAL-CELLS AMERICAN JOURNAL OF PHYSIOLOGY Rabkin, R., Tsao, T., Elliot, S. J., Striker, L. J., Striker, G. E. 1993; 265 (2): C453-C459

    Abstract

    Endothelial cells isolated from a variety of vascular beds bind and transport insulin but exhibit relatively low insulin degrading activity. Because endothelial cells exhibit heterogeneity and since kidney is a major site of insulin degradation, we studied the processing of insulin by glomerular endothelial cells (GEC). When exposed to 2 x 10(-10) M 125I-labeled insulin, GEC associated with the hormone in a specific manner. This interaction was inhibited by insulin but not by a number of unrelated peptide hormones. Over a 90-min period, GEC degraded 42 +/- 3% of the 125I-insulin, as measured by solubility in trichloroacetic acid (TCA). Degradation was inhibited 90% by an excess of insulin or adrenocorticotropic hormone (10(-6) M) and 57% by glucagon, whereas growth hormone and calcitonin were without effect. Separation of plasma membrane bound from internalized insulin was achieved by decreasing extracellular pH. In the steady state, 43% of cell-associated insulin was membrane bound and 57% internalized. The fate of the internalized 125I-insulin was examined by incubating acid-washed cells at 37 degrees C for 60 min. Over this time 18% of the radioactivity was released as TCA insoluble- and 72% as TCA-soluble radioactivity. Release was increased by insulin (10(-6) M) but not by unrelated peptide hormones. In the presence of chloroquine, 125I-insulin release increased by one third while degradation fell. High-performance liquid chromatography revealed that GEC released both intact insulin and large intermediates and that chloroquine inhibited intermediate formation.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1993LW03100019

    View details for PubMedID 8368274

  • AMMONIUM-CHLORIDE ALTERS RENAL TUBULAR CELL-GROWTH AND PROTEIN-TURNOVER LABORATORY INVESTIGATION Rabkin, R., PALATHUMPAT, M., Tsao, T. 1993; 68 (4): 427-438

    Abstract

    Since renal hypertrophy occurs in conditions associated with increased renal ammonia genesis it has been suggested that ammonia may stimulate renal growth. Indeed, quiescent cultured tubular epithelial cells exposed to NH4Cl undergo hypertrophy. However, ammonia inhibits liver regeneration in vivo and proliferation of cultured fibroblasts. This study was designed to evaluate the effect of ammonia (NH3/NH4+) as a regulator of kidney cell hypertrophy and also kidney cell proliferation.Cultured opossum kidney cells and primary rabbit proximal tubular epithelial cells were grown with or without (controls) NH4Cl present.After 3 days exposure to 5-20 mM NH4Cl, there was a dose-dependent depression of cell replication that ranged between 8 and 63% compared with controls (p < 0.017). In contrast, cell volume and protein content were significantly greater in the NH4Cl-treated cells. At 20 mM NH4Cl the protein content of treated cells exceeded that of controls by as much of 75%. This difference in protein content could, in part, be related to the disparity in cell density. However, experiments performed with cells at similar density revealed that NH4Cl also has a direct effect on cell protein content that increased by 25%; this appeared to be a consequence of depressed protein breakdown and was not due to altered protein synthesis. Experiments with rabbit kidney cells revealed that inhibition of replication was associated with a decrease in DNA [3H]thymidine incorporation. Cell cycle analysis revealed a fall in the proportion of cells in the S + G2 + M phase compared with controls (22 versus 30%, respectively; p < 0.01). NH4Cl also inhibited the burst of replication that followed chemically induced hypoxic injury of quiescent opossum kidney cells.We conclude that in addition to inducing hypertrophy, NH4Cl can inhibit tubular cell proliferation. Thus, while heightened ammoniagenesis in vivo may favor hypertrophy, this in vitro study raises the question whether an elevated intrarenal ammonia content might be harmful when cell replication is required. Acute tubular necrosis is a condition in which elevated ammonia levels and a requirement for cell replication coexist and could serve as an important model to study this question.

    View details for Web of Science ID A1993KY59200008

    View details for PubMedID 8479151

  • ENDOSOMES DEGRADE INSULIN 7TH INTERNATIONAL SYMP ON NEPHROLOGY Fawcett, J., Rabkin, R. KARGER. 1993: 61–65

    View details for Web of Science ID A1993BX78S00011

    View details for PubMedID 8467689

  • RENAL CORTICAL ENDOSOMES PARTICIPATE IN THE DEGRADATION OF INSULIN 6TH INTERNATIONAL CONF IN RENAL NUTRITION AND METABOLISM Fawcett, J., Frank, B., Rabkin, R. KARGER. 1992: 80–83

    Abstract

    It has long been thought that metabolism of insulin by kidney proximal tubule cells takes place only in lysosomes. But previous studies with the perfused rat kidney and a proximal tubule-like cultured kidney cell line suggested that insulin degradation occurred in a nonlysosomal compartment. In this study we show that endosomes isolated from rat kidney cortex degrade insulin and that this process is ATP and pH dependent.

    View details for Web of Science ID A1992JY99600003

    View details for PubMedID 1334526

  • AMMONIUM-CHLORIDE INCREASES KIDNEY-CELL PROTEIN-CONTENT 6TH INTERNATIONAL CONF IN RENAL NUTRITION AND METABOLISM Dahl, D. C., Tsao, T., Rabkin, R. KARGER. 1992: 104–7

    Abstract

    Augmented renal ammoniagenesis and renal hypertrophy often occur together. Ammonia may increase cell protein content by modulating protein synthesis, protein degradation, or both. We conducted experiments to examine the effect of ammonium chloride on the synthesis and degradation of protein in cultured kidney cells. Quiescent opossum kidney cells were exposed to 20 mM NH4Cl for two days. Monolayers were then analyzed for cell number, protein content, protein synthesis ([14C]valine incorporation), protein degradation ([14C]valine release) and DNA synthesis ([3H]thymidine incorporation). Cell protein content was increased by 18% in NH4Cl-treated cells (190 +/- 6 pg/cell control vs. 225 +/- 7 pg/cell NH4Cl, p < 0.001). NH4Cl suppressed protein degradation (1.36 +/- 0.02%/h control vs. 1.12 +/- 0.04%/h NH4Cl, p < 0.001) but did not change protein synthesis, DNA synthesis, or cell number. Thus, reduced protein degradation accounts entirely for enhanced protein accumulation at 2 days in this in vitro model of kidney cell hypertrophy.

    View details for Web of Science ID A1992JY99600009

    View details for PubMedID 1465042

  • ALTERED INTESTINAL AND RENAL BRUSH-BORDER AMINO-OLIGOPEPTIDASE STRUCTURE IN DIABETES AND METABOLIC-ACIDOSIS - NORMAL AND BIOBREED (BB) RATS METABOLISM-CLINICAL AND EXPERIMENTAL Najjar, S. M., Hampp, L. T., Rabkin, R., Gray, G. M. 1992; 41 (1): 76-84

    Abstract

    Amino-oligopeptidase (AOP, aminopeptidase N), a major glycoprotein hydrolase in intestinal and kidney brush border membranes, plays a crucial role in digesting peptide nutrients and salvaging filtered peptides. The molecular structure of rat intestinal and kidney AOP was compared for normal Wistar and congenitally diabetic BB Wistar (BBd) rats. Brush border membranes were isolated, solubilized with Triton X-100, and the AOP specifically immunoprecipitated with polyvalent rabbit antiserum and analyzed on 7% sodium dodecyl sulfate (SDS)-acrylamide electrophoresis. While the specific hydrolytic activity was maintained, BBd rats displayed an altered migration of AOP on SDS gels. Intestinal AOP migrated as a smaller species (130 kd) in the BBd than in the normal Wistar (135 to 140 kd). In some BBd rats, additional intestinal AOP species were observed (a 130- to 135-kd doublet or a 125-, 130-, or 135-kd triplet). Kidney AOP migrated as a broader band (125 to 140 kd) than intestine for all rat groups, probably due to carbohydrate chain heterogeneity, and was approximately 5 kd smaller in the BBd rat than in the normal Wistar. In contrast, no mass change was found in diabetes induced by streptozotocin (STZ). The altered intestinal AOP in the BBd rat was present when first inserted into the brush border membrane (6 hours after intraperitoneal [35S]methionine labeling), and hence was not due to nonenzymatic glycosylation (NEG). Abnormal intestinal and kidney AOP structure appeared in early diabetes, irrespective of high plasma glucose levels or ketoacidosis, and was reversed following evolution of the diabetes under prolonged (21 to 120 days) insulin treatment.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1992GZ10700014

    View details for PubMedID 1538646

  • AMINO-ACIDS REGULATE KIDNEY-CELL PROTEIN BREAKDOWN JOURNAL OF LABORATORY AND CLINICAL MEDICINE Rabkin, R., Tsao, T., Shi, J. D., Mortimore, G. 1991; 117 (6): 505-513

    Abstract

    Amino acids inhibit breakdown of long-lived intracellular proteins in some but not all tissues studied. Because no information is available relating to the effect of amino acids on kidney cell proteolysis, this study was conducted with cultured proximal-like opossum kidney (OK) cells and primary cultured rabbit proximal tubular cells in which long-lived cell proteins were labeled with carbon 14-labeled valine. These cultured cells were acutely deprived of amino acids; this was followed by a 57% to 66% increase in the proteolytic rate in OK cells and a 22% rate increase in the rabbit kidney cells. In cultured OK cells incubated in serum-free minimal essential medium containing 13 amino acids, proteolysis averaged 4.62% +/- 0.28%/2 hr and increased to 7.66% +/- 0.38%/2 hr when amino acids were deleted. Each amino acid was then added alone. Leucine, phenylalanine, and lysine had significant effects in inhibiting the deprivation response by 40%, 26%, and 22%, respectively. Leucine appears to inhibit proteolysis directly and not through its metabolites, since alpha-ketoisocaproate, the leucine transamination product, was without effect. Similarly, failure of tyrosine to inhibit proteolysis suggests a direct phenylalanine action. When leucine, phenylalanine, and lysine were simultaneously deleted from the incubation medium, the increase in proteolysis corresponded to 56% of the response after deletion of all amino acids. Thus to maximally affect proteolysis, amino acids, which on their own have little effect on protein breakdown, also appear to play a role. From this study we conclude that amino acids seem to play an important and direct role in the regulation of kidney epithelial cell protein breakdown.

    View details for Web of Science ID A1991FQ78100012

    View details for PubMedID 1904473

  • SUCRASE-ALPHA-DEXTRINASE IN DIABETIC BIOBREED RATS - REVERSIBLE ALTERATION OF SUBUNIT STRUCTURE AMERICAN JOURNAL OF PHYSIOLOGY Najjar, S. M., Hampp, L. T., Rabkin, R., Gray, G. M. 1991; 260 (2): G275-G283

    Abstract

    BioBreed (BB) Wistar rats develop diabetes mellitus, which closely resembles the human disease, in 50% of progeny. Intestinal sucrase-alpha-dextrinase, a glycoprotein hydrolase of the enterocyte's brush border consisting of 140-kDa alpha-dextrinase and 125-kDa sucrase subunits, is essential for surface digestion of carbohydrate nutrients. Although its catalytic characteristics were found to be maintained in the diabetic state, the structure of the subunits, as compared with normal Wistar rats, was altered in the BB rat within 2 days of the onset of diabetes. Its capacity to react in a solid-phase immunoassay was reduced by 50%; when examined by 6% acrylamide electrophoresis, the sucrase subunit was increased in mass by 5 kDa and, in some BB rats, the dextrinase subunit was reduced by 5 kDa. Intact rats labeled intraintestinally with [35S]methionine displayed the alteration within 6 h of synthesis, indicating that nonenzymatic glycosylation could not account for the structural change. This mass change was not seen in streptozotocin-induced diabetes and was independent of the plasma glucose concentration or the degree of acidosis. Deglycosylation with peptide N-glycosidase indicated that the N-linked chains of the normal dextrinase subunit (11 kDa) have twice the mass of those in the BB rat (6 kDa) and that the sucrase subunit may have an increased mass of O-linked chains. Overall, these experiments point to changes in glycosylation as a mechanism of structural alteration in congenital diabetes. Despite persistence of the insulin-dependent diabetes, the subunit pattern eventually became indistinguishable from normal, but at differential rates (21 days and 35 days, respectively, for sucrase and dextrinase subunits).

    View details for PubMedID 1996646

  • EFFECT OF BACITRACIN ON RETROENDOCYTOSIS AND DEGRADATION OF INSULIN IN CULTURED KIDNEY EPITHELIAL-CELL LINE DIABETES Dahl, D. C., Tsao, T., Duckworth, W. C., Frank, B. H., Rabkin, R. 1990; 39 (11): 1339-1346

    Abstract

    In an earlier study, we described the presence of a retroendocytotic pathway for insulin in a cultured kidney epithelial cell line. Derived from the opossum kidney (OK), these cells possess many features of proximal tubule epithelium, which is the major site of kidney insulin metabolism. We studied the interaction between the retroendocytotic and the degradative pathways with bacitracin as a pharmacological probe. Monolayers of OK cells were loaded with 125I-labeled insulin over 30 min, acid washed to remove membrane-bound insulin, then incubated in fresh medium for 60 min while the release of intracellular radioactivity was monitored. In experiments carried out in the presence of bacitracin (2 mM), there was a two-thirds increase in intracellular radioactivity at the end of the loading phase. Measurements made during the subsequent release phase showed that bacitracin reduced the release of degradation products. Thus, although controls released 72.1 +/- 8.1% of the internalized radioactivity as trichloroacetic acid (TCA)-soluble products, bacitracin-treated cells released 59.2 +/- 9.4% (P less than 0.02). In contrast, release of TCA-precipitable insulin increased from 15.2 +/- 4.6% in controls to 25.8 +/- 3.7% in bacitracin-treated cells (P less than 0.01). In separate experiments analyzed by gel-exclusion chromatography, 6.4 +/- 0.6% of radioactivity released from preloaded control cells into medium over 60 min was insulin sized compared to 29.7 +/- 1.4% in bacitracin-treated cells. High-performance liquid chromatography revealed that 61.5 +/- 3.5% of this insulin-sized material released from control cells preloaded with A14-insulin eluted as intact insulin and the remainder as unidentified intermediate degradation products.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1990EE88700005

    View details for PubMedID 2227108

  • MODULATION OF KIDNEY-CELL PROTEIN-DEGRADATION BY INSULIN JOURNAL OF LABORATORY AND CLINICAL MEDICINE Tsao, T. C., Shi, J. D., Mortimore, G. E., Cragoe, E. J., Rabkin, R. 1990; 116 (3): 369-376

    Abstract

    Insulin is an established regulator of intracellular proteolysis in several mammalian tissues but little is known about its role in the kidney. The present study was undertaken to determine whether insulin influences protein degradation in isolated rat renal proximal tubules and to investigate its mechanism of action in cultured proximal-like tubular epithelial cells from the opossum kidney. Long-lived protein degradation was determined from the release of carbon 14-labeled valine from previously labeled cellular protein under conditions designed to minimize label reutilization. In isolated tubules, the mean control rate of proteolysis was 2.18% per hour, indicating an appreciable turnover of cellular protein. Insulin (10(-6) mol/L) decreased the rate by 23%. In cultured kidney cells, the rate of protein degradation averaged 1.25% per hour in the presence of serum and 1.68% per hour in its absence, an increase of 34%. High insulin concentrations suppressed this acceleration completely, and physiologic levels inhibited it partially. No evidence was obtained to indicate that insulin action is mediated through stimulation of Na(+)-H+ antiport or through increased amino acid utilization. Ammonium chloride, however, strongly attenuated the serum deprivation response and the inhibitory effect of insulin. The exact mechanisms whereby insulin inhibits proteolysis is not known, but these findings are consistent with an inhibitory action of insulin on the lysosomal pathway.

    View details for Web of Science ID A1990DZ68800014

    View details for PubMedID 2169515

  • FACTORS CONTROLLING INTRACELLULAR PROTEIN-TURNOVER IN THE KIDNEY SEMINARS IN NEPHROLOGY Rabkin, R., Dahl, D. C. 1990; 10 (5): 472-480

    View details for Web of Science ID A1990EA33000006

    View details for PubMedID 2236989

  • ALTERNATE PATHWAYS FOR THE RENAL PROCESSING OF INSULIN 6TH INTERNATIONAL SYMP OF NEPHROLOGY : KIDNEY, PROTEINS, AND DRUGS Dahl, D. C., Tsao, T., Rabkin, R. KARGER. 1990: 53–59

    View details for Web of Science ID A1990BS89A00009

    View details for PubMedID 2100722

  • BASOLATERAL AND APICAL BINDING, INTERNALIZATION, AND DEGRADATION OF INSULIN BY CULTURED KIDNEY EPITHELIAL-CELLS AMERICAN JOURNAL OF PHYSIOLOGY Rabkin, R., Yagil, C., Frank, B. 1989; 257 (6): E895-E902

    Abstract

    In vivo, filtered insulin is absorbed and degraded in proximal tubules after binding to the apical membrane. Peritubular removal also occurs and involves basolateral receptor binding and degradation. Whether basolateral degradation proceeds within the cell or on the cell surface is unknown. Because of the difficulties in addressing this question in vivo, this study was carried out with a cultured opossum kidney epithelium cell line with proximal-like features and insulin receptors. Cells were grown in partitioned wells on polycarbonate filters and, when confluent, the monolayer effectively separated the culture well into apical and basolateral compartments. Apical and basolateral binding, internalization, and degradation were studied separately by incubating monolayers with 125I-insulin added to either the apical or basal compartment. At 37 degrees C insulin associated with either pole in a time-dependent manner. This interaction was specific, for it was competitively inhibited by cold insulin but not by unrelated peptides. Separation of surface-bound from internalized insulin was achieved by lowering extracellular pH. At 4 degrees C, 92% of the radioactivity added to either side of the monolayer was surface-bound, whereas at 37 degrees C and after 1 h, 57% was surface-bound and 43% internalized. Affinity of apical and basolateral receptors were similar (1-2 nM), but basolateral receptor number was greater, for at high insulin concentrations (5 x 10(-8) M) basolateral membrane binding exceeded apical by fivefold (250 +/- 81 vs. 56 +/- 11 fm/10(6) cells). Degradation followed exposure to either pole of the cell.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1989CG26200038

    View details for PubMedID 2558575

  • Protein catabolism in cultured kidney cells. Kidney international. Supplement Rabkin, R., Dahl, D. C., Mahoney, C. A., Tsao, T. 1989; 27: S11-4

    View details for PubMedID 2699993

  • PROTEIN CATABOLISM IN CULTURED KIDNEY-CELLS KIDNEY INTERNATIONAL Rabkin, R., Dahl, D. C., Mahoney, C. A., Tsao, T. 1989; 36: S11-S14
  • ABNORMAL INSULIN METABOLISM BY SPECIFIC ORGANS FROM RATS WITH SPONTANEOUS HYPERTENSION AMERICAN JOURNAL OF PHYSIOLOGY Mondon, C. E., Reaven, G. M., Azhar, S., Lee, C. M., Rabkin, R. 1989; 257 (4): E491-E498

    Abstract

    Spontaneously hypertensive rats (SHR) have been shown to be both insulin resistant and hyperinsulinemic after oral glucose administration or infusion of exogenous insulin during an insulin suppression test. To determine if this hyperinsulinemia may be due to decreased removal of insulin, the metabolic clearance (k) of insulin was measured in isolated perfused liver, kidney, and hindlimb skeletal muscle from SHR and Wistar-Kyoto (WKY) control rats. The data indicate that the k for insulin removal by liver was similar in SHR and WKY rats, averaging 287 +/- 18 and 271 +/- 10 microliters.min-1.g-1 liver, respectively. In contrast, the k for insulin removal by hindlimbs from SHR was decreased 37% (P less than 0.001) compared with WKY rats (8.6 +/- 0.5 vs. 13.7 +/- 0.7 microliters.min-1.g-1 muscle), and this decrease was not accompanied by decreased binding of insulin to its receptor in plantaris muscle. Although the removal of insulin by glomerular filtration was similar in SHR and WKY rats (653 +/- 64 microliters/min vs. 665 +/- 90 microliters.min-1.kidney-1), total insulin removal by kidney was significantly lower (P less than 0.05) in SHR (710 +/- 78 microliters/min) compared with WKY rats (962 +/- 67 microliters/min), due to decreased peritubular clearance of insulin in SHR (56 +/- 73 vs. 297 +/- 59 microliters/min, P less than 0.05). These findings suggest that the decreased clearance of insulin in SHR rats was possibly not due to impaired hepatic removal of insulin but rather to decreased removal by skeletal muscle and kidneys.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1989CC64500037

    View details for PubMedID 2679128

  • RETROENDOCYTOSIS OF INSULIN IN A CULTURED KIDNEY EPITHELIAL-CELL LINE AMERICAN JOURNAL OF PHYSIOLOGY Dahl, D. C., Tsao, T., Duckworth, W. C., Mahoney, M. J., Rabkin, R. 1989; 257 (2): C190-C196

    Abstract

    It has been generally accepted that in renal tubular epithelium endocytosed proteohormones are transported to lysosomes where they undergo complete hydrolysis. En route, as endosomal pH falls, the proteohormone uncouples from the endocytosed membrane binding site, which recycles to the cell surface. However, studies in other tissues have uncovered alternate intracellular pathways for proteins. One such pathway is retroendocytosis (endocytosis then exocytosis). To determine whether a retroendocytotic pathway exists for insulin in renal epithelium, a study was carried out with confluent monolayers of a proximal-like opossum kidney cell line that exhibits receptor-mediated endocytosis of insulin. Cells were preloaded with 125I-labeled insulin (4 X 10(-10) M) for 30 min, surface-bound insulin was then removed by acid washing, and over the next 60 min the release of intracellular radioactivity into the medium was monitored. At 37 degrees C, control cells released on average 7-15% of the intracellular radioactivity as intact insulin [trichloroacetic acid (TCA)-precipitable radioactivity] and approximately 62% as TCA-soluble degradation products. In the presence of 0.1 mM chloroquine (an acidotropic agent) the release of intact insulin increased approximately twofold while degradation fell by nearly one-half. With Sephadex G-50 chromatography we found that the released radioactivity included insulin-size material that increased in the presence of chloroquine. High-performance liquid chromatography revealed that 53 (controls) and 81% (chloroquine treatment) of this latter material consisted of intact insulin. We conclude that, in addition to a major degradative pathway, cultured kidney epithelial cells exhibit a retroendocytotic pathway for insulin. Chloroquine inhibits degradation and appears to divert insulin from the degradative into the retroendocytotic pathway.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1989AL12600006

    View details for PubMedID 2669506

  • INSULIN DEGRADATION PRODUCTS FROM PERFUSED RAT-KIDNEY AMERICAN JOURNAL OF PHYSIOLOGY Duckworth, W. C., Hamel, F. G., Liepnieks, J., PEAVY, D., Frank, B., Rabkin, R. 1989; 256 (2): E208-E214

    Abstract

    The kidney is a major site for insulin metabolism, but the enzymes involved and the products generated have not been established. To examine the products, we have perfused rat kidneys with insulin specifically iodinated on either the A14 or the B26 tyrosine. Labeled material from both the perfusate and kidney extract was examined by Sephadex G50 and high-performance liquid chromatography (HPLC). In perfusate from a filtering kidney, 22% of the insulin-sized material was not intact insulin on HPLC. With the nonfiltering kidney, 10.6% was not intact insulin. Labeled material from HPLC was sulfitolyzed and reinjected on HPLC. By use of 125I-iodo(A14)-insulin, almost all the degradation products contained an intact A-chain. By use of 125I-iodo(B26)-insulin, several different B-chain-cleaved products were obtained. The material extracted from the perfused kidney was different from perfusate products but similar to intracellular products from hepatocytes, suggesting that cellular metabolism by kidney and liver are similar. The major intracellular product had characteristics consistent with a cleavage between the B16 and B17 amino acids. This product and several of the perfusate products are also produced by insulin protease suggesting that this enzyme is involved in the degradation of insulin by kidney.

    View details for Web of Science ID A1989T343500035

    View details for PubMedID 2645781

  • HIGH-PERFORMANCE LIQUID-CHROMATOGRAPHIC ANALYSIS OF INSULIN DEGRADATION PRODUCTS FROM A CULTURED KIDNEY-CELL LINE ENDOCRINOLOGY Duckworth, W. C., Hamel, F. G., Liepnieks, J., Frank, B. H., Yagil, C., Rabkin, R. 1988; 123 (6): 2701-2708

    Abstract

    The kidney is a major site for insulin removal and degradation, but the subcellular processes and enzymes involved have not been established. We have examined this process by analyzing insulin degradation products by HPLC. Monoiodoinsulin specifically labeled on either the A14 or B26 tyrosine residue was incubated with a cultured kidney epithelial cell line, and both intracellular and extracellular products were examined on HPLC. The products were then compared with products of known structure generated by hepatocytes and the enzyme insulin protease. Intracellular and extracellular products were different, suggesting two different degradative pathways, as previously shown in liver. The extracellular degradation products eluted from HPLC both before and after sulfitolysis similarly with hepatocyte products and products generated by insulin protease. The intracellular products also eluted identically with hepatocyte products. Based on comparisons with identified products, the kidney cell generates two fragments from the A chain of intact insulin, one with a cleavage at A13-A14 and the other at A14-A15. The B chain of intact insulin is cleaved in a number of different sites, resulting in peptides that elute identically with B chain peptides cleaved at B9-B10, B13-B14, B16-B17, B24-B25, and B25-B26. These similarities with hepatocytes and insulin protease suggest that liver and kidney have similar mechanisms for insulin degradation and that insulin protease or a very similar enzyme is involved in both tissues.

    View details for Web of Science ID A1988R218000015

    View details for PubMedID 3058457

  • EFFECT OF INSULIN THERAPY ON ESTABLISHED DIABETIC NEPHROPATHY IN RATS DIABETES Petersen, J., Ross, J., Rabkin, R. 1988; 37 (10): 1346-1350

    Abstract

    The effectiveness of insulin therapy on early diabetic nephropathy has not been established. In this study we examined the influence of continuous subcutaneous insulin on the progression of established nephropathy in streptozocin-induced diabetic rats. Normal controls and diabetic rats were studied for 11 mo. During the first 6 mo, all the diabetic rats received 2 U protamine zinc insulin s.c. twice weekly. During the last 5 mo of study, diabetic rats either continued on the occasional subcutaneous insulin regimen or received regular insulin by continuous subcutaneous infusion. Six months after the initiation of the study, the diabetic rats were severely hyperglycemic, and their relative mesangial areas had increased. Continued poor glycemic control in the rats receiving occasional insulin was associated with relative increased mesangial area (25.2 +/- 1.0% of glomerular area) and significant proteinuria (148 +/- 17 mg/24 h) compared with normal controls. In contrast, the use of continuous subcutaneous insulin therapy with improved glycemic control arrested mesangial changes (19.5 +/- 1.4% of glomerular area) and prevented the excessive proteinuria (71 +/- 13 mg/24 h). Indeed, these parameters did not differ from age-matched controls. We conclude that in the rat, continuous subcutaneous insulin therapy instituted after the development of early glomerular pathology is effective in arresting the disease process.

    View details for Web of Science ID A1988Q291300007

    View details for PubMedID 2970981

  • EFFECT OF BACITRACIN ON BINDING AND PROCESSING OF INSULIN BY ESTABLISHED RENAL-CELL LINE DIABETES Yagil, C., Frank, B. H., Rabkin, R. 1988; 37 (6): 800-805

    Abstract

    The effect of bacitracin on the binding and processing of 125I-labeled insulin was studied in a proximal tubular epithelium-like opossum kidney cell line. This cultured cell line handles insulin in a manner comparable to the in vivo situation, which requires membrane binding, internalization, and intracellular degradation. The addition of bacitracin inhibited insulin degradation significantly and delayed the time of appearance of products in the medium (22 min) compared with control cells (14 min). Maximum total cell-associated radioactivity increased from 1.5 +/- 0.19% in the control cells to 2.5 +/- 0.17% in the treated cells. Separation of cell membrane from internalized radioactivity was achieved by acid washing and showed no change in membrane-bound radioactivity or rate of internalization, but a significant increase in intracellular radioactivity was noted. Gel-filtration chromatography revealed that this was due to an accumulation of chromatographically intact insulin. Accordingly, we conclude that bacitracin inhibits insulin degradation in cultured kidney cells by perturbing the intracellular processing of insulin, not by altering the binding or internalization of the hormone or by inhibiting the release of small degradation products. Because of the multiple actions of this agent, the exact site in these kidney cells at which intracellular degradation is inhibited remains to be established. However, in contrast to studies with lysosomes isolated from cells of other tissues, this study showed that when lysosomes isolated from rat renal cortex were exposed to bacitracin, insulin degradation was inhibited markedly (81%).

    View details for Web of Science ID A1988N605900020

    View details for PubMedID 3290001

  • INTERNALIZATION AND CATABOLISM OF INSULIN BY AN ESTABLISHED RENAL-CELL LINE AMERICAN JOURNAL OF PHYSIOLOGY Yagil, C., Frank, B. H., Rabkin, R. 1988; 254 (6): C822-C828

    Abstract

    Proximal renal tubules are a key site of insulin metabolism. To explore the kinetics and metabolic requirements of insulin internalization and catabolism, we used the opossum kidney cell line, which has proximal tubular-like features and possesses insulin-specific receptors. Internalization was determined by separating membrane-bound insulin from intracellular insulin by exposure to an acidified medium. Internalization of membrane-bound insulin was rapid, and half-maximal internalization occurred within 2.5 min. Degradation products did not accumulate in the cell but appeared in the medium after a delay of 5 min from the onset of internalization. In other experiments, addition of KCN (2 mM) or omission of glucose did not alter degradation, but KCN, combined with the omission of glucose, inhibited degradation by 64%. This was associated with a 240% increase in membrane-bound insulin and an 81% decrease in intracellular insulin. Accordingly, it appears that under these circumstances impaired degradation was a consequence of impaired internalization. In contrast, although 0.1 mM chloroquine, an endosomal-lysosomal inhibitor, also depressed degradation (by 57%), intracellular insulin increased fourfold, indicating failure of intracellular processing. We conclude that these cultured kidney cells rapidly internalize and degrade insulin and that internalization, a prerequisite for degradation, is dependent on energy that can be derived from anaerobic glycolysis or oxidative metabolism. Furthermore, the intracellular degradative processing of insulin by these cells involves a chloroquine-sensitive pathway.

    View details for Web of Science ID A1988N862400014

    View details for PubMedID 3287948

  • INSULIN BINDING, INTERNALIZATION, AND DEGRADATION BY A CULTURED KIDNEY-CELL LINE AMERICAN JOURNAL OF PHYSIOLOGY Yagil, C., Ehmann, U. K., Frank, B. H., Rabkin, R. 1988; 254 (5): E601-E608

    Abstract

    Proximal tubules are a key site of insulin metabolism, which is in part a receptor-mediated process. To explore the interaction between insulin and the kidney and to evaluate the role of receptors in insulin uptake and processing, a study was carried out with a cultured proximal-like opossum kidney (OK) cell line. 125I-insulin associated with confluent monolayers in a specific manner, and this interaction was competitively inhibited by insulin; unrelated peptides were relatively ineffective. Insulin degradation exhibited time and temperature dependency and up to a concentration of 5 X 10(-8) M was not saturable. Degradation exhibited partial hormone specificity. Separation of plasma membrane bound from internalized insulin was achieved by lowering extracellular pH. At 4 degrees C, 94% of cell-associated radioactivity was membrane bound, whereas at 37 degrees C, in the steady state, 33% was membrane bound and 67% was internalized. There was a significant correlation between membrane-bound insulin and the rate of degradation. These findings reveal that the binding and processing of insulin by the kidney cell line are compatible with the description of the uptake of filtered insulin by the proximal tubule in the intact kidney. Accordingly we conclude that this cell line provides a good model for studying renal epithelial uptake and metabolism of insulin.

    View details for Web of Science ID A1988N390700028

    View details for PubMedID 2834959

  • RENAL METABOLISM OF CALCITONIN AMERICAN JOURNAL OF PHYSIOLOGY Simmons, R. E., Hjelle, J. T., Mahoney, C., Deftos, L. J., Lisker, W., Kato, P., Rabkin, R. 1988; 254 (4): F593-F600

    Abstract

    The kidneys account for approximately two-thirds of the metabolism of calcitonin, but relatively little is known regarding the details thereof. To further characterize this process, we examined the renal handling and metabolism of human calcitonin (hCT) by the isolated perfused rat kidney. We also studied the degradation of radiolabeled salmon calcitonin (sCT) by subcellular fractions prepared from isolated rabbit proximal tubules. The total renal (organ) clearance of immunoreactive hCT by the isolated kidney was 1.96 +/- 0.18 ml/min. This was independent of the perfusate total calcium concentration from 5.5 to 10.2 mg/dl. Total renal clearance exceeded the glomerular filtration rate (GFR, 0.68 +/- 0.05 ml/min), indicating filtration-independent removal. Urinary calcitonin clearance as a fraction of GFR averaged 2.6%. Gel filtration chromatography of medium from isolated kidneys perfused with 125I-labeled sCT showed the principal degradation products to be low molecular weight forms eluting with monoiodotyrosine. Intermediate size products were not detected. In the subcellular fractionation experiments, when carried out at pH 5.0, calcitonin hydrolysis exclusively followed the activities of the lysosomal enzyme N-acetyl-beta-glucosaminidase. Typically, at pH 7.5, 42% of total degradation occurred in the region of the brush-border enzyme alanyl aminopeptidase and 29% occurred in the region of the cytosolic enzyme phosphoglucomutase. Although 9% of the calcitonin-degrading activity was associated with basolateral membrane fractions, most of this activity could be accounted for by the presence of brush-border membranes.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1988N029100074

    View details for PubMedID 2833122

  • DIFFERENCES IN RENAL METABOLISM OF INSULIN AND CYTOCHROME-C AMERICAN JOURNAL OF PHYSIOLOGY Herrman, J., Simmons, R. E., Frank, B. H., Rabkin, R. 1988; 254 (4): E419-E428

    Abstract

    Kidneys degrade small proteins such as cytochrome c (CYT c) by the classic lysosomal pathway. However, because alternate routes for the transport and degradation of protein hormones have been identified in other tissues, we set out to determine whether extralysosomal sites might participate in the renal degradation of insulin. First, we compared the effect of the lysosomal inhibitor NH4Cl on insulin and CYT c degradation by isolated perfused rat kidneys. After kidneys were loaded with radiolabeled proteins to allow for absorption and transport to lysosomes, degradation was measured in the presence or absence of inhibitors. Control kidneys degraded 45 +/- 1.5% of the trapped CYT c per hour, and this was inhibited 62 +/- 1.3% by NH4Cl. In contrast, 86 +/- 2.4% of the trapped insulin was degraded per hour, and this was inhibited 26 +/- 4% by NH4Cl. Next we followed the subcellular distribution of 125I-labeled insulin in kidneys exposed to 125I-labeled insulin in vivo or when isolated and perfused. Under both circumstances the distribution of insulin on a linear sucrose gradient differed from that of the lysosomal enzyme N-acetyl-beta-glucosaminidase. In contrast, [14CH3]CYT c, injected in vivo, distributed over a density similar to the lysosomal marker. Thus important differences exist between the renal metabolism of CYT c, which proceeds in lysosomes, and the renal metabolism of insulin. These include rate of degradation, sensitivity to NH4Cl, and subcellular sites of localization. Accordingly, we suggest that insulin degradation may occur, at least in part, in a different compartment from the classic lysosomal site of protein degradation.

    View details for Web of Science ID A1988N029000026

    View details for PubMedID 2833109

  • Intrarenal pathways of calcitonin degradation. Contributions to nephrology Mahoney, C. A., Simmons, R. E., Hjelle, J. T., Rabkin, R. 1988; 68: 12-18

    View details for PubMedID 3233989

  • HORMONE RECEPTOR INTERACTIONS - DISCUSSION KIDNEY INTERNATIONAL BRENNER, B. M., Schrier, R. W., Striker, G. E., Brown, D., Rabkin, R., Jamison, R. L., Hammerman, M. R., Seifter, J. L., Daniel, T. O., SKORECKI, C. L., FINE, L. G. 1987; 32: S61-S67
  • EFFECT OF EXPERIMENTAL DIABETES ON INSULIN BINDING BY RENAL BASOLATERAL MEMBRANES KIDNEY INTERNATIONAL Rabkin, R., Hirayama, P., Roth, R. A., Frank, B. H. 1986; 30 (3): 348-354

    Abstract

    Removal of insulin from the peritubular vessels involves binding of insulin to specific receptors in the basolateral membranes (BLM); this is followed by phosphorylation of the receptor which may mediate the actions of the hormone. In most tissues receptor number is regulated by plasma insulin levels and is increased in insulinopenic diabetics. To determine whether cortical BLM insulin receptors are similarly regulated, we studied insulin binding to receptors in BLM from normal control rats and rats with streptozotocin diabetes of varying severity. Specific binding of insulin did not differ between control and modestly insulinopenic diabetics but was increased significantly in the severely insulinopenic diabetics. Insulin treatment returned binding to normal. Scatchard analysis suggested an increase in the binding capacity of the severe diabetic BLM rather than an increase in affinity for insulin. This latter was confirmed by competitive experiments in which similar displacement curves were obtained with control and diabetic membranes. Insulin removed by glomerular filtration binds to specific receptors in the luminal membranes but unlike BLM receptors, phosphorylation of these luminal receptors has not been observed. To determine whether luminal and BLM receptors differ structurally, binding sites in both membranes were affinity labelled with 125I-insulin and the cross linking agent, disuccinimidyl suberate, and subjected to SDS-polyacrylamide gel electrophoresis in the presence of a reducing agent. Autoradiograms revealed that the major specifically labelled subunit in both membranes is a 135,000 Mr species which is more abundant in the BLM. We conclude that insulin receptors in cortical BLM respond to severe insulinopenic diabetes as do receptors in most other tissues.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1986D725200009

    View details for PubMedID 3537445

  • Mechanisms and consequences of proteinuria. Laboratory investigation; a journal of technical methods and pathology Kaysen, G. A., Myers, B. D., Couser, W. G., Rabkin, R., FELTS, J. M. 1986; 54 (5): 479-498

    Abstract

    The glomerulus is a complex structure containing a remarkable capillary bed which is freely permeable to water and solutes up to the size of inulin. Many small proteins are filtered, reabsorbed, and catabolized by the kidney; but most large proteins, such as albumin or immunoglobulins, are almost entirely excluded from the glomerular ultrafiltrate due to the charge-size permselectivity of the glomerular capillary basement membrane. These large proteins appear in the urine when diseases reduce the charge selectivity or result in the development of large pores in this membrane. The reabsorptive capacity of the renal tubules for these proteins is overwhelmed. Hypoalbuminemia results when increased synthetic and decreased catabolic rates of albumin fail to compensate for the urinary loss of the protein. The resulting decrease in serum oncotic pressure increases the flux of fluid out of systemic capillaries into the interstitial space, a process that increases lymphatic flow and returns the relatively protein-poor ultrafiltrate to the plasma compartment. Interstitial proteins are swept into the plasma by the increased lymphatic flow, leading to a depletion of the extravascular pool of albumin even more severe than the depletion of albumin in the plasma compartment. The rate of albumin synthesis is increased but not sufficiently to replace losses and restore the serum concentration to normal. The rate of albumin catabolism is decreased. This decrease from the normal catabolic rate is as important as the increased rate of albumin synthesis in maintenance of albumin homeostasis in nephrosis. Whereas the reduced serum oncotic pressure certainly contributes to edema formation, sodium retention may result from processes intrinsic to the kidney itself; and plasma volume may actually be expanded despite hypoalbuminemia. The hyperlipemia that occurs in nephrosis is due to a combined defect in lipoprotein metabolism: increased hepatic synthesis of VLDL and decreased removal of TG and highly atherogenic remnants of incompletely metabolized CMs. The defects in lipoprotein metabolism may in part be the end result of the urinary loss of highly negative-charged macromolecules of the mucopolysaccharide called orosomucoid, which carries with it heparan sulfate, and important cofactor for LPL.

    View details for PubMedID 3517485

  • INSULIN METABOLISM BY LIVER, MUSCLE, AND KIDNEYS FROM SPONTANEOUSLY DIABETIC RATS AMERICAN JOURNAL OF PHYSIOLOGY Rabkin, R., Reaven, G. M., Mondon, C. E. 1986; 250 (5): E530-E537

    Abstract

    The in vivo metabolism of insulin is a complex process in which liver, kidney, and muscle are major participants. In this study we evaluated the effect of spontaneous hyperglycemic nonketoacidotic diabetes (DH) and ketoacidotic diabetes (DKA) on insulin clearance and degradation by these organs. Livers, hindlimbs, and kidneys from nondiabetic controls and DH and DKA Bio-Breed rats were isolated and perfused with artificial media. Liver clearance of immunoreactive insulin (ml/min) was significantly higher in DH rats, 6.0 +/- 0.2, but significantly lower in DKA rats, 3.4 +/- 0.5, compared with controls, 4.6 +/- 0.2. Acidosis alone induced by ammonium chloride loading, did not impair liver insulin clearance (4.8 +/- 0.4 ml/min). Muscle responded differently to the diabetic state in that insulin clearance was not altered by DH and DKA. Renal (organ) clearance of insulin was significantly depressed in the DKA state when compared with controls (0.52 +/- 0.04 and 0.75 +/- 0.07 ml X min-1 X g-1, respectively). This could largely be explained by a lower glomerular filtration rate. Fractional urinary insulin clearance was increased twofold above control values in DH kidneys and fourfold in DKA kidneys, indicating that tubular luminal absorption of insulin was impaired in both states. By contrast contraluminal uptake (peritubular clearance) did not differ significantly from controls. 125I-insulin degrading activity of the 100,000 g supernate fraction from muscle homogenates was similar in the diabetic and control groups. However in liver and kidney, degrading activity did not correspond to whole organ insulin clearance in a consistent manner.(ABSTRACT TRUNCATED AT 250 WORDS)

    View details for Web of Science ID A1986C334800025

    View details for PubMedID 3085515

  • BIOLOGY OF DISEASE - MECHANISMS AND CONSEQUENCES OF PROTEINURIA LABORATORY INVESTIGATION Kaysen, G. A., Myers, B. D., Couser, W. G., Rabkin, R., FELTS, J. M. 1986; 54 (5): 479-498
  • PATHWAYS OF RENAL INSULIN DEGRADATION Simmons, A., Herrman, J., Frank, B., Rabkin, R. SLACK INC. 1986: A701–A701
  • HEPATORENAL-SYNDROME MANAGED WITH HEMODIALYSIS, THEN REVERSED BY PERITONEOVENOUS SHUNTING JOURNAL OF CLINICAL GASTROENTEROLOGY Kearns, P. J., POLHEMUS, R. J., Oakes, D., Rabkin, R. 1985; 7 (4): 341-343

    Abstract

    A patient with acute decompensated chronic liver disease developed acute tubular necrosis after an episode of hypotension. Renal failure was managed by hemodialysis for 11 weeks during which period hepatic function improved. Despite persistently severe oliguria, tubular function recovered as judged by a fall in urine sodium content and a rise in specific gravity, suggesting the development of the hepato-renal syndrome. Therefore, a peritoneovenous shunt was inserted. This was followed by a prompt diuresis; further dialysis was not required. This case suggests potential roles for hemodialysis and peritoneovenous shunting in patients with advanced, but potentially reversible hepatic and renal failure and draws attention to the need for formal evaluation of such a possibility.

    View details for Web of Science ID A1985APN2800014

    View details for PubMedID 4045179

  • Peritubular uptake and processing of insulin. Contributions to nephrology Rabkin, R., Petersen, J. 1984; 42: 38-48

    View details for PubMedID 6398196

  • THE RENAL METABOLISM OF INSULIN DIABETOLOGIA Rabkin, R., Ryan, M. P., Duckworth, W. C. 1984; 27 (3): 351-357

    Abstract

    The kidney plays a pivotal role in the clearance and degradation of circulating insulin and is also an important site of insulin action. The kidney clears insulin via two distinct routes. The first route entails glomerular filtration and subsequent luminal reabsorption of insulin by proximal tubular cells by means of endocytosis. The second involves diffusion of insulin from peritubular capillaries and subsequent binding of insulin to the contraluminal membranes of tubular cells, especially those lining the distal half of the nephron. Insulin delivered to the latter sites stimulates several important processes, including reabsorption of sodium, phosphate, and glucose. In contrast, insulin delivered to proximal tubular cells is degraded to oligopeptides and amino-acids by one of two poorly delineated enzymatic pathways. One pathway probably involves the sequential action of insulin protease and either GIT or non-specific proteases; the other probably involves the sequential action of GIT and lysosomal proteases. The products of insulin degradation are reabsorbed into the peritubular capillaries, apparently via simple diffusion. Impairment of the renal clearance of insulin prolongs the half-life of circulating insulin by a number of mechanisms and often results in a decrease in the insulin requirement of diabetic patients. Much needs to be learned about these metabolic events at the subcellular level and how they are affected by disease states. Owing to the heterogeneity of cell types within the kidney and to their anatomical and functional polarity, investigation of these areas will be challenging indeed.

    View details for Web of Science ID A1984TM82100001

    View details for PubMedID 6389240

  • RENAL METABOLISM OF PEPTIDE-HORMONES MINERAL AND ELECTROLYTE METABOLISM Rabkin, R., KITAJI, J. 1983; 9 (4-6): 212-226

    View details for Web of Science ID A1983RQ85300004

    View details for PubMedID 6361510

  • FATE OF [I-125]-LABELED INSULIN REMOVED FROM THE PERITUBULAR CIRCULATION OF ISOLATED PERFUSED RAT-KIDNEY AMERICAN JOURNAL OF PHYSIOLOGY Petersen, J., KITAJI, J., Duckworth, W. C., Rabkin, R. 1982; 243 (2): F126-F132

    Abstract

    Although there is considerable evidence that insulin is removed from the peritubular circulation of the mammalian kidney, it is unclear whether binding to insulin-specific receptors is involved in this process, whether after peritubular removal the hormone is degraded to small fragments with release into the circulation, or whether it merely undergoes a minor modification with loss of immunoreactivity. We examined the metabolism of [125I]insulin removed from the peritubular circulation of the nonfiltering isolated perfused rat kidney and compared it to that of [125I]insulin metabolized by filtering isolated kidneys and kidney homogenates. The results indicate that after peritubular removal, a small amount of insulin is degraded to form low-molecular-weight products similar to those seen with filtering kidneys and kidney homogenates. However, most of the insulin removed from the peritubular circulation is processed either to nonimmunoreactive products of molecular weight similar to that of insulin or, to a lesser extent, to products of larger molecular weight. Both these products are also formed by filtering kidneys. In the filtering kidney, the products having molecular weight similar to that of insulin probably originate from the peritubular process, because it is unlikely that material of this size could be derived from the filtration-absorption pathway. Of particular note was the finding that [125I]insulin trapped in the peritubular compartment of nonfiltering kidneys was displaced severalfold more effectively by unlabeled insulin than by several peptide hormones (P less than 0.01); the latter were no more effective than vehicle alone. The findings suggest the presence of peritubular insulin-specific receptors.

    View details for Web of Science ID A1982PC51800053

    View details for PubMedID 7051850

  • THE INFLUENCE OF AMINO-ACIDS (AA) ON TUBULAR ABSORPTION OF INSULIN AND GROWTH-HORMONE Rabkin, R., GOTTHEINER, T., Tsao, T. BLACKWELL SCIENCE INC. 1982: 254–54
  • AMINO-ACIDS ENHANCE RENAL TUBULAR ABSORPTION OF THE LOW-MOLECULAR-WEIGHT PROTEINS INSULIN AND GROWTH-HORMONE AMERICAN JOURNAL OF PHYSIOLOGY Rabkin, R., GOTTHEINER, T. I., Tsao, T. S. 1982; 242 (6): F745-F749

    Abstract

    The effect of amino acids(AA) on the tubular absorption of low-molecular-weight (LMW) proteins was studied in isolated rat kidneys. Kidneys were perfused with an albumin-electrolyte solution that contained insulin or human growth hormone (hGH) and, unless otherwise stated, the following L-amino acids: glycine, isoleucine, serine, alanine, methionine, proline, arginine, and aspartic acid. In kidneys perfused without AA, fractional urinary insulin clearance (FCi) averaged 7.4 +/- 1.54%, whereas in the presence of multiple AA the FCi was significantly lower (0.68 +/- 0.2%, P less than 0.01). Addition of glycine or alpha-aminoisobutyric acid (AIB) alone also reduced the FCi significantly (1.79 +/- 0.66 and 1.59 +/-1.06%, respectively). By contrast, perfusion with the other AA individually did not alter the FCi. The fractional urinary hGH clearance was also significantly lower in kidneys perfused with multiple AA (0.94 +/- 0.47%) than in those perfused without AA (9.07 +/- 1.2%). We conclude that tubular absorption of filtered insulin and hGH is enhanced by the presence of AA. The mechanism is unclear, but enhancement of insulin absorption can be produced by glycine and AIB alone. This raises the possibility of a link between the absorption of insulin and the glycine and AIB shared transport system, but excludes a primary metabolic effect because AIB is nonmetabolizable.

    View details for Web of Science ID A1982NX19200088

    View details for PubMedID 7046464

  • BINDING AND DEGRADATION OF INSULIN BY ISOLATED RENAL BRUSH-BORDER MEMBRANES DIABETES Rabkin, R., Petersen, J., Mamelok, R. 1982; 31 (7): 618-623

    Abstract

    Filtered proteins including insulin are absorbed in the proximal tubule by means of pinocytosis. The first step in this process is binding of the protein to brush border membrane. As it is not known whether absorption exhibits specificity, we set out to determine whether specific binding sites for insulin are present in brush border membranes. Rabbit-isolated brush border membranes were incubated with 125I-insulin and varying concentrations of cold insulin or other peptide hormones. Binding and degradation of 125I-insulin occurred in a time- and temperature-dependent manner. Native insulin competitively inhibited 125I-insulin binding, but calcitonin, arginine vasopressin, glucagon, and growth hormone (10(-6) M) were relatively ineffective. Nonspecific binding averaged one-third of the total radioactivity bound. Scatchard analysis of binding data revealed two classes of insulin receptors: high affinity, low capacity receptors and low affinity, high capacity receptors. Gel filtration analysis of 125I-insulin exposed to brush border membrane revealed the formation of low-molecular-weight products similar to that produced by intact kidneys. The degrading process exhibited some specificity, for cold insulin (10(-6) M) was more effective than calcitonin, vasopressin, glucagon, or growth hormone in inhibiting degradation (32% versus less than 13% inhibition; P less than 0.01). Whether this reflects inhibition of insulin specific binding before exposure to degradation or inhibition of specific enzymes is unclear. In summary, it appears that renal brush border membranes have a major insulin-specific receptor component that could potentially mediate tubular insulin absorption. In addition, there is a smaller nonspecific component that may also have the potential to mediate insulin absorption. Finally, it appears that brush border membranes have the ability to degrade insulin to low-molecular-weight products by a process that exhibits some specificity for insulin.

    View details for Web of Science ID A1982NV92300010

    View details for PubMedID 6761200

  • AMINO-ACIDS (AA) ENHANCE TUBULAR LOW-MOLECULAR WEIGHT PROTEIN (LMWP) ABSORPTION GOTTHEINER, T. I., Tsao, T., Rabkin, R. SLACK INC. 1981: A72–A72
  • METABOLIC CHARACTERISTICS OF RENAL INSULIN UPTAKE GOTTHEINER, T. I., Tsao, T., Rabkin, R. NATURE PUBLISHING GROUP. 1981: 222–22
  • AMINO-ACIDS (AA) ENHANCE RENAL TUBULAR LOW-MOLECULAR WEIGHT PROTEIN (LMWP) ABSORPTION GOTTHEINER, T. I., Tsao, T., Rabkin, R. SLACK INC. 1981: A554–A554
  • REMOVAL AND EXCRETION OF IMMUNOREACTIVE RAT GROWTH-HORMONE BY THE ISOLATED KIDNEY AMERICAN JOURNAL OF PHYSIOLOGY Rabkin, R., GOTTHEINER, T. I., Fang, V. S. 1981; 240 (4): F282-F287

    Abstract

    The renal uptake of immunoreactive rat growth hormone (rGH), molecular weight 21,500 daltons, was examined in the isolated perfused rat kidney to determine whether peritubular removal of a protein greater than 12,000 daltons occurs and to assess the functional characteristics of renal GH uptake. Organ clearance of rGH (OCGH) in control kidneys was 1,039 +/- 99 microliters/min and was unaffected by an excess of insulin but markedly depressed by col (10 degrees C( and KCN. Although glomerular filtration rate (GFR) did not differ significantly from OCGH in the control rats, we suspected that filtration could not account for all the rGH removed because of glomerular protein sieving. However, GFR was significantly less than OCGH with cold and KCN treatment, indicating the occurrence of peritubular removal. In nonfiltering kidneys, rGH removal exceeded that of [14C]inulin (P less than 0.05), demonstrating peritubular rGH removal. Tubular absorption of rGH was unaffected by insulin but markedly depressed by cold and KCN. We conclude that rGH is removed from the renal circulation mainly by the glomerular filtration-tubular absorptive pathway, but, in addition, as with smaller proteins, that peritubular removal occurs.

    View details for Web of Science ID A1981LL45100065

    View details for PubMedID 7013498

  • METABOLIC CHARACTERISTICS OF RENAL INSULIN UPTAKE DIABETES Rabkin, R., GOTTHEINER, T. I., Tsao, T. S. 1981; 30 (11): 929-934

    View details for Web of Science ID A1981MQ22700007

    View details for PubMedID 7028545

  • REVERSIBLE HYPERINSULINURIA IN DIABETIC-KETOACIDOSIS IN MAN AMERICAN JOURNAL OF PHYSIOLOGY Sacks, H., Rabkin, R., Kitabchi, A. E. 1981; 241 (5): E396-E405

    Abstract

    Urinary clearance and fractional urinary clearance of immunoreactive insulin (IRI) and beta 2-microglobulin (I beta 2M) were studied in patients with diabetic ketoacidosis (DKA) before, during, and after treatment. Our results indicate that in DKA in man a) there is an approximate 250-fold increase in urinary and fractional urinary clearance of IRI and a 600-fold increase in urinary and fractional urinary I beta 2M clearance, which suggests that the hyperinsulinuria is secondary to a nonspecific defect in tubular luminal uptake of low-molecular-weight proteins, although decreased IRI degradation cannot be excluded; b) because increased IRI clearance is not changed by the pharmacologic plasma IRI levels achieved, the residual tubular absorptive capacity is not saturable; c) I beta 2M clearance but not IRI clearance is significantly improved by the time metabolic control is attained, suggesting separate tubular transport systems; d) a small, therapeutically insignificant fraction of the infused insulin is lost in the urine during therapy of DKA; and e) defective renal tubular luminal uptake (and possibly degradation) of IRI is reversible.

    View details for Web of Science ID A1981MS96200023

    View details for PubMedID 6795930

  • DETERMINANTS OF RENAL INSULIN UPTAKE AND EXCRETION Rabkin, R., GOTTHEINER, T. I., Chen, T. S. SLACK INC. 1980: A536–A536
  • REMOVAL OF IMMUNOREACTIVE ARGININE VASOPRESSIN BY THE PERFUSED-RAT-LIVER ENDOCRINOLOGY Rabkin, R., GHAZELEH, S., Share, L., Crofton, J., UNTERHALTER, S. A. 1980; 106 (3): 930-934

    Abstract

    To assess the ability of the liver to remove immunoreactive arginine vasopressin (AVP) from the circulation and to determine the effect of certain metabolic factors on the process, a study was carried out with rat livers perfused at 37 C with an oxygenated albumin--electrolyte solution containing AVP (117 +/- 4.5 muU/ml). In controls, the hepatic clearance of AVP was 795 +/- 120 microliter/min (SEM). The addition of AVP in concentrations up to 9029 microU/ml, perfusion with a glucose-free medium, or perfusion without oxygen did not significantly alter the hepatic clearance of AVP. However, perfusion with cold medium (11 C) significantly altered AVP removal in that initially AVP removal increased, while later on AVP removal became completely inhibited. This phenomenon may possibly be a consequence of a cold-induced increase in hepatic AVP trapping which is rapidly saturated due to a cold-induced depression of AVP transport and degradation. Support for this thesis was provided by finding that high AVP concentrations depressed the cold-endhancing removal phase.

    View details for Web of Science ID A1980JH68500043

    View details for PubMedID 7353552

  • HANDLING OF IMMUNOREACTIVE VASOPRESSIN BY THE ISOLATED PERFUSED RAT-KIDNEY JOURNAL OF CLINICAL INVESTIGATION Rabkin, R., Share, L., Payne, P. A., Young, J., Crofton, J. 1979; 63 (1): 6-13

    Abstract

    Using the isolated rat kidney perfused with an artificial medium containing glucose as the sole fuel, we studied the renal handling of immunoreactive arginine vasopressin (AVP) and determined the effect of various factors on the ability of the kidney to remove AVP. In control kidneys perfused with AVP at concentrations below 116 muU/ml, the organ clearance of AVP (OC(AVP)) was 1,145+/-47 (SE) mul/min, whereas glomerular filtration rate (GFR) averaged 515+/-37 mul/min. Filtration could thus account for up to 45% of the OC(AVP), the balance presumably being cleared from the peritubular circulation. Of the AVP filtered, only 38% could be recovered in the urine (urinary clearance AVP averaged 205+/-12 mul/min) suggesting that the balance was taken up by the tubular epithelium and degraded. Fractional excretion of filtered AVP rose significantly in the presence of anoxia and cold (10 degrees C) to 49 and 59%, respectively, but was not affected by ouabain or high levels of AVP (458+/-58 muU/ml). The OC(AVP) was not significantly altered by the absence of glucose in the perfusate, anoxia, or ureteral ligation, maneuvers that were associated with significant reductions in GFR. In these and the control experiments, there was a significant inverse correlation between GFR and peritubular clearance emphasizing the importance of the latter (r = -0.749; P < 0.001). Cold, ouabain, and high concentrations of AVP reduced the clearance of AVP by the kidneys. On the basis of these studies we conclude that the kidney clears AVP from the circulation via two pathways, glomerular clearance and peritubular clearance. This exposes both the luminal and contraluminal surfaces of the tubular cells to the hormone, allowing these cells to remove AVP from the filtrate and the peritubular compartment. Noteworthy is the observation that under several conditions when GFR falls reducing the glomerular clearance of AVP, peritubular clearance increases and the total clearance of AVP by the kidney remains constant.

    View details for Web of Science ID A1979GC73400002

    View details for PubMedID 762248

  • RENAL HANDLING OF IMMUNOREACTIVE RAT GROWTH-HORMONE (RGH) GOTTHEINER, T. I., Rabkin, R., Fang, V. NATURE PUBLISHING GROUP. 1979: 816–16
  • EFFECT OF PROLONGED UREMIA ON INSULIN METABOLISM BY ISOLATED LIVER AND MUSCLE KIDNEY INTERNATIONAL Rabkin, R., UNTERHALTER, S. A., Duckworth, W. C. 1979; 16 (4): 433-439

    Abstract

    As the prolonged metabolic clearance rate of insulin in chronic uremia cannot be entirely explained by impaired removal and degradation of insulin by the kidney, we set out to determine whether prolonged uremia depresses other major sites of insulin degradation. The study was conducted with livers and skeletal muscle obtained from normal control rats and uremic rats 4 weeks after 80% nephrectomy. Despite a significant difference between renal function in the control and uremic rats (BUN, 18 vs. 46 mg/dl), there was no significant difference in the clearance of insulin by isolated uremic or control livers perfused with a bloodless medium. Similarly, the 125I-insulin degrading activity of liver homogenates was not depressed by uremia. In contrast, binding and degradation by uremic liver cell membranes was significantly reduced to 58% and 85% of the controls, respectively. Degradation by homogenates of skeletal muscle and by intact epitrochlaris muscle was significantly less in uremics than in controls. These results indicate that chronic uremia depresses skeletal muscle insulin degradation but not hepatic insulin removal or degradation despite a decrease in insulin binding and degradation by liver plasma membranes. It thus appears that depression of insulin degradation by muscle may contribute to the prolonged insulin metabolic clearance rate seen in chronic uremia. Furthermore, it is possible that the impaired binding of insulin to liver membranes may play a role in the insulin resistance of uremia.

    View details for Web of Science ID A1979HR63800001

    View details for PubMedID 398415

  • HANDLING OF INSULIN, PROINSULIN, AND C-PEPTIDE BY ISOLATED RAT-KIDNEY DIABETES Rabkin, R., Ross, B. D., MAKO, M. E., Rubenstein, A. H. 1978; 27: 192-196

    View details for Web of Science ID A1978ER62200007

    View details for PubMedID 631440

  • EFFECT OF RENAL-TRANSPLANTATION ON PITUITARY GONADAL FUNCTION METABOLISM-CLINICAL AND EXPERIMENTAL Morley, J. E., Distiller, L. A., UNTERHALTER, S., Myers, J. B., Rabkin, R., Katz, M. 1978; 27 (7): 781-785

    Abstract

    Twelve adult male patients who had undergone successful renal transplantation were investigated. The gonadotropin responses to 100 microgram luteinizing hormone-releasing hormone (LRH) were studied, and basal serum testosterone and prolactin assayed. Significantly elevated mean basal levels of luteinizing hormone (LH) and follicle stimulating hormone (FSH) were found, associated with a correspondingly excessive LH and FSH response to LRH. Mean basal serum testosterone levels in the posttransplant patients were significantly lower than in normal controls, while the mean basal prolactin levels were similar in the two groups. The results were not influenced by the varying degrees of renal function found in the posttransplant patients.

    View details for Web of Science ID A1978FF70100004

    View details for PubMedID 351325

  • FACTORS INFLUENCING HANDLING OF INSULIN BY ISOLATED RAT-KIDNEY JOURNAL OF CLINICAL INVESTIGATION Rabkin, R., Kitabchi, A. E. 1978; 62 (1): 169-175

    Abstract

    The renal handling of immunoreactive insulin was studied in the isolated perfused normothermic rat kidney to determine (a) the relative contributions of glomerular clearance and peritubular clearance to the renal clearance of insulin under different conditions, (b) what metabolic factors influence the ability of tubular cells to remove insulin from the glomerular filtrate and the peritubular circulation, and (c) whether the same factors influence the luminal and contraluminal uptake of insulin.In control kidneys the organ clearance of insulin (OCi) was 974+/-63 mul/min (SEM), of which a maximum of 46% could theoretically be accounted for by filtration. OCi was not altered by fasting, lack of exogenous fuel (glucose), or the addition of cyanide. The glomerular filtration rate did not correlate with the OCi, but there was a significant (P < 0.001) negative correlation (r = -0.828) between the peritubular clearance and glomerular filtration rate. Both N-ethylmaleimide and cold (10 degrees C) reduced the rate of insulin removal. Fractional excretion of filtered insulin (9.7+/-1.7% in controls) was not significantly altered by fasting or perfusing without glucose. In contrast, KCN increased fractional excretion of insulin to 41.9+/-3.7% whereas cold increased fractional excretion to 69.0+/-3.3%. This study indicates that renal tubular cells remove insulin from the tubular lumen and the peritubular compartment. Furthermore, the data suggest that insulin removal by tubular cells is a temperature-sensitive process consisting of two different systems. The system associated with the luminal aspect of the cell appears to be dependent on oxidative metabolism, whereas the system associated with the contraluminal aspects of the cell appears to be independent thereof. Under several circumstances when the glomerular clearance of insulin falls thereby reducing the amount of insulin absorbed by the luminal aspect of the cell, contraluminal uptake increases, and a constant rate of insulin removal is maintained by the kidney.

    View details for Web of Science ID A1978FE08800022

    View details for PubMedID 659630

  • INSULIN EXTRACTION FROM RENAL PERITUBULAR CIRCULATION IN CHICKEN ENDOCRINOLOGY Rabkin, R., Jones, J., Kitabchi, A. E. 1977; 101 (6): 1828-1833

    View details for Web of Science ID A1977EE28600023

    View details for PubMedID 590194

  • Post-transplantation analgesic dependence in patients who formerly suffered from analgesic neophropathy. Clinical nephrology FURMAN, K. I., Galasko, G. T., Meyers, A. M., Rabkin, R. 1976; 5 (2): 54-56

    Abstract

    Twenty-two patients with functioning grafts who originally developed renal failure due to analgesic nephropathy ("analgesic" group), and 84 patients with various other causes of renal failure ("non-analgesic" group) were studied over one year to assess the extent of analgesic use and abuse. On each occasion that one of these patients reported to hospital a urine sample was collected and analyzed for N-acetylparaminophenol (NAPA). Of the "analgesic" group of patients, 14% had consistently negative urine samples while 41% showed NAPA in more than half the urines collected. In the "non-analgesic" group 29% of patients had entirely negative urine tests, and in only 7% were more than half of the urines NAPA-positive. Patients in the "non-analgesic" group readily admitted taking anaglesics at frequencies compatible with the observed number of positive tests. This contrasted with the "analgesic" group in which 14 of 19 patients with NAPA-positive urines denied analgesic intake. This is considered to be a guilt manifestation in patients who have developed a psychological dependence on analgesics and have recidivated with full knowledge of the possible harm these drugs may inflict on their grafts.

    View details for PubMedID 767023

  • Lymphocoeles in renal homograft recipients. Proceedings of the European Dialysis and Transplant Association. European Dialysis and Transplant Association Meyers, A., Salant, D., Rabkin, R., Milne, J., Botha, R., Myburgh, J. 1976; 12: 452-460

    Abstract

    Seven lymphocoeles have complicated 158 cadaver and 18 related live donor transplants performed over six years. The purpose of this study is to review the clinical, diagnostic and therapeutic problems encountered with lymphocoeles at this centre. Presentation was insidious, delayed (50 days post-op), and manifested by one or more of the following: palpable rectal or supra-pubic mass; unilateral leg oedema; recurrent urine infections with radiographic evidence of obstruction; filling defect in bladder on routine IVP. Possible pathogenetic factors: transplants performed on side of functioning Scribner shunts (6/7 cases); severe rejection episodes with graft lymphatic leak (1/7 cases). Differentiation from urinomas, haematomas, perinephric abscesses or other causes of obstructive uropathy were facilitated by: needle aspiration; IVP and cystogram, serial ultrasound sonography. Lymphangiography was not used. Treatment was conservative in three, repeated aspirations were performed in two and formal drainage procedures were required in three patients. Repeated cyst aspirations resulted in serious infections in both patients. Marsupialisation into the peritoneal cavity failed in one. Conclusions: 1) The graft should be anastomosed on side opposite a functioning shunt. 2) Lymphocoeles have an occult presentation and should be actively sought for as they may produce urinary obstruction. 3) Serial ultrasound is an excellent method of diagnosis and follow-up. 4) Unless urinary obstruction is present management should be conservative as spontaneous resolution may occur. 5) Ideal surgical management is problematical. Repeated needle aspirations should be approached with caution. Lymphocoeles are an uncommon but well documented complication of renal transplantation (Schweizer et al, 1972; Koehler et al, 1972; Christiansen et al, 1974; Rashid et al, 1974). They represent definite hazards and can either directly or indirectly contribute to morbidity or even mortality in the post-transplant patient. They must be differentiated from other pelvic masses such as urinomas, haematomas and perinephric abscesses as they all require different modes of treatment. As a rule, the diagnosis of one from another is straightforward, but as certain clinical features are shared, differentiation can on occasions be more difficult. In order to clarify some of these problems and outline a mode of therapy, a study was carried out in patients who have presented with lymphocoeles in our unit.

    View details for PubMedID 778837

  • KAPOSIS SARCOMA IN AN IMMUNOSUPPRESSED RENAL-ALLOGRAFT RECIPIENT SOUTH AFRICAN MEDICAL JOURNAL Meyers, A. M., RICE, G. C., Kaye, S., Myburgh, J. A., Rabkin, R. 1976; 50 (33): 1299-1300

    Abstract

    A 47-year-old White man developed a buccal Kaposi's sarcoma 15 months after receiving a renal allograft. Atypical clinical and histological features were noted. A previous viral infection might have played a role in the occurrence of this patient's tumour but other possibilities of tumour induction in the immunosuppressed host are discussed. Although the condition is lethal when diagnosed after renal transplantation, this case report demonstrates that prompt diagnosis and radiotherapy may result in permanent cure.

    View details for Web of Science ID A1976BZ53000015

    View details for PubMedID 785625

  • HYPERLIPEMIA IN RENAL-TRANSPLANT PATIENTS/TS SOUTH AFRICAN MEDICAL JOURNAL Goldberg, R. B., JUDELMAN, J. J., Mindel, A., Meyers, A. M., Salant, D. J., Myburgh, J. A., Rabkin, R., Joffe, B. I., Seftel, H. C. 1976; 50 (33): 1291-1294

    Abstract

    The prevalence, nature and possible aetiology of post-transplantation hyperlipidaemia were studied in 94 renal allograft recipients. Sixty-five per cent of the group had hypercholesterolaemia or hypertriglyceridaemia, and types IIb and IV were the commonest forms of hyperlipoproteinaemia encountered. The pathogenesis of post-transplantation hyperlipidaemia is complex. Hypertriglyceridaemia was maximal in the first year after transplantation and could be correlated with high corticosteroid dosage during this period. Thereafter hypertriglyceridaemia was less marked and was related to obesity, corbohydrate intolerance and basal insulin levels. Mean cholesterol levels were elevated throughout the post-transplantation period, varied little and could not be related to corticosteroid dosage, body weight, carbohydrate intolerance, basal insulin levels or renal dysfunction.

    View details for Web of Science ID A1976BZ53000013

    View details for PubMedID 785623

  • DEPRESSED NEUTROPHIL CHEMOTAXIS IN PATIENTS WITH CHRONIC RENAL-FAILURE AND AFTER RENAL-TRANSPLANTATION JOURNAL OF LABORATORY AND CLINICAL MEDICINE Salant, D. J., Glover, A. M., Anderson, R., Meyers, A. M., Rabkin, R., Myburgh, J. A., RABSON, A. R. 1976; 88 (4): 536-545

    Abstract

    Polymorphonuclear leukocyte chemotaxis, phagocytosis, and bactericidal ability were tested with neutrophlis from patients with chronic renal failure on conservative therapy, from others of regular peritoneal dialysis or hemodialysis, and from a group of patients who had received renal allografts. Chemotaxis was depressed in all groups. Phagocytosis was mildly impaired in the two groups on dialysis. The leukocytes of patients on conservative therapy had mildly decreased killing ability, whereas this function was normal in other groups tested. Sera from all patients, when activated with endotoxin, were poorly chemotactic to normal neutrophils, especially in those on conservative therapy and hemodialysis, but these sera functioned normally as opsonins for phagocytosis. The observed defect in chemotaxis could account, at least partly, for the frequency of bacterial infections in these patients.

    View details for Web of Science ID A1976CF99500003

    View details for PubMedID 787456

  • Polymorphonuclear leucocyte function in chronic renal failure and after renal transplantation. Proceedings of the European Dialysis and Transplant Association. European Dialysis and Transplant Association Salant, D. J., Glover, A. M., Anderson, R., Meyers, A. M., Rabkin, R., Myburgh, J. A., RABSON, A. R. 1976; 12: 370-379

    Abstract

    Neutrophil chemotaxis, phagocytosis and bactericidal ability were assessed in patients with chronic renal failure on conservative therapy, in others on regular peritoneal and haemodialysis, and in a group of patients who had received renal allografts. Chemotaxis was abnormal in all groups. The groups on peritoneal and haemodialysis had mildly impaired phagocytosis, while killing ability was normal in all but the group on conservative therapy who showed a mild defect. Serum from groups when activated with endotoxin was poorly chemotactic to normal neutrophils, but functioned normally as opsonins for phagocytosis.

    View details for PubMedID 778831

  • EFFECT OF DIFFERENT PREPARATIONS OF HUMAN GROWTH-HORMONE ON PLASMA-RENIN ACTIVITY IN NORMAL MALES JOURNAL OF CLINICAL ENDOCRINOLOGY & METABOLISM Epstein, S., LeRoith, D., Rabkin, R. 1976; 42 (2): 390-392

    Abstract

    The effect of acute administration of 2 preparations of growth hormone (hGH) on plasma renin activity (PRA) was studied in normal volunteers. 4 IU of standard, commercially available hGH II, Kabi, Sweden) were injected im into each of four normal subjects, and the PRA was determined in the basal state and at 30, 60, 120, 180, and 240 min after injection. Free fatty acid (FFA) was determined basally and at 15 and 210 min post-injection. The study was repeated on a different day in the same group using highly purified hGH (hGH I, 4 IU) virtually free of arginine-vasopressin. Four other normal subjects received 12 IU standard hGH (hGH II, Kabi, Sweden). There was no significant difference in PRA values with 4 IU of either preparation or with 12 IU of hGH II in any of the groups, although mean PRA was elevated in two of the patients receiving 12 IU hGH II. A rise in FFA occurred in all subjects, indicating the biological activity of hGH preparations. The possible significance of these findings to the renin-angiotensin system found in acromegaly is considered.

    View details for Web of Science ID A1976BG96800024

    View details for PubMedID 1262436

  • STRONGYLOIDES-STERCORALIS HYPERINFECTION IN A RENAL-ALLOGRAFT RECIPIENT SOUTH AFRICAN MEDICAL JOURNAL Meyers, A. M., Shapiro, D. J., Milne, F. J., Myburgh, J. A., Rabkin, R. 1976; 50 (33): 1301-1302

    Abstract

    A patient who had received a renal allograft required intensive immunosuppression for a severe rejection episode. Four months after the graft he died of septicaemia and respiratory failure caused indirectly by a Strongyloides stercoralis hyperinfection. Patients from endemic areas who are to undergo a renal transplant should be screened for the parasite before receiving immunosuppressive therapy. Should this infection occur after transplantation, early diagnosis and treatment with thiabendazole is essential to prevent the high mortality rate associated with Strongyloides hyperinfection.

    View details for Web of Science ID A1976BZ53000016

    View details for PubMedID 785626

  • The qualitative nitroblue tetrazolium (NBT) test as a means to differentiate between infection and rejection in renal transplant patients. Clinical nephrology Sher, R., Anderson, R., Meyers, A. M., Rabkin, R., Koornhof, H. J. 1975; 4 (1): 8-12

    Abstract

    In order to determine whether immunosupression depresses the response of the NBT test to bacterial infections and to note the effect of allograft rejection on this test, a prospective study was carried out on 30 renal transplant recipients. 12 of 30 renal transplant patients developed bacterial infections and in these patients NBT readings were elevated. 12 of the remianing 18 patients who developed rejection episodes showed normal NBT results. All patients were on high doses of steroids and other immunsuppressive agents. We conclude that the NBT test may be of value in diagnosing bacterial infection in the immunsuppressed allograft recipient, and may also be an useful adjunct in the differentiation between allograft infection and bacterial infection.

    View details for PubMedID 1098815

  • Changes in oxygen delivery during hemodialysis. Clinical nephrology TORRANCE, J. D., Milne, F. J., Hurwitz, S., ZWI, S., Rabkin, R. 1975; 3 (2): 54-59

    Abstract

    The effect of hemodialysis on oxygen delivery has been assessed in nine patients. During hemodialysis the arterial PO2 decreased from 76.89 mm Hg to 69.03 mm Hg. There was also an increase in pH from 7.39 to 7.49 which resulted in an increase in the affinity of the blood for oxygen. In the absence of compensatory changes this could result in a 17.9% decrease in oxygen delivery. This could be hazardous in patients who are already anemic particularly in the presence of cardiac or respiratory insufficiency.

    View details for PubMedID 235391

  • PITUITARY-GONADAL FUNCTION IN CHRONIC RENAL-FAILURE - EFFECT OF LUTEINIZING HORMONE-RELEASING HORMONE AND INFLUENCE OF DIALYSIS METABOLISM-CLINICAL AND EXPERIMENTAL Distiller, L. A., Morley, J. E., Sagel, J., POKROY, M., Rabkin, R. 1975; 24 (6): 711-720

    Abstract

    Sixteen adult male patients with chronic renal failure undergoing either chronic intermittent hemodialysis (HD) or chronic intermittent peritoneal dialysis (PD) were studied both before and immediately after dialysis. The gonadotropin responses to luteinizing hormone-releasing hormone (LH-RH) was determined, revealing an excessive luteinizing hormone (LH) response with a delayed return to normal in both dialysis groups. No significant alteration in follicle-stimulating hormone (FSH) kinetics was observed. There was no significant difference in the mean gonadotropin responses to LH-RH between the HD and PD groups, and dialysis had no effect on either mean LH or FSH responses. The chronic renal failure patients with testicular atrophy had an excessive FSH response to LH-RH when compared to those patients without testosterone was significantly lower than normal. Chronic renal failure effects testicular function and testicular atrophy is associated with seminiferous tubular destruction and an excessive FSH response. Poor renal clearance may play a role in the abnormal LH response observed.

    View details for Web of Science ID A1975AE60400005

    View details for PubMedID 1092955

  • EFFECT OF GROWTH-HORMONE ON RENAL SODIUM AND WATER EXCRETION HORMONE AND METABOLIC RESEARCH Rabkin, R., Epstein, S., Swann, M. 1975; 7 (2): 139-142
  • ANALGESIC RENAL PAPILLARY NECROSIS SOUTH AFRICAN MEDICAL JOURNAL Smith, J. D., Rabkin, R., Stables, D., THATCHER, G. N., EALES, L. 1975; 49 (44): 1819-1822

    Abstract

    Analgesic nephropathy is well recognised. This is a retrospective review of 19 patients with the disease, who presented at Groote Schuur Hospital over a 4-year period. The diagnosis was made on historical and clinical grounds and on the radiological manifestations of papillary necrosis. The mean age was 49 years (26 - 74) with a sex ratio of 3,8 female : 1 male. The period of abuse varied between 3 and 20 years. Aspirin, phenacetin, codeine, and antipyrine were the compounds mainly ingested. If the abuse is stopped, renal deterioration can be halted. However, factors which lead to dehydration and infection tend to cause further decompensation. As a means of preventing this disease greater control over analgescis is needed.

    View details for Web of Science ID A1975AT45200008

    View details for PubMedID 1188544

  • UNUSUAL HORMONAL CAUSE OF HYPERTENSION AND HYPOKALEMIA SOUTH AFRICAN MEDICAL JOURNAL Salant, D. J., Rabkin, R. 1975; 49 (37): 1525-1526

    Abstract

    A patient with primary hyperparathyroidism who presented with hypokalaemia and hypertension is described. Renal potassium wasting was documented and cured by removal of a parathyroid adenoma. Possible mechanisms for this unusual manifestation of hyperparathyroidism are mentioned. Other features of the case were severe anaemia, nephrocalcinosis, pseudogout and postoperative acidosis.

    View details for Web of Science ID A1975AN26200012

    View details for PubMedID 1162532

  • EFFECT OF GROWTH-HORMONE ON SODIUM-TRANSPORT AND OSMOTIC WATER-FLOW ACROSS TOAD SKIN HORMONE AND METABOLIC RESEARCH Rabkin, R., Swann, M., Shapiro, D. J., Isaacson, L. 1974; 6 (2): 129-132

    View details for Web of Science ID A1974S554900008

    View details for PubMedID 4828272

  • PRESENSITIZATION AND CLINICAL KIDNEY-TRANSPLANTATION .2. CORRELATIVE STUDIES OF HUMORAL AND CELLULAR IMMUNITY AND CLINICAL COURSE TRANSPLANTATION Myburgh, J. A., Maier, G., Smit, J. A., Shapiro, M., Meyers, A. M., Rabkin, R., VANBLERK, P. J., JERSKY, J. 1974; 18 (3): 213-222

    View details for Web of Science ID A1974U077600003

    View details for PubMedID 4607999

  • PRESENSITIZATION AND CLINICAL KIDNEY-TRANSPLANTATION .1. FAVORABLE COURSE OF A SUBSTANTIAL NUMBER OF PATIENTS TRANSPLANTATION Myburgh, J. A., Maier, G., Smit, J. A., Shapiro, M., Meyers, A. M., Rabkin, R., VANBLERK, P. J., JERSKY, J. 1974; 18 (3): 206-212

    View details for Web of Science ID A1974U077600002

    View details for PubMedID 4607998

  • EXPERIENCE WITH CHRONIC HEMODIALYSIS IN JOHANNESBURG SOUTH AFRICAN MEDICAL JOURNAL Milne, F. J., Goldberg, B., Meyers, A. M., FURMAN, K. I., Cohen, I., GECELTER, L., RANSOME, O., VANDENHE, T. O., Davis, R., Rabkin, R. 1974; 48 (43): 1821-1825

    View details for Web of Science ID A1974U014300008

    View details for PubMedID 4609021

  • NEPHROTIC SYNDROME, MALIGNANCY AND IMMUNOSUPPRESSION SOUTH AFRICAN MEDICAL JOURNAL Rabkin, R., THATCHER, G. N., Diamond, L. H., EALES, L. 1973; 47 (14): 605-606

    View details for Web of Science ID A1973P309900009

    View details for PubMedID 4739810

  • AUTORADIOGRAPHIC DEMONSTRATION OF GLOMERULAR FILTRATION AND PROXIMAL TUBULAR ABSORPTION OF GROWTH-HORMONE I-125 IN MOUSE HORMONE AND METABOLIC RESEARCH Rabkin, R., PIMSTONE, B. L., EALES, L. 1973; 5 (3): 172-175

    View details for Web of Science ID A1973P746300006

    View details for PubMedID 4714543

  • RENAL TUBULAR FUNCTION IN SYSTEMIC LUPUS-ERYTHEMATOSUS SOUTH AFRICAN MEDICAL JOURNAL Jessop, S., Rabkin, R., MUMFORD, G., EALES, L. 1973; 47 (4): 132-135

    View details for Web of Science ID A1973O561300010

    View details for PubMedID 4132237

  • Glomerular filtration and proximal tubular absorption of insulin 125 I. The American journal of physiology Rabkin, R., Rubenstein, A. H., COLWELL, J. A. 1972; 223 (5): 1093-1096

    View details for PubMedID 4654343

  • BILATERAL TRAUMATIC RENAL-ARTERY OCCLUSION WITH SURVIVAL AFTER LATE RECONSTITUTION OF ARTERIAL FLOW BRITISH JOURNAL OF SURGERY BIRKENST, W. E., STABLES, D. P., Rabkin, R. 1972; 59 (11): 915-?

    View details for Web of Science ID A1972O038800090

    View details for PubMedID 4637144

  • SIGNIFICANCE OF SERUM CREATININE AND BLOOD UREA SERUM CREATININE RATIO IN AZOTEMIA SOUTH AFRICAN MEDICAL JOURNAL Kahn, S., Sagel, J., Rabkin, R., EALES, L. 1972; 46 (47): 1828-?

    View details for Web of Science ID A1972O026700008

    View details for PubMedID 4539756

  • GROWTH-HORMONE STIMULATION OF IONIC TRANSPORT ACROSS TOAD SKIN Rabkin, R., ISAACSON, L. C., Swann, M. MED ASSOC S AFRICA. 1972: 851-?

    View details for Web of Science ID A1972M699300035

    View details for PubMedID 4672795

  • DISAPPEARANCE OF HUMAN GROWTH-HORMONE I-125 IN ANEPHRIC NON-UREMIC AND UREMIC RAT HORMONE AND METABOLIC RESEARCH Rabkin, R., PIMSTONE, B. L., Marks, T., EALES, L. 1972; 4 (6): 467-469

    View details for Web of Science ID A1972O420800012

    View details for PubMedID 4655502

  • TRAUMATIC RENAL-ARTERY OCCLUSION BIRKENST, W. E., STABLES, D. P., Rabkin, R. MED ASSOC S AFRICA. 1972: 849-?

    View details for Web of Science ID A1972M699300023

    View details for PubMedID 5076075

  • SHORT-TERM FOLLOW-UP OF ACUTE TUBULAR NECROSIS Keeton, G. R., Miller, D., Rabkin, R., EALES, L. MED ASSOC S AFRICA. 1972: 849-?

    View details for Web of Science ID A1972M699300021

    View details for PubMedID 5076073

  • RENAL TUBULAR FUNCTION IN SYSTEMIC LUPUS ERYTHEMATOSIS Jessop, S., EALES, L., MUMFORD, G., Rabkin, R. MED ASSOC S AFRICA. 1972: 848-?

    View details for Web of Science ID A1972M699300017

    View details for PubMedID 5076069

  • BLOOD PRIMING FOR HAEMODIALYSIS LANCET Keeton, G. R., Rabkin, R., Thatcher, G., EALES, L. 1971; 2 (7719): 311-?

    View details for Web of Science ID A1971J980300020

    View details for PubMedID 4104993

  • EFFECT OF RENAL DISEASE ON RENAL UPTAKE AND EXCRETION OF INSULIN IN MAN NEW ENGLAND JOURNAL OF MEDICINE Rabkin, R., Simon, N. M., Steiner, S., COLWELL, J. A. 1970; 282 (4): 182-?

    View details for Web of Science ID A1970F128700002

    View details for PubMedID 5409813

  • RENAL UPTAKE AND EXCRETION OF INSULIN IN DOG JOURNAL OF LABORATORY AND CLINICAL MEDICINE Rabkin, R., COLWELL, J. A. 1969; 73 (6): 893-?

    View details for Web of Science ID A1969D397200002

    View details for PubMedID 5786458

  • Intermittent peritoneal dialysis in chronic renal failure. South African medical journal = Suid-Afrikaanse tydskrif vir geneeskunde Rabkin, R. H., Goldberg, B. 1968; 42 (41): 1095-1098

    View details for PubMedID 5726067

  • PROPHYLACTIC VALUE OF PROPRANOLOL IN ANGINA PECTORIS AMERICAN JOURNAL OF CARDIOLOGY Rabkin, R., STABLES, D. P., Levin, N. W., SUZMAN, M. M. 1966; 18 (3): 370-?

    View details for Web of Science ID A19668197400016

    View details for PubMedID 4958813