44 results on '"Finberg KE"'
Search Results
2. Bone marrow sinusoidal endothelial cells are a site of Fgf23 upregulation in a mouse model of iron deficiency anemia.
- Author
-
Li X, Lozovatsky L, Tommasini SM, Fretz J, and Finberg KE
- Subjects
- Animals, Mice, Bone Marrow metabolism, Disease Models, Animal, Endothelial Cells metabolism, Iron, RNA, Messenger genetics, Up-Regulation, Anemia, Iron-Deficiency genetics, Anemia, Iron-Deficiency metabolism, Erythropoietin genetics, Erythropoietin metabolism
- Abstract
Iron deficiency is a potent stimulator of fibroblast growth factor 23 (FGF23), a hormonal regulator of phosphate and vitamin D metabolism, that is classically thought to be produced by bone-embedded osteocytes. Here, we show that iron-deficient transmembrane serine protease 6 knockout (Tmprss6-/-) mice exhibit elevated circulating FGF23 and Fgf23 messenger RNA (mRNA) upregulation in the bone marrow (BM) but not the cortical bone. To clarify sites of Fgf23 promoter activity in Tmprss6-/- mice, we introduced a heterozygous enhanced green fluorescent protein (eGFP) reporter allele at the endogenous Fgf23 locus. Heterozygous Fgf23 disruption did not alter the severity of systemic iron deficiency or anemia in the Tmprss6-/- mice. Tmprss6-/-Fgf23+/eGFP mice showed green fluorescence in the vascular regions of BM sections and showed a subset of BM endothelial cells that were GFPbright by flow cytometry. Mining of transcriptomic data sets from mice with normal iron balance revealed higher Fgf23 mRNA in BM sinusoidal endothelial cells (BM-SECs) than that in other BM endothelial cell populations. Anti-GFP immunohistochemistry of fixed BM sections from Tmprss6-/-Fgf23+/eGFP mice revealed GFP expression in BM-SECs, which was more intense than in nonanemic controls. In addition, in mice with intact Tmprss6 alleles, Fgf23-eGFP reporter expression increased in BM-SECs following large-volume phlebotomy and also following erythropoietin treatment both ex vivo and in vivo. Collectively, our results identified BM-SECs as a novel site for Fgf23 upregulation in both acute and chronic anemia. Given the elevated serum erythropoietin in both anemic models, our findings raise the possibility that erythropoietin may act directly on BM-SECs to promote FGF23 production during anemia., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
3. Hepatocellular Adenoma: Report of 2 Cases That Highlight the Relevance of Phenotype-Genotype Correlation in the Pediatric Population.
- Author
-
Jiao J, Finberg KE, Jain D, and Morotti R
- Subjects
- Male, Humans, Child, Adolescent, beta Catenin genetics, Hedgehog Proteins, Phenotype, Genotype, Adenoma, Liver Cell diagnosis, Adenoma, Liver Cell genetics, Adenoma, Liver Cell pathology, Liver Neoplasms diagnosis, Liver Neoplasms genetics, Liver Neoplasms pathology, Carcinoma, Hepatocellular
- Abstract
Background: Hepatocellular adenoma (HCA) in the pediatric population is very rare and there are only limited studies, especially with molecular characterization of the tumors. Main HCA subtypes recognized in the current WHO classification include HNF1A -inactivated HCA (H-HCA), inflammatory HCA (IHCA), β-catenin-activated HCA (b-HCA), and β-catenin-activated IHCA (b-IHCA) and sonic hedgehog HCA (shHCA) is reported as an emerging subtype., Methods: Clinical history, pathological information, and molecular studies for a series of 2 cases of pediatric HCA were reviewed., Results: Case 1 was a b-HCA characterized by somatic CTNNB1 S45 mutation in a 11-year-old male with Abernethy malformation. Case 2 was a H-HCA characterized by germline HNF1A variant (c.526+1G>A) in a 15-year-old male associated with maturity-onset diabetes of the young type 3 (MODY3)., Conclusion: Our findings highlight the rarity of these 2 cases associated with adenomatosis, and the contribution of molecular/genetic analysis for proper sub-typing, prognosis and family surveillance., Competing Interests: Declaration of Conflicting InterestsThe author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
- Published
- 2023
- Full Text
- View/download PDF
4. Endometrial Carcinoma as the Presenting Malignancy in a Teenager With a Pathogenic TP53 Germline Mutation: A Case Report and Literature Review.
- Author
-
Irshaid L, Clark M, Fadare O, Finberg KE, and Parkash V
- Subjects
- Adolescent, Female, Genes, p53 genetics, Genetic Predisposition to Disease, Germ-Line Mutation, Humans, Tumor Suppressor Protein p53 genetics, Endometrial Neoplasms diagnosis, Endometrial Neoplasms genetics, Li-Fraumeni Syndrome complications, Li-Fraumeni Syndrome genetics
- Abstract
Patients with germline TP53 mutations are characterized by the occurrence of multiple early-onset malignancies. The characteristic syndrome is Li-Fraumeni syndrome (OMIM # 151623), an autosomal dominant disorder typified by premenopausal breast carcinoma, adrenal cortical tumors, bone and soft tissue sarcomas, leukemias, and tumors of the brain and spinal cord. Gynecologic malignancies are uncommonly reported in families harboring TP53 mutations, and the predominant tumor type reported is ovarian. Uterine carcinoma has been reported only a handful of times in patients with germline TP53 mutations, none as a presenting tumor in a teenager. We report on an 18-year-old patient who presented with grade 3, high-stage endometrioid endometrial carcinoma. Sequencing detected a single-nucleotide substitution in the TP53 gene (NM_000546.6:c.818G>A), encoding the missense substitution p.Arg273His (R273H) in both the tumor and normal tissue, consistent with a germline mutation. We discuss the biology of the TP53 gene and p53 protein, with emphasis on the R273H mutation. We also review the literature on endometrial carcinoma in patients with germline TP53 mutations., Competing Interests: The authors declare no conflict of interest., (Copyright © 2021 by the International Society of Gynecological Pathologists.)
- Published
- 2022
- Full Text
- View/download PDF
5. Blocking (iron) traffic in the gut.
- Author
-
Finberg KE
- Subjects
- Dietary Supplements, Gastrointestinal Microbiome, Iron
- Published
- 2022
- Full Text
- View/download PDF
6. Yale Precision Medicine Tumor Board: reawakening the guardian of the genome.
- Author
-
Grant MJ, Finberg KE, Walther Z, Stein SM, Lacy J, Eder JP, and Goldberg SB
- Subjects
- Genome, Humans, Molecular Targeted Therapy, Neoplasms genetics, Neoplasms pathology, Neoplasms therapy, Precision Medicine
- Published
- 2022
- Full Text
- View/download PDF
7. Pathogenic BRCA2 germline variants in combined hepatocellular-cholangiocarcinoma.
- Author
-
Li H, Zhang X, Finberg KE, Walther Z, Jain D, and Gibson J
- Subjects
- Aged, Carcinoma, Hepatocellular pathology, Cholangiocarcinoma pathology, Female, Germ-Line Mutation, Humans, Liver Neoplasms pathology, Loss of Heterozygosity, Male, Middle Aged, BRCA2 Protein genetics, Carcinoma, Hepatocellular genetics, Cholangiocarcinoma genetics, Liver Neoplasms genetics
- Published
- 2022
- Full Text
- View/download PDF
8. Functional characterization of a novel SLC40A1 Arg88Ile mutation in a kindred with familial iron overload treated by phlebotomy.
- Author
-
Womack J, Sukumaran A, Li X, Lozovatsky L, Gallagher PG, Seid JE, and Finberg KE
- Subjects
- Cation Transport Proteins chemistry, Humans, Iron Overload congenital, Iron Overload therapy, Male, Middle Aged, Models, Molecular, Phlebotomy, Cation Transport Proteins genetics, Iron Overload genetics, Point Mutation
- Published
- 2021
- Full Text
- View/download PDF
9. Yale Cancer Center Precision Medicine Tumor Board: molecular findings alter a diagnosis and treatment plan.
- Author
-
Gibson JA, Finberg KE, Nalbantoglu I, Cecchini M, Ganzak A, Walther Z, Sklar JL, Eder JP, and Goldberg SB
- Subjects
- Adenocarcinoma diagnosis, Adenocarcinoma genetics, Diagnosis, Differential, Gallbladder Neoplasms diagnosis, Gallbladder Neoplasms genetics, High-Throughput Nucleotide Sequencing, Humans, Male, Mesothelioma diagnosis, Mesothelioma genetics, Middle Aged, Molecular Targeted Therapy, Mutation, Peritoneal Neoplasms diagnosis, Peritoneal Neoplasms genetics, Prognosis, Adenocarcinoma therapy, Biomarkers, Tumor genetics, Gallbladder Neoplasms therapy, Mesothelioma therapy, Patient Care Planning, Peritoneal Neoplasms therapy, Precision Medicine
- Published
- 2021
- Full Text
- View/download PDF
10. Ironing out an approach to alleviate the hypoferremia of acute inflammation.
- Author
-
Finberg KE
- Subjects
- C-Peptide, Humans, Inflammation, Hepcidins, Iron
- Published
- 2021
- Full Text
- View/download PDF
11. NCOA4 is regulated by HIF and mediates mobilization of murine hepatic iron stores after blood loss.
- Author
-
Li X, Lozovatsky L, Sukumaran A, Gonzalez L, Jain A, Liu D, Ayala-Lopez N, and Finberg KE
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Cell Line, Female, Gene Expression Regulation, Gene Knockdown Techniques, Hemorrhage genetics, Hemorrhage pathology, Hepatocytes pathology, Humans, Hypoxia-Inducible Factor 1, alpha Subunit genetics, Liver pathology, Mice, Nuclear Receptor Coactivators genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Hemorrhage metabolism, Hepatocytes metabolism, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Iron metabolism, Liver metabolism, Nuclear Receptor Coactivators biosynthesis
- Abstract
The mechanisms by which phlebotomy promotes the mobilization of hepatic iron stores are not well understood. NCOA4 (nuclear receptor coactivator 4) is a widely expressed intracellular protein previously shown to mediate the autophagic degradation of ferritin. Here, we investigate a local requirement for NCOA4 in the regulation of hepatic iron stores and examine mechanisms of NCOA4 regulation. Hepatocyte-targeted Ncoa4 knockdown in nonphlebotomized mice had only modest effects on hepatic ferritin subunit levels and nonheme iron concentration. After phlebotomy, mice with hepatocyte-targeted Ncoa4 knockdown exhibited anemia and hypoferremia similar to control mice with intact Ncoa4 regulation but showed a markedly impaired ability to lower hepatic ferritin subunit levels and hepatic nonheme iron concentration. This impaired hepatic response was observed even when dietary iron was limited. In both human and murine hepatoma cell lines, treatment with chemicals that stabilize hypoxia inducible factor (HIF), including desferrioxamine, cobalt chloride, and dimethyloxalylglycine, raised NCOA4 messenger RNA. This NCOA4 messenger RNA induction occurred within 3 hours, preceded a rise in NCOA4 protein, and was attenuated in the setting of dual HIF-1α and HIF-2α knockdown. In summary, we show for the first time that NCOA4 plays a local role in facilitating iron mobilization from the liver after blood loss and that HIF regulates NCOA4 expression in cells of hepatic origin. Because the prolyl hydroxylases that regulate HIF stability are oxygen- and iron-dependent enzymes, our findings suggest a novel mechanism by which hypoxia and iron deficiency may modulate NCOA4 expression to impact iron homeostasis., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
12. EBV-Positive Primary Large B-Cell Lymphoma: The Role of Immunohistochemistry and XPO1 in the Diagnosis of Mediastinal Lymphomas.
- Author
-
Maracaja DLV, Puthenpura V, Pels SG, O'Malley DP, Sklar JL, Finberg KE, and Xu ML
- Subjects
- Adult, Diagnosis, Differential, Epstein-Barr Virus Infections genetics, Epstein-Barr Virus Infections pathology, High-Throughput Nucleotide Sequencing, Humans, Immunohistochemistry, Immunophenotyping, Karyopherins genetics, Karyopherins immunology, Lymphoma, Large B-Cell, Diffuse genetics, Lymphoma, Large B-Cell, Diffuse pathology, Male, Mutation genetics, Receptors, Cytoplasmic and Nuclear genetics, Receptors, Cytoplasmic and Nuclear immunology, Thymus Neoplasms genetics, Thymus Neoplasms pathology, Young Adult, Exportin 1 Protein, Biomarkers, Tumor metabolism, Epstein-Barr Virus Infections diagnosis, Herpesvirus 4, Human physiology, Karyopherins metabolism, Lymphoma, Large B-Cell, Diffuse diagnosis, Mediastinal Neoplasms diagnosis, Receptors, Cytoplasmic and Nuclear metabolism, Thymus Neoplasms diagnosis
- Abstract
Primary mediastinal (thymic) large B-cell lymphoma (PMBL) is described as almost always negative for Epstein-Barr virus (EBV). In the context of a mediastinal lymphoma, the distinction between PMBL, classical Hodgkin lymphoma, diffuse large B-cell lymphoma, and mediastinal gray-zone lymphoma can be very difficult; hence, EBV positivity often argues against PMBL. We present a 19-year-old man with mediastinal mass morphologically consistent with PMBL. The tumor expressed classic immunophenotype, including positivity for CD20, CD19, MAL, OCT2, BOB1, BCL6, CD79a, and subset positivity for CD30. However, the tumor was EBV-positive by in situ hybridization. Next-generation sequencing detected somatic mutations in XPO1 (E571K), SMARCB1 (L356fs), and MYCC (T73A). Although the immunophenotype and XPO1 mutation are characteristic of PMBL, EBV expression is uncommon. Since EBV positivity can occur in rare PMBLs, it should not be the deciding factor in the diagnosis. This is the first EBV-positive PMBL in which mutational profiling has been reported. Aside from providing diagnostic support, the finding of the XPO1 E571K mutation may suggest a targeted therapeutic option.
- Published
- 2020
- Full Text
- View/download PDF
13. IL-1β Drives Production of FGF-23 at the Onset of Chronic Kidney Disease in Mice.
- Author
-
McKnight Q, Jenkins S, Li X, Nelson T, Marlier A, Cantley LG, Finberg KE, and Fretz JA
- Subjects
- Animals, Female, Fibroblast Growth Factor-23, Fibroblast Growth Factors, Forkhead Transcription Factors, Humans, Interleukin-1beta, Kidney, Male, Mice, Minerals, Parathyroid Hormone, Phosphates, Renal Insufficiency, Chronic, Tumor Necrosis Factor-alpha
- Abstract
FGF-23 has arisen as an early biomarker of renal dysfunction, but at the onset of chronic kidney disease (CKD), data suggest that FGF-23 may be produced independently of the parathyroid hormone (PTH), 1,25(OH)
2 -vitamin D3 signaling axis. Iron status is inversely correlated to the level of circulating FGF-23, and improvement in iron bioavailability within patients correlates with a decrease in FGF-23. Alternately, recent evidence also supports a regulatory role of inflammatory cytokines in the modulation of FGF-23 expression. To determine the identity of the signal from the kidney-inducing upregulation of osteocytic FGF-23 at the onset of CKD, we utilized a mouse model of congenital CKD that fails to properly mature the glomerular capillary tuft. We profiled the sequential presentation of indicators of renal dysfunction, phosphate imbalance, and iron bioavailability and transport to identify the events that initiate osteocytic production of FGF-23 during the onset of CKD. We report here that elevations in circulating intact-FGF-23 coincide with the earliest indicators of renal dysfunction (P14), and precede changes in serum phosphate or iron homeostasis. Serum PTH was also not changed within the first month. Instead, production of the inflammatory protein IL-1β from the kidney and systemic elevation of it in the circulation matched the induction of FGF-23. IL-1β's ability to induce FGF-23 was confirmed on bone chips in culture and within mice in vivo. Furthermore, neutralizing antibody to IL-1β blocked FGF-23 expression in both our congenital model of CKD and a second nephrotoxic serum-mediated model. We conclude that early CKD resembles a situation of primary FGF-23 excess mediated by inflammation. These findings do not preclude that altered mineral availability or anemia can later modulate FGF-23 levels but find that in early CKD they are not the driving stimulus for the initial upregulation of FGF-23. © 2020 American Society for Bone and Mineral Research., (© 2020 American Society for Bone and Mineral Research.)- Published
- 2020
- Full Text
- View/download PDF
14. Yale Cancer Center Precision Medicine Tumor Board: new technology, new drugs, and the value of repeat testing.
- Author
-
Hafez N, Walther Z, Eder JP, Sklar JL, Gettinger SN, Finberg KE, and Goldberg SB
- Subjects
- Humans, Molecular Targeted Therapy trends, Neoplasms epidemiology, Neoplasms genetics, Neoplasms pathology, Neoplasms drug therapy, Oncology Service, Hospital trends, Precision Medicine trends
- Published
- 2020
- Full Text
- View/download PDF
15. Low iron promotes megakaryocytic commitment of megakaryocytic-erythroid progenitors in humans and mice.
- Author
-
Xavier-Ferrucio J, Scanlon V, Li X, Zhang PX, Lozovatsky L, Ayala-Lopez N, Tebaldi T, Halene S, Cao C, Fleming MD, Finberg KE, and Krause DS
- Subjects
- Anemia, Iron-Deficiency complications, Animals, Cell Proliferation, Humans, Iron, Megakaryocyte Progenitor Cells cytology, Megakaryocytes cytology, Mice, Mice, Knockout, Thrombocytosis etiology, Thrombocytosis metabolism, Anemia, Iron-Deficiency metabolism, Cell Differentiation physiology, Megakaryocyte Progenitor Cells metabolism, Megakaryocytes metabolism
- Abstract
The mechanisms underlying thrombocytosis in patients with iron deficiency anemia remain unknown. Here, we present findings that support the hypothesis that low iron biases the commitment of megakaryocytic (Mk)-erythroid progenitors (MEPs) toward the Mk lineage in both human and mouse. In MEPs of transmembrane serine protease 6 knockout (Tmprss6-/-) mice, which exhibit iron deficiency anemia and thrombocytosis, we observed a Mk bias, decreased labile iron, and decreased proliferation relative to wild-type (WT) MEPs. Bone marrow transplantation assays suggest that systemic iron deficiency, rather than a local role for Tmprss6-/- in hematopoietic cells, contributes to the MEP lineage commitment bias observed in Tmprss6-/- mice. Nontransgenic mice with acquired iron deficiency anemia also show thrombocytosis and Mk-biased MEPs. Gene expression analysis reveals that messenger RNAs encoding genes involved in metabolic, vascular endothelial growth factor, and extracellular signal-regulated kinase (ERK) pathways are enriched in Tmprss6-/- vs WT MEPs. Corroborating our findings from the murine models of iron deficiency anemia, primary human MEPs exhibit decreased proliferation and Mk-biased commitment after knockdown of transferrin receptor 2, a putative iron sensor. Signal transduction analyses reveal that both human and murine MEP have lower levels of phospho-ERK1/2 in iron-deficient conditions compared with controls. These data are consistent with a model in which low iron in the marrow environment affects MEP metabolism, attenuates ERK signaling, slows proliferation, and biases MEPs toward Mk lineage commitment., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
16. Going solo in iron transport.
- Author
-
Finberg KE
- Subjects
- Animals, Homeostasis, Iron, Mice, Erythropoiesis, Transferrin
- Published
- 2019
- Full Text
- View/download PDF
17. Genetic loss of Tmprss6 alters terminal erythroid differentiation in a mouse model of β-thalassemia intermedia.
- Author
-
Stagg DB, Whittlesey RL, Li X, Lozovatsky L, Gardenghi S, Rivella S, and Finberg KE
- Subjects
- Animals, Disease Models, Animal, Erythroid Cells pathology, Membrane Proteins metabolism, Mice, Mice, Knockout, Serine Endopeptidases metabolism, beta-Thalassemia genetics, beta-Thalassemia pathology, Cell Differentiation, Erythroid Cells metabolism, Membrane Proteins deficiency, Serine Endopeptidases deficiency, beta-Thalassemia metabolism
- Published
- 2019
- Full Text
- View/download PDF
18. Normalizing hepcidin predicts TMPRSS6 mutation status in patients with chronic iron deficiency.
- Author
-
Heeney MM, Guo D, De Falco L, Campagna DR, Olbina G, Kao PP, Schmitz-Abe K, Rahimov F, Gutschow P, Westerman K, Ostland V, Jackson T, Klaassen RJ, Markianos K, Finberg KE, Iolascon A, Westerman M, London WB, and Fleming MD
- Subjects
- Adult, Humans, Middle Aged, Prognosis, Anemia, Iron-Deficiency blood, Anemia, Iron-Deficiency genetics, Hepcidins blood, Iron blood, Membrane Proteins genetics, Mutation, Serine Endopeptidases genetics
- Published
- 2018
- Full Text
- View/download PDF
19. Analytical Validation of the Next-Generation Sequencing Assay for a Nationwide Signal-Finding Clinical Trial: Molecular Analysis for Therapy Choice Clinical Trial.
- Author
-
Lih CJ, Harrington RD, Sims DJ, Harper KN, Bouk CH, Datta V, Yau J, Singh RR, Routbort MJ, Luthra R, Patel KP, Mantha GS, Krishnamurthy S, Ronski K, Walther Z, Finberg KE, Canosa S, Robinson H, Raymond A, Le LP, McShane LM, Polley EC, Conley BA, Doroshow JH, Iafrate AJ, Sklar JL, Hamilton SR, and Williams PM
- Subjects
- Clinical Trials as Topic, Computational Biology methods, Genetic Variation, Genomics methods, Genomics standards, Humans, Quality Assurance, Health Care, Quality Control, Reproducibility of Results, Sensitivity and Specificity, Workflow, High-Throughput Nucleotide Sequencing methods, High-Throughput Nucleotide Sequencing standards, Neoplasms diagnosis, Neoplasms genetics
- Abstract
The National Cancer Institute-Molecular Analysis for Therapy Choice (NCI-MATCH) trial is a national signal-finding precision medicine study that relies on genomic assays to screen and enroll patients with relapsed or refractory cancer after standard treatments. We report the analytical validation processes for the next-generation sequencing (NGS) assay that was tailored for regulatory compliant use in the trial. The Oncomine Cancer Panel assay and the Personal Genome Machine were used in four networked laboratories accredited for the Clinical Laboratory Improvement Amendments. Using formalin-fixed paraffin-embedded clinical specimens and cell lines, we found that the assay achieved overall sensitivity of 96.98% for 265 known mutations and 99.99% specificity. High reproducibility in detecting all reportable variants was observed, with a 99.99% mean interoperator pairwise concordance across the four laboratories. The limit of detection for each variant type was 2.8% for single-nucleotide variants, 10.5% for insertion/deletions, 6.8% for large insertion/deletions (gap ≥4 bp), and four copies for gene amplification. The assay system from biopsy collection through reporting was tested and found to be fully fit for purpose. Our results indicate that the NCI-MATCH NGS assay met the criteria for the intended clinical use and that high reproducibility of a complex NGS assay is achievable across multiple clinical laboratories. Our validation approaches can serve as a template for development and validation of other NGS assays for precision medicine., (Copyright © 2017 American Society for Investigative Pathology and the Association for Molecular Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
20. Ironing out the role of Toll-like receptors.
- Author
-
Finberg KE
- Subjects
- Animals, Humans, Male, Cation Transport Proteins physiology, Hepcidins pharmacology, Inflammation physiopathology, Iron Deficiencies, Toll-Like Receptor 2 physiology, Toll-Like Receptor 6 physiology
- Published
- 2015
- Full Text
- View/download PDF
21. Iron-refractory iron deficiency anemia (IRIDA).
- Author
-
Heeney MM and Finberg KE
- Subjects
- Anemia, Iron-Deficiency genetics, Diagnosis, Differential, Humans, Anemia, Iron-Deficiency diagnosis, Anemia, Iron-Deficiency therapy
- Abstract
Iron deficiency anemia is a common global problem whose etiology is typically attributed to acquired inadequate dietary intake and/or chronic blood loss. However, in several kindreds multiple family members are affected with iron deficiency anemia that is unresponsive to oral iron supplementation and only partially responsive to parenteral iron therapy. The discovery that many of these cases harbor mutations in the TMPRSS6 gene led to the recognition that they represent a single clinical entity: iron-refractory iron deficiency anemia (IRIDA). This article reviews clinical features of IRIDA, recent genetic studies, and insights this disorder provides into the regulation of systemic iron homeostasis., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
22. Fibrosis-associated single-nucleotide polymorphisms in TGFB1 and CAV1 are not associated with the development of nephrogenic systemic fibrosis.
- Author
-
Le LP, Garibyan L, Lara D, Finberg KE, Iafrate AJ, Duncan LM, Kay J, and Nazarian RM
- Subjects
- Adult, Aged, Case-Control Studies, Chi-Square Distribution, Codon, Contrast Media adverse effects, Female, Fibrosis, Gadolinium adverse effects, Gene Frequency, Genetic Predisposition to Disease, Humans, Introns, Male, Middle Aged, Multivariate Analysis, Nephrogenic Fibrosing Dermopathy chemically induced, Nephrogenic Fibrosing Dermopathy pathology, Phenotype, Risk Factors, Caveolin 1 genetics, Nephrogenic Fibrosing Dermopathy genetics, Polymorphism, Single Nucleotide, Transforming Growth Factor beta1 genetics
- Abstract
Objective: To test the hypothesis that the subset of patients with impaired renal function who are exposed to gadolinium-containing contrast agents (GCCAs) and develop nephrogenic systemic fibrosis (NSF) have a genetic predisposition for disease., Methods: We examined whether an intronic single-nucleotide polymorphism (SNP) in caveolin-1 (CAV1 rs4730751) and 2 coding SNPs in transforming growth factor-beta 1 (TGFB1 rs1800471, codon 25; and rs1800470, codon 10) were associated with the NSF phenotype., Results: Forty-one patients with a history of chronic kidney disease and GCCA administration were studied, including NSF cases (n = 17) and control subjects (n = 24) without clinical or histological evidence of NSF. No significant differences in the genotype frequencies at these SNPs in TGFB1 and CAV1 were found between patients with NSF and subjects without NSF., Conclusions: We conclude that polymorphisms in the genes encoding TGFB1 and CAV1 previously associated with the development and progression of fibrosis in several organ systems are not associated with development of NSF in this cohort of patients with renal impairment after GCCA exposure.
- Published
- 2013
- Full Text
- View/download PDF
23. Regulation of systemic iron homeostasis.
- Author
-
Finberg KE
- Subjects
- Erythropoiesis physiology, Hepcidins, Humans, Signal Transduction physiology, Antimicrobial Cationic Peptides physiology, Bone Morphogenetic Proteins physiology, Homeostasis physiology, Iron metabolism, Iron Metabolism Disorders physiopathology, Iron, Dietary metabolism
- Abstract
Purpose of Review: The circulating peptide hepcidin modulates systemic iron balance by limiting the absorption of dietary iron and the release of iron from macrophage stores. Recent studies conducted in humans, animal models, and tissue culture systems have enhanced our understanding of the molecular mechanisms by which hepcidin levels are altered in response to iron stores, inflammation, and erythropoietic activity., Recent Findings: The bone morphogenetic protein (BMP) type I receptors ALK2 and ALK3 play key, nonredundant roles in mediating hepcidin synthesis through the BMP signaling pathway. Actions of the hereditary hemochromatosis proteins HFE and transferrin receptor 2 may intersect with the BMP pathway. Hepcidin induction in response to inflammation requires cooperative BMP signaling. A variety of innate immune and infectious stimuli induce hepcidin expression. The hypoxia inducible factor pathway appears to suppress hepcidin indirectly through the capacity of erythropoietin to stimulate erythropoiesis., Summary: Study of the molecular mechanisms underlying the regulation of hepcidin synthesis has revealed complex biology. Improved understanding of the signaling pathways involved in hepcidin regulation may contribute to improved therapeutic outcomes for patients with genetic and acquired disorders that impact systemic iron balance.
- Published
- 2013
- Full Text
- View/download PDF
24. Striking the target in iron overload disorders.
- Author
-
Finberg KE
- Subjects
- Animals, Female, Male, Hemochromatosis therapy, Membrane Proteins genetics, Serine Endopeptidases genetics, beta-Thalassemia therapy
- Abstract
The liver, a major site of body iron stores, mediates key responses that preserve systemic iron homeostasis. In this issue of the JCI, Guo et al. demonstrate that administration of antisense oligonucleotides that reduce expression of Tmprss6, a hepatic protein that plays an essential role in maintaining iron balance, can attenuate disease severity in mouse models of human iron overload disorders. These data reveal the potential of novel TMPRSS6-targeted therapies for the treatment of clinical conditions such as hereditary hemochromatosis and β-thalassemia.
- Published
- 2013
- Full Text
- View/download PDF
25. Altered V-ATPase expression in renal intercalated cells isolated from B1 subunit-deficient mice by fluorescence-activated cell sorting.
- Author
-
Vedovelli L, Rothermel JT, Finberg KE, Wagner CA, Azroyan A, Hill E, Breton S, Brown D, and Paunescu TG
- Subjects
- Animals, Cell Membrane metabolism, Epithelial Cells cytology, Flow Cytometry, Kidney cytology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Protein Subunits genetics, Protein Subunits metabolism, Up-Regulation, Vacuolar Proton-Translocating ATPases metabolism, Epithelial Cells metabolism, Kidney metabolism, Vacuolar Proton-Translocating ATPases genetics
- Abstract
Unlike human patients with mutations in the 56-kDa B1 subunit isoform of the vacuolar proton-pumping ATPase (V-ATPase), B1-deficient mice (Atp6v1b1(-/-)) do not develop metabolic acidosis under baseline conditions. This is due to the insertion of V-ATPases containing the alternative B2 subunit isoform into the apical membrane of renal medullary collecting duct intercalated cells (ICs). We previously reported that quantitative Western blots (WBs) from whole kidneys showed similar B2 protein levels in Atp6v1b1(-/-) and wild-type mice (Păunescu TG, Russo LM, Da Silva N, Kovacikova J, Mohebbi N, Van Hoek AN, McKee M, Wagner CA, Breton S, Brown D. Am J Physiol Renal Physiol 293: F1915-F1926, 2007). However, WBs from renal medulla (including outer and inner medulla) membrane and cytosol fractions reveal a decrease in the levels of the ubiquitous V-ATPase E1 subunit. To compare V-ATPase expression specifically in ICs from wild-type and Atp6v1b1(-/-) mice, we crossed mice in which EGFP expression is driven by the B1 subunit promoter (EGFP-B1(+/+) mice) with Atp6v1b1(-/-) mice to generate novel EGFP-B1(-/-) mice. We isolated pure IC populations by fluorescence-assisted cell sorting from EGFP-B1(+/+) and EGFP-B1(-/-) mice to compare their V-ATPase subunit protein levels. We report that V-ATPase A, E1, and H subunits are all significantly downregulated in EGFP-B1(-/-) mice, while the B2 protein level is considerably increased in these animals. We conclude that under baseline conditions B2 upregulation compensates for the lack of B1 and is sufficient to maintain basal acid-base homeostasis, even when other V-ATPase subunits are downregulated.
- Published
- 2013
- Full Text
- View/download PDF
26. Iron refractory iron deficiency anemia: presentation with hyperferritinemia and response to oral iron therapy.
- Author
-
Khuong-Quang DA, Schwartzentruber J, Westerman M, Lepage P, Finberg KE, Majewski J, and Jabado N
- Subjects
- Administration, Oral, Adolescent, Anemia, Hypochromic diagnosis, Anemia, Iron-Deficiency diagnosis, Anemia, Refractory diagnosis, Child, Child, Preschool, Exome genetics, Female, Follow-Up Studies, Genetic Carrier Screening, Genotype, Humans, Long-Term Care, Male, Mutation, Missense, Anemia, Hypochromic drug therapy, Anemia, Hypochromic genetics, Anemia, Iron-Deficiency drug therapy, Anemia, Iron-Deficiency genetics, Anemia, Refractory drug therapy, Anemia, Refractory genetics, Chromosome Aberrations, DNA Mutational Analysis, Ferritins blood, Iron administration & dosage, Membrane Proteins genetics, Serine Endopeptidases genetics
- Abstract
Iron-refractory iron-deficiency anemia (IRIDA) is an autosomal recessive disorder caused by mutations in TMPRSS6. Patients have hypochromic microcytic anemia refractory to oral iron and are only partially responsive to parenteral iron administration. We report a French-Canadian kindred in which 2 siblings presented in early childhood with severe microcytic anemia, hypoferremia, and hyperferritinemia. Both children have been successfully treated solely with low-dose oral iron since diagnosis. Clinical and biological presentation did not fit any previously described genetic iron-deficiency anemia. Whole exome sequencing identified in both patients compound heterozygous mutations of TMPRSS6 leading to p.G442R and p.E522K, 2 mutations previously reported to cause classic IRIDA, and no additional mutations in known iron-regulatory genes. Thus, the phenotype associated with the unique combination of mutations uncovered in both patients expands the spectrum of disease associated with TMPRSS6 mutations to include iron deficiency anemia that is accompanied by hyperferritinemia at initial presentation and is responsive to continued oral iron therapy. Our results have implications for genetic testing in early childhood iron deficiency anemia. Importantly, they emphasize that whole exome sequencing can be used as a diagnostic tool and greatly facilitate the elucidation of the genetic basis of unusual clinical presentations, including hypomorphic mutations or compound heterozygosity leading to different phenotypes in known Mendelian diseases.
- Published
- 2013
- Full Text
- View/download PDF
27. Clinicopathologic and molecular profiles of microsatellite unstable Barrett Esophagus-associated adenocarcinoma.
- Author
-
Farris AB 3rd, Demicco EG, Le LP, Finberg KE, Miller J, Mandal R, Fukuoka J, Cohen C, Gaissert HA, Zukerberg LR, Lauwers GY, Iafrate AJ, and Mino-Kenudson M
- Subjects
- Adenocarcinoma pathology, Aged, Barrett Esophagus complications, Barrett Esophagus genetics, Esophageal Neoplasms pathology, Female, Humans, Immunohistochemistry, In Situ Hybridization, Lymphocytes, Tumor-Infiltrating pathology, Male, Neoplasm Staging, Polymerase Chain Reaction, Adenocarcinoma genetics, Barrett Esophagus pathology, Esophageal Neoplasms genetics, Microsatellite Instability
- Abstract
Microsatellite instability (MSI) has been reported in various tumors, with colon cancer as the prototype. However, little is known about MSI in Barrett esophagus (BE)-associated adenocarcinoma. Thus, the aim of this study was to compare the clinicopathologic and molecular features of BE-associated adenocarcinomas with and without MSI. The study cohort consisted of 76 patients with BE-associated adenocarcinomas (66 male, 10 female), with a mean age of 65.1 years. Immunohistochemistry (IHC) for MLH1, MSH2, MSH6, PMS2, and CD3 and in situ hybridization for Epstein-Barr virus-encoded RNA were performed. MLH1 and PMS2 expression was lost by IHC in 5 cases (6.6%); of these, 5 showed high-level MSI (MSI-H) by polymerase chain reaction assay, and 4 showed hMLH1 promoter methylation. Histologically, tumors with MSI-H were heterogenous and included conventional adenocarcinomas with tumor-infiltrating lymphocytes (n=1), medullary carcinoma (n=2), signet ring cells (n=1), and signet ring cell and mucinous components (n=1). Compared with tumors negative for MSI by IHC, BE-associated adenocarcinomas with MSI-H were associated with older patient age (P=0.0060), lymphovascular invasion (P=0.027), and significantly larger numbers of tumor-infiltrating lymphocytes (P<0.0001). However, there was no statistical difference in overall survival between the 2 groups (P=0.285). In conclusion, MSI-H is uncommon in BE-associated adenocarcinomas, but is associated with clinicopathologic features fairly similar to sporadic microsatellite unstable colorectal cancers. Given the growing evidence that indicates lack of benefits from adjuvant therapy with fluorouracil in the colonic counterpart, it may be important to identify MSI-H in BE-associated adenocarcinomas.
- Published
- 2011
- Full Text
- View/download PDF
28. Tmprss6 is a genetic modifier of the Hfe-hemochromatosis phenotype in mice.
- Author
-
Finberg KE, Whittlesey RL, and Andrews NC
- Subjects
- Animals, Antimicrobial Cationic Peptides metabolism, Female, Genotype, Hemochromatosis Protein, Hepcidins, Heterozygote, Histocompatibility Antigens Class I metabolism, Homozygote, Humans, Liver physiology, Male, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, Phenotype, Serine Endopeptidases metabolism, Signal Transduction physiology, Up-Regulation physiology, Hemochromatosis genetics, Hemochromatosis metabolism, Hemochromatosis physiopathology, Histocompatibility Antigens Class I genetics, Iron metabolism, Membrane Proteins genetics, Serine Endopeptidases genetics
- Abstract
The hereditary hemochromatosis protein HFE promotes the expression of hepcidin, a circulating hormone produced by the liver that inhibits dietary iron absorption and macrophage iron release. HFE mutations are associated with impaired hepatic bone morphogenetic protein (BMP)/SMAD signaling for hepcidin production. TMPRSS6, a transmembrane serine protease mutated in iron-refractory iron deficiency anemia, inhibits hepcidin expression by dampening BMP/SMAD signaling. In the present study, we used genetic approaches in mice to examine the relationship between Hfe and Tmprss6 in the regulation of systemic iron homeostasis. Heterozygous loss of Tmprss6 in Hfe(-/-) mice reduced systemic iron overload, whereas homozygous loss caused systemic iron deficiency and elevated hepatic expression of hepcidin and other Bmp/Smad target genes. In contrast, neither genetic loss of Hfe nor hepatic Hfe overexpression modulated the hepcidin elevation and systemic iron deficiency of Tmprss6(-/-) mice. These results indicate that genetic loss of Tmprss6 increases Bmp/Smad signaling in an Hfe-independent manner that can restore Bmp/Smad signaling in Hfe(-/-) mice. Furthermore, these results suggest that natural genetic variation in the human ortholog TMPRSS6 might modify the clinical penetrance of HFE-associated hereditary hemochromatosis, raising the possibility that pharmacologic inhibition of TMPRSS6 could attenuate iron loading in this disorder.
- Published
- 2011
- Full Text
- View/download PDF
29. Unraveling mechanisms regulating systemic iron homeostasis.
- Author
-
Finberg KE
- Subjects
- Antimicrobial Cationic Peptides metabolism, Erythropoiesis, Hepcidins, Humans, Inflammation pathology, Models, Biological, Homeostasis, Iron metabolism
- Abstract
Systemic iron balance must be tightly regulated to prevent the deleterious effects of iron deficiency and iron overload. Hepcidin, a circulating hormone that is synthesized by the liver, has emerged as a key regulator of systemic iron homeostasis. Hepcidin inhibits the absorption of dietary iron from the intestine and the release of iron derived from red blood cells from macrophages. Therefore, variation in hepcidin levels modifies the total amount of iron stored in the body and the availability of iron for erythropoiesis. The production of hepcidin by the liver is modulated by multiple physiological stimuli, including iron loading, inflammation, and erythropoietic activity. Investigation of the functions of the gene products mutated in inherited iron disorders using tissue-culture systems and animal models has provided valuable insights into the mechanisms by which these hepcidin responses are mediated. This review focuses on recent advances in our understanding of the molecular mechanisms underlying the regulation of systemic iron homeostasis.
- Published
- 2011
- Full Text
- View/download PDF
30. Down-regulation of Bmp/Smad signaling by Tmprss6 is required for maintenance of systemic iron homeostasis.
- Author
-
Finberg KE, Whittlesey RL, Fleming MD, and Andrews NC
- Subjects
- Anemia, Iron-Deficiency metabolism, Anemia, Iron-Deficiency pathology, Animals, Blotting, Western, Bone Morphogenetic Proteins genetics, Down-Regulation, Female, GPI-Linked Proteins, Hemochromatosis Protein, Hepatocytes metabolism, Hepcidins, Homeostasis, Inhibitor of Differentiation Protein 1 metabolism, Liver metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, RNA, Messenger genetics, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Antimicrobial Cationic Peptides metabolism, Bone Morphogenetic Proteins metabolism, Iron metabolism, Membrane Proteins physiology, Serine Endopeptidases physiology, Smad Proteins metabolism
- Abstract
Iron-refractory, iron-deficiency anemia (IRIDA) is a familial disorder characterized by iron deficiency anemia unresponsive to oral iron treatment but partially responsive to intravenous iron therapy. Previously, we showed that IRIDA patients harbor loss-of-function mutations in TMPRSS6, a type II transmembrane serine protease primarily expressed by the liver. Both humans and mice with TMPRSS6 mutations show inappropriately elevated levels of the iron-regulatory hormone hepcidin, suggesting that TMPRSS6 acts to negatively regulate hepcidin expression. Here we investigate the relationship between Tmprss6 and the bone morphogenetic protein (BMP)-Smad signaling pathway, a key pathway promoting hepcidin transcription in hepatocytes. We show that livers from mice deficient for Tmprss6 have decreased iron stores and decreased Bmp6 mRNA, but markedly increased mRNA for Id1, a target gene of Bmp6 signaling. In contrast, mice deficient for both Tmprss6 and hemojuvelin (Hjv), a BMP coreceptor that augments hepcidin expression in hepatocytes, showed markedly decreased hepatic levels of hepcidin and Id1 mRNA, markedly increased hepatic Bmp6 mRNA levels, and systemic iron overload similar to mice deficient for Hjv alone. These findings suggest that down-regulation of Bmp/Smad signaling by Tmprss6 is required for regulation of hepcidin expression and maintenance of systemic iron homeostasis.
- Published
- 2010
- Full Text
- View/download PDF
31. Iron-refractory iron deficiency anemia.
- Author
-
Finberg KE
- Subjects
- Administration, Oral, Anemia, Iron-Deficiency blood, Anemia, Iron-Deficiency genetics, Anemia, Iron-Deficiency physiopathology, Anemia, Refractory blood, Anemia, Refractory genetics, Anemia, Refractory physiopathology, Antimicrobial Cationic Peptides blood, Child, Child, Preschool, Female, GPI-Linked Proteins, Hemochromatosis Protein, Hepcidins, Humans, Infant, Infusions, Parenteral, Iron blood, Male, Membrane Proteins genetics, Membrane Proteins metabolism, Mutation, Serine Endopeptidases genetics, Treatment Failure, Up-Regulation, Anemia, Iron-Deficiency drug therapy, Anemia, Refractory drug therapy, Hematinics administration & dosage, Iron metabolism, Iron Compounds administration & dosage, Liver metabolism
- Abstract
Iron-refractory iron deficiency anemia (IRIDA) is an autosomal recessive disorder characterized by iron deficiency anemia unresponsive to oral iron treatment but partially responsive to parenteral iron therapy. IRIDA has recently been shown to be caused by mutations in the gene TMPRSS6, which encodes a transmembrane serine protease (also known as matriptase-2) expressed by the liver. IRIDA patients show inappropriately elevated levels of hepcidin, a circulating hormone produced by the liver that inhibits both iron absorption from the intestine and iron release from macrophage stores. Recent studies suggest that TMPRSS6 normally acts to downregulate hepcidin expression by cleaving hemojuvelin, a membrane-bound protein that promotes hepcidin signaling in hepatocytes. A discussion of the clinical presentation of IRIDA, the molecular genetics of this disorder, and recent studies elucidating the underlying pathophysiology are presented.
- Published
- 2009
- Full Text
- View/download PDF
32. Iron homeostasis: casting new roles.
- Author
-
Finberg KE
- Published
- 2008
- Full Text
- View/download PDF
33. Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA).
- Author
-
Finberg KE, Heeney MM, Campagna DR, Aydinok Y, Pearson HA, Hartman KR, Mayo MM, Samuel SM, Strouse JJ, Markianos K, Andrews NC, and Fleming MD
- Subjects
- Adolescent, Anemia, Iron-Deficiency drug therapy, Anemia, Iron-Deficiency metabolism, Child, Child, Preschool, Female, Homeostasis genetics, Humans, Infant, Iron therapeutic use, Male, Protein Structure, Tertiary genetics, Anemia, Iron-Deficiency genetics, Germ-Line Mutation, Iron metabolism, Membrane Proteins genetics, Serine Endopeptidases genetics
- Abstract
Iron deficiency is usually attributed to chronic blood loss or inadequate dietary intake. Here, we show that iron deficiency anemia refractory to oral iron therapy can be caused by germline mutations in TMPRSS6, which encodes a type II transmembrane serine protease produced by the liver that regulates the expression of the systemic iron regulatory hormone hepcidin. These findings demonstrate that TMPRSS6 is essential for normal systemic iron homeostasis in humans.
- Published
- 2008
- Full Text
- View/download PDF
34. Mucinous differentiation correlates with absence of EGFR mutation and presence of KRAS mutation in lung adenocarcinomas with bronchioloalveolar features.
- Author
-
Finberg KE, Sequist LV, Joshi VA, Muzikansky A, Miller JM, Han M, Beheshti J, Chirieac LR, Mark EJ, and Iafrate AJ
- Subjects
- Adenocarcinoma, Bronchiolo-Alveolar diagnosis, Adenocarcinoma, Bronchiolo-Alveolar genetics, Adenocarcinoma, Mucinous diagnosis, Aged, Aged, 80 and over, Cell Differentiation genetics, DNA Mutational Analysis, Disease Progression, Female, Gene Dosage, Humans, In Situ Hybridization, Fluorescence, Lung Neoplasms diagnosis, Male, Middle Aged, Prognosis, Retrospective Studies, Adenocarcinoma, Bronchiolo-Alveolar pathology, Adenocarcinoma, Mucinous genetics, Adenocarcinoma, Mucinous pathology, Genes, erbB-1, Genes, ras, Lung Neoplasms genetics, Lung Neoplasms pathology, Mutation
- Abstract
Somatic mutations in the epidermal growth factor receptor gene (EGFR) are detected in a subset of lung adenocarcinomas, particularly bronchioloalveolar carcinoma (BAC) and adenocarcinoma with bronchioloalveolar features (AWBF), and correlate with clinical response to tyrosine kinase inhibitors (TKIs). In contrast, lung adenocarcinomas refractory to TKIs often have activating mutations in KRAS but lack EGFR mutations. Some adenocarcinomas have mucinous histology, but the clinical and molecular significance of the mucinous pattern is less well studied. We analyzed 43 BAC and AWBF tumors submitted for EGFR mutation testing to identify histopathological features that predicted EGFR or KRAS mutations. EGFR mutations were detected in 14 of 30 (47%) nonmucinous tumors, whereas 0 of 13 mucinous tumors harbored an EGFR mutation (P = 0.003). Missense mutations in KRAS codon 12 were detected in six of seven (86%) mucinous adenocarcinomas but only 3 of 18 (17%) nonmucinous adenocarcinomas (P = 0.003). Thus, in BAC/AWBF mucinous differentiation was significantly correlated with the absence of EGFR mutation and presence of KRAS mutation, suggesting that mucinous BACs/AWBFs are unlikely to respond to TKIs. Therefore, our data suggest that EGFR sequence analysis could be avoided in BAC/AWBF when true mucinous morphology is identified, avoiding the associated testing costs.
- Published
- 2007
- Full Text
- View/download PDF
35. The connecting tubule is the main site of the furosemide-induced urinary acidification by the vacuolar H+-ATPase.
- Author
-
Kovacikova J, Winter C, Loffing-Cueni D, Loffing J, Finberg KE, Lifton RP, Hummler E, Rossier B, and Wagner CA
- Subjects
- Acid-Base Equilibrium physiology, Amiloride pharmacokinetics, Amiloride pharmacology, Animals, Diuretics pharmacokinetics, Epithelial Sodium Channels genetics, Epithelial Sodium Channels metabolism, Furosemide pharmacokinetics, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Gene Expression Regulation, Enzymologic drug effects, Gene Expression Regulation, Enzymologic genetics, Hydrochlorothiazide pharmacokinetics, Hydrochlorothiazide pharmacology, Hydrogen-Ion Concentration, Kidney Tubules, Collecting metabolism, Kidney Tubules, Distal metabolism, Metabolic Clearance Rate drug effects, Metabolic Clearance Rate physiology, Mice, Mice, Knockout, Nephrons drug effects, Nephrons physiology, Proton-Translocating ATPases genetics, Water-Electrolyte Balance physiology, Acid-Base Equilibrium drug effects, Diuretics pharmacology, Furosemide pharmacology, Kidney Tubules, Distal drug effects, Proton-Translocating ATPases metabolism
- Abstract
Final urinary acidification is achieved by electrogenic vacuolar H(+)-ATPases expressed in acid-secretory intercalated cells (ICs) in the connecting tubule (CNT) and the cortical (CCD) and initial medullary collecting duct (MCD), respectively. Electrogenic Na(+) reabsorption via epithelial Na(+) channels (ENaCs) in the apical membrane of the segment-specific CNT and collecting duct cells may promote H(+)-ATPases-mediated proton secretion by creating a more lumen-negative voltage. The exact localization where this supposed functional interaction takes place is unknown. We used several mouse models performing renal clearance experiments and assessed the furosemide-induced urinary acidification. Increasing Na(+) delivery to the CNT and CCD by blocking Na(+) reabsorption in the thick ascending limb with furosemide enhanced urinary acidification and net acid excretion. This effect of furosemide was abolished with amiloride or benzamil blocking ENaC action. In mice deficient for the IC-specific B1 subunit of the vacuolar H(+)-ATPase, furosemide led to only a small urinary acidification. In contrast, in mice with a kidney-specific inactivation of the alpha subunit of ENaC in the CCD and MCD, but not in the CNT, furosemide alone and in combination with hydrochlorothiazide induced normal urinary acidification. These results suggest that the B1 vacuolar H(+)-ATPase subunit is necessary for the furosemide-induced acute urinary acidification. Loss of ENaC channels in the CCD and MCD does not affect this acidification. Thus, functional expression of ENaC channels in the CNT is sufficient for furosemide-stimulated urinary acidification and identifies the CNT as a major segment in electrogenic urinary acidification.
- Published
- 2006
- Full Text
- View/download PDF
36. The B1-subunit of the H(+) ATPase is required for maximal urinary acidification.
- Author
-
Finberg KE, Wagner CA, Bailey MA, Paunescu TG, Breton S, Brown D, Giebisch G, Geibel JP, and Lifton RP
- Subjects
- Acidosis, Renal Tubular genetics, Acidosis, Renal Tubular urine, Acids administration & dosage, Animals, Humans, Hydrogen-Ion Concentration, Kidney Medulla enzymology, Mice, Mice, Knockout, Protein Subunits genetics, Protein Subunits metabolism, Protons, Vacuolar Proton-Translocating ATPases deficiency, Acidosis, Renal Tubular enzymology, Acids urine, Cell Membrane enzymology, Kidney Tubules, Collecting enzymology, Vacuolar Proton-Translocating ATPases metabolism
- Abstract
The multisubunit vacuolar-type H(+)ATPases mediate acidification of various intracellular organelles and in some tissues mediate H(+) secretion across the plasma membrane. Mutations in the B1-subunit of the apical H(+)ATPase that secretes protons in the distal nephron cause distal renal tubular acidosis in humans, a condition characterized by metabolic acidosis with an inappropriately alkaline urine. To examine the detailed cellular and organismal physiology resulting from this mutation, we have generated mice deficient in the B1-subunit (Atp6v1b1(-/-) mice). Urine pH is more alkaline and metabolic acidosis is more severe in Atp6v1b1(-/-) mice after oral acid challenge, demonstrating a failure of normal urinary acidification. In Atp6v1b1(-/-) mice, the normal urinary acidification induced by a lumen-negative potential in response to furosemide infusion is abolished. After an acute intracellular acidification, Na(+)-independent pH recovery rates of individual Atp6v1b1(-/-) intercalated cells of the cortical collecting duct are markedly reduced and show no further decrease after treatment with the selective H(+)ATPase inhibitor concanamycin. Apical expression of the alternative B-subunit isoform, B2, is increased in Atp6v1b1(-/-) medulla and colocalizes with the H(+)ATPase E-subunit; however, the greater severity of metabolic acidosis in Atp6v1b1(-/-) mice after oral acid challenge indicates that the B2-subunit cannot fully functionally compensate for the loss of B1. Our results indicate that the B1 isoform is the major B-subunit isoform that incorporates into functional, plasma membrane H(+)ATPases in intercalated cells of the cortical collecting duct and is required for maximal urinary acidification.
- Published
- 2005
- Full Text
- View/download PDF
37. Renal vacuolar H+-ATPase.
- Author
-
Wagner CA, Finberg KE, Breton S, Marshansky V, Brown D, and Geibel JP
- Subjects
- Animals, Cell Membrane physiology, Cell Membrane ultrastructure, Humans, Kidney ultrastructure, Kidney enzymology, Vacuolar Proton-Translocating ATPases physiology
- Abstract
Vacuolar H(+)-ATPases are ubiquitous multisubunit complexes mediating the ATP-dependent transport of protons. In addition to their role in acidifying the lumen of various intracellular organelles, vacuolar H(+)-ATPases fulfill special tasks in the kidney. Vacuolar H(+)-ATPases are expressed in the plasma membrane in the kidney almost along the entire length of the nephron with apical and/or basolateral localization patterns. In the proximal tubule, a high number of vacuolar H(+)-ATPases are also found in endosomes, which are acidified by the pump. In addition, vacuolar H(+)-ATPases contribute to proximal tubular bicarbonate reabsorption. The importance in final urinary acidification along the collecting system is highlighted by monogenic defects in two subunits (ATP6V0A4, ATP6V1B1) of the vacuolar H(+)-ATPase in patients with distal renal tubular acidosis. The activity of vacuolar H(+)-ATPases is tightly regulated by a variety of factors such as the acid-base or electrolyte status. This regulation is at least in part mediated by various hormones and protein-protein interactions between regulatory proteins and multiple subunits of the pump.
- Published
- 2004
- Full Text
- View/download PDF
38. Localization and regulation of the ATP6V0A4 (a4) vacuolar H+-ATPase subunit defective in an inherited form of distal renal tubular acidosis.
- Author
-
Stehberger PA, Schulz N, Finberg KE, Karet FE, Giebisch G, Lifton RP, Geibel JP, and Wagner CA
- Subjects
- Acid-Base Equilibrium, Acidosis, Renal Tubular enzymology, Animals, Humans, Kidney chemistry, Male, Mice, Mice, Inbred C57BL, Proteins, Vacuolar Proton-Translocating ATPases analysis, Acidosis, Renal Tubular genetics, Proton-Translocating ATPases, Vacuolar Proton-Translocating ATPases metabolism
- Abstract
Vacuolar-type H(+)-ATPases (V-H(+)-ATPases) are the major H(+)-secreting protein in the distal portion of the nephron and are involved in net H(+) secretion (bicarbonate generation) or H(+) reabsorption (net bicarbonate secretion). In addition, V-H(+)-ATPases are involved in HCO(3)(-) reabsorption in the proximal tubule and distal tubule. V-H(+)-ATPases consist of at least 13 subunits, the functions of which have not all been elucidated. Mutations in the accessory ATP6V0A4 (a4 isoform) subunit have recently been shown to cause an inherited form of distal renal tubular acidosis in humans. Here, the localization of this subunit in human and mouse kidney was studied and the regulation of expression and localization of this subunit in mouse kidney in response to acid-base and electrolyte intake was investigated. Reverse transcription-PCR on dissected mouse nephron segments amplified a4-specific transcripts in proximal tubule, loop of Henle, distal convoluted tubule, and cortical and medullary collecting duct. a4 protein was localized by immunohistochemistry to the apical compartment of the proximal tubule (S1/S2 segment), the loop of Henle, the intercalated cells of the distal convoluted tubule, the connecting segment, and all intercalated cells of the entire collecting duct in human and mouse kidney. All types of intercalated cells expressed a4. NH(4)Cl or NaHCO(3) loading for 24 h, 48 h, or 7 d as well as K(+) depletion for 7 and 14 d had no influence on a4 protein expression levels in either cortex or medulla as determined by Western blotting. Immunohistochemistry, however, demonstrated a subcellular redistribution of a4 in response to the different stimuli. NH(4)Cl and K(+) depletion led to a pronounced apical staining in the connecting segment, cortical collecting duct, and outer medullary collecting duct, whereas NaHCO(3) loading caused a stronger bipolar staining in the cortical collecting duct. Taken together, these results demonstrate a4 expression in the proximal tubule, loop of Henle, distal tubule, and collecting duct and suggest that under conditions in which increased V-H(+)-ATPase activity is required, a4 is regulated by trafficking but not protein expression. This may allow for the rapid adaptation of V-H(+)-ATPase activity to altered acid-base intake to achieve systemic pH homeostasis. The significance of a4 expression in the proximal tubule in the context of distal renal tubular acidosis will require further clarification.
- Published
- 2003
- Full Text
- View/download PDF
39. Molecular cloning and characterization of Atp6v1b1, the murine vacuolar H+ -ATPase B1-subunit.
- Author
-
Finberg KE, Wagner CA, Stehberger PA, Geibel JP, and Lifton RP
- Subjects
- 5' Flanking Region genetics, Amino Acid Sequence, Animals, Antibody Specificity, Base Sequence, Cloning, Molecular, DNA chemistry, DNA genetics, DNA, Complementary chemistry, DNA, Complementary genetics, Epididymis enzymology, Gene Expression Regulation, Enzymologic, Humans, Immune Sera immunology, Immunohistochemistry, Isoenzymes genetics, Isoenzymes metabolism, Kidney enzymology, Male, Mice, Mice, Inbred Strains, Molecular Sequence Data, Phylogeny, Protein Subunits genetics, Protein Subunits immunology, Protein Subunits metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Sequence Alignment, Sequence Analysis, DNA, Sequence Homology, Amino Acid, Sequence Homology, Nucleic Acid, Vacuolar Proton-Translocating ATPases metabolism, Vacuolar Proton-Translocating ATPases genetics
- Abstract
The multisubunit vacuolar-type proton-translocating ATPases (H(+)-ATPases) mediate the acidification of various intracellular organelles. In a subset of tissues, they also mediate H(+) secretion at the plasma membrane. Two isoforms of the H(+)-ATPase B-subunit exist in humans; we have shown that mutations in ATP6V1B1, encoding the B1-isoform, cause the clinical condition distal renal tubular acidosis. Here we report the cloning and characterization of murine Atp6v1b1, which encodes a 513-amino acid (aa) protein with 93% identity to human ATP6V1B1. Genomic organization is conserved between the murine and human H(+)-ATPase B1-subunits, and Atp6v1b1 maps to a region of mouse chromosome 6 syntenic to human 2p13, the location of ATP6V1B1. Northern blotting detects a 2.2-kb Atp6v1b1 transcript in the kidney and testis, but not other major organs. In mouse kidney, the B1-subunit localizes to intercalated cells of the cortical and medullary collecting duct. B1 protein levels were not increased in either mouse renal cortex or medulla after either 2 or 7 days of oral acid loading. These results demonstrate that Atp6v1b1 encodes the murine ortholog of human ATP6V1B1 and provides a tool for future development of animal models based on manipulation of the Atp6v1b1 genomic locus.
- Published
- 2003
- Full Text
- View/download PDF
40. Regulation of the expression of the Cl-/anion exchanger pendrin in mouse kidney by acid-base status.
- Author
-
Wagner CA, Finberg KE, Stehberger PA, Lifton RP, Giebisch GH, Aronson PS, and Geibel JP
- Subjects
- Alkalosis chemically induced, Alkalosis metabolism, Animals, Aquaporin 2, Aquaporin 6, Aquaporins analysis, Aquaporins biosynthesis, Carrier Proteins analysis, Cell Polarity, Cytosol metabolism, Kidney Cortex cytology, Kidney Tubules, Collecting cytology, Kidney Tubules, Collecting metabolism, Male, Mice, Mice, Inbred C57BL, Potassium, Dietary pharmacology, Sodium Bicarbonate pharmacology, Sulfate Transporters, Acid-Base Equilibrium physiology, Carrier Proteins biosynthesis, Kidney Cortex metabolism, Membrane Transport Proteins
- Abstract
Background: Pendrin belongs to a superfamily of Cl-/anion exchangers and is expressed in the inner ear, the thyroid gland, and the kidney. In humans, mutations in pendrin cause Pendred syndrome characterized by sensorineural deafness and goiter. Recently pendrin has been localized to the apical side of non-type A intercalated cells of the cortical collecting duct, and reduced bicarbonate secretion was demonstrated in a pendrin knockout mouse model. To investigate a possible role of pendrin in modulating acid-base transport in the cortical collecting duct, we examined the regulation of expression of pendrin by acid-base status in mouse kidney., Methods: Mice were treated orally either with an acid or bicarbonate load (0.28 mol/L NH4Cl or NaHCO3) or received a K+-deficient diet for one week. Immunohistochemistry and Western blotting was performed., Results: Acid-loading caused a reduction in pendrin protein expression levels within one day and decreased expression to 23% of control levels after one week. Concomitantly, pendrin protein was shifted from the apical membrane to the cytosol, and the relative abundance of pendrin positive cells declined. Similarly, in chronic K+-depletion, known to elicit a metabolic alkalosis, pendrin protein levels decreased and pendrin expression was shifted to an intracellular pool with the relative number of pendrin positive cells reduced. In contrast, following oral bicarbonate loading pendrin was found exclusively in the apical membrane and the relative number of pendrin positive cells increased., Conclusions: These results are in agreement with a potential role of pendrin in bicarbonate secretion and regulation of acid-base transport in the cortical collecting duct.
- Published
- 2002
- Full Text
- View/download PDF
41. Molecular cloning and characterization of Atp6n1b: a novel fourth murine vacuolar H+-ATPase a-subunit gene.
- Author
-
Smith AN, Finberg KE, Wagner CA, Lifton RP, Devonald MA, Su Y, and Karet FE
- Subjects
- Amino Acid Sequence, Animals, Aquaporin 2, Aquaporin 6, Aquaporins analysis, Chromosome Mapping, Cloning, Molecular, Mice, Molecular Sequence Data, Phylogeny, Polymorphism, Genetic, Protein Subunits, Proton Pumps analysis, Proton Pumps chemistry, Reverse Transcriptase Polymerase Chain Reaction, Vacuolar Proton-Translocating ATPases, Pregnancy Proteins, Proton Pumps genetics, Proton-Translocating ATPases, Suppressor Factors, Immunologic
- Abstract
The 116-kDa a-subunit of the vacuolar proton pump (H(+)-ATPase) exists as several isoforms encoded by different genes and with different patterns of tissue expression. Its function within the multisubunit pump complex has yet to be elucidated. To date, three isoforms have been identified in mouse (designated a1-a3). We now report the cloning and characterization of Atp6n1b, encoding a novel fourth murine isoform (a4). Murine a4 has 833 residues and shows 85% amino acid identity to the human kidney-specific ATP6N1B protein in which loss-of-function alterations cause autosomal recessive distal renal tubular acidosis. The human and murine genes have similar genomic organization; furthermore, Atp6n1b maps to a region of mouse chromosome 6 that is syntenic with the segment of human 7q33-34 containing ATP6N1B. Together these findings establish the two genes as orthologs. The mouse a4 protein is 61, 52, and 47% identical to a1, a2, and a3, respectively. Phylogenetic analysis confirms that among vertebrates there are four a-subunit families, with a4 most resembling a1. Northern blot analysis of Atp6n1b reveals a 3.7-kilobase a4 transcript in kidney but not other major organs, and a reverse transcription polymerase chain reaction in 12 mouse tissues detects expression in kidney alone. Immunofluorescence studies in murine kidney demonstrate high intensity a4 staining at the surface of intercalated cells, with additional expression in the proximal tubule (not previously reported in human kidney). Similar apical a4 immunostaining is also present in male genital tissue. Identification of this novel murine kidney-enriched 116-kDa a-subunit provides a molecular tool for investigation of the currently unknown role of this protein, which is essential for proper function of the apical renal vacuolar H(+)-ATPase in man.
- Published
- 2001
- Full Text
- View/download PDF
42. Localization of a gene for autosomal recessive distal renal tubular acidosis with normal hearing (rdRTA2) to 7q33-34.
- Author
-
Karet FE, Finberg KE, Nayir A, Bakkaloglu A, Ozen S, Hulton SA, Sanjad SA, Al-Sabban EA, Medina JF, and Lifton RP
- Subjects
- Acidosis, Renal Tubular physiopathology, Adenosine Triphosphatases genetics, Adult, Child, Child, Preschool, Chromosomes, Human, Pair 2 genetics, Consanguinity, DNA Mutational Analysis, Female, Hearing genetics, Humans, Hydrogen-Ion Concentration, Infant, Infant, Newborn, Lod Score, Male, Membrane Proteins genetics, Middle East, Molecular Sequence Data, Pakistan, Pedigree, Polymorphism, Single-Stranded Conformational, SLC4A Proteins, Acidosis, Renal Tubular genetics, Anion Transport Proteins, Antiporters, Chromosome Mapping, Chromosomes, Human, Pair 7 genetics, Genes, Recessive genetics, Hearing physiology
- Abstract
Failure of distal nephrons to excrete excess acid results in the "distal renal tubular acidoses" (dRTA). Early childhood features of autosomal recessive dRTA include severe metabolic acidosis with inappropriately alkaline urine, poor growth, rickets, and renal calcification. Progressive bilateral sensorineural hearing loss (SNHL) is evident in approximately one-third of patients. We have recently identified mutations in ATP6B1, encoding the B-subunit of the collecting-duct apical proton pump, as a cause of recessive dRTA with SNHL. We now report the results of genetic analysis of 13 kindreds with recessive dRTA and normal hearing. Analysis of linkage and molecular examination of ATP6B1 indicated that mutation in ATP6B1 rarely, if ever, accounts for this phenotype, prompting a genomewide linkage search for loci underlying this trait. The results strongly supported linkage with locus heterogeneity to a segment of 7q33-34, yielding a maximum multipoint LOD score of 8.84 with 68% of kindreds linked. The LOD-3 support interval defines a 14-cM region flanked by D7S500 and D7S688. That 4 of these 13 kindreds do not support linkage to rdRTA2 and ATP6B1 implies the existence of at least one additional dRTA locus. These findings establish that genes causing recessive dRTA with normal and impaired hearing are different, and they identify, at 7q33-34, a new locus, rdRTA2, for recessive dRTA with normal hearing.
- Published
- 1999
- Full Text
- View/download PDF
43. Mutations in the gene encoding B1 subunit of H+-ATPase cause renal tubular acidosis with sensorineural deafness.
- Author
-
Karet FE, Finberg KE, Nelson RD, Nayir A, Mocan H, Sanjad SA, Rodriguez-Soriano J, Santos F, Cremers CW, Di Pietro A, Hoffbrand BI, Winiarski J, Bakkaloglu A, Ozen S, Dusunsel R, Goodyer P, Hulton SA, Wu DK, Skvorak AB, Morton CC, Cunningham MJ, Jha V, and Lifton RP
- Subjects
- Acidosis, Renal Tubular complications, Acidosis, Renal Tubular genetics, Base Sequence, Child, Preschool, Cochlea metabolism, Female, Genes, Recessive, Genetic Linkage, Hearing Loss, Sensorineural complications, Hearing Loss, Sensorineural genetics, Humans, Infant, Male, Molecular Sequence Data, Pedigree, Proton-Translocating ATPases metabolism, Acidosis, Renal Tubular enzymology, Chromosomes, Human, Pair 2, Hearing Loss, Sensorineural enzymology, Mutation, Proton-Translocating ATPases genetics
- Abstract
H+-ATPases are ubiquitous in nature; V-ATPases pump protons against an electrochemical gradient, whereas F-ATPases reverse the process, synthesizing ATP. We demonstrate here that mutations in ATP6B1, encoding the B-subunit of the apical proton pump mediating distal nephron acid secretion, cause distal renal tubular acidosis, a condition characterized by impaired renal acid secretion resulting in metabolic acidosis. Patients with ATP6B1 mutations also have sensorineural hearing loss; consistent with this finding, we demonstrate expression of ATP6B1 in cochlea and endolymphatic sac. Our data, together with the known requirement for active proton secretion to maintain proper endolymph pH, implicate ATP6B1 in endolymph pH homeostasis and in normal auditory function. ATP6B1 is the first member of the H+-ATPase gene family in which mutations are shown to cause human disease.
- Published
- 1999
- Full Text
- View/download PDF
44. Interactions of VirB9, -10, and -11 with the membrane fraction of Agrobacterium tumefaciens: solubility studies provide evidence for tight associations.
- Author
-
Finberg KE, Muth TR, Young SP, Maken JB, Heitritter SM, Binns AN, and Banta LM
- Subjects
- Agrobacterium tumefaciens genetics, Bacterial Proteins genetics, Bacterial Proteins metabolism, Biological Transport, Cell Compartmentation, DNA, Bacterial metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Mutation, Protein Binding, Sequence Deletion, Virulence genetics, Agrobacterium tumefaciens pathogenicity, Bacterial Proteins isolation & purification, Cell Membrane chemistry, Membrane Proteins isolation & purification, Plants microbiology, Virulence Factors
- Abstract
The eleven predicted gene products of the Agrobacterium tumefaciens virB operon are believed to form a transmembrane pore complex through which T-DNA export occurs. The VirB10 protein is required for virulence and is a component of an aggregate associated with the membrane fraction of A. tumefaciens. Removal of the putative membrane-spanning domain (amino acids 22 through 55) disrupts the membrane topology of VirB10 (J. E. Ward, E. M. Dale, E. W. Nester, and A. N. Binns, J. Bacteriol. 172:5200-5210, 1990). Deletion of the sequences encoding amino acids 22 to 55 abolishes the ability of plasmid-borne virB10 to complement a null mutation in the virB10 gene, suggesting that the proper topology of VirB10 in the membrane may indeed play a crucial role in T-DNA transfer to the plant cell. Western blot (immunoblot) analysis indicated that the observed loss of virulence could not be attributed to a decrease in the steady-state levels of the mutant VirB10 protein. Although the deletion of the single transmembrane domain would be expected to perturb membrane association, VirB10 delta 22-55 was found exclusively in the membrane fraction. Urea extraction studies suggested that this membrane localization might be the result of a peripheral membrane association; however, the mutant protein was found in both inner and outer membrane fractions separated by sucrose density gradient centrifugation. Both wild-type VirB10 and wild-type VirB9 were only partially removed from the membranes by extraction with 1% Triton X-100, while VirB5 and VirB8 were Triton X-100 soluble. VirB11 was stripped from the membranes by 6 M urea but not by a more mild salt extraction. The fractionation patterns of VirB9, VirB10, and VirB11 were not dependent on each other or on VirB8 or VirD4. The observed tight association of VirB9, VirB10, and VirB11 with the membrane fraction support the notion that these proteins may exist as components of multiprotein pore complexes, perhaps spanning both the inner and outer membranes of Agrobacterium cells.
- Published
- 1995
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.