Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Biol Chem ; 293(23): 8802-8811, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29653945

ABSTRACT

Control of protein homeostasis is an essential cellular process that, when perturbed, can result in the deregulation or toxic accumulation of proteins. Owing to constant mechanical stress, striated muscle proteins are particularly prone to wear and tear and require several protein quality-control mechanisms to coordinate protein turnover and removal of damaged proteins. Kelch-like proteins, substrate adapters for the Cullin-3 (Cul3)-RING ligase (CRL3) complex, are emerging as critical regulators of striated muscle development and function, highlighting the importance of Cul3-mediated proteostasis in muscle function. To explore the role of Cul3-mediated proteostasis in striated muscle, here we deleted Cul3 specifically in either skeletal muscle (SkM-Cul3 KO) or cardiomyocytes (CM-Cul3 KO) of mice. The loss of Cul3 caused neonatal lethality and dramatic alterations in the proteome, which were unique to each striated muscle type. Many of the proteins whose expression was significantly changed in the SkM-Cul3 KO were components of the extracellular matrix and sarcomere, whereas proteins altered in the CM-Cul3 KO were involved in metabolism. These findings highlight the requirement for striated muscle-specific CRL3 activity and indicate how the CRL3 complex can control different nodes of protein interaction networks in different types of striated muscle. Further identification of Cul3 substrates, and how these substrates are targeted, may reveal therapeutic targets and treatment regimens for striated muscle diseases.


Subject(s)
Cullin Proteins/genetics , Gene Deletion , Muscle, Striated/pathology , Myocytes, Cardiac/pathology , Animals , Cells, Cultured , Cullin Proteins/metabolism , Gene Expression Regulation, Developmental , Metabolome , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Striated/embryology , Muscle, Striated/metabolism , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Protein Interaction Maps
2.
Am J Physiol Endocrinol Metab ; 317(2): E250-E260, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31084489

ABSTRACT

Fetuin-A (Fet-A), a hepatokine associated with insulin resistance, obesity, and incident type 2 diabetes, is shown to exist in both phosphorylated and dephosphorylated forms in circulation. However, studies on fetuin-A phosphorylation status in insulin-resistant conditions and its functional significance are limited. We demonstrate that serum phosphofetuin-A (Ser312) levels were significantly elevated in high-fat diet-induced obese mice, insulin-resistant Zucker diabetic fatty rats, and in individuals with obesity who are insulin resistant. Unlike serum total fetuin-A, serum phosphofetuin-A was associated with body weight, insulin, and markers of insulin resistance. To characterize potential mechanisms, fetuin-A was purified from Hep3B human hepatoma cells. Hep3B Fet-A was phosphorylated (Ser312) and inhibited insulin-stimulated glucose uptake and glycogen synthesis in L6GLUT4 myoblasts. Furthermore, single (Ser312Ala) and double (Ser312Ala + Ser120Ala) phosphorylation-defective Fet-A mutants were without effect on glucose uptake and glycogen synthesis in L6GLUT4 myoblasts. Together, our studies demonstrate that phosphorylation status of Fet-A (Ser312) is associated with obesity and insulin resistance and raise the possibility that Fet-A phosphorylation may play a role in regulation of insulin action.


Subject(s)
Insulin Resistance/physiology , Obesity/metabolism , Protein Kinases/metabolism , alpha-2-HS-Glycoprotein/metabolism , 3T3-L1 Cells , Adult , Aged , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Insulin/metabolism , Insulin Antagonists/metabolism , Insulin Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phosphorylation , Rats , Rats, Zucker , alpha-2-HS-Glycoprotein/pharmacology
3.
Genes Dev ; 25(21): 2291-305, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22056672

ABSTRACT

Regulation of cell differentiation programs requires complex interactions between transcriptional and epigenetic networks. Elucidating the principal molecular events responsible for the establishment and maintenance of cell fate identities will provide important insights into how cell lineages are specified and maintained and will improve our ability to recapitulate cell differentiation events in vitro. In this study, we demonstrate that Nkx2.2 is part of a large repression complex in pancreatic ß cells that includes DNMT3a, Grg3, and HDAC1. Mutation of the endogenous Nkx2.2 tinman (TN) domain in mice abolishes the interaction between Nkx2.2 and Grg3 and disrupts ß-cell specification. Furthermore, we demonstrate that Nkx2.2 preferentially recruits Grg3 and HDAC1 to the methylated Aristaless homeobox gene (Arx) promoter in ß cells. The Nkx2.2 TN mutation results in ectopic expression of Arx in ß cells, causing ß-to-α-cell transdifferentiation. A corresponding ß-cell-specific deletion of DNMT3a is also sufficient to cause Arx-dependent ß-to-α-cell reprogramming. Notably, subsequent removal of Arx in the ß cells of Nkx2.2(TNmut/TNmut) mutant mice reverts the ß-to-α-cell conversion, indicating that the repressor activities of Nkx2.2 on the methylated Arx promoter in ß cells are the primary regulatory events required for maintaining ß-cell identity.


Subject(s)
Glucagon-Secreting Cells/cytology , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Co-Repressor Proteins , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Diabetes Mellitus/physiopathology , Gene Expression Regulation , Ghrelin/metabolism , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Insulin/metabolism , Mice , Mutation , Nuclear Proteins , Organ Specificity/genetics , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Transcription Factors/genetics , Zebrafish Proteins
4.
PLoS Genet ; 9(2): e1003278, 2013.
Article in English | MEDLINE | ID: mdl-23408910

ABSTRACT

During pancreatic development, transcription factor cascades gradually commit precursor populations to the different endocrine cell fate pathways. Although mutational analyses have defined the functions of many individual pancreatic transcription factors, the integrative transcription factor networks required to regulate lineage specification, as well as their sites of action, are poorly understood. In this study, we investigated where and how the transcription factors Nkx2.2 and Neurod1 genetically interact to differentially regulate endocrine cell specification. In an Nkx2.2 null background, we conditionally deleted Neurod1 in the Pdx1+ pancreatic progenitor cells, the Neurog3+ endocrine progenitor cells, or the glucagon+ alpha cells. These studies determined that, in the absence of Nkx2.2 activity, removal of Neurod1 from the Pdx1+ or Neurog3+ progenitor populations is sufficient to reestablish the specification of the PP and epsilon cell lineages. Alternatively, in the absence of Nkx2.2, removal of Neurod1 from the Pdx1+ pancreatic progenitor population, but not the Neurog3+ endocrine progenitor cells, restores alpha cell specification. Subsequent in vitro reporter assays demonstrated that Nkx2.2 represses Neurod1 in alpha cells. Based on these findings, we conclude that, although Nkx2.2 and Neurod1 are both necessary to promote beta cell differentiation, Nkx2.2 must repress Neurod1 in a Pdx1+ pancreatic progenitor population to appropriately commit a subset of Neurog3+ endocrine progenitor cells to the alpha cell lineage. These results are consistent with the proposed idea that Neurog3+ endocrine progenitor cells represent a heterogeneous population of unipotent cells, each restricted to a particular endocrine lineage.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Homeodomain Proteins , Pancreas , Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Lineage , Gene Expression Regulation, Developmental , Glucagon-Secreting Cells/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Mice , Nuclear Proteins , Pancreas/cytology , Pancreas/growth & development , Pancreas/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish Proteins
5.
Mucosal Immunol ; 16(4): 551-562, 2023 08.
Article in English | MEDLINE | ID: mdl-37290501

ABSTRACT

Astroviruses cause a spectrum of diseases spanning asymptomatic infections to severe diarrhea, but little is understood about their pathogenesis. We previously determined that small intestinal goblet cells were the main cell type infected by murine astrovirus-1. Here, we focused on the host immune response to infection and inadvertently discovered a role for indoleamine 2,3-dioxygenase 1 (Ido1), a host tryptophan catabolizing enzyme, in the cellular tropism of murine and human astroviruses. We identified that Ido1 expression was highly enriched among infected goblet cells, and spatially corresponded to the zonation of infection. Because Ido1 can act as a negative regulator of inflammation, we hypothesized it could dampen host antiviral responses. Despite robust interferon signaling in goblet cells, as well as tuft cell and enterocyte bystanders, we observed delayed cytokine induction and suppressed levels of fecal lipocalin-2. Although we found Ido-/- animals were more resistant to infection, this was not associated with fewer goblet cells nor could it be rescued by knocking out interferon responses, suggesting that IDO1 instead regulates cell permissivity. We characterized IDO1-/- Caco-2 cells and observed significantly reduced human astrovirus-1 infection. Together this study highlights a role for Ido1 in astrovirus infection and epithelial cell maturation.


Subject(s)
Astroviridae Infections , Indoleamine-Pyrrole 2,3,-Dioxygenase , Animals , Humans , Mice , Caco-2 Cells , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferons , Tryptophan/metabolism
7.
J Med Chem ; 64(11): 7296-7311, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34042448

ABSTRACT

Whereas the PROTAC approach to target protein degradation greatly benefits from rational design, the discovery of small-molecule degraders relies mostly on phenotypic screening and retrospective target identification efforts. Here, we describe the design, synthesis, and screening of a large diverse library of thalidomide analogues against a panel of patient-derived leukemia and medulloblastoma cell lines. These efforts led to the discovery of potent and novel GSPT1/2 degraders displaying selectivity over classical IMiD neosubstrates, such as IKZF1/3, and high oral bioavailability in mice. Taken together, this study offers compound 6 (SJ6986) as a valuable chemical probe for studying the role of GSPT1/2 in vitro and in vivo, and it supports the utility of a diverse library of CRBN binders in the pursuit of targeting undruggable oncoproteins.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Peptide Termination Factors/metabolism , Proteolysis/drug effects , Small Molecule Libraries/pharmacology , Ubiquitin-Protein Ligases/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Administration, Oral , Animals , Binding Sites , Cell Line, Tumor , Half-Life , Humans , Ikaros Transcription Factor/metabolism , Mice , Molecular Dynamics Simulation , Retrospective Studies , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Structure-Activity Relationship , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/metabolism , Thalidomide/pharmacology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Leukemia ; 35(11): 3078-3091, 2021 11.
Article in English | MEDLINE | ID: mdl-33714976

ABSTRACT

Acute lymphoblastic leukemia (ALL) is a hematopoietic malignancy comprised of molecular subtypes largely characterized by aneuploidy or recurring chromosomal rearrangements. Despite extensive information on the ALL transcriptome and methylome, there is limited understanding of the ALL chromatin landscape. We therefore mapped accessible chromatin in 24 primary ALL cell biospecimens comprising three common molecular subtypes (DUX4/ERG, ETV6-RUNX1 and hyperdiploid) from patients treated at St. Jude Children's Research Hospital. Our findings highlight extensive chromatin reprogramming in ALL, including the identification ALL subtype-specific chromatin landscapes that are additionally modulated by genetic variation. Chromatin accessibility differences between ALL and normal B-cells implicate the activation of B-cell repressed chromatin domains and detail the disruption of normal B-cell development in ALL. Among ALL subtypes, we uncovered roles for basic helix-loop-helix, homeodomain and activator protein 1 transcription factors in promoting subtype-specific chromatin accessibility and distinct gene regulatory networks. In addition to chromatin subtype-specificity, we further identified over 3500 DNA sequence variants that alter the ALL chromatin landscape and contribute to inter-individual variability in chromatin accessibility. Collectively, our data suggest that subtype-specific chromatin landscapes and gene regulatory networks impact ALL biology and contribute to transcriptomic differences among ALL subtypes.


Subject(s)
Chromatin/genetics , Chromosome Aberrations , DNA Methylation , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Precursor Cell Lymphoblastic Leukemia-Lymphoma/classification , Transcription Factors/metabolism , Chromatin/metabolism , Epigenomics , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Transcription Factors/genetics , Transcriptome
9.
J Biomed Res ; 35(2): 115-134, 2020 Nov 09.
Article in English | MEDLINE | ID: mdl-33349624

ABSTRACT

With advancements in gene editing technologies, our ability to make precise and efficient modifications to the genome is increasing at a remarkable rate, paving the way for scientists and clinicians to uniquely treat a multitude of previously irremediable diseases. CRISPR-Cas9, short for clustered regularly interspaced short palindromic repeats and CRISPR-associated protein 9, is a gene editing platform with the ability to alter the nucleotide sequence of the genome in living cells. This technology is increasing the number and pace at which new gene editing treatments for genetic disorders are moving toward the clinic. The ß-hemoglobinopathies are a group of monogenic diseases, which despite their high prevalence and chronic debilitating nature, continue to have few therapeutic options available. In this review, we will discuss our existing comprehension of the genetics and current state of treatment for ß-hemoglobinopathies, consider potential genome editing therapeutic strategies, and provide an overview of the current state of clinical trials using CRISPR-Cas9 gene editing.

11.
J Clin Invest ; 127(10): 3730-3740, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28872460

ABSTRACT

Maintenance of muscle structure and function depends on the precise organization of contractile proteins into sarcomeres and coupling of the contractile apparatus to the sarcoplasmic reticulum (SR), which serves as the reservoir for calcium required for contraction. Several members of the Kelch superfamily of proteins, which modulate protein stability as substrate-specific adaptors for ubiquitination, have been implicated in sarcomere formation. The Kelch protein Klhl31 is expressed in a muscle-specific manner under control of the transcription factor MEF2. To explore its functions in vivo, we created a mouse model of Klhl31 loss of function using the CRISPR-Cas9 system. Mice lacking Klhl31 exhibited stunted postnatal skeletal muscle growth, centronuclear myopathy, central cores, Z-disc streaming, and SR dilation. We used proteomics to identify several candidate Klhl31 substrates, including Filamin-C (FlnC). In the Klhl31-knockout mice, FlnC protein levels were highly upregulated with no change in transcription, and we further demonstrated that Klhl31 targets FlnC for ubiquitination and degradation. These findings highlight a role for Klhl31 in the maintenance of skeletal muscle structure and provide insight into the mechanisms underlying congenital myopathies.


Subject(s)
Muscle, Skeletal/metabolism , Myotonia Congenita/genetics , Myotonia Congenita/metabolism , Transcription Factors/deficiency , Animals , Filamins/genetics , Filamins/metabolism , MEF2 Transcription Factors/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/pathology , Myotonia Congenita/pathology , Transcription Factors/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL