LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 127

Search options

  1. Article: Investing in the Advancement of Science.

    Perlmutter, David H

    Missouri medicine

    2022  Volume 119, Issue 5, Page(s) 429–431

    MeSH term(s) Humans ; Science ; Investments
    Language English
    Publishing date 2022-11-01
    Publishing country United States
    Document type Journal Article
    ZDB-ID 427362-x
    ISSN 0026-6620
    ISSN 0026-6620
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: Current and Emerging Treatments for Alpha-1 Antitrypsin Deficiency.

    Perlmutter, David H

    Gastroenterology & hepatology

    2016  Volume 12, Issue 7, Page(s) 446–448

    Language English
    Publishing date 2016-05-16
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2386402-3
    ISSN 1554-7914
    ISSN 1554-7914
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article: α1-antitrypsin Deficiency: A Misfolded Secretory Protein Variant with Unique Effects on the Endoplasmic Reticulum.

    Perlmutter, David H

    Endoplasmic reticulum stress in diseases

    2016  Volume 3, Issue 1, Page(s) 63–72

    Abstract: In the classical form of α1-antitrypsin deficiency (ATD) a point mutation leads to accumulation of a misfolded secretory glycoprotein in the endoplasmic reticulum (ER) of liver cells and so ATD has come to be considered a prototypical ER storage disease. ...

    Abstract In the classical form of α1-antitrypsin deficiency (ATD) a point mutation leads to accumulation of a misfolded secretory glycoprotein in the endoplasmic reticulum (ER) of liver cells and so ATD has come to be considered a prototypical ER storage disease. It is associated with two major types of clinical disorders, chronic obstructive pulmonary disease (COPD) by loss-of-function mechanisms and hepatic cirrhosis and carcinogenesis by gain-of-function mechanisms. The lung disease predominantly results from proteolytic damage to the pulmonary connective tissue matrix because of reduced levels of protease inhibitor activity of α1-anitrypsin (AT) in the circulating blood and body fluids. Cigarette smoking is a powerful disease-promoting modifier but other modifiers are known to exist because variation in the lung disease phenotype is still found in smoking and non-smoking homozygotes. The liver disease is highly likely to be caused by the proteotoxic effects of intracellular misfolded protein accumulation and a high degree of variation in the hepatic phenotype among affected homozygotes has been hypothetically attributed to genetic and environmental modifiers that alter proteostasis responses. Liver biopsies of homozygotes show intrahepatocytic inclusions with dilation and expansion of the ER and recent studies of iPS-derived hepatocyte-like cells from individuals with ATD indicate that the changes in the ER directly vary with the hepatic phenotype i.e there is much lesser alteration in the ER in cells derived from homozygotes that do not have clinically significant liver disease. From a signaling perspective, studies in mammalian cell line and animal models expressing the classical α1-antitrypsin Z variant (ATZ) have found that ER signaling is perturbed in a relatively unique way with powerful activation of autophagy and the NFκB pathway but relatively limited, if any, UPR signaling. It is still not known how much these unique structural and functional changes and the variation among affected homozygotes relate to the tendency of this variant to polymerize and aggregate and/or to the repertoire of proteostasis mechanisms that are activated.
    Language English
    Publishing date 2016-09-19
    Publishing country Poland
    Document type Journal Article
    ISSN 2300-4266
    ISSN 2300-4266
    DOI 10.1515/ersc-2016-0004
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Variants in autophagy genes MTMR12 and FAM134A are putative modifiers of the hepatic phenotype in α1-antitrypsin deficiency.

    Tafaleng, Edgar N / Li, Jie / Wang, Yan / Hidvegi, Tunda / Soto-Gutierrez, Alex / Locke, Adam E / Nicholas, Thomas J / Wang, Yung-Chun / Pak, Stephen / Cho, Michael H / Silverman, Edwin K / Silverman, Gary A / Jin, Sheng Chih / Fox, Ira J / Perlmutter, David H

    Hepatology (Baltimore, Md.)

    2024  

    Abstract: Background and aims: In the classical form of α1-antitrypsin deficiency, a misfolded variant α1-antitrypsin Z accumulates in the endoplasmic reticulum of liver cells and causes liver cell injury by gain-of-function proteotoxicity in a sub-group of ... ...

    Abstract Background and aims: In the classical form of α1-antitrypsin deficiency, a misfolded variant α1-antitrypsin Z accumulates in the endoplasmic reticulum of liver cells and causes liver cell injury by gain-of-function proteotoxicity in a sub-group of affected homozygotes but relatively little is known about putative modifiers. Here, we carried out genomic sequencing in a uniquely affected family with an index case of liver failure and 2 homozygous siblings with minimal or no liver disease. Their sequences were compared to sequences in well-characterized cohorts of homozygotes with or without liver disease, and then candidate sequence variants were tested for changes in the kinetics of α1-antitrypsin variant Z degradation in iPS-derived hepatocyte-like cells derived from the affected siblings themselves.
    Approach and results: Specific variants in autophagy genes MTMR12 and FAM134A could each accelerate the degradation of α1-antitrypsin variant Z in cells from the index patient, but both MTMR12 and FAM134A variants were needed to slow the degradation of α1-antitrypsin variant Z in cells from a protected sib, indicating that inheritance of both variants is needed to mediate the pathogenic effects of hepatic proteotoxicity at the cellular level. Analysis of homozygote cohorts showed that multiple patient-specific variants in proteostasis genes are likely to explain liver disease susceptibility at the population level.
    Conclusions: These results validate the concept that genetic variation in autophagy function can determine susceptibility to liver disease in α1-antitrypsin deficiency and provide evidence that polygenic mechanisms and multiple patient-specific variants are likely needed for proteotoxic pathology.
    Language English
    Publishing date 2024-04-01
    Publishing country United States
    Document type Journal Article
    ZDB-ID 604603-4
    ISSN 1527-3350 ; 0270-9139
    ISSN (online) 1527-3350
    ISSN 0270-9139
    DOI 10.1097/HEP.0000000000000865
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Alpha-1-antitrypsin deficiency: importance of proteasomal and autophagic degradative pathways in disposal of liver disease-associated protein aggregates.

    Perlmutter, David H

    Annual review of medicine

    2011  Volume 62, Page(s) 333–345

    Abstract: Alpha-1-antitrypsin (AT) deficiency is the most common genetic cause of liver disease in children. The primary pathological issue is a point mutation that renders an abundant hepatic secretory glycoprotein prone to altered folding and a tendency to ... ...

    Abstract Alpha-1-antitrypsin (AT) deficiency is the most common genetic cause of liver disease in children. The primary pathological issue is a point mutation that renders an abundant hepatic secretory glycoprotein prone to altered folding and a tendency to polymerize and aggregate. However, the expression of serious liver damage among homozygotes is dependent on genetic and/or environmental modifiers. Several studies have validated the concept that endogenous hepatic pathways for disposal of aggregation-prone proteins, including the proteasomal and autophagic degradative pathways, could play a key role in the variation in hepatic damage and be the target of the modifiers. Exciting recent results have shown that a drug that enhances autophagy can reduce the hepatic load of aggregated protein and reverse fibrosis in a mouse model of this disease.
    MeSH term(s) Animals ; Autophagy ; Carbamazepine/metabolism ; Carbamazepine/pharmacology ; Cell Transformation, Neoplastic/metabolism ; Humans ; Liver/drug effects ; Liver/metabolism ; Liver/pathology ; Liver Diseases/etiology ; Liver Diseases/metabolism ; Liver Diseases/pathology ; Lung Diseases/drug therapy ; Lung Diseases/metabolism ; Mice ; Point Mutation ; Proteasome Endopeptidase Complex/metabolism ; Protein Folding ; alpha 1-Antitrypsin/blood ; alpha 1-Antitrypsin/metabolism ; alpha 1-Antitrypsin Deficiency/complications ; alpha 1-Antitrypsin Deficiency/metabolism
    Chemical Substances alpha 1-Antitrypsin ; Carbamazepine (33CM23913M) ; Proteasome Endopeptidase Complex (EC 3.4.25.1)
    Language English
    Publishing date 2011
    Publishing country United States
    Document type Journal Article ; Review
    ZDB-ID 207930-6
    ISSN 1545-326X ; 0066-4219
    ISSN (online) 1545-326X
    ISSN 0066-4219
    DOI 10.1146/annurev-med-042409-151920
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Multiple Genes Core to ERAD, Macroautophagy and Lysosomal Degradation Pathways Participate in the Proteostasis Response in α1-Antitrypsin Deficiency.

    Li, Jie / Moretti, Francesca / Hidvegi, Tunda / Sviben, Sanja / Fitzpatrick, James A J / Sundaramoorthi, Hemalatha / Pak, Stephen C / Silverman, Gary A / Knapp, Britta / Filipuzzi, Ireos / Alford, John / Reece-Hoyes, John / Nigsch, Florian / Murphy, Leon O / Nyfeler, Beat / Perlmutter, David H

    Cellular and molecular gastroenterology and hepatology

    2024  Volume 17, Issue 6, Page(s) 1007–1024

    Abstract: Background & aims: In the classic form of α1-antitrypsin deficiency (ATD), the misfolded α1-antitrypsin Z (ATZ) variant accumulates in the endoplasmic reticulum (ER) of liver cells. A gain-of-function proteotoxic mechanism is responsible for chronic ... ...

    Abstract Background & aims: In the classic form of α1-antitrypsin deficiency (ATD), the misfolded α1-antitrypsin Z (ATZ) variant accumulates in the endoplasmic reticulum (ER) of liver cells. A gain-of-function proteotoxic mechanism is responsible for chronic liver disease in a subgroup of homozygotes. Proteostatic response pathways, including conventional endoplasmic reticulum-associated degradation and autophagy, have been proposed as the mechanisms that allow cellular adaptation and presumably protection from the liver disease phenotype. Recent studies have concluded that a distinct lysosomal pathway called endoplasmic reticulum-to-lysosome completely supplants the role of the conventional macroautophagy pathway in degradation of ATZ. Here, we used several state-of-the-art approaches to characterize the proteostatic responses more fully in cellular systems that model ATD.
    Methods: We used clustered regularly interspaced short palindromic repeats (CRISPR)-mediated genome editing coupled to a cell selection step by fluorescence-activated cell sorter to perform screening for proteostasis genes that regulate ATZ accumulation and combined that with selective genome editing in 2 other model systems.
    Results: Endoplasmic reticulum-associated degradation genes are key early regulators and multiple autophagy genes, from classic as well as from ER-to-lysosome and other newly described ER-phagy pathways, participate in degradation of ATZ in a manner that is temporally regulated and evolves as ATZ accumulation persists. Time-dependent changes in gene expression are accompanied by specific ultrastructural changes including dilation of the ER, formation of globular inclusions, budding of autophagic vesicles, and alterations in the overall shape and component parts of mitochondria.
    Conclusions: Macroautophagy is a critical component of the proteostasis response to cellular ATZ accumulation and it becomes more important over time as ATZ synthesis continues unabated. Multiple subtypes of macroautophagy and nonautophagic lysosomal degradative pathways are needed to respond to the high concentrations of misfolded protein that characterizes ATD and these pathways are attractive candidates for genetic variants that predispose to the hepatic phenotype.
    Language English
    Publishing date 2024-02-07
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2819778-1
    ISSN 2352-345X ; 2352-345X
    ISSN (online) 2352-345X
    ISSN 2352-345X
    DOI 10.1016/j.jcmgh.2024.02.006
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article: Longitudinal modeling of human neuronal aging identifies RCAN1-TFEB pathway contributing to neurodegeneration of Huntington's disease.

    Lee, Seong Won / Oh, Young Mi / Victor, Matheus B / Strunilin, Ilya / Chen, Shawei / Dahiya, Sonika / Dolle, Roland E / Pak, Stephen C / Silverman, Gary A / Perlmutter, David H / Yoo, Andrew S

    Research square

    2023  

    Abstract: Aging is a common risk factor in neurodegenerative disorders and the ability to investigate aging of neurons in an isogenic background would facilitate discovering the interplay between neuronal aging and onset of neurodegeneration. Here, we perform ... ...

    Abstract Aging is a common risk factor in neurodegenerative disorders and the ability to investigate aging of neurons in an isogenic background would facilitate discovering the interplay between neuronal aging and onset of neurodegeneration. Here, we perform direct neuronal reprogramming of longitudinally collected human fibroblasts to reveal genetic pathways altered at different ages. Comparative transcriptome analysis of longitudinally aged striatal medium spiny neurons (MSNs), a primary neuronal subtype affected in Huntington's disease (HD), identified pathways associated with RCAN1, a negative regulator of calcineurin. Notably, RCAN1 undergoes age-dependent increase at the protein level detected in reprogrammed MSNs as well as in human postmortem striatum. In patient-derived MSNs of adult-onset HD (HD-MSNs), counteracting
    Language English
    Publishing date 2023-05-09
    Publishing country United States
    Document type Preprint
    DOI 10.21203/rs.3.rs-2815300/v1
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article ; Online: Longitudinal modeling of human neuronal aging reveals the contribution of the RCAN1-TFEB pathway to Huntington's disease neurodegeneration.

    Lee, Seong Won / Oh, Young Mi / Victor, Matheus B / Yang, Yan / Chen, Shawei / Strunilin, Ilya / Dahiya, Sonika / Dolle, Roland E / Pak, Stephen C / Silverman, Gary A / Perlmutter, David H / Yoo, Andrew S

    Nature aging

    2023  Volume 4, Issue 1, Page(s) 95–109

    Abstract: Aging is a common risk factor in neurodegenerative disorders. Investigating neuronal aging in an isogenic background stands to facilitate analysis of the interplay between neuronal aging and neurodegeneration. Here we perform direct neuronal ... ...

    Abstract Aging is a common risk factor in neurodegenerative disorders. Investigating neuronal aging in an isogenic background stands to facilitate analysis of the interplay between neuronal aging and neurodegeneration. Here we perform direct neuronal reprogramming of longitudinally collected human fibroblasts to reveal genetic pathways altered at different ages. Comparative transcriptome analysis of longitudinally aged striatal medium spiny neurons (MSNs) in Huntington's disease identified pathways involving RCAN1, a negative regulator of calcineurin. Notably, RCAN1 protein increased with age in reprogrammed MSNs as well as in human postmortem striatum and RCAN1 knockdown rescued patient-derived MSNs of Huntington's disease from degeneration. RCAN1 knockdown enhanced chromatin accessibility of genes involved in longevity and autophagy, mediated through enhanced calcineurin activity, leading to TFEB's nuclear localization by dephosphorylation. Furthermore, G2-115, an analog of glibenclamide with autophagy-enhancing activities, reduced the RCAN1-calcineurin interaction, phenocopying the effect of RCAN1 knockdown. Our results demonstrate that targeting RCAN1 genetically or pharmacologically can increase neuronal resilience in Huntington's disease.
    MeSH term(s) Humans ; Aged ; Calcineurin/genetics ; Huntington Disease/genetics ; Aging/genetics ; Transcription Factors/metabolism ; Corpus Striatum/metabolism ; DNA-Binding Proteins/metabolism ; Muscle Proteins/genetics ; Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
    Chemical Substances Calcineurin (EC 3.1.3.16) ; Transcription Factors ; RCAN1 protein, human ; DNA-Binding Proteins ; Muscle Proteins ; TFEB protein, human ; Basic Helix-Loop-Helix Leucine Zipper Transcription Factors
    Language English
    Publishing date 2023-12-08
    Publishing country United States
    Document type Journal Article
    ISSN 2662-8465
    ISSN (online) 2662-8465
    DOI 10.1038/s43587-023-00538-3
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: How did the 2022 global mpox outbreak happen? A travel-associated case 6 months earlier may provide important clues.

    Kreuze, Molly A / Minhaj, Faisal S / Duwell, Monique / Gigante, Crystal M / Kim, Alexander M / Crum, David / Perlmutter, Rebecca / Rubin, Jamie H / Myers, Robert / Lukula, Salimatu L / Ravi-Caldwell, Nivedita / Sockwell, Denise / Chen, Tai-Ho / de Perio, Marie A / Hughes, Christine M / Davidson, Whitni B / Wilkins, Kim / Baird, Nicolle / Lowe, David /
    Li, Yu / McCollum, Andrea M / Blythe, David / Rao, Agam K

    Travel medicine and infectious disease

    2023  Volume 55, Page(s) 102618

    MeSH term(s) Humans ; Mpox (monkeypox) ; Travel
    Language English
    Publishing date 2023-07-26
    Publishing country Netherlands
    Document type Letter
    ZDB-ID 2170891-5
    ISSN 1873-0442 ; 1477-8939
    ISSN (online) 1873-0442
    ISSN 1477-8939
    DOI 10.1016/j.tmaid.2023.102618
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Resolution of hepatic fibrosis after ZFN-mediated gene editing in the PiZ mouse model of human α1-antitrypsin deficiency.

    Li, Yanfeng / Guha, Chandan / Asp, Patrik / Wang, Xia / Tchaikovskya, Tatyana L / Kim, Kenneth / Mendel, Matthew / Cost, Gregory J / Perlmutter, David H / Roy-Chowdhury, Namita / Fox, Ira J / Conway, Anthony / Roy-Chowdhury, Jayanta

    Hepatology communications

    2023  Volume 7, Issue 3, Page(s) e0070

    Abstract: Background: α1-antitrypsin deficiency is most commonly caused by a mutation in exon-7 of SERPINA1 (SA1-ATZ), resulting in hepatocellular accumulation of a misfolded variant (ATZ). Human SA1-ATZ-transgenic (PiZ) mice exhibit hepatocellular ATZ ... ...

    Abstract Background: α1-antitrypsin deficiency is most commonly caused by a mutation in exon-7 of SERPINA1 (SA1-ATZ), resulting in hepatocellular accumulation of a misfolded variant (ATZ). Human SA1-ATZ-transgenic (PiZ) mice exhibit hepatocellular ATZ accumulation and liver fibrosis. We hypothesized that disrupting the SA1-ATZ transgene in PiZ mice by in vivo genome editing would confer a proliferative advantage to the genome-edited hepatocytes, enabling them to repopulate the liver.
    Methods: To create a targeted DNA break in exon-7 of the SA1-ATZ transgene, we generated 2 recombinant adeno-associated viruses (rAAV) expressing a zinc-finger nuclease pair (rAAV-ZFN), and another rAAV for gene correction by targeted insertion (rAAV-TI). PiZ mice were injected i.v. with rAAV-TI alone or the rAAV-ZFNs at a low (7.5×1010vg/mouse, LD) or a high dose (1.5×1011vg/mouse, HD), with or without rAAV-TI. Two weeks and 6 months after treatment, livers were harvested for molecular, histological, and biochemical analyses.
    Results: Two weeks after treatment, deep sequencing of the hepatic SA1-ATZ transgene pool showed 6%±3% or 15%±4% nonhomologous end joining in mice receiving LD or HD rAAV-ZFN, respectively, which increased to 36%±12% and 36%±12%, respectively, 6 months after treatment. Two weeks postinjection of rAAV-TI with LD or HD of rAAV-ZFN, repair by targeted insertion occurred in 0.10%±0.09% and 0.25%±0.14% of SA1-ATZ transgenes, respectively, which increased to 5.2%±5.0% and 33%±13%, respectively, 6 months after treatment. Six months after rAAV-ZFN administration, there was a marked clearance of ATZ globules from hepatocytes, and resolution of liver fibrosis, along with reduction of hepatic TAZ/WWTR1, hedgehog ligands, Gli2, a TIMP, and collagen content.
    Conclusions: ZFN-mediated SA1-ATZ transgene disruption provides a proliferative advantage to ATZ-depleted hepatocytes, enabling them to repopulate the liver and reverse hepatic fibrosis.
    MeSH term(s) Humans ; Animals ; Mice ; Gene Editing ; Zinc Finger Nucleases ; Liver Cirrhosis/genetics ; Liver Cirrhosis/therapy ; Hepatocytes ; Disease Models, Animal ; Intracellular Signaling Peptides and Proteins
    Chemical Substances Zinc Finger Nucleases (EC 3.1.-) ; Intracellular Signaling Peptides and Proteins
    Language English
    Publishing date 2023-02-27
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ISSN 2471-254X
    ISSN (online) 2471-254X
    DOI 10.1097/HC9.0000000000000070
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top