159 results on '"Seth L. Alper"'
Search Results
2. Can KCa3.1 channel activators serve as novel inhibitors of platelet aggregation?
- Author
-
Heike Wulff, Andrew P. Braun, and Seth L. Alper
- Subjects
Platelet Aggregation ,Platelet Function Tests ,Humans ,Pyrazoles ,Hematology ,Intermediate-Conductance Calcium-Activated Potassium Channels - Published
- 2022
3. ADAR regulates APOL1 via A-to-I RNA editing by inhibition of MDA5 activation in a paradoxical biological circuit
- Author
-
Cristian V, Riella, Michelle, McNulty, Guilherme T, Ribas, Calum F, Tattersfield, Chandra, Perez-Gill, Felix, Eichinger, Jessica, Kelly, Justin, Chun, Balajikarthick, Subramanian, Dieval, Guizelini, Seth L, Alper, Martin R, Pollak, Matthew G, Sampson, and David J, Friedman
- Subjects
Interferon-Induced Helicase, IFIH1 ,Adenosine ,Multidisciplinary ,Adenosine Deaminase ,Apolipoprotein L1 ,Inosine ,Mice ,Interferon-gamma ,Interferon Type I ,Humans ,Animals ,RNA Editing ,RNA, Messenger ,3' Untranslated Regions ,RNA, Double-Stranded - Abstract
APOL1 risk variants are associated with increased risk of kidney disease in patients of African ancestry, but not all individuals with the APOL1 high-risk genotype develop kidney disease. As APOL1 gene expression correlates closely with the degree of kidney cell injury in both cell and animal models, the mechanisms regulating APOL1 expression may be critical determinants of risk allele penetrance. The APOL1 messenger RNA includes Alu elements at the 3′ untranslated region that can form a double-stranded RNA structure (Alu-dsRNA) susceptible to posttranscriptional adenosine deaminase acting on RNA (ADAR)–mediated adenosine-to-inosine (A-to-I) editing, potentially impacting gene expression. We studied the effects of ADAR expression and A-to-I editing on APOL1 levels in podocytes, human kidney tissue, and a transgenic APOL1 mouse model. In interferon-γ (IFN-γ)–stimulated human podocytes, ADAR down-regulates APOL1 by preventing melanoma differentiation-associated protein 5 (MDA5) recognition of dsRNA and the subsequent type I interferon (IFN-I) response. Knockdown experiments showed that recognition of APOL1 messenger RNA itself is an important contributor to the MDA5-driven IFN-I response. Mathematical modeling suggests that the IFN–ADAR–APOL1 network functions as an incoherent feed-forward loop, a biological circuit capable of generating fast, transient responses to stimuli. Glomeruli from human kidney biopsies exhibited widespread editing of APOL1 Alu-dsRNA, while the transgenic mouse model closely replicated the edited sites in humans. APOL1 expression in mice was inversely correlated with Adar1 expression under IFN-γ stimuli, supporting the idea that ADAR regulates APOL1 levels in vivo. ADAR-mediated A-to-I editing is an important regulator of APOL1 expression that could impact both penetrance and severity of APOL1-associated kidney disease.
- Published
- 2022
4. Nobel prize in physiology or medicine 2021, receptors for temperature and touch: Implications for hematology
- Author
-
Achille Iolascon, Seth L. Alper, Immacolata Andolfo, Andolfo, I., Alper, S. L., and Iolascon, A.
- Subjects
medicine.medical_specialty ,Hematology ,business.industry ,Temperature ,MEDLINE ,TRPV Cation Channels ,Anemia, Sickle Cell ,Bioinformatics ,Ion Channels ,Nobel Prize ,Touch ,Internal medicine ,medicine ,Animals ,Humans ,Calcium ,Thermosensing ,business ,Receptor - Published
- 2021
5. New drugs on the horizon for cerebral edema: what’s in the clinical development pipeline?
- Author
-
Jinwei Zhang, Kristopher T. Kahle, Stephanie M. Robert, Seth L. Alper, and Benjamin C. Reeves
- Subjects
0301 basic medicine ,Pharmacology ,animal structures ,business.industry ,Pipeline (computing) ,Drug Repositioning ,Brain Edema ,Drugs, Investigational ,General Medicine ,medicine.disease ,Article ,Cerebral edema ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Drug Development ,030220 oncology & carcinogenesis ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Molecular Targeted Therapy ,Current (fluid) ,business ,Neuroscience - Abstract
INTRODUCTION: Research has advanced our understanding of the molecular and cellular mechanisms of cerebral edema and has propelled the development of novel anti-edema therapeutics. Current evidence supports aberrant neuro-glial ion transport as a central mechanism that underlies pathological fluid accumulation after central nervous system injury. AREAS COVERED: Novel agents in clinical development show potential in altering the natural history and treatment of cerebral edema. Using the PubMed and Google Scholar databases, we review recent advances in our understanding of cerebral edema and describe agents under active investigation, their mechanism, and their application in recent and ongoing clinical trials. EXPERT OPINION: Pharmacotherapies that target molecular mechanisms underlying the compensatory post-injury response of ion channels and transporters that lead to pathological alteration of osmotic gradients, are the most promising therapeutic strategies. Repurposing of drugs such as glyburide that inhibit the aberrant upregulation of ion channels such as SUR1-TRPM4, and novel agents, such as ZT-a1, which re-establish physiological regulation of ion channels such as NKCC1/KCC, could be useful adjuvants to prevent and even reverse fluid accumulation in the brain parenchyma.
- Published
- 2020
6. Disruption of Cav1.2-mediated signaling is a pathway for ketamine-induced pathology
- Author
-
Seth L. Alper, Xiang Xie, Weiqun Yu, Huan Chen, David H. Vandorpe, and Mark L. Zeidel
- Subjects
0301 basic medicine ,Xenopus ,General Physics and Astronomy ,02 engineering and technology ,Pharmacology ,Kidney ,Cav1.2 ,chemistry.chemical_compound ,Mice ,Cystitis ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Voltage-dependent calcium channel ,biology ,Smooth muscle contraction ,Urological manifestations ,021001 nanoscience & nanotechnology ,Bay K8644 ,Ketamine ,0210 nano-technology ,medicine.drug ,Signal Transduction ,Agonist ,Calcium Channels, L-Type ,medicine.drug_class ,Science ,Urinary Bladder ,Receptors, N-Methyl-D-Aspartate ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,medicine ,Animals ,Humans ,Cell Proliferation ,business.industry ,Antagonist ,Muscle, Smooth ,General Chemistry ,Disease Models, Animal ,030104 developmental biology ,chemistry ,Anesthetic ,biology.protein ,Oocytes ,lcsh:Q ,Calcium ,business - Abstract
The general anesthetic ketamine has been repurposed by physicians as an anti-depressant and by the public as a recreational drug. However, ketamine use can cause extensive pathological changes, including ketamine cystitis. The mechanisms of ketamine’s anti-depressant and adverse effects remain poorly understood. Here we present evidence that ketamine is an effective L-type Ca2+ channel (Cav1.2) antagonist that directly inhibits calcium influx and smooth muscle contractility, leading to voiding dysfunction. Ketamine prevents Cav1.2-mediated induction of immediate early genes and transcription factors, and inactivation of Cav1.2 in smooth muscle mimics the ketamine cystitis phenotype. Our results demonstrate that ketamine inhibition of Cav1.2 signaling is an important pathway mediating ketamine cystitis. In contrast, Cav1.2 agonist Bay k8644 abrogates ketamine-induced smooth muscle dysfunction. Indeed, Cav1.2 activation by Bay k8644 decreases voiding frequency while increasing void volume, indicating Cav1.2 agonists might be effective drugs for treatment of bladder dysfunction., Ketamine is a general anesthetic that is used also as an anti-depressant, but its use is associated with cystitis. Here, the authors show that ketamine is an antagonist of the Cav1.2 channel in bladder smooth muscle cells, that ablation of this channel in mice mimics the cystitis induced by ketamine, and show that this effect can be abrogated by an agonist of this ion channel.
- Published
- 2020
7. Inflammation in acquired hydrocephalus: pathogenic mechanisms and therapeutic targets
- Author
-
Jason K. Karimy, Benjamin C. Reeves, Eyiyemisi Damisah, Phan Q. Duy, Prince Antwi, Wyatt David, Kevin Wang, Steven J. Schiff, David D. Limbrick, Seth L. Alper, Benjamin C. Warf, Maiken Nedergaard, J. Marc Simard, and Kristopher T. Kahle
- Subjects
0301 basic medicine ,Inflammation ,Disease ,Article ,Pathogenesis ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Cerebrospinal fluid ,Immune system ,medicine ,Animals ,Humans ,business.industry ,medicine.disease ,Hydrocephalus ,Acquired Hydrocephalus ,030104 developmental biology ,Immunology ,Encephalitis ,Choroid plexus ,Neurology (clinical) ,Inflammation Mediators ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Hydrocephalus is the most common neurosurgical disorder worldwide and is characterized by enlargement of the cerebrospinal fluid (CSF)-filled brain ventricles resulting from failed CSF homeostasis. Since the 1840s, physicians have observed inflammation in the brain and the CSF spaces in both posthaemorrhagic hydrocephalus (PHH) and postinfectious hydrocephalus (PIH). Reparative inflammation is an important protective response that eliminates foreign organisms, damaged cells and physical irritants; however, inappropriately triggered or sustained inflammation can respectively initiate or propagate disease. Recent data have begun to uncover the molecular mechanisms by which inflammation - driven by Toll-like receptor 4-regulated cytokines, immune cells and signalling pathways - contributes to the pathogenesis of hydrocephalus. We propose that therapeutic approaches that target inflammatory mediators in both PHH and PIH could address the multiple drivers of disease, including choroid plexus CSF hypersecretion, ependymal denudation, and damage and scarring of intraventricular and parenchymal (glia-lymphatic) CSF pathways. Here, we review the evidence for a prominent role of inflammation in the pathogenic mechanism of PHH and PIH and highlight promising targets for therapeutic intervention. Focusing research efforts on inflammation could shift our view of hydrocephalus from that of a lifelong neurosurgical disorder to that of a preventable neuroinflammatory condition.
- Published
- 2020
8. Whole exome sequencing identified ATP6V1C2 as a novel candidate gene for recessive distal renal tubular acidosis
- Author
-
Tilman Jobst-Schwan, Marcella Greco, Patricia M. Kane, Michelle A. Baum, Verena Klämbt, Shrikant Mane, Seema Hashmi, Seth L. Alper, Jutta Gellermann, Richard P. Lifton, Amar J. Majmundar, Florian Buerger, John F. Heneghan, Friedhelm Hildebrandt, Guido F. Laube, Shirlee Shril, Francesco Emma, Farkhanda Hafeez, Hanan M. Fathy, Rezan Topaloglu, Isabel Ottlewski, Martin Pohl, Danko Milosevic, Boris E. Shmukler, and Maureen Tarsio
- Subjects
0301 basic medicine ,Vacuolar Proton-Translocating ATPases ,Candidate gene ,Pathology ,medicine.medical_specialty ,DNA Mutational Analysis ,030232 urology & nephrology ,medicine.disease_cause ,Renal tubular acidosis ,03 medical and health sciences ,0302 clinical medicine ,Renal tubular dysfunction ,Distal renal tubular acidosis ,Anion Exchange Protein 1, Erythrocyte ,Exome Sequencing ,medicine ,Humans ,Chloride-Bicarbonate Antiporters ,Child ,Exome sequencing ,Kidney ,Mutation ,business.industry ,Forkhead Transcription Factors ,Acidosis, Renal Tubular ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,FOXI1 ,Nephrology ,business - Abstract
Distal renal tubular acidosis is a rare renal tubular disorder characterized by hyperchloremic metabolic acidosis and impaired urinary acidification. Mutations in three genes (ATP6V0A4, ATP6V1B1 and SLC4A1) constitute a monogenic causation in 58-70% of familial cases of distal renal tubular acidosis. Recently, mutations in FOXI1 have been identified as an additional cause. Therefore, we hypothesized that further monogenic causes of distal renal tubular acidosis remain to be discovered. Panel sequencing and/or whole exome sequencing was performed in a cohort of 17 families with 19 affected individuals with pediatric onset distal renal tubular acidosis. A causative mutation was detected in one of the three "classical" known distal renal tubular acidosis genes in 10 of 17 families. The seven unsolved families were then subjected to candidate whole exome sequencing analysis. Potential disease causing mutations in three genes were detected: ATP6V1C2, which encodes another kidney specific subunit of the V-type proton ATPase (1 family); WDR72 (2 families), previously implicated in V-ATPase trafficking in cells; and SLC4A2 (1 family), a paralog of the known distal renal tubular acidosis gene SLC4A1. Two of these mutations were assessed for deleteriousness through functional studies. Yeast growth assays for ATP6V1C2 revealed loss-of-function for the patient mutation, strongly supporting ATP6V1C2 as a novel distal renal tubular acidosis gene. Thus, we provided a molecular diagnosis in a known distal renal tubular acidosis gene in 10 of 17 families (59%) with this disease, identified mutations in ATP6V1C2 as a novel human candidate gene, and provided further evidence for phenotypic expansion in WDR72 mutations from amelogenesis imperfecta to distal renal tubular acidosis.
- Published
- 2020
9. FSGS-Causing INF2 Mutation Impairs Cleaved INF2 N-Fragment Functions in Podocytes
- Author
-
Balajikarthick Subramanian, Henry N. Higgs, Seth L. Alper, Chandra Perez-Gill, Justin Chun, Johannes Schlondorff, Martin R. Pollak, Isaac E. Stillman, and Paul Yan
- Subjects
0301 basic medicine ,Gene isoform ,030232 urology & nephrology ,Formins ,Podocyte foot ,Cleavage (embryo) ,Podocyte ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Up Front Matters ,medicine ,Animals ,Humans ,Protein Isoforms ,Cells, Cultured ,Actin ,biology ,Glomerulosclerosis, Focal Segmental ,Podocytes ,Chemistry ,General Medicine ,Cathepsins ,Peptide Fragments ,Cell biology ,Mice, Inbred C57BL ,INF2 ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,Nephrology ,Mutation ,biology.protein ,MDia1 - Abstract
Background Mutations in the gene encoding inverted formin-2 (INF2), a member of the formin family of actin regulatory proteins, are among the most common causes of autosomal dominant FSGS. INF2 is regulated by interaction between its N-terminal diaphanous inhibitory domain (DID) and its C-terminal diaphanous autoregulatory domain (DAD). INF2 also modulates activity of other formins, such as the mDIA subfamily, and promotes stable microtubule assembly. Why the disease-causing mutations are restricted to the N terminus and how they cause human disease has been unclear. Methods We examined INF2 isoforms present in podocytes and evaluated INF2 cleavage as an explanation for immunoblot findings. We evaluated the expression of INF2 N- and C-terminal fragments in human kidney disease conditions. We also investigated the localization and functions of the DID-containing N-terminal fragment in podocytes and assessed whether the FSGS-associated R218Q mutation impairs INF2 cleavage or the function of the N-fragment. Results The INF2-CAAX isoform is the predominant isoform in podocytes. INF2 is proteolytically cleaved, a process mediated by cathepsin proteases, liberating the N-terminal DID to function independently. Although the N-terminal region normally localizes to podocyte foot processes, it does not do so in the presence of FSGS-associated INF2 mutations. The C-terminal fragment localizes to the cell body irrespective of INF2 mutations. In podocytes, the N-fragment localizes to the plasma membrane, binds mDIA1, and promotes cell spreading in a cleavage-dependent way. The disease-associated R218Q mutation impairs these N-fragment functions but not INF2 cleavage. Conclusions INF2 is cleaved into an N-terminal DID-containing fragment and a C-terminal DAD-containing fragment. Cleavage allows the N-terminal fragment to function independently and helps explain the clustering of FSGS-associated mutations.
- Published
- 2020
10. Hereditary anemia caused by multilocus inheritance of
- Author
-
Barbara Eleni, Rosato, Seth L, Alper, Giovanna, Tomaiuolo, Roberta, Russo, Achille, Iolascon, and Immacolata, Andolfo
- Subjects
Anion Exchange Protein 1, Erythrocyte ,Hydrops Fetalis ,Mutation ,Humans ,ATP-Binding Cassette Transporters ,Female ,Anemia, Hemolytic, Congenital ,Ion Channels - Published
- 2022
11. The choroid plexus links innate immunity to CSF dysregulation in hydrocephalus
- Author
-
Stephanie M. Robert, Benjamin C. Reeves, Emre Kiziltug, Phan Q. Duy, Jason K. Karimy, M. Shahid Mansuri, Arnaud Marlier, Garrett Allington, Ana B.W. Greenberg, Tyrone DeSpenza, Amrita K. Singh, Xue Zeng, Kedous Y. Mekbib, Adam J. Kundishora, Carol Nelson-Williams, Le Thi Hao, Jinwei Zhang, TuKiet T. Lam, Rashaun Wilson, William E. Butler, Michael L. Diluna, Philip Feinberg, Dorothy P. Schafer, Kiavash Movahedi, Allen Tannenbaum, Sunil Koundal, Xinan Chen, Helene Benveniste, David D. Limbrick, Steven J. Schiff, Bob S. Carter, Murat Gunel, J. Marc Simard, Richard P. Lifton, Seth L. Alper, Eric Delpire, Kristopher T. Kahle, Laboratory of Molecullar and Cellular Therapy, and Basic (bio-) Medical Sciences
- Subjects
surgery ,Cellular and Molecular Neuroscience ,Hydrocephalus/cerebrospinal fluid ,Cytokine Release Syndrome/pathology ,Neuroscience(all) ,Humans ,Brain/metabolism ,Blood-Brain Barrier/metabolism ,Choroid Plexus/metabolism ,Immunity, Innate ,General Biochemistry, Genetics and Molecular Biology - Abstract
The choroid plexus (ChP) is the blood-cerebrospinal fluid (CSF) barrier and the primary source of CSF. Acquired hydrocephalus, caused by brain infection or hemorrhage, lacks drug treatments due to obscure pathobiology. Our integrated, multi-omic investigation of post-infectious hydrocephalus (PIH) and post-hemorrhagic hydrocephalus (PHH) models revealed that lipopolysaccharide and blood breakdown products trigger highly similar TLR4-dependent immune responses at the ChP-CSF interface. The resulting CSF "cytokine storm", elicited from peripherally derived and border-associated ChP macrophages, causes increased CSF production from ChP epithelial cells via phospho-activation of the TNF-receptor-associated kinase SPAK, which serves as a regulatory scaffold of a multi-ion transporter protein complex. Genetic or pharmacological immunomodulation prevents PIH and PHH by antagonizing SPAK-dependent CSF hypersecretion. These results reveal the ChP as a dynamic, cellularly heterogeneous tissue with highly regulated immune-secretory capacity, expand our understanding of ChP immune-epithelial cell cross talk, and reframe PIH and PHH as related neuroimmune disorders vulnerable to small molecule pharmacotherapy.
- Published
- 2023
12. Apolipoprotein L1 (APOL1) risk variant toxicity depends on the haplotype background
- Author
-
Daniel Blackler, Martin R. Pollak, Leny Dias, Herbert Lannon, David J. Friedman, Seth L. Alper, and Shrijal S Shah
- Subjects
0301 basic medicine ,Genetics ,High rate ,Apolipoprotein L1 ,Haplotype ,030232 urology & nephrology ,Genetic variants ,Biology ,Article ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Human disease ,Risk variant ,Haplotypes ,Nephrology ,biology.protein ,Humans ,Kidney Diseases ,Genetic Background - Abstract
The apolipoprotein L1 (APOL1) risk variants G1 and G2 are associated with high rates of kidney disease in African Americans in genetic studies. However, our understanding of APOL1 biology has lagged far behind. Here we report that engineering G1 and G2 mutations on unnatural haplotype backgrounds instead of on the specific G1 and G2 haplotype backgrounds that occur in nature profoundly alters APOL1-mediated cytotoxicity in experimental systems. Thus, in addition to helping resolve some important controversies in the APOL1 field, our demonstration of the critical influence of haplotype background may apply more generally to the study of other genetic variants that cause or predispose to human disease.
- Published
- 2019
13. Monitoring Daily Ultrafiltration in Automated Peritoneal Dialysis
- Author
-
Fabian Eibensteiner, Krystell Oviedo Flores, Markus Unterwurzacher, Rebecca Herzog, Klaus Kratochwill, Seth L. Alper, Christoph Aufricht, Franz König, and Andreas Vychytil
- Subjects
Adult ,Male ,Transplantation ,Epidemiology ,Hemodiafiltration ,Middle Aged ,Critical Care and Intensive Care Medicine ,Nephrology ,Research Letter ,Humans ,Female ,Peritoneal Dialysis ,Aged ,Monitoring, Physiologic - Published
- 2021
14. Impaired neurogenesis alters brain biomechanics in a neuroprogenitor-based genetic subtype of congenital hydrocephalus
- Author
-
Phan Q. Duy, Stefan C. Weise, Claudia Marini, Xiao-Jun Li, Dan Liang, Peter J. Dahl, Shaojie Ma, Ana Spajic, Weilai Dong, Jane Juusola, Emre Kiziltug, Adam J. Kundishora, Sunil Koundal, Maysam Z. Pedram, Lucia A. Torres-Fernández, Kristian Händler, Elena De Domenico, Matthias Becker, Thomas Ulas, Stefan A. Juranek, Elisa Cuevas, Le Thi Hao, Bettina Jux, André M. M. Sousa, Fuchen Liu, Suel-Kee Kim, Mingfeng Li, Yiying Yang, Yutaka Takeo, Alvaro Duque, Carol Nelson-Williams, Yonghyun Ha, Kartiga Selvaganesan, Stephanie M. Robert, Amrita K. Singh, Garrett Allington, Charuta G. Furey, Andrew T. Timberlake, Benjamin C. Reeves, Hannah Smith, Ashley Dunbar, Tyrone DeSpenza, June Goto, Arnaud Marlier, Andres Moreno-De-Luca, Xin Yu, William E. Butler, Bob S. Carter, Evelyn M. R. Lake, R. Todd Constable, Pasko Rakic, Haifan Lin, Engin Deniz, Helene Benveniste, Nikhil S. Malvankar, Juvianee I. Estrada-Veras, Christopher A. Walsh, Seth L. Alper, Joachim L. Schultze, Katrin Paeschke, Angelika Doetzlhofer, F. Gregory Wulczyn, Sheng Chih Jin, Richard P. Lifton, Nenad Sestan, Waldemar Kolanus, and Kristopher T. Kahle
- Subjects
cerebrospinal fluid [Hydrocephalus] ,genetics [Ubiquitin-Protein Ligases] ,General Neuroscience ,Neurogenesis ,Ubiquitin-Protein Ligases ,Brain ,genetics [Tripartite Motif Proteins] ,Whole Exome Sequencing ,Article ,Biomechanical Phenomena ,Tripartite Motif Proteins ,metabolism [Cerebrospinal Fluid] ,Mice ,metabolism [Tripartite Motif Proteins] ,metabolism [Brain] ,genetics [Hydrocephalus] ,genetics [Neurogenesis] ,Exome Sequencing ,Animals ,Humans ,ddc:610 ,TRIM71 protein, human ,Cerebrospinal Fluid ,Hydrocephalus - Abstract
Hydrocephalus, characterized by cerebral ventricular dilatation, is routinely attributed to primary defects in cerebrospinal fluid (CSF) homeostasis. This fosters CSF shunting as the leading reason for brain surgery in children despite considerable disease heterogeneity. In this study, by integrating human brain transcriptomics with whole-exome sequencing of 483 patients with congenital hydrocephalus (CH), we found convergence of CH risk genes in embryonic neuroepithelial stem cells. Of all CH risk genes, TRIM71/lin-41 harbors the most de novo mutations and is most specifically expressed in neuroepithelial cells. Mice harboring neuroepithelial cell-specific Trim71 deletion or CH-specific Trim71 mutation exhibit prenatal hydrocephalus. CH mutations disrupt TRIM71 binding to its RNA targets, causing premature neuroepithelial cell differentiation and reduced neurogenesis. Cortical hypoplasia leads to a hypercompliant cortex and secondary ventricular enlargement without primary defects in CSF circulation. These data highlight the importance of precisely regulated neuroepithelial cell fate for normal brain-CSF biomechanics and support a clinically relevant neuroprogenitor-based paradigm of CH.
- Published
- 2021
15. Brain ventricles as windows into brain development and disease
- Author
-
Phan Q. Duy, Pasko Rakic, Seth L. Alper, William E. Butler, Christopher A. Walsh, Nenad Sestan, Daniel H. Geschwind, Sheng Chih Jin, and Kristopher T. Kahle
- Subjects
Neural Stem Cells ,General Neuroscience ,Humans ,Article ,Cerebral Ventricles ,Hydrocephalus - Abstract
Dilation of the fluid-filled cerebral ventricles (ventriculomegaly) characterizes hydrocephalus and is frequently seen in autism and schizophrenia. Recent work suggests that the genomic study of congenital hydrocephalus may be unexpectedly fertile ground for revealing insights into neural stem cell regulation, human cerebrocortical development, and pathogenesis of neuropsychiatric disease.
- Published
- 2021
16. Purinergic signaling is essential for full Psickle activation by hypoxia and by normoxic acid pH in mature human sickle red cells and in vitro-differentiated cultured human sickle reticulocytes
- Author
-
David H, Vandorpe, Alicia, Rivera, Markus, Ganter, Selasi, Dankwa, Jay G, Wohlgemuth, Jeffrey S, Dlott, L Michael, Snyder, Carlo, Brugnara, Manoj, Duraisingh, and Seth L, Alper
- Subjects
Adenosine Triphosphate ,Erythrocytes ,Reticulocytes ,Probenecid ,Cations ,Apyrase ,Humans ,Anemia, Sickle Cell ,Suramin ,Hydrogen-Ion Concentration ,Hypoxia ,Cells, Cultured - Abstract
Paracrine ATP release by erythrocytes has been shown to regulate endothelial cell function via purinergic signaling, and this erythoid-endothelial signaling network is pathologically dysregulated in sickle cell disease. We tested the role of extracellular ATP-mediated purinergic signaling in the activation of Psickle, the mechanosensitive Ca
- Published
- 2021
17. Haplotype-resolved germline and somatic alterations in renal medullary carcinomas
- Author
-
Elizabeth J. Perlman, Matthew Meyerson, Rameen Beroukhim, Won Jun Kim, Yueh Yun Chi, Jeremiah A. Wala, Seth L. Alper, Guillaume Kugener, Hyunji Kim, Thomas P. Howard, Elizabeth Mullen, Gavin Ha, William C. Hahn, Andrew L. Hong, Heng Li, Jian Carrot-Zhang, Kar Tong Tan, and Yuxiang Zhang
- Subjects
Male ,Oncogene Proteins, Fusion ,QH426-470 ,medicine.disease_cause ,Germline ,0302 clinical medicine ,Databases, Genetic ,Child ,Genetics (clinical) ,Genetics ,0303 health sciences ,Mutation ,Genomics ,Kidney Neoplasms ,Gene Expression Regulation, Neoplastic ,Carcinoma, Medullary ,Child, Preschool ,030220 oncology & carcinogenesis ,Medicine ,Molecular Medicine ,Female ,Sickle cell trait ,Genotype ,Locus (genetics) ,Biology ,Polymorphism, Single Nucleotide ,Renal medullary carcinoma ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,Molecular Biology ,Alleles ,Genetic Association Studies ,Germ-Line Mutation ,030304 developmental biology ,Whole Genome Sequencing ,Research ,DNA Breaks ,Linked-read sequencing ,Haplotype ,Computational Biology ,medicine.disease ,Haplotypes ,Founder effect - Abstract
Background Renal medullary carcinomas (RMCs) are rare kidney cancers that occur in adolescents and young adults of African ancestry. Although RMC is associated with the sickle cell trait and somatic loss of the tumor suppressor, SMARCB1, the ancestral origins of RMC remain unknown. Further, characterization of structural variants (SVs) involving SMARCB1 in RMC remains limited. Methods We used linked-read genome sequencing to reconstruct germline and somatic haplotypes in 15 unrelated patients with RMC registered on the Children’s Oncology Group (COG) AREN03B2 study between 2006 and 2017 or from our prior study. We performed fine-mapping of the HBB locus and assessed the germline for cancer predisposition genes. Subsequently, we assessed the tumor samples for mutations outside of SMARCB1 and integrated RNA sequencing to interrogate the structural variants at the SMARCB1 locus. Results We find that the haplotype of the sickle cell mutation in patients with RMC originated from three geographical regions in Africa. In addition, fine-mapping of the HBB locus identified the sickle cell mutation as the sole candidate variant. We further identify that the SMARCB1 structural variants are characterized by blunt or 1-bp homology events. Conclusions Our findings suggest that RMC does not arise from a single founder population and that the HbS allele is a strong candidate germline allele which confers risk for RMC. Furthermore, we find that the SVs that disrupt SMARCB1 function are likely repaired by non-homologous end-joining. These findings highlight how haplotype-based analyses using linked-read genome sequencing can be applied to identify potential risk variants in small and rare disease cohorts and provide nucleotide resolution to structural variants.
- Published
- 2021
18. DIAPH1 Variants in Non–East Asian Patients With Sporadic Moyamoya Disease
- Author
-
Carol Nelson-Williams, Nicholas S. Diab, Qiongshi Lu, Danielle F Miyagishima, Phan Q. Duy, Daniel H. Geschwind, Shih Shan Lang Chen, Dean Thumkeo, James R. Knight, Bo Zhang, Shozeb Haider, Adam J. Kundishora, Francesc López-Giráldez, Darren B. Orbach, Boyang Li, Samuel T. Peters, Sheng Chih Jin, Tanyeri Barak, Arnaud Marlier, Charles C. Matouk, Seth L. Alper, Richard P. Lifton, Michael J. Bamshad, Shrikant Mane, Shujuan Zhao, Jack Ocken, Weilai Dong, Hannah Smith, Ashley Dunbar, Amber N. Stratman, Daniel Duran, Kristopher T. Kahle, Shreyas Panchagnula, Christopher Castaldi, Dianna M. Milewicz, Benjamin C. Reeves, Rebecca L. Walker, Michael L. DiLuna, Phillip B. Storm, Isabelle Roszko, Nanthiya Sujijantarat, Yi Hsien Chen, Amélie Pinard, Murat Gunel, Stephanie M. Robert, Deborah A. Nickerson, Hongyu Zhao, and Edward R. Smith
- Subjects
Proband ,Oncology ,Adult ,Male ,medicine.medical_specialty ,Candidate gene ,Formins ,Genome-wide association study ,Compound heterozygosity ,White People ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Exome Sequencing ,medicine ,Humans ,Computer Simulation ,Exome ,030212 general & internal medicine ,Age of Onset ,Child ,Exome sequencing ,Genetic association ,Original Investigation ,business.industry ,Sequence Analysis, RNA ,Genetic Variation ,Infant ,Middle Aged ,Actin cytoskeleton ,Magnetic Resonance Imaging ,Phenotype ,Child, Preschool ,Cohort ,Female ,Neurology (clinical) ,Moyamoya Disease ,business ,Cell Adhesion Molecules ,030217 neurology & neurosurgery - Abstract
Importance Moyamoya disease (MMD), a progressive vasculopathy leading to narrowing and ultimate occlusion of the intracranial internal carotid arteries, is a cause of childhood stroke. The cause of MMD is poorly understood, but genetic factors play a role. Several familial forms of MMD have been identified, but the cause of most cases remains elusive, especially among non-East Asian individuals. Objective To assess whether ultrarare de novo and rare, damaging transmitted variants with large effect sizes are associated with MMD risk. Design, setting, and participants A genetic association study was conducted using whole-exome sequencing case-parent MMD trios in a small discovery cohort collected over 3.5 years (2016-2019); data were analyzed in 2020. Medical records from US hospitals spanning a range of 1 month to 1.5 years were reviewed for phenotyping. Exomes from a larger validation cohort were analyzed to identify additional rare, large-effect variants in the top candidate gene. Participants included patients with MMD and, when available, their parents. All participants who met criteria and were presented with the option to join the study agreed to do so; none were excluded. Twenty-four probands (22 trios and 2 singletons) composed the discovery cohort, and 84 probands (29 trios and 55 singletons) composed the validation cohort. Main outcomes and measures Gene variants were identified and filtered using stringent criteria. Enrichment and case-control tests assessed gene-level variant burden. In silico modeling estimated the probability of variant association with protein structure. Integrative genomics assessed expression patterns of MMD risk genes derived from single-cell RNA sequencing data of human and mouse brain tissue. Results Of the 24 patients in the discovery cohort, 14 (58.3%) were men and 18 (75.0%) were of European ancestry. Three of 24 discovery cohort probands contained 2 do novo (1-tailed Poisson P = 1.1 × 10-6) and 1 rare, transmitted damaging variant (12.5% of cases) in DIAPH1 (mammalian diaphanous-1), a key regulator of actin remodeling in vascular cells and platelets. Four additional ultrarare damaging heterozygous DIAPH1 variants (3 unphased) were identified in 3 other patients in an 84-proband validation cohort (73.8% female, 77.4% European). All 6 patients were non-East Asian. Compound heterozygous variants were identified in ena/vasodilator-stimulated phosphoproteinlike protein EVL, a mammalian diaphanous-1 interactor that regulates actin polymerization. DIAPH1 and EVL mutant probands had severe, bilateral MMD associated with transfusion-dependent thrombocytopenia. DIAPH1 and other MMD risk genes are enriched in mural cells of midgestational human brain. The DIAPH1 coexpression network converges in vascular cell actin cytoskeleton regulatory pathways. Conclusions and relevance These findings provide the largest collection to date of non-East Asian individuals with sporadic MMD harboring pathogenic variants in the same gene. The results suggest that DIAPH1 is a novel MMD risk gene and impaired vascular cell actin remodeling in MMD pathogenesis, with diagnostic and therapeutic ramifications.
- Published
- 2021
19. Recruitment of APOL1 kidney disease risk variants to lipid droplets attenuates cell toxicity
- Author
-
Jose M. Magraner, Seth L. Alper, Cristian Riella, Maris S. Wilkins, David J. Friedman, Balajikarthick Subramanian, Justin Chun, Martin R. Pollak, and Jia-Yue Zhang
- Subjects
Programmed cell death ,Endosome ,Black People ,Endosomes ,Biology ,Mitochondrion ,Endoplasmic Reticulum ,Kidney ,Podocyte ,Risk Factors ,Lipid droplet ,Autophagy ,medicine ,Humans ,Multidisciplinary ,Podocytes ,Endoplasmic reticulum ,Genetic Variation ,Lipid Droplets ,Apolipoprotein L1 ,Cell biology ,HEK293 Cells ,medicine.anatomical_structure ,PNAS Plus ,Kidney Diseases ,Lysosomes ,Intracellular - Abstract
Two coding variants in the apolipoprotein L1 (APOL1) gene (termed G1 and G2) are strongly associated with increased risk of nondiabetic kidney disease in people of recent African ancestry. The mechanisms by which the risk variants cause kidney damage, although not well-understood, are believed to involve injury to glomerular podocytes. The intracellular localization and function of APOL1 in podocytes remain unclear, with recent studies suggesting possible roles in the endoplasmic reticulum (ER), mitochondria, endosomes, lysosomes, and autophagosomes. Here, we demonstrate that APOL1 also localizes to intracellular lipid droplets (LDs). While a large fraction of risk variant APOL1 (G1 and G2) localizes to the ER, a significant proportion of wild-type APOL1 (G0) localizes to LDs. APOL1 transiently interacts with numerous organelles, including the ER, mitochondria, and endosomes. Treatment of cells that promote LD formation with oleic acid shifted the localization of G1 and G2 from the ER to LDs, with accompanying reduction of autophagic flux and cytotoxicity. Coexpression of G0 APOL1 with risk variant APOL1 enabled recruitment of G1 and G2 from the ER to LDs, accompanied by reduced cell death. The ability of G0 APOL1 to recruit risk variant APOL1 to LDs may help explain the recessive pattern of kidney disease inheritance. These studies establish APOL1 as a bona fide LD-associated protein, and reveal that recruitment of risk variant APOL1 to LDs reduces cell toxicity, autophagic flux, and cell death. Thus, interventions that divert APOL1 risk variants to LDs may serve as a novel therapeutic strategy to alleviate their cytotoxic effects.
- Published
- 2019
20. Inflammatory hydrocephalus
- Author
-
Stephanie M. Robert, Benjamin C. Reeves, Arnaud Marlier, Phan Q. Duy, Tyrone DeSpenza, Adam Kundishora, Emre Kiziltug, Amrita Singh, Garrett Allington, Seth L. Alper, and Kristopher T. Kahle
- Subjects
Inflammation ,Pediatrics, Perinatology and Child Health ,Cytokines ,Humans ,Hemorrhage ,Neurology (clinical) ,General Medicine ,Hydrocephalus - Abstract
Reparative inflammation is an important protective response that eliminates foreign organisms, damaged cells, and physical irritants. However, inappropriately triggered or sustained inflammation can respectively initiate, propagate, or prolong disease. Post-hemorrhagic (PHH) and post-infectious hydrocephalus (PIH) are the most common forms of hydrocephalus worldwide. They are treated using neurosurgical cerebrospinal fluid (CSF) diversion techniques with high complication and failure rates. Despite their distinct etiologies, clinical studies in human patients have shown PHH and PIH share similar CSF cytokine and immune cell profiles. Here, in light of recent work in model systems, we discuss the concept of "inflammatory hydrocephalus" to emphasize potential shared mechanisms and potential therapeutic vulnerabilities of these disorders. We propose that this change of emphasis could shift our thinking of PHH and PIH from a framework of life-long neurosurgical disorders to that of preventable conditions amenable to immunomodulation.
- Published
- 2021
21. PTEN mutations in autism spectrum disorder and congenital hydrocephalus: developmental pleiotropy and therapeutic targets
- Author
-
Nell Mermin-Bunnell, Seth L. Alper, Phan Q. Duy, Jack Ocken, Benjamin C. Reeves, Ellen J. Hoffman, Aladine A. Elsamadicy, Marina Carlson, Adam J. Kundishora, Shreyas Panchagnula, Tyrone DeSpenza, Kristopher T. Kahle, Hannah Smith, Sheng Chih Jin, and Stephanie M. Robert
- Subjects
Autism Spectrum Disorder ,Bioinformatics ,behavioral disciplines and activities ,Article ,Phosphatidylinositol 3-Kinases ,mental disorders ,Medicine ,PTEN ,Tensin ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,Mammals ,biology ,business.industry ,Mechanism (biology) ,General Neuroscience ,Macrocephaly ,PTEN Phosphohydrolase ,medicine.disease ,Phenotype ,Pleiotropy (drugs) ,Autism spectrum disorder ,Neurodevelopmental Disorders ,Mutation ,biology.protein ,medicine.symptom ,business ,Hydrocephalus - Abstract
The lack of effective treatments for autism spectrum disorder (ASD) and congenital hydrocephalus (CH) reflects the limited understanding of the biology underlying these common neurodevelopmental disorders. Although ASD and CH have been extensively studied as independent entities, recent human genomic and pre-clinical animal studies have uncovered shared molecular pathophysiology. Here, we review and discuss phenotypic, genomic, and molecular similarities between ASD and CH, and identify the PTEN-PI3K-mTOR (phosphatase and tensin homolog-phosphoinositide 3-kinase-mammalian target of rapamycin) pathway as a common underlying mechanism that holds diagnostic, prognostic, and therapeutic promise for individuals with ASD and CH.
- Published
- 2021
22. Genomics of human congenital hydrocephalus
- Author
-
Kristopher T. Kahle, Jian Ryou, Seth L. Alper, Phan Q. Duy, Adam J. Kundishora, Amrita K. Singh, Sheng Chih Jin, and Garrett Allington
- Subjects
medicine.medical_specialty ,business.industry ,Genetic counseling ,Brain ,General Medicine ,Disease ,Human brain ,Genomics ,medicine.disease ,Bioinformatics ,Hydrocephalus ,Cerebral Ventricles ,Pathogenesis ,Cerebrospinal fluid ,medicine.anatomical_structure ,Pediatrics, Perinatology and Child Health ,Exome Sequencing ,Medicine ,Humans ,Neurology (clinical) ,Neurosurgery ,business ,Ventriculomegaly - Abstract
Congenital hydrocephalus (CH), characterized by enlarged brain ventricles, is considered a disease of pathological cerebrospinal fluid (CSF) accumulation and, therefore, treated largely by neurosurgical CSF diversion. The persistence of ventriculomegaly and poor neurodevelopmental outcomes in some post-surgical patients highlights our limited knowledge of disease mechanisms. Recent whole-exome sequencing (WES) studies have shown that rare, damaging de novo and inherited mutations with large effect contribute to ~ 25% of sporadic CH. Interestingly, multiple CH genes are key regulators of neural stem cell growth and differentiation and converge in human transcriptional networks and cell types pertinent to fetal neurogliogenesis. These data implicate genetic disruption of early brain development as the primary pathomechanism in a substantial minority of patients with sporadic CH, shedding new light on human brain development and the pathogenesis of hydrocephalus. These data further suggest WES as a clinical tool with potential to re-classify CH according to a molecular nomenclature of increased precision and utility for genetic counseling, outcome prognostication, and treatment stratification.
- Published
- 2021
23. A
- Author
-
David H, Vandorpe, Boris E, Shmukler, Yann, Ilboudo, Swati, Bhasin, Beena, Thomas, Alicia, Rivera, Jay G, Wohlgemuth, Jeffrey S, Dlott, L Michael, Snyder, Colin, Sieff, Manoj, Bhasin, Guillaume, Lettre, Carlo, Brugnara, and Seth L, Alper
- Subjects
Cations ,Humans ,Spherocytosis, Hereditary ,Letters to the Editor ,Permeability - Published
- 2021
24. Countermeasures against COVID-19: how to navigate medical practice through a nascent, evolving evidence base - a European multicentre mixed methods study
- Author
-
Günter Klaus, Tanja Stamm, Christoph Aufricht, Augustina Jankauskiene, Claus Peter Schmitt, Seth L. Alper, Aleksandra Zurowska, Valentin Ritschl, Constantinos J. Stefanidis, Asil Cetin, Karel Vondrak, Sevcan A. Bakkaloglu, Fabian Eibensteiner, Enrico Verrina, Fabio Paglialonga, Gema Ariceta, Alberto Edefonti, Bruno Ranchin, Rukshana Shroff, Johan Vandewalle, Institut Català de la Salut, [Eibensteiner F] Division of Pediatric Nephrology and Gastroenterology, Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics, Medical University of Vienna, Vienna, Austria. Ludwig Boltzmann Institute Digital Health and Patient Safety, Medical University of Vienna, Wien, Austria. [Ritschl V, Stamm T] Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria. Ludwig Boltzmann Institute for Arthritis and Rehabilitation, Vienna, Austria. [Cetin A] Research Platform Data Science, University of Vienna, Vienna, Austria. [Schmitt CP] Pediatric Nephrology, Center for Child and Adolescent Medicine, Heidelberg University, Heidelberg, Germany. [Ariceta G] Servei de Nefrologia Pediàtrica, Vall d'Hebron Hospital Universitari, Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
medicine.medical_specialty ,nephrology ,COVID-19 ,health & safety ,health policy ,paediatrics ,qualitative research ,030204 cardiovascular system & hematology ,Pediatrics ,03 medical and health sciences ,0302 clinical medicine ,Statutory law ,Renal Dialysis ,medicine ,virosis::infecciones por virus ARN::infecciones por Nidovirales::infecciones por Coronaviridae::infecciones por Coronavirus [ENFERMEDADES] ,Humans ,Pandèmia de COVID-19, 2020 ,030212 general & internal medicine ,Child ,instalaciones, servicios y personal de asistencia sanitaria::centros sanitarios::hospitales::centros de asistencia terciaria [ATENCIÓN DE SALUD] ,Personal protective equipment ,Pandemics ,Health policy ,Health Care Facilities, Manpower, and Services::Health Facilities::Hospitals::Tertiary Care Centers [HEALTH CARE] ,administración de los servicios de salud::organización y administración::medidas de seguridad [ATENCIÓN DE SALUD] ,computer.programming_language ,Response rate (survey) ,Health Services Administration::Organization and Administration::Security Measures [HEALTH CARE] ,Infection Control ,business.industry ,Nefrologia pediàtrica ,Paediatrics ,Virus Diseases::RNA Virus Infections::Nidovirales Infections::Coronaviridae Infections::Coronavirus Infections [DISEASES] ,General Medicine ,Centres sanitaris - Mesures de seguretat ,Europe ,Countermeasure ,Work (electrical) ,Family medicine ,Medicine ,business ,computer ,Delivery of Health Care ,Delphi ,Qualitative research - Abstract
ObjectivesIn a previously published Delphi exercise the European Pediatric Dialysis Working Group (EPDWG) reported widely variable counteractive responses to COVID-19 during the first week of statutory public curfews in 12 European countries with case loads of 4–680 infected patients per million. To better understand these wide variations, we assessed different factors affecting countermeasure implementation rates and applied the capability, opportunity, motivation model of behaviour to describe their determinants.DesignWe undertook this international mixed methods study of increased depth and breadth to obtain more complete data and to better understand the resulting complex evidence.SettingThis study was conducted in 14 paediatric nephrology centres across 12 European countries during the COVID-19 pandemic.ParticipantsThe 14 participants were paediatric nephrologists and EPDWG members from 12 European centres.Main outcome measures52 countermeasures clustered into eight response domains (access control, patient testing, personnel testing, personal protective equipment policy, patient cohorting, personnel cohorting, suspension of routine care, remote work) were categorised by implementation status, drivers (expert opinion, hospital regulations) and resource dependency. Governmental strictness and media attitude were independently assessed for each country and correlated with relevant countermeasure implementation factors.ResultsImplementation rates varied widely among response domains (median 49.5%, range 20%–71%) and centres (median 46%, range 31%–62%). Case loads were insufficient to explain response rate variability. Increasing case loads resulted in shifts from expert opinion-based to hospital regulation-based decisions to implement additional countermeasures despite increased resource dependency. Higher governmental strictness and positive media attitude towards countermeasure implementation were associated with higher implementation rates.ConclusionsCOVID-19 countermeasure implementation by paediatric tertiary care centres did not reflect case loads but rather reflected heterogeneity of local rules and of perceived resources. These data highlight the need of ongoing reassessment of current practices, facilitating rapid change in ‘institutional behavior’ in response to emerging evidence of countermeasure efficacy.
- Published
- 2021
25. Plasma Mucin-1 (CA15-3) Levels in Autosomal Dominant Tubulointerstitial Kidney Disease due to MUC1 Mutations
- Author
-
Petr, Vylet'al, Kendrah, Kidd, Hannah C, Ainsworth, Drahomíra, Springer, Alena, Vrbacká, Anna, Přistoupilová, Rebecca P, Hughey, Seth L, Alper, Niall, Lennon, Steven, Harrison, Maegan, Harden, Victoria, Robins, Abbigail, Taylor, Lauren, Martin, Katrice, Howard, Ibrahim, Bitar, Carl D, Langefeld, Veronika, Barešová, Hana, Hartmannová, Kateřina, Hodaňová, Tomáš, Zima, Martina, Živná, Stanislav, Kmoch, and Anthony J, Bleyer
- Subjects
Adult ,Male ,Patient-Oriented, Translational Research: Research Article ,Mucin-1 ,Middle Aged ,Prognosis ,digestive system ,biological factors ,digestive system diseases ,Healthy Volunteers ,Cross-Sectional Studies ,Case-Control Studies ,Mutation ,Uromodulin ,Humans ,Nephritis, Interstitial ,Female ,skin and connective tissue diseases ,neoplasms ,Alleles ,Biomarkers ,Aged - Abstract
INTRODUCTION: Patients with ADTKD-MUC1 have one allele producing normal mucin-1 (MUC1) and one allele producing mutant MUC1, which remains intracellular. We hypothesized that ADTKD-MUC1 patients, who have only 1 secretory-competent wild-type MUC1 allele, should exhibit decreased plasma mucin-1 (MUC1) levels. To test this hypothesis, we repurposed the serum CA15-3 assay used to measure MUC1 in breast cancer to measure plasma MUC1 levels in ADTKD-MUC1. METHODS: This cross-sectional study analyzed CA15-3 levels in a reference population of 6,850 individuals, in 85 individuals with ADTKD-MUC1, and in a control population including 135 individuals with ADTKD-UMOD and 114 healthy individuals. RESULTS: Plasma CA15-3 levels (mean ± standard deviation) were 8.6 ± 4.3 U/mL in individuals with ADTKD-MUC1 and 14.6 ± 5.6 U/mL in controls (p < 0.001). While there was a significant difference in mean CA15-3 levels, there was substantial overlap between the 2 groups. Plasma CA15-3 levels were 20 U/mL in 1/85 ADTKD-MUC1 patients, in 18% of control individuals, and in 25% of the reference population. Segregation of plasma CA15-3 levels by the rs4072037 genotype did not significantly improve differentiation between affected and unaffected individuals. CA15-3 levels were minimally affected by gender and estimated glomerular filtration rate. DISCUSSION/CONCLUSIONS: Plasma CA15-3 levels in ADTKD-MUC1 patients are approximately 40% lower than levels in healthy individuals, though there is significant overlap between groups. Further investigations need to be performed to see if plasma CA15-3 levels would be useful in diagnosis, prognosis, or assessing response to new therapies in this disorder.
- Published
- 2020
26. Exome sequencing implicates genetic disruption of prenatal neuro-gliogenesis in sporadic congenital hydrocephalus
- Author
-
William J. Sullivan, Bermans J. Iskandar, Yasar Bayri, James R. Knight, James M. Johnston, Michael L.J. Apuzzo, Kristopher T. Kahle, Boyang Li, Andrew T. Timberlake, Sheng Chih Jin, Steven J. Schiff, Shreyas Panchagnula, Rebecca L. Walker, Shozeb Haider, Li Ge, Daniel H. Geschwind, Hannah Smith, Richard P. Lifton, Seth L. Alper, Carol Nelson-Williams, Boris Keren, Christine Hehnly, Arnaud Marlier, Bulent Guclu, Laura R. Ment, Ellen J. Hoffman, Francesco T. Mangano, Xue Zeng, Edith Mbabazi Kabachelor, Kaya Bilguvar, August A Allocco, Ashley Dunbar, James R. Broach, Benjamin C. Warf, William E. Butler, Helena Perez Pena, Sierra B Conine, David D. Limbrick, Qiongshi Lu, Edward R. Smith, Jason K. Karimy, Christopher Castaldi, Eric M. Jackson, Yener Sahin, Murat Gunel, Adam J. Kundishora, Charles C. Duncan, Michael L. DiLuna, Shrikant Mane, Michael C. Sierant, Gregory G. Heuer, June Goto, Charuta G. Furey, Andres Moreno-De-Luca, Peter Ssenyonga, Weilai Dong, Nenad Sestan, Phan Q. Duy, Benjamin C. Reeves, Tyrone DeSpenza, Irina Tikhonova, Jin, Sheng Chih, Dong, Weilai, Kundishora, Adam J., Panchagnula, Shreyas, Moreno-De-Luca, Andres, Furey, Charuta G., Allocco, August A., Walker, Rebecca L., Nelson-Williams, Carol, Smith, Hannah, Dunbar, Ashley, Conine, Sierra, Lu, Qiongshi, Zeng, Xue, Sierant, Michael C., Knight, James R., Sullivan, William, Duy, Phan Q., DeSpenza, Tyrone, Reeves, Benjamin C., Karimy, Jason K., Marlier, Arnaud, Castaldi, Christopher, Tikhonova, Irina R., Li, Boyang, Pena, Helena Perez, Broach, James R., Kabachelor, Edith M., Ssenyonga, Peter, Hehnly, Christine, Ge, Li, Keren, Boris, Timberlake, Andrew T., Goto, June, Mangano, Francesco T., Johnston, James M., Butler, William E., Warf, Benjamin C., Smith, Edward R., Schiff, Steven J., Limbrick, David D., Jr., Heuer, Gregory, Jackson, Eric M., Iskandar, Bermans J., Mane, Shrikant, Haider, Shozeb, Guclu, Bulent, Bayri, Yasar, Sahin, Yener, Duncan, Charles C., Apuzzo, Michael L. J., DiLuna, Michael L., Hoffman, Ellen J., Sestan, Nenad, Ment, Laura R., Alper, Seth L., Bilguvar, Kaya, Geschwind, Daniel H., Gunel, Murat, Lifton, Richard P., and Kahle, Kristopher T.
- Subjects
EXPRESSION ,0301 basic medicine ,Male ,PTEN ,Cell type ,Neurogenesis ,Ubiquitin-Protein Ligases ,CHILDREN ,Disease ,VENTRICULAR ZONE DISRUPTION ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,Article ,COWDEN-SYNDROME ,Cerebral Ventricles ,PATHWAY ,Tripartite Motif Proteins ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,RARE ,Neural Stem Cells ,Pregnancy ,Exome Sequencing ,Medicine ,Humans ,Genetic Predisposition to Disease ,Exome ,AUTISM ,Exome sequencing ,Gliogenesis ,SPECTRUM ,Fetus ,business.industry ,Brain ,General Medicine ,medicine.disease ,Neural stem cell ,DE-NOVO MUTATION ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Female ,business ,Neuroglia ,Ventriculomegaly ,Transcription Factors ,Hydrocephalus - Abstract
Congenital hydrocephalus (CH), characterized by enlarged brain ventricles, is considered a disease of excessive cerebrospinal fluid (CSF) accumulation and thereby treated with neurosurgical CSF diversion with high morbidity and failure rates. The poor neurodevelopmental outcomes and persistence of ventriculomegaly in some post-surgical patients highlight our limited knowledge of disease mechanisms. Through whole-exome sequencing of 381 patients (232 trios) with sporadic, neurosurgically treated CH, we found that damaging de novo mutations account for >17% of cases, with five different genes exhibiting a significant de novo mutation burden. In all, rare, damaging mutations with large effect contributed to similar to 22% of sporadic CH cases. Multiple CH genes are key regulators of neural stem cell biology and converge in human transcriptional networks and cell types pertinent for fetal neuro-gliogenesis. These data implicate genetic disruption of early brain development, not impaired CSF dynamics, as the primary pathomechanism of a significant number of patients with sporadic CH.
- Published
- 2020
27. Phosphorylation of ACTN4 Leads to Podocyte Vulnerability and Proteinuric Glomerulosclerosis
- Author
-
Isaac E. Stillman, Mukesh Kumar, Jan Muntel, Di Feng, Seth L. Alper, Susan B. Gurley, Johannes Schlondorff, Donald E. Ingber, Hanno Steen, Susan Marquez, Saima Ahmed, Gabriel Birrane, Martin R. Pollak, Carlos F. Ng, Richard M. Novak, Tom Ferrante, Lai Ding, and Minxian Wang
- Subjects
Male ,macromolecular substances ,Nephrectomy ,Podocyte ,Mice ,Focal segmental glomerulosclerosis ,Transforming Growth Factor beta ,Lab-On-A-Chip Devices ,medicine ,Extracellular ,Serine ,Albuminuria ,Animals ,Humans ,Actinin ,Phosphorylation ,Cells, Cultured ,Kidney ,Chemistry ,Glomerulosclerosis, Focal Segmental ,Podocytes ,Wild type ,Glomerulosclerosis ,General Medicine ,medicine.disease ,Actins ,Cell biology ,medicine.anatomical_structure ,Basic Research ,Glucose ,Renal pathology ,Nephrology ,Female ,Peptidomimetics ,Protein Processing, Post-Translational ,Protein Binding - Abstract
Background Genetic mutations in α-actinin-4 (ACTN4)-an important actin crosslinking cytoskeletal protein that provides structural support for kidney podocytes-have been linked to proteinuric glomerulosclerosis in humans. However, the effect of post-translational modifications of ACTN4 on podocyte integrity and kidney function is not known. Methods Using mass spectrometry, we found that ACTN4 is phosphorylated at serine (S) 159 in human podocytes. We used phosphomimetic and nonphosphorylatable ACTN4 to comprehensively study the effects of this phosphorylation in vitro and in vivo. We conducted x-ray crystallography, F-actin binding and bundling assays, and immunofluorescence staining to evaluate F-actin alignment. Microfluidic organ-on-a-chip technology was used to assess for detachment of podocytes simultaneously exposed to fluid flow and cyclic strain. We then used CRISPR/Cas9 to generate mouse models and assessed for renal injury by measuring albuminuria and examining kidney histology. We also performed targeted mass spectrometry to determine whether high extracellular glucose or TGF-β levels increase phosphorylation of ACTN4. Results Compared with the wild type ACTN4, phosphomimetic ACTN4 demonstrated increased binding and bundling activity with F-actin in vitro. Phosphomimetic Actn4 mouse podocytes exhibited more spatially correlated F-actin alignment and a higher rate of detachment under mechanical stress. Phosphomimetic Actn4 mice developed proteinuria and glomerulosclerosis after subtotal nephrectomy. Moreover, we found that exposure to high extracellular glucose or TGF-β stimulates phosphorylation of ACTN4 at S159 in podocytes. Conclusions These findings suggest that increased phosphorylation of ACTN4 at S159 leads to biochemical, cellular, and renal pathology that is similar to pathology resulting from human disease-causing mutations in ACTN4. ACTN4 may mediate podocyte injury as a consequence of both genetic mutations and signaling events that modulate phosphorylation.
- Published
- 2020
28. Peritoneal Dialysis Fluid Supplementation with Alanyl-Glutamine Attenuates Conventional Dialysis Fluid-Mediated Endothelial Cell Injury by Restoring Perturbed Cytoprotective Responses
- Author
-
Claus Peter Schmitt, Betti Schaefer, Maria Bartosova, Klaus Kratochwill, Lilian Kuster, Anja Wagner, Seth L. Alper, Rebecca Herzog, Silvia Tarantino, Anton Lichtenauer, Juan Manuel Sacnun, Markus Unterwurzacher, and Christoph Aufricht
- Subjects
0301 basic medicine ,Proteomics ,Endothelium ,medicine.medical_treatment ,lcsh:QR1-502 ,030232 urology & nephrology ,Pharmacology ,Biochemistry ,Models, Biological ,lcsh:Microbiology ,Umbilical vein ,Article ,Peritoneal dialysis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,vascular damage ,Dialysis Solutions ,medicine ,Extracellular ,Human Umbilical Vein Endothelial Cells ,Humans ,l-Alanyl-l-glutamine ,Child ,Molecular Biology ,vasculopathy ,Chemistry ,Dipeptides ,Endothelial stem cell ,Arterioles ,030104 developmental biology ,medicine.anatomical_structure ,peritoneal dialysis ,Cytoprotection ,Proteome ,Alanyl glutamine - Abstract
Long-term clinical outcome of peritoneal dialysis (PD) depends on adequate removal of small solutes and water. The peritoneal endothelium represents the key barrier and peritoneal transport dysfunction is associated with vascular changes. Alanyl-glutamine (AlaGln) has been shown to counteract PD-induced deteriorations but the effect on vascular changes has not yet been elucidated. Using multiplexed proteomic and bioinformatic analyses we investigated the molecular mechanisms of vascular pathology in-vitro (primary human umbilical vein endothelial cells, HUVEC) and ex-vivo (arterioles of patients undergoing PD) following exposure to PD-fluid. An overlap of 1813 proteins (40%) of over 3100 proteins was identified in both sample types. PD-fluid treatment significantly altered 378 in endothelial cells and 192 in arterioles. The HUVEC proteome resembles the arteriolar proteome with expected sample specific differences of mainly immune system processes only present in arterioles and extracellular region proteins primarily found in HUVEC. AlaGln-addition to PD-fluid revealed 359 differentially abundant proteins and restored the molecular process landscape altered by PD fluid. This study provides evidence on validity and inherent limitations of studying endothelial pathomechanisms in-vitro compared to vascular ex-vivo findings. AlaGln could reduce PD-associated vasculopathy by reducing endothelial cellular damage, restoring perturbed abundances of pathologically important proteins and enriching protective processes.
- Published
- 2020
- Full Text
- View/download PDF
29. Trpv1 and Trpa1 are not essential for Psickle-like activity in red cells of the SAD mouse model of sickle cell disease
- Author
-
Boris E. Shmukler, Marie Trudel, L. Michael Snyder, David H. Vandorpe, Jay Wohlgemuth, Seth L. Alper, Alicia Rivera, Jeffrey S. Dlott, and Carlo Brugnara
- Subjects
Agonist ,medicine.medical_specialty ,Erythrocytes ,medicine.drug_class ,Cell ,TRPV1 ,TRPV Cation Channels ,chemistry.chemical_element ,Anemia, Sickle Cell ,Disease ,Calcium ,behavioral disciplines and activities ,Mice ,Cations ,Internal medicine ,mental disorders ,medicine ,Animals ,Humans ,TRPA1 Cation Channel ,Molecular Biology ,Mice, Knockout ,Erythrocyte fragility ,food and beverages ,Cell Biology ,Hematology ,Hypoxia (medical) ,Disease Models, Animal ,medicine.anatomical_structure ,Endocrinology ,chemistry ,behavior and behavior mechanisms ,Molecular Medicine ,medicine.symptom ,Gene Deletion ,psychological phenomena and processes - Abstract
The molecular identity of Psickle, the deoxygenation-activated cation conductance of the human sickle erythrocyte, remains unknown. We observed in human sickle red cells that inhibitors of TRPA1 and TRPV1 inhibited Psickle, whereas a TRPV1 agonist activated a Psickle-like cation current. These observations prompted us to test the roles of TRPV1 and TRPA1 in Psickle in red cells of the SAD mouse model of sickle cell disease. We generated SAD mice genetically deficient in either TRPV1 or TRPA1. SAD;Trpv1-/- and SAD;Trpa1-/- mice were indistinguishable in appearance, hematological indices, and osmotic fragility from SAD mice. We found that deoxygenation-activated cation currents remained robust in SAD;Trpa1-/- and SAD;Trpv1-/- mice. In addition, 45Ca2+ influx into SAD mouse red cells during prolonged deoxygenation was not reduced in red cells from SAD;Trpa1-/- and SAD;Trpv1-/- mice. We conclude that the nonspecific cation channels TRPA1 and TRPV1 are not required for deoxygenation to stimulate Psickle-like activity in red cells of the SAD mouse model of sickle cell disease. (159).
- Published
- 2021
30. Genome-wide association study of erythrocyte density in sickle cell disease patients
- Author
-
Pablo Bartolucci, Seth L. Alper, Guillaume Lettre, Marie Trudel, Carlo Brugnara, Alicia Rivera, Yann Ilboudo, Josepha-Clara Sedzro, Mélissa Beaudoin, and Frédéric Galactéros
- Subjects
Adult ,Erythrocyte Indices ,Male ,0301 basic medicine ,medicine.medical_specialty ,Erythrocytes ,Genotype ,Calcium pump ,Hemoglobin, Sickle ,Quantitative Trait Loci ,Genome-wide association study ,Anemia, Sickle Cell ,Biology ,Polymorphism, Single Nucleotide ,Plasma Membrane Calcium-Transporting ATPases ,Young Adult ,03 medical and health sciences ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Humans ,Molecular Biology ,Alleles ,Mean corpuscular hemoglobin concentration ,medicine.diagnostic_test ,Genetic Variation ,Cell Biology ,Hematology ,medicine.disease ,Sickle cell anemia ,Red blood cell ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,ATP2B4 ,Immunology ,Molecular Medicine ,Female ,Hemoglobin ,Intracellular ,Genome-Wide Association Study - Abstract
Deoxy-hemoglobin S polymerization into rigid fibers is the direct cause of the clinical sequelae observed in sickle cell disease (SCD). The rate of polymerization of sickle hemoglobin is determined primarily by intracellular hemoglobin concentration, itself dependent on the amount of sickle hemoglobin and on red blood cell (RBC) volume. Dense, dehydrated RBC (DRBC) are observed in SCD patients, and their number correlates with hemolytic parameters and complications such as renal dysfunction, leg ulcers and priapism. To identify new genes involved in RBC hydration in SCD, we performed the first genome-wide association study for DRBC in 374 sickle cell anemia (HbSS) patients. We did not find genome-wide significant results, indicating that variants that modulate DRBC have modest-to-weak effects. A secondary analysis demonstrated a nominal association (P=0.003) between DRBC in SCD patients and a variant associated with mean corpuscular hemoglobin concentration (MCHC) in non-anemic individuals. This intronic variant controls the expression of ATP2B4, the main calcium pump in erythrocytes. Our study highlights ATP2B4 as a promising target for modulation of RBC hydration in SCD patients.
- Published
- 2017
31. Lithium preserves peritoneal membrane integrity by suppressing mesothelial cell αB-crystallin
- Author
-
Klaus Kratochwill, Lisa Daniel-Fischer, Anja Wagner, Andreas Vychytil, Seth L. Alper, Markus Unterwurzacher, Rebecca Herzog, Katarzyna Bialas, Lucía Pascual-Antón, Christoph Aufricht, Maria Bartosova, Guadalupe González-Mateo, Krisztina Rusai, Juan Manuel Sacnun, Klaus Kaczirek, Isabel J. Sobieszek, Manuel López-Cabrera, and Claus Peter Schmitt
- Subjects
Proteomics ,Vascular Endothelial Growth Factor A ,Angiogenesis ,medicine.medical_treatment ,030232 urology & nephrology ,Lithium ,Peritoneal dialysis ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Fibrosis ,medicine ,Animals ,Humans ,Child ,Peritoneal Fibrosis ,030304 developmental biology ,0303 health sciences ,Chemistry ,Epithelial Cells ,General Medicine ,medicine.disease ,Crystallins ,3. Good health ,Vascular endothelial growth factor ,Vascular endothelial growth factor A ,Cancer research ,Peritoneum ,Homeostasis ,Mesothelial Cell - Abstract
Life-saving renal replacement therapy by peritoneal dialysis (PD) is limited in use and duration by progressive impairment of peritoneal membrane integrity and homeostasis. Preservation of peritoneal membrane integrity during chronic PD remains an urgent but long unmet medical need. PD therapy failure results from peritoneal fibrosis and angiogenesis caused by hypertonic PD fluid (PDF)-induced mesothelial cytotoxicity. However, the pathophysiological mechanisms involved are incompletely understood, limiting identification of therapeutic targets. We report that addition of lithium chloride (LiCl) to PDF is a translatable intervention to counteract PDF-induced mesothelial cell death, peritoneal membrane fibrosis, and angiogenesis. LiCl improved mesothelial cell survival in a dose-dependent manner. Combined transcriptomic and proteomic characterization of icodextrin-based PDF-induced mesothelial cell injury identified αB-crystallin as the mesothelial cell protein most consistently counter-regulated by LiCl. In vitro and in vivo overexpression of αB-crystallin triggered a fibrotic phenotype and PDF-like up-regulation of vascular endothelial growth factor (VEGF), CD31-positive cells, and TGF-β-independent activation of TGF-β-regulated targets. In contrast, αB-crystallin knockdown decreased VEGF expression and early mesothelial-to-mesenchymal transition. LiCl reduced VEGF release and counteracted fibrosis- and angiogenesis-associated processes. αB-crystallin in patient-derived mesothelial cells was specifically up-regulated in response to PDF and increased in peritoneal mesothelial cells from biopsies from pediatric patients undergoing PD, correlating with markers of angiogenesis and fibrosis. LiCl-supplemented PDF promoted morphological preservation of mesothelial cells and the submesothelial zone in a mouse model of chronic PD. Thus, repurposing LiCl as a cytoprotective PDF additive may offer a translatable therapeutic strategy to combat peritoneal membrane deterioration during PD therapy.
- Published
- 2019
32. APOL1 risk variants induce opening of the mitochondrial permeability transition pore
- Author
-
Shrijal S, Shah, Herbert, Lannon, Leny, Dias, Jia-Yue, Zhang, Seth L, Alper, Martin R, Pollak, and David J, Friedman
- Subjects
Cell Death ,Mitochondrial Permeability Transition Pore ,Cell Respiration ,Apolipoprotein L1 ,Kidney ,Mitochondrial Membrane Transport Proteins ,Recombinant Proteins ,Mitochondrial Transmembrane Permeability-Driven Necrosis ,Protein Transport ,HEK293 Cells ,Basic Research ,Gain of Function Mutation ,Humans ,Kidney Failure, Chronic ,Genetic Predisposition to Disease ,RNA Interference ,Protein Multimerization ,RNA, Small Interfering - Abstract
BACKGROUND: Genetic Variants in Apolipoprotein L1 (APOL1) are associated with large increases in CKD rates among African Americans. Experiments in cell and mouse models suggest that these risk-related polymorphisms are toxic gain-of-function variants that cause kidney dysfunction, following a recessive mode of inheritance. Recent data in trypanosomes and in human cells indicate that such variants may cause toxicity through their effects on mitochondria. METHODS: To examine the molecular mechanisms underlying APOL1 risk variant–induced mitochondrial dysfunction, we generated tetracycline-inducible HEK293 T-REx cells stably expressing the APOL1 nonrisk G0 variant or APOL1 risk variants. Using these cells, we mapped the molecular pathway from mitochondrial import of APOL1 protein to APOL1-induced cell death with small interfering RNA knockdowns, pharmacologic inhibitors, blue native PAGE, mass spectrometry, and assessment of mitochondrial permeability transition pore function. RESULTS: We found that the APOL1 G0 and risk variant proteins shared the same import pathway into the mitochondrial matrix. Once inside, G0 remained monomeric, whereas risk variant proteins were prone to forming higher-order oligomers. Both nonrisk G0 and risk variant proteins bound components of the mitochondrial permeability transition pore, but only risk variant proteins activated pore opening. Blocking mitochondrial import of APOL1 risk variants largely eliminated oligomer formation and also rescued toxicity. CONCLUSIONS: Our study illuminates important differences in the molecular behavior of APOL1 nonrisk and risk variants, and our observations suggest a mechanism that may explain the very different functional effects of these variants, despite the lack of consistently observed differences in trafficking patterns, intracellular localization, or binding partners. Variant-dependent differences in oligomerization pattern may underlie APOL1’s recessive, gain-of-function biology.
- Published
- 2019
33. Glymphatic System Impairment in Alzheimer's Disease and Idiopathic Normal Pressure Hydrocephalus
- Author
-
Benjamin C. Reeves, Jason K. Karimy, Adam J. Kundishora, Humberto Mestre, H. Mert Cerci, Charles Matouk, Seth L. Alper, Iben Lundgaard, Maiken Nedergaard, and Kristopher T. Kahle
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Disease ,03 medical and health sciences ,0302 clinical medicine ,Cerebrospinal fluid ,Alzheimer Disease ,medicine ,Dementia ,Animals ,Humans ,Molecular Biology ,Brain Ventricle ,Aquaporin 4 ,Amyloid beta-Peptides ,business.industry ,Brain ,medicine.disease ,Hydrocephalus, Normal Pressure ,Astrogliosis ,030104 developmental biology ,medicine.anatomical_structure ,Astrocytes ,Molecular Medicine ,Glymphatic system ,business ,Glymphatic System ,030217 neurology & neurosurgery ,Homeostasis ,Astrocyte - Abstract
Approximately 10% of dementia patients have idiopathic normal pressure hydrocephalus (iNPH), an expansion of the cerebrospinal fluid (CSF)-filled brain ventricles. iNPH and Alzheimer's disease (AD) both exhibit sleep disturbances, build-up of brain metabolic wastes and amyloid-β (Aβ) plaques, perivascular reactive astrogliosis, and mislocalization of astrocyte aquaporin-4 (AQP4). The glia–lymphatic (glymphatic) system facilitates brain fluid clearance and waste removal during sleep via glia-supported perivascular channels. Human studies have implicated impaired glymphatic function in both AD and iNPH. Continued investigation into the role of glymphatic system biology in AD and iNPH models could lead to new strategies to improve brain health by restoring homeostatic brain metabolism and CSF dynamics.
- Published
- 2019
34. Combined genetic disruption of K-Cl cotransporters and Gardos channel KCNN4 rescues erythrocyte dehydration in the SAD mouse model of sickle cell disease
- Author
-
Carlo Brugnara, Edward H. Kim, Marie Trudel, Ann Hsu, Parul Bhargava, Alicia Rivera, Katherine Nishimura, Seth L. Alper, Boris E. Shmukler, Christian A. Hübner, and Marco B. Rust
- Subjects
Lysis ,Erythrocytes ,Cell ,Anemia, Sickle Cell ,Article ,KCNN4 ,Mice ,medicine ,Animals ,Humans ,Molecular Biology ,Cell Size ,Red Cell ,Dehydration ,Symporters ,Chemistry ,Erythrocyte fragility ,Cell Biology ,Hematology ,Intermediate-Conductance Calcium-Activated Potassium Channels ,Molecular biology ,Potassium channel ,Red blood cell ,Disease Models, Animal ,medicine.anatomical_structure ,Molecular Medicine ,Cotransporter - Abstract
Excessive red cell dehydration contributes to the pathophysiology of sickle cell disease (SCD). The densest fraction of sickle red cells (with the highest corpuscular hemoglobin concentration) undergoes the most rapid polymerization of deoxy-hemoglobin S, leading to accelerated cell sickling and increased susceptibility to endothelial activation, red cell adhesion, and vaso-occlusion. Increasing red cell volume in order to decrease red cell density can thus serve as an adjunct therapeutic goal in SCD. Regulation of circulating mouse red cell volume and density is mediated largely by the Gardos channel, KCNN4, and the K-Cl cotransporters, KCC3 and KCC1. Whereas inhibition of the Gardos channel in subjects with sickle cell disease increased red cell volume, decreased red cell density, and improved other hematological indices in subjects with SCD, specific KCC inhibitors have not been available for testing. We therefore investigated the effect of genetic inactivation of KCC3 and KCC1 in the SAD mouse model of sickle red cell dehydration, finding decreased red cell density and improved hematological indices. We describe here generation of mice genetically deficient in the three major red cell volume regulatory gene products, KCNN4, KCC3, and KCC1 in C57BL6 non-sickle and SAD sickle backgrounds. We show that combined loss-of-function of all three gene products in SAD mice leads to incrementally increased MCV, decreased CHCM and % hyperchromic cells, decreased red cell density (phthalate method), increased resistance to hypo-osmotic lysis, and increased cell K content. The data show that combined genetic deletion of the Gardos channel and K-Cl cotransporters in a mouse SCD model decreases red cell density and improves several hematological parameters, supporting the strategy of combined pharmacological inhibition of these ion transport pathways in the adjunct treatment of human SCD.
- Published
- 2019
35. Transmembrane insertases and
- Author
-
Brianna E, Talbot, David H, Vandorpe, Brian R, Stotter, Seth L, Alper, and Johannes S, Schlondorff
- Subjects
Glycosylation ,Cell Death ,Podocytes ,Cell Membrane ,Membrane Proteins ,Cell Biology ,N-Acetylglucosaminyltransferases ,HEK293 Cells ,Gain of Function Mutation ,TRPC6 Cation Channel ,Humans ,Calcium ,CRISPR-Cas Systems ,Protein Binding ,RNA, Guide, Kinetoplastida - Abstract
Transient receptor potential cation channel subfamily C member 6 (TRPC6) is a widely expressed ion channel. Gain–of–function mutations in the human TRPC6 channel cause autosomal-dominant focal segmental glomerulosclerosis, but the molecular components involved in disease development remain unclear. Here, we found that overexpression of gain–of–function TRPC6 channel variants is cytotoxic in cultured cells. Exploiting this phenotype in a genome-wide CRISPR/Cas screen for genes whose inactivation rescues cells from TRPC6-associated cytotoxicity, we identified several proteins essential for TRPC6 protein expression, including the endoplasmic reticulum (ER) membrane protein complex transmembrane insertase. We also identified transmembrane protein 208 (TMEM208), a putative component of a signal recognition particle-independent (SND) ER protein-targeting pathway, as being necessary for expression of TRPC6 and several other ion channels and transporters. TRPC6 expression was also diminished by loss of the previously uncharacterized WD repeat domain 83 opposite strand (WDR83OS), which interacted with both TRPC6 and TMEM208. Additionally enriched among the screen hits were genes involved in N-linked protein glycosylation. Deletion of the mannosyl (α-1,3-)-glycoprotein β-1,2-N-acetylglucosaminyltransferase (MGAT1), necessary for the generation of complex N-linked glycans, abrogated TRPC6 gain–of–function variant-mediated Ca(2+) influx and extracellular signal-regulated kinase activation in HEK cells, but failed to diminish cytotoxicity in cultured podocytes. However, mutating the two TRPC6 N-glycosylation sites abrogated the cytotoxicity of mutant TRPC6 and reduced its surface expression. These results expand the targets of TMEM208-mediated ER translocation to include multipass transmembrane proteins and suggest that TRPC6 N-glycosylation plays multiple roles in modulating channel trafficking and activity.
- Published
- 2019
36. Erythrocyte ion content and dehydration modulate maximal gardos channel activity in KCNN4 V282M/+ hereditary xerocytosis red cells
- Author
-
Boris E. Shmukler, Michael Auerbach, Jay Wohlgemuth, Immacolata Andolfo, Carlo Brugnara, Natasha M. Archer, Seth L. Alper, James Morton, David H. Vandorpe, Nelson Hamerschlak, Achille Iolascon, Alicia Rivera, L. Michael Snyder, Estela Shabani, Rivera, A., Vandorpe, D. H., Shmukler, B. E., Andolfo, I., Iolascon, A., Archer, N. M., Shabani, E., Auerbach, M., Hamerschlak, N., Morton, J., Wohlgemuth, J. G., Brugnara, C., Snyder, L. M., and Alper, S. L.
- Subjects
Erythrocyte Indices ,0301 basic medicine ,Erythrocytes ,Physiology ,Hydrops Fetalis ,Senicapoc ,Mutant ,Mutation, Missense ,Anemia, Hemolytic, Congenital ,Ion Channels ,Membrane Potentials ,03 medical and health sciences ,KCNN4 ,chemistry.chemical_compound ,0302 clinical medicine ,Ionophore ,medicine ,Humans ,Missense mutation ,Genetic Predisposition to Disease ,Calcium Signaling ,Potassium channel ,PIEZO1 ,Cell Biology ,Dehydrated stomatocytosi ,Intermediate-Conductance Calcium-Activated Potassium Channels ,Osmotic Fragility ,Red blood cell ,Phenotype ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Case-Control Studies ,Potassium ,Biophysics ,Mechanosensitive channels ,030215 immunology - Abstract
Hereditary xerocytosis (HX) is caused by missense mutations in either the mechanosensitive cation channel PIEZO1 or the Ca2+-activated K+channel KCNN4. All HX-associated KCNN4 mutants studied to date have revealed increased current magnitude and red cell dehydration. Baseline KCNN4 activity was increased in HX red cells heterozygous for KCNN4 mutant V282M. However, HX red cells maximally stimulated by Ca2+ionophore A23187 or by PMCA Ca2+-ATPase inhibitor orthovanadate displayed paradoxically reduced KCNN4 activity. This reduced Ca2+-stimulated mutant KCNN4 activity in HX red cells was associated with unchanged sensitivity to KCNN4 inhibitor senicapoc and KCNN4 activator Ca2+, with slightly elevated Ca2+uptake and reduced PMCA activity, and with decreased KCNN4 activation by calpain inhibitor PD150606. The altered intracellular monovalent cation content of HX red cells prompted experimental nystatin manipulation of red cell Na and K contents. Nystatin-mediated reduction of intracellular K+with corresponding increase in intracellular Na+in wild-type cells to mimic conditions of HX greatly suppressed vanadate-stimulated and A23187 -stimulated KCNN4 activity in those wild-type cells. However, conferral of wild-type cation contents on HX red cells failed to restore wild-type-stimulated KCNN4 activity to those HX cells. The phenotype of reduced, maximally stimulated KCNN4 activity was shared by HX erythrocytes expressing heterozygous PIEZO1 mutants R2488Q and V598M, but not by HX erythrocytes expressing heterozygous KCNN4 mutant R352H or PIEZO1 mutant R2456H. Our data suggest that chronic KCNN4-driven red cell dehydration and intracellular cation imbalance can lead to reduced KCNN4 activity in HX and wild-type red cells.
- Published
- 2019
37. Loss of Cystic Fibrosis Transmembrane Regulator Impairs Intestinal Oxalate Secretion
- Author
-
Felix Knauf, Adedotun Adebamiro, Zhirong Jiang, John F. Heneghan, Marie E. Egan, R. B. Thomson, John R. Asplin, Seth L. Alper, Christina Barone, Claire L. Thomson, and Peter S. Aronson
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Cystic Fibrosis ,Urology ,Regulator ,030232 urology & nephrology ,Calcium oxalate ,Cystic Fibrosis Transmembrane Conductance Regulator ,Cystic fibrosis ,Antiporters ,Oxalate ,Intestinal absorption ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Intestinal mucosa ,Internal medicine ,medicine ,SLC26A6 ,Animals ,Humans ,Intestinal Mucosa ,Transcellular ,Oxalate secretion ,Ion transporter ,Mice, Knockout ,Hyperoxaluria ,Oxalates ,Ion Transport ,Calcium Oxalate ,biology ,business.industry ,General Medicine ,medicine.disease ,Transmembrane protein ,Cystic fibrosis transmembrane conductance regulator ,Cell biology ,Intestines ,Basic Research ,030104 developmental biology ,Endocrinology ,chemistry ,Sulfate Transporters ,Nephrology ,biology.protein ,business - Abstract
Patients with cystic fibrosis have an increased incidence of hyperoxaluria and calcium oxalate nephrolithiasis. Net intestinal absorption of dietary oxalate results from passive paracellular oxalate absorption as modified by oxalate back secretion mediated by the SLC26A6 oxalate transporter. We used mice deficient in the cystic fibrosis transmembrane conductance regulator gene (Cftr) to test the hypothesis that SLC26A6–mediated oxalate secretion is defective in cystic fibrosis. We mounted isolated intestinal tissue from C57BL/6 (wild-type) and Cftr−/− mice in Ussing chambers and measured transcellular secretion of [14C]oxalate. Intestinal tissue isolated from Cftr−/− mice exhibited significantly less transcellular oxalate secretion than intestinal tissue of wild-type mice. However, glucose absorption, another representative intestinal transport process, did not differ in Cftr−/− tissue. Compared with wild-type mice, Cftr−/− mice showed reduced expression of SLC26A6 in duodenum by immunofluorescence and Western blot analysis. Furthermore, coexpression of CFTR stimulated SLC26A6–mediated Cl−–oxalate exchange in Xenopus oocytes. In association with the profound defect in intestinal oxalate secretion, Cftr−/− mice had serum and urine oxalate levels 2.5-fold greater than those of wild-type mice. We conclude that defective intestinal oxalate secretion mediated by SLC26A6 may contribute to the hyperoxaluria observed in this mouse model of cystic fibrosis. Future studies are needed to address whether similar mechanisms contribute to the increased risk for calcium oxalate stone formation observed in patients with cystic fibrosis.
- Published
- 2016
38. Structural characterization of the C-terminal coiled-coil domains of wild-type and kidney disease-associated mutants of apolipoprotein L1
- Author
-
Seth L. Alper, Martin R. Pollak, David J. Friedman, and Alok K. Sharma
- Subjects
Trypanosoma brucei rhodesiense ,0301 basic medicine ,Circular dichroism ,Protein Conformation ,Apolipoprotein L1 ,Mutant ,Mutation, Missense ,030232 urology & nephrology ,Molecular Dynamics Simulation ,Biochemistry ,Article ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Missense mutation ,Amino Acid Sequence ,Cloning, Molecular ,Nuclear Magnetic Resonance, Biomolecular ,Molecular Biology ,Genetics ,Coiled coil ,Sequence Homology, Amino Acid ,biology ,Circular Dichroism ,Wild type ,Cell Biology ,Apolipoproteins ,030104 developmental biology ,Lytic cycle ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,biology.protein ,Kidney Diseases ,Lipoproteins, HDL - Abstract
Trypanosomes that cause sleeping sickness endocytose apolipoprotein L1 (APOL1)-containing trypanolytic factors from human serum, leading to trypanolytic death through generation of APOL1-associated lytic pores in trypanosomal membranes. The trypanosome Trypanosoma brucei rhodesiense counteracts trypanolysis by expressing the surface protein serum response-associated (SRA), which can bind APOL1 common variant G0 to block its trypanolytic activity. However, two missense variants in the C terminal predicted coiled-coil (CC) domains of human APOL1 G1 (S342G/I384M) and G2 (ΔN388Y389) decrease or abrogate APOL1 binding to T. brucei rhodesiense SRA, thus preserving APOL1 trypanolytic activity. These evolutionarily selected APOL1 missense variants, found at a high frequency in some populations of African descent, also confer elevated risk of kidney disease. Understanding the SRA-APOL1 interaction and the role of APOL1 G1 and G2 variants in kidney disease demands structural characterization of the APOL1 CC domain. Using CD, heteronuclear NMR, and molecular dynamics (MD) simulation on structural homology models, we report here unique and dynamic solution conformations of nephropathy variants G1 and G2 as compared with the common variant G0. Conformational plasticity in G1 and G2 CC domains led to interhelical α1-α2 approximation coupled with secondary structural changes and delimited motional properties absent in the G0 CC domain. The G1 substitutions conferred local structural changes principally along helix α1, whereas the G2 deletion altered the structure of both helix α2 and helix α1. These dynamic features of APOL1 CC variants likely reflect their intrinsic structural properties, and should help interpret future APOL1 structural studies and define the contribution of APOL1 risk variants to kidney disease.
- Published
- 2016
39. APOL1 kidney disease risk variants cause cytotoxicity by depleting cellular potassium and inducing stress-activated protein kinases
- Author
-
David J. Friedman, Salvatore DiBartolo, Seth L. Alper, Paul Yan, John F. Heneghan, Martin R. Pollak, Lynn VerPlank, Johannes Schlondorff, Opeyemi A. Olabisi, Jia-Yue Zhang, Nathan Zahler, and Jung Hee Suh
- Subjects
Risk ,STAT3 Transcription Factor ,0301 basic medicine ,Programmed cell death ,MAP Kinase Signaling System ,Apolipoprotein L1 ,Recombinant Fusion Proteins ,p38 mitogen-activated protein kinases ,Mutation, Missense ,030232 urology & nephrology ,Black People ,Biology ,Transfection ,03 medical and health sciences ,0302 clinical medicine ,Gene Frequency ,Cytokine Receptor gp130 ,Humans ,Genetic Predisposition to Disease ,Phosphorylation ,Protein kinase A ,Cytotoxicity ,Cell Size ,Ion Transport ,Multidisciplinary ,Cell Death ,Kinase ,Biological Sciences ,Cell biology ,Enzyme Activation ,HEK293 Cells ,Apolipoproteins ,030104 developmental biology ,Amino Acid Substitution ,Disease Progression ,Potassium ,biology.protein ,Kidney Diseases ,Efflux ,Mitogen-Activated Protein Kinases ,Lipoproteins, HDL ,Protein Processing, Post-Translational ,Intracellular - Abstract
Two specific genetic variants of the apolipoprotein L1 (APOL1) gene are responsible for the high rate of kidney disease in people of recent African ancestry. Expression in cultured cells of these APOL1 risk variants, commonly referred to as G1 and G2, results in significant cytotoxicity. The underlying mechanism of this cytotoxicity is poorly understood. We hypothesized that this cytotoxicity is mediated by APOL1 risk variant-induced dysregulation of intracellular signaling relevant for cell survival. To test this hypothesis, we conditionally expressed WT human APOL1 (G0), the APOL1 G1 variant, or the APOL1 G2 variant in human embryonic kidney cells (T-REx-293) using a tetracycline-mediated (Tet-On) system. We found that expression of either G1 or G2 APOL1 variants increased apparent cell swelling and cell death compared with G0-expressing cells. These manifestations of cytotoxicity were preceded by G1 or G2 APOL1-induced net efflux of intracellular potassium as measured by X-ray fluorescence, resulting in the activation of stress-activated protein kinases (SAPKs), p38 MAPK, and JNK. Prevention of net K(+) efflux inhibited activation of these SAPKs by APOL1 G1 or G2. Furthermore, inhibition of SAPK signaling and inhibition of net K(+) efflux abrogated cytotoxicity associated with expression of APOL1 risk variants. These findings in cell culture raise the possibility that nephrotoxicity of APOL1 risk variants may be mediated by APOL1 risk variant-induced net loss of intracellular K(+) and subsequent induction of stress-activated protein kinase pathways.
- Published
- 2015
40. Erythrocytes from hereditary xerocytosis patients heterozygous for KCNN4 V282M exhibit increased spontaneous Gardos channel-like activity inhibited by senicapoc
- Author
-
Denis R. Gallagher, Boris E. Shmukler, Seth L. Alper, Carlo Brugnara, Frans A. Kuypers, Christopher C. Fikry, L. Michael Snyder, Alicia Rivera, and David H. Vandorpe
- Subjects
Heterozygote ,Erythrocytes ,Patch-Clamp Techniques ,Hydrops Fetalis ,Drug Evaluation, Preclinical ,Mutation, Missense ,Cell Count ,In Vitro Techniques ,Anemia, Hemolytic, Congenital ,03 medical and health sciences ,chemistry.chemical_compound ,KCNN4 ,0302 clinical medicine ,Erythrocyte Deformability ,Acetamides ,Potassium Channel Blockers ,Humans ,Ion Transport ,Hereditary xerocytosis ,Senicapoc ,Trityl Compounds ,Hematology ,Intermediate-Conductance Calcium-Activated Potassium Channels ,Potassium channel ,Osmotic Fragility ,chemistry ,Gain of Function Mutation ,030220 oncology & carcinogenesis ,Potassium ,Biophysics ,Channel (broadcasting) ,030215 immunology - Published
- 2017
41. Modulation of brain cation-Cl
- Author
-
Jinwei, Zhang, Mohammad Iqbal H, Bhuiyan, Ting, Zhang, Jason K, Karimy, Zhijuan, Wu, Victoria M, Fiesler, Jingfang, Zhang, Huachen, Huang, Md Nabiul, Hasan, Anna E, Skrzypiec, Mariusz, Mucha, Daniel, Duran, Wei, Huang, Robert, Pawlak, Lesley M, Foley, T Kevin, Hitchens, Margaret B, Minnigh, Samuel M, Poloyac, Seth L, Alper, Bradley J, Molyneaux, Andrew J, Trevelyan, Kristopher T, Kahle, Dandan, Sun, and Xianming, Deng
- Subjects
Brain ,Kinases ,Protein Serine-Threonine Kinases ,Article ,Mice, Inbred C57BL ,Stroke ,Mice ,Nitriles ,Hydrocarbons, Chlorinated ,Animals ,Humans ,Solute Carrier Family 12, Member 2 ,Enzyme Inhibitors ,Phosphorylation - Abstract
The SLC12A cation-Cl− cotransporters (CCC), including NKCC1 and the KCCs, are important determinants of brain ionic homeostasis. SPAK kinase (STK39) is the CCC master regulator, which stimulates NKCC1 ionic influx and inhibits KCC-mediated efflux via phosphorylation at conserved, shared motifs. Upregulation of SPAK-dependent CCC phosphorylation has been implicated in several neurological diseases. Using a scaffold-hybrid strategy, we develop a novel potent and selective SPAK inhibitor, 5-chloro-N-(5-chloro-4-((4-chlorophenyl)(cyano)methyl)-2-methylphenyl)-2-hydroxybenzamide (“ZT-1a”). ZT-1a inhibits NKCC1 and stimulates KCCs by decreasing their SPAK-dependent phosphorylation. Intracerebroventricular delivery of ZT-1a decreases inflammation-induced CCC phosphorylation in the choroid plexus and reduces cerebrospinal fluid (CSF) hypersecretion in a model of post-hemorrhagic hydrocephalus. Systemically administered ZT-1a reduces ischemia-induced CCC phosphorylation, attenuates cerebral edema, protects against brain damage, and improves outcomes in a model of stroke. These results suggest ZT-1a or related compounds may be effective CCC modulators with therapeutic potential for brain disorders associated with impaired ionic homeostasis., SPAK kinase is a master regulator of cation-Cl− cotransporters. Here the authors describe a new SPAK inhibitor with robust protective effects in rodent hydrocephalous and ischemic stroke models.
- Published
- 2018
42. Noninvasive Immunohistochemical Diagnosis and Novel
- Author
-
Martina, Živná, Kendrah, Kidd, Anna, Přistoupilová, Veronika, Barešová, Mathew, DeFelice, Brendan, Blumenstiel, Maegan, Harden, Peter, Conlon, Peter, Lavin, Dervla M, Connaughton, Hana, Hartmannová, Kateřina, Hodaňová, Viktor, Stránecký, Alena, Vrbacká, Petr, Vyleťal, Jan, Živný, Miroslav, Votruba, Jana, Sovová, Helena, Hůlková, Victoria, Robins, Rebecca, Perry, Andrea, Wenzel, Bodo B, Beck, Tomáš, Seeman, Ondřej, Viklický, Sylvie, Rajnochová-Bloudíčková, Gregory, Papagregoriou, Constantinos C, Deltas, Seth L, Alper, Anna, Greka, Anthony J, Bleyer, and Stanislav, Kmoch
- Subjects
Male ,Incidence ,Biopsy, Needle ,Mucin-1 ,Polycystic Kidney, Autosomal Dominant ,Prognosis ,Immunohistochemistry ,Risk Assessment ,Pedigree ,Case-Control Studies ,Mutation ,Humans ,Female ,Genetic Predisposition to Disease ,Registries ,Erratum ,Retrospective Studies - Abstract
Autosomal dominant tubulointerstitial kidney disease caused by mucin-1 gene (We performed MUC1fs immunostaining on urinary cell smears and various tissues from ADTKD-After technique refinement, the sensitivity and specificity for MUC1fs immunostaining of urinary cell smears were 94.2% and 88.6%, respectively. Further genetic testing on 17 families with positive MUC1fs immunostaining revealed six families with five novelWe developed a noninvasive immunohistochemical method to detect MUC1fs that, after further validation, may be useful in the future for diagnostic testing. Production of the MUC1fs protein may be central to the pathogenesis of ADTKD
- Published
- 2018
43. Innate immunity pathways regulate the nephropathy gene Apolipoprotein L1
- Author
-
Prachi Jog, Brendan Nichols, Seth L. Alper, Michael Wilmot, Jeffrey B. Kopp, David J. Friedman, Jessica H. Lee, Vivette D. D'Agati, Martin R. Pollak, Glen S. Markowitz, and Daniel Blackler
- Subjects
Adult ,Male ,Genotype ,Apolipoprotein L1 ,Molecular Sequence Data ,Biology ,Cohort Studies ,Small hairpin RNA ,Risk Factors ,Interferon ,medicine ,Humans ,Amino Acid Sequence ,Aged ,Gene knockdown ,Glomerulosclerosis, Focal Segmental ,Pattern recognition receptor ,Middle Aged ,Immunity, Innate ,Toll-Like Receptor 3 ,3. Good health ,Black or African American ,Apolipoproteins ,HEK293 Cells ,IRF1 ,Gene Expression Regulation ,Nephrology ,Immunology ,TLR3 ,biology.protein ,Female ,Lipoproteins, HDL ,IRF3 ,Signal Transduction ,medicine.drug - Abstract
Apolipoprotein L1 (APOL1) risk variants greatly elevate the risk of kidney disease in African Americans. Here we report a cohort of patients who developed collapsing focal segmental glomerulosclerosis while receiving therapeutic interferon, all of whom carried the APOL1 high-risk genotype. This finding raised the possibility that interferons and the molecular pattern recognition receptors that stimulate interferon production may contribute to APOL1-associated kidney disease. In cell culture, interferons and Toll-like receptor (TLR) agonists increased APOL1 expression by up to 200-fold, in some cases with the appearance of transcripts not detected under basal conditions. PolyI:C, a double-stranded RNA TLR3 agonist, increased APOL1 expression by upregulating interferons directly or through an interferon-independent, IFN-regulatory factor 3 (IRF3)-dependent pathway. Using pharmacological inhibitors, small hairpin RNA knockdown, and chromatin immunoprecipitation, we found that the interferon-independent TLR3 pathway relied on signaling through TBK1, NF-κB, and Jak kinases, and on binding of IRF1, IRF2, and STAT2 at the APOL1 transcription start site. We also demonstrate that overexpression of the APOL1 risk variants is more injurious to cells than overexpression of the wild-type APOL1 protein. Our study illustrates that antiviral pathways may be important inducers of kidney disease in individuals with the APOL1 high-risk genotype and identifies potential targets for prevention or treatment.
- Published
- 2015
44. Revised prevalence estimate of possible Hereditary Xerocytosis as derived from a large U.S. Laboratory database
- Author
-
Carlo Brugnara, Justin K. Niles, L. Michael Snyder, Seth L. Alper, Denis R. Gallagher, Harvey W. Kaufman, and Alicia Rivera
- Subjects
Pediatrics ,medicine.medical_specialty ,Databases, Factual ,business.industry ,Anemia ,Hereditary xerocytosis ,Hydrops Fetalis ,MEDLINE ,Hematology ,medicine.disease ,Anemia, Hemolytic, Congenital ,United States ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Hydrops fetalis ,medicine ,Prevalence ,Humans ,business ,030215 immunology - Published
- 2017
45. Functional and Transcriptomic Characterization of Peritoneal Immune-Modulation by Addition of Alanyl-Glutamine to Dialysis Fluid
- Author
-
Tobias Gluexam, Andreas Spittler, Andreas Vychytil, Christoph Aufricht, Seth L. Alper, Julia Becker, Lilian Kuster, Manoj Bhasin, Rebecca Herzog, Dietmar Pils, and Klaus Kratochwill
- Subjects
Adult ,Male ,0301 basic medicine ,Science ,medicine.medical_treatment ,Peritonitis ,Pilot Projects ,Peritoneal equilibration test ,Pharmacology ,Article ,03 medical and health sciences ,Peritoneal cavity ,Immune system ,Renal Dialysis ,Dialysis Solutions ,Humans ,Medicine ,Aged ,Cross-Over Studies ,Multidisciplinary ,business.industry ,Gene Expression Profiling ,Interleukin ,Dipeptides ,Middle Aged ,medicine.disease ,3. Good health ,Glutamine ,030104 developmental biology ,Cytokine ,medicine.anatomical_structure ,Immunology ,Cytokines ,Feasibility Studies ,Female ,Tumor necrosis factor alpha ,Peritoneum ,Transcriptome ,business - Abstract
Peritonitis remains a major cause of morbidity and mortality during chronic peritoneal dialysis (PD). Glucose-based PD fluids reduce immunological defenses in the peritoneal cavity. Low concentrations of peritoneal extracellular glutamine during PD may contribute to this immune deficit. For these reasons we have developed a clinical assay to measure the function of the immune-competent cells in PD effluent from PD patients. We then applied this assay to test the impact on peritoneal immune-competence of PD fluid supplementation with alanyl-glutamine (AlaGln) in 6 patients in an open-label, randomized, crossover pilot trial (EudraCT 2012-004004-36), and related the functional results to transcriptome changes in PD effluent cells. Ex-vivo stimulation of PD effluent peritoneal cells increased release of interleukin (IL) 6 and tumor necrosis factor (TNF) α. Both IL-6 and TNF-α were lower at 1 h than at 4 h of the peritoneal equilibration test but the reductions in cytokine release were attenuated in AlaGln-supplemented samples. AlaGln-supplemented samples exhibited priming of IL-6-related pathways and downregulation of TNF-α upstream elements. Results from measurement of cytokine release and transcriptome analysis in this pilot clinical study support the conclusion that suppression of PD effluent cell immune function in human subjects by standard PD fluid is attenuated by AlaGln supplementation.
- Published
- 2017
46. Effects of Alanyl-Glutamine Treatment on the Peritoneal Dialysis Effluent Proteome Reveal Pathomechanism-Associated Molecular Signatures
- Author
-
Christoph Aufricht, Andreas Vychytil, Keiryn L. Bennett, Rebecca Herzog, Anton Lichtenauer, André C. Mueller, Anja Wagner, Peter Májek, Seth L. Alper, Michael Boehm, Katja Parapatics, Markus Unterwurzacher, and Klaus Kratochwill
- Subjects
0301 basic medicine ,Male ,Proteome ,medicine.medical_treatment ,Pharmacology ,Biochemistry ,Analytical Chemistry ,Peritoneal dialysis ,03 medical and health sciences ,Fibrosis ,medicine ,Humans ,Renal replacement therapy ,Molecular Biology ,Cellular proteins ,Cross-Over Studies ,Chemistry ,Research ,Blood Proteins ,Dipeptides ,medicine.disease ,Blood proteins ,3. Good health ,Isobaric labeling ,030104 developmental biology ,Female ,Alanyl glutamine ,Peritoneal Dialysis - Abstract
Peritoneal dialysis (PD) is a modality of renal replacement therapy in which the high volumes of available PD effluent (PDE) represents a rich source of biomarkers for monitoring disease and therapy. Although this information could help guide the management of PD patients, little is known about the potential of PDE to define pathomechanism-associated molecular signatures in PD. We therefore subjected PDE to a high-performance multiplex proteomic analysis after depletion of highly-abundant plasma proteins and enrichment of low-abundance proteins. A combination of label-free and isobaric labeling strategies was applied to PDE samples from PD patients (n = 20) treated in an open-label, randomized, two-period, cross-over clinical trial with standard PD fluid or with a novel PD fluid supplemented with alanyl-glutamine (AlaGln). With this workflow we identified 2506 unique proteins in the PDE proteome, greatly increasing coverage beyond the 171 previously-reported proteins. The proteins identified range from high abundance plasma proteins to low abundance cellular proteins, and are linked to larger numbers of biological processes and pathways, some of which are novel for PDE. Interestingly, proteins linked to membrane remodeling and fibrosis are overrepresented in PDE compared with plasma, whereas the proteins underrepresented in PDE suggest decreases in host defense, immune-competence and response to stress. Treatment with AlaGln-supplemented PD fluid is associated with reduced activity of membrane injury-associated mechanisms and with restoration of biological processes involved in stress responses and host defense. Our study represents the first application of the PDE proteome in a randomized controlled prospective clinical trial of PD. This novel proteomic workflow allowed detection of low abundance biomarkers to define pathomechanism-associated molecular signatures in PD and their alterations by a novel therapeutic intervention.
- Published
- 2017
47. Mutations in Chromatin Modifier and Ephrin Signaling Genes in Vein of Galen Malformation
- Author
-
Jungmin Choi, Seth L. Alper, James R. Knight, Alejandro Berenstein, Edward R. Smith, Christopher Castaldi, Ava Hunt, Carol Nelson-Williams, Michael L. DiLuna, Erin Loring, Kaya Bilguvar, Brian P. Walcott, Jinwei Zhang, Michelle Sorscher, August A Allocco, Kristopher T. Kahle, Mark W. Youngblood, Andrew T. Timberlake, Shrikant Mane, Murat Gunel, Jennifer Klein, Beverly Aagaard-Kienitz, Sheng Chih Jin, Charles C. Matouk, Jason K. Karimy, Richard P. Lifton, Alan Dardik, Francesc López-Giráldez, Eric M. Jackson, Georges Rodesch, Shozeb Haider, Xue Zeng, Charuta G. Furey, Jonathan Gaillard, Mariya Soban, Qiongshi Lu, Daniel Duran, Sierra B Conine, Joseph M. Zabramski, Weilai Dong, Christopher J Stapleton, Darren B. Orbach, Miikka Vikkula, Bogdan Yatsula, Irina Tikhonova, Masaki Komiyama, Andrew F. Ducruet, and UCL - SSS/DDUV - Institut de Duve
- Subjects
Male ,0301 basic medicine ,Proband ,Receptor, EphB4 ,de novo mutations ,Penetrance ,Biology ,medicine.disease_cause ,whole exome sequencing ,03 medical and health sciences ,0302 clinical medicine ,medicine ,arterio-venous malformation ,Humans ,Ephrin ,EPHB4 ,Vein ,Gene ,Exome sequencing ,ephrin signaling ,Genetics ,Mutation ,Membrane Glycoproteins ,General Neuroscience ,Metalloendopeptidases ,Chromatin Assembly and Disassembly ,pediatric neurosurgery ,Pedigree ,Chromatin ,Vein of Galen malformation ,chromatin modifier ,030104 developmental biology ,medicine.anatomical_structure ,Vein of Galen Malformations ,Female ,Ephrins ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Summary Normal vascular development includes the formation and specification of arteries, veins, and intervening capillaries. Vein of Galen malformations (VOGMs) are among the most common and severe neonatal brain arterio-venous malformations, shunting arterial blood into the brain’s deep venous system through aberrant direct connections. Exome sequencing of 55 VOGM probands, including 52 parent-offspring trios, revealed enrichment of rare damaging de novo mutations in chromatin modifier genes that play essential roles in brain and vascular development. Other VOGM probands harbored rare inherited damaging mutations in Ephrin signaling genes, including a genome-wide significant mutation burden in EPHB4. Inherited mutations showed incomplete penetrance and variable expressivity, with mutation carriers often exhibiting cutaneous vascular abnormalities, suggesting a two-hit mechanism. The identified mutations collectively account for ∼30% of studied VOGM cases. These findings provide insight into disease biology and may have clinical implications for risk assessment.
- Published
- 2019
48. Multiple clinical forms of dehydrated hereditary stomatocytosis arise from mutations in PIEZO1
- Author
-
Maria D'Armiento, Lucia De Franceschi, Seth L. Alper, Boris E. Shmukler, Stanley L. Schrier, David H. Vandorpe, Annalisa Vetro, Maria Rosaria Esposito, Bertil Glader, Achille Iolascon, Orsetta Zuffardi, Jean Delaunay, Donatella Montanaro, Carla Auriemma, Gordon W. Stewart, Immacolata Andolfo, Ivan Limongelli, Roberta Russo, Carlo Brugnara, Luigia De Falco, Rupa Narayan, Fara Vallefuoco, Andolfo, I, Alper, Sl, De Franceschi, L, Auriemma, C, Russo, Roberta, De Falco, L, Vallefuoco, F, Esposito, Mr, Vandorpe, Dh, Shmukler, Be, Narayan, R, Montanaro, D, D'Armiento, Maria, Vetro, A, Limongelli, I, Zuffardi, O, Glader, Be, Schrier, Sl, Brugnara, C, Stewart, Gw, Delaunay, J, and Iolascon, Achille
- Subjects
Adult ,Hemolytic anemia ,Hydrops Fetalis ,Molecular Sequence Data ,Immunology ,Mice, Transgenic ,Anemia, Hemolytic, Congenital ,Transfection ,medicine.disease_cause ,Models, Biological ,Biochemistry ,Ion Channels ,Mice ,Xenopus laevis ,MISSENSE MUTATION ,medicine ,Animals ,Humans ,Missense mutation ,HEMOLYTIC ANEMIA ,Amino Acid Sequence ,Genetics ,Mutation ,Sequence Homology, Amino Acid ,business.industry ,PIEZO1 ,Gene Expression Regulation, Developmental ,Cell Biology ,Hematology ,Embryo, Mammalian ,medicine.disease ,Molecular biology ,Pedigree ,medicine.anatomical_structure ,stomatocytosis ,Dehydrated hereditary stomatocytosis ,Female ,Bone marrow ,business ,Cation transport ,Stomatocytosis - Abstract
Autosomal dominant dehydrated hereditary stomatocytosis (DHSt) usually presents as a compensated hemolytic anemia with macrocytosis and abnormally shaped red blood cells (RBCs). DHSt is part of a pleiotropic syndrome that may also exhibit pseudohyperkalemia and perinatal edema. We identified PIEZO1 as the disease gene for pleiotropic DHSt in a large kindred by exome sequencing analysis within the previously mapped 16q23-q24 interval. In 26 affected individuals among 7 multigenerational DHSt families with the pleiotropic syndrome, 11 heterozygous PIEZO1 missense mutations cosegregated with disease. PIEZO1 is expressed in the plasma membranes of RBCs and its messenger RNA, and protein levels increase during in vitro erythroid differentiation of CD34(+) cells. PIEZO1 is also expressed in liver and bone marrow during human and mouse development. We suggest for the first time a correlation between a PIEZO1 mutation and perinatal edema. DHSt patient red cells with the R2456H mutation exhibit increased ion-channel activity. Functional studies of PIEZO1 mutant R2488Q expressed in Xenopus oocytes demonstrated changes in ion-channel activity consistent with the altered cation content of DHSt patient red cells. Our findings provide direct evidence that R2456H and R2488Q mutations in PIEZO1 alter mechanosensitive channel regulation, leading to increased cation transport in erythroid cells.
- Published
- 2013
49. Strain-specific variations in cation content and transport in mouse erythrocytes
- Author
-
Robert Y.L. Zee, Carlo Brugnara, Alicia Rivera, Seth L. Alper, and Luanne L. Peters
- Subjects
Male ,Erythrocytes ,Sodium-Hydrogen Exchangers ,Physiology ,Sodium-Potassium-Exchanging ATPase ,Biology ,Mice ,KCNN4 ,Model Organisms ,Species Specificity ,Cations ,Genetics ,Animals ,Humans ,Na+/K+-ATPase ,Ion transporter ,Sex Characteristics ,Ion Transport ,Symporters ,Sodium ,Membrane transport ,Sodium–hydrogen antiporter ,Biochemistry ,Symporter ,Potassium ,Female ,Cotransporter - Abstract
Studies of ion transport pathophysiology in hematological disorders and tests of possible new therapeutic agents for these disorders have been carried out in various mouse models because of close functional similarities between mouse and human red cells. We have explored strain-specific differences in erythrocyte membrane physiology in 10 inbred mouse strains by determining erythrocyte contents of Na+, K+, and Mg2+, and erythrocyte transport of ions via the ouabain-sensitive Na-K pump, the amiloride-sensitive Na-H exchanger (NHE1), the volume and chloride-dependent K-Cl cotransporter (KCC), and the charybdotoxin-sensitive Gardos channel (KCNN4). Our data reveal substantial strain-specific and sex-specific differences in both ion content and trans-membrane ion transport in mouse erythrocytes. These differences demonstrate the feasibility of identifying specific quantitative trait loci for erythroid ion transport and content in genetically standardized inbred mouse strains.
- Published
- 2013
50. The SLC26 gene family of anion transporters and channels
- Author
-
Seth L. Alper and Alok K. Sharma
- Subjects
Models, Molecular ,Protein Conformation ,Anion Transport Proteins ,Clinical Biochemistry ,Arabidopsis ,Saccharomyces cerevisiae ,SLC26A3 ,medicine.disease_cause ,Models, Biological ,Biochemistry ,Article ,Mice ,Protein structure ,Species Specificity ,Protein Interaction Mapping ,medicine ,SLC26A6 ,Animals ,Humans ,Gene family ,Molecular Biology ,Gene ,Phylogeny ,Mutation ,biology ,General Medicine ,Transmembrane protein ,Transport protein ,Bicarbonates ,Phenotype ,Sulfate Transporters ,Multigene Family ,biology.protein ,Molecular Medicine - Abstract
The phylogenetically ancient SLC26 gene family encodes multifunctional anion exchangers and anion channels transporting a broad range of substrates, including Cl(-), HCO3(-), sulfate, oxalate, I(-), and formate. SLC26 polypeptides are characterized by N-terminal cytoplasmic domains, 10-14 hydrophobic transmembrane spans, and C-terminal cytoplasmic STAS domains, and appear to be homo-oligomeric. SLC26-related SulP proteins of marine bacteria likely transport HCO3(-) as part of oceanic carbon fixation. SulP genes present in antibiotic operons may provide sulfate for antibiotic biosynthetic pathways. SLC26-related Sultr proteins transport sulfate in unicellular eukaryotes and in plants. Mutations in three human SLC26 genes are associated with congenital or early onset Mendelian diseases: chondrodysplasias for SLC26A2, chloride diarrhea for SLC26A3, and deafness with enlargement of the vestibular aqueduct for SLC26A4. Additional disease phenotypes evident only in mouse knockout models include oxalate urolithiasis for Slc26a6 and Slc26a1, non-syndromic deafness for Slc26a5, gastric hypochlorhydria for Slc26a7 and Slc26a9, distal renal tubular acidosis for Slc26a7, and male infertility for Slc26a8. STAS domains are required for cell surface expression of SLC26 proteins, and contribute to regulation of the cystic fibrosis transmembrane regulator in complex, cell- and tissue-specific ways. The protein interactomes of SLC26 polypeptides are under active investigation.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.