Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Biol Chem ; 300(3): 105732, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336290

ABSTRACT

The manganese (Mn) export protein SLC30A10 is essential for Mn excretion via the liver and intestines. Patients with SLC30A10 deficiency develop Mn excess, dystonia, liver disease, and polycythemia. Recent genome-wide association studies revealed a link between the SLC30A10 variant T95I and markers of liver disease. The in vivo relevance of this variant has yet to be investigated. Using in vitro and in vivo models, we explore the impact of the T95I variant on SLC30A10 function. While SLC30A10 I95 expressed at lower levels than T95 in transfected cell lines, both T95 and I95 variants protected cells similarly from Mn-induced toxicity. Adeno-associated virus 8-mediated expression of T95 or I95 SLC30A10 using the liver-specific thyroxine binding globulin promoter normalized liver Mn levels in mice with hepatocyte Slc30a10 deficiency. Furthermore, Adeno-associated virus-mediated expression of T95 or I95 SLC30A10 normalized red blood cell parameters and body weights and attenuated Mn levels and differential gene expression in livers and brains of mice with whole body Slc30a10 deficiency. While our in vivo data do not indicate that the T95I variant significantly compromises SLC30A10 function, it does reinforce the notion that the liver is a key site of SLC30A10 function. It also supports the idea that restoration of hepatic SLC30A10 expression is sufficient to attenuate phenotypes in SLC30A10 deficiency.


Subject(s)
Amino Acid Substitution , Cation Transport Proteins , Dependovirus , Liver , Manganese , Mutation , Animals , Mice , Body Weight , Brain/metabolism , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line , Dependovirus/genetics , Erythrocytes , Genome-Wide Association Study , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Liver Diseases/genetics , Liver Diseases/metabolism , Manganese/metabolism , Manganese Poisoning/metabolism , Phenotype , Promoter Regions, Genetic , Thyroxine-Binding Globulin/genetics
2.
Arterioscler Thromb Vasc Biol ; 43(12): 2256-2264, 2023 12.
Article in English | MEDLINE | ID: mdl-37855126

ABSTRACT

Blood pressure management involves antihypertensive therapies blocking the renin-angiotensin system (RAS). Yet, it might be inadequate due to poor patient adherence or the so-called RAS escape phenomenon, elicited by the compensatory renin elevation upon RAS blockade. Recently, evidence points toward targeting hepatic AGT (angiotensinogen) as a novel approach to block the RAS pathway that could circumvent the RAS escape phenomenon. Removing AGT, from which all angiotensins originate, should prevent further angiotensin generation, even when renin rises. Furthermore, by making use of a trivalent N-acetylgalactosamine ligand-conjugated small interfering RNA that specifically targets the degradation of hepatocyte-produced mRNAs in a highly potent and specific manner, it may be possible in the future to manage hypertension with therapy that is administered 1 to 2× per year, thereby supporting medication adherence. This review summarizes all current findings on AGT small interfering RNA in preclinical models, making a comparison versus classical RAS blockade with either ACE (angiotensin-converting enzyme) inhibitors or AT1 (angiotensin II type 1) receptor antagonists and AGT suppression with antisense oligonucleotides. It ends with discussing the first-in-human study with AGT small interfering RNA.


Subject(s)
Angiotensinogen , Hypertension , Humans , Acetylgalactosamine , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Angiotensinogen/genetics , Angiotensinogen/metabolism , Blood Pressure , Hypertension/therapy , Hypertension/drug therapy , Renin/metabolism , Renin-Angiotensin System/physiology , RNA, Small Interfering/pharmacology
3.
Genes Dev ; 29(10): 1074-86, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25956904

ABSTRACT

Colorectal cancer (CRC) remains a major contributor to cancer-related mortality. LIN28A and LIN28B are highly related RNA-binding protein paralogs that regulate biogenesis of let-7 microRNAs and influence development, metabolism, tissue regeneration, and oncogenesis. Here we demonstrate that overexpression of either LIN28 paralog cooperates with the Wnt pathway to promote invasive intestinal adenocarcinoma in murine models. When LIN28 alone is induced genetically, half of the resulting tumors harbor Ctnnb1 (ß-catenin) mutation. When overexpressed in Apc(Min/+) mice, LIN28 accelerates tumor formation and enhances proliferation and invasiveness. In conditional genetic models, enforced expression of a LIN28-resistant form of the let-7 microRNA reduces LIN28-induced tumor burden, while silencing of LIN28 expression reduces tumor volume and increases tumor differentiation, indicating that LIN28 contributes to tumor maintenance. We detected aberrant expression of LIN28A and/or LIN28B in 38% of a large series of human CRC samples (n = 595), where LIN28 expression levels were associated with invasive tumor growth. Our late-stage CRC murine models and analysis of primary human tumors demonstrate prominent roles for both LIN28 paralogs in promoting CRC growth and progression and implicate the LIN28/let-7 pathway as a therapeutic target.


Subject(s)
Adenocarcinoma/physiopathology , Colorectal Neoplasms/physiopathology , RNA-Binding Proteins/metabolism , Signal Transduction , Wnt Proteins/metabolism , Animals , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/physiopathology , RNA-Binding Proteins/genetics
4.
J Biol Chem ; 296: 100446, 2021.
Article in English | MEDLINE | ID: mdl-33617884

ABSTRACT

Aggregation of the circulating protein leukocyte-cell-derived chemotaxin 2 (LECT2) causes amyloidosis of LECT2 (ALECT2), one of the most prevalent forms of systemic amyloidosis affecting the kidney and liver. The I40V mutation is thought to be necessary but not sufficient for ALECT2, with a second, as-yet undetermined condition being required for the disease. EM, X-ray diffraction, NMR, and fluorescence experiments demonstrate that LECT2 forms amyloid fibrils in vitro in the absence of other proteins. Removal of LECT2's single bound Zn2+ appears to be obligatory for fibril formation. Zinc-binding affinity is strongly dependent on pH: 9-13 % of LECT2 is calculated to exist in the zinc-free state over the normal pH range of blood, with this fraction rising to 80 % at pH 6.5. The I40V mutation does not alter zinc-binding affinity or kinetics but destabilizes the zinc-free conformation. These results suggest a mechanism in which loss of zinc together with the I40V mutation leads to ALECT2.


Subject(s)
Amyloid/chemistry , Intercellular Signaling Peptides and Proteins/chemistry , Zinc/chemistry , Amyloid/metabolism , Humans , Hydrogen-Ion Concentration , Intercellular Signaling Peptides and Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , X-Ray Diffraction , Zinc/metabolism
5.
Nature ; 535(7611): 246-51, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27383785

ABSTRACT

Poor prognosis in neuroblastoma is associated with genetic amplification of MYCN. MYCN is itself a target of let-7, a tumour suppressor family of microRNAs implicated in numerous cancers. LIN28B, an inhibitor of let-7 biogenesis, is overexpressed in neuroblastoma and has been reported to regulate MYCN. Here we show, however, that LIN28B is dispensable in MYCN-amplified neuroblastoma cell lines, despite de-repression of let-7. We further demonstrate that MYCN messenger RNA levels in amplified disease are exceptionally high and sufficient to sponge let-7, which reconciles the dispensability of LIN28B. We found that genetic loss of let-7 is common in neuroblastoma, inversely associated with MYCN amplification, and independently associated with poor outcomes, providing a rationale for chromosomal loss patterns in neuroblastoma. We propose that let-7 disruption by LIN28B, MYCN sponging, or genetic loss is a unifying mechanism of neuroblastoma development with broad implications for cancer pathogenesis.


Subject(s)
Gene Amplification/genetics , MicroRNAs/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , RNA-Binding Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Chromosome Deletion , Female , Gene Deletion , Genes, Neoplasm/genetics , Humans , Mice , MicroRNAs/metabolism , Models, Genetic , N-Myc Proto-Oncogene Protein , Neuroblastoma/pathology , Xenograft Model Antitumor Assays
6.
J Struct Biol ; 207(3): 260-269, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31170474

ABSTRACT

ALECT2 (leukocyte chemotactic factor 2) amyloidosis is one of the most recently identified amyloid-related diseases, with LECT2 amyloids commonly found in different types of tissues. Under physiological conditions, LECT2 is a 16 kDa multifunctional protein produced by the hepatocytes and secreted into circulation. The pathological mechanisms causing LECT2 transition into the amyloid state are still largely unknown. In the case of ALECT2 patients, there is no disease-causing mutation, yet almost all patients carry a common polymorphism that appears to be necessary but not sufficient to directly trigger amyloidogenesis. In this work, we followed a reductionist methodology in order to detect critical amyloidogenic "hot-spots" during the fibrillation of LECT2. By associating experimental and computational assays, this approach reveals the explicit amyloidogenic core of human LECT2 and pinpoints regions with distinct amyloidogenic properties. The fibrillar architecture of LECT2 polymers, based on our results, provides a wealth of detailed information about the amyloidogenic "hot-spot" interactions and represents a starting point for future peptide-driven intervention in ALECT2 amyloidosis.


Subject(s)
Amyloid/chemistry , Amyloidosis/genetics , Intercellular Signaling Peptides and Proteins/chemistry , Polymorphism, Single Nucleotide , Amino Acid Sequence , Amyloid/metabolism , Amyloid/ultrastructure , Amyloidosis/diagnosis , Amyloidosis/metabolism , Binding Sites/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Microscopy, Electron , Models, Molecular , Protein Aggregates , Protein Aggregation, Pathological , Protein Binding , Protein Conformation
7.
Gastroenterology ; 154(5): 1421-1434, 2018 04.
Article in English | MEDLINE | ID: mdl-29274368

ABSTRACT

BACKGROUND & AIMS: Cytokinesis can fail during normal postnatal liver development, leading to polyploid hepatocytes. We investigated whether inhibiting cytokinesis in the liver slows tumor growth without compromising the health of normal hepatocytes. We inhibited cytokinesis in cancer cells by knocking down ANLN, a cytoskeletal scaffolding protein that regulates cytokinesis and might promote tumorigenesis, in mice with liver disease. METHODS: We analyzed clinical and gene expression data from The Cancer Genome Atlas, Oncomine, PrognoScan, and a hepatocellular carcinoma (HCC) tissue microarray. We knocked down ANLN with small interfering RNAs (siRNAs) in H2.35 liver cells and performed image analyses of cells undergoing cytokinesis. siRNAs were delivered to LAP-MYC mice, which develop hepatoblastoma, using lipid nanoparticles. H2.35 cells with knockdown of ANLN or control cells were injected into FRG mice, which develop chronic liver damage, and tumor growth was monitored. We also developed mice with inducible expression of transgenes encoding small hairpin RNAs (shRNAs) against Anln messenger RNA and studied liver tumorigenesis after administration of diethylnitrosamine and carbon tetrachloride. siRNAs against Anln messenger RNA were conjugated to N-acetylgalactosamine to reduce toxicity and increase hepatocyte tropism; their effects were studied in mouse models of liver cancer and regeneration. RESULTS: Levels of ANLN messenger RNA were increased in human HCC tissues compared to non-tumor liver tissues. siRNA knockdown of ANLN blocked cytokinesis in H2.35 liver cells. Administration of siRNA against ANLN increased survival times of LAP-MYC mice, compared to mice given a control siRNA. H2.35 liver cells with shRNA knockdown of ANLN formed tumors more slowly in FRG mice than control H2.35 cells. Mice with inducible expression of shRNAs against Anln mRNA developed fewer liver tumors after administration of diethylnitrosamine and carbon tetrachloride than control mice. Knockdown of ANLN did not affect liver regeneration after acute and chronic liver injuries. CONCLUSIONS: Knockdown of ANLN in liver cells blocks cytokinesis and inhibits development of liver tumors in mice. Agents that inhibit ANLN in the liver might be effective for prevention or treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carrier Proteins/genetics , Cell Transformation, Neoplastic/metabolism , Cytokinesis , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Liver Regeneration , Microfilament Proteins/deficiency , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/prevention & control , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Genetic Predisposition to Disease , Hepatectomy , Hepatocytes/pathology , Hepatocytes/transplantation , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/prevention & control , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Phenotype , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Time Factors , Transfection
8.
Blood ; 128(2): 204-16, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27095787

ABSTRACT

In the developing mouse embryo, the first hematopoietic stem cells (HSCs) arise in the aorta-gonad-mesonephros (AGM) and mature as they transit through the fetal liver (FL). Compared with FL and adult HSCs, AGM HSCs have reduced repopulation potential in irradiated adult transplant recipients but mechanisms underlying this deficiency in AGM HSCs are poorly understood. By co-expression gene network analysis, we deduced that AGM HSCs show lower levels of interferon-α (IFN-α)/Jak-Stat1-associated gene expression than FL HSCs. Treatment of AGM HSCs with IFN-α enhanced long-term hematopoietic engraftment and donor chimerism. Conversely, IFN-α receptor-deficient AGMs (Ifnαr1(-/-)), had significantly reduced donor chimerism. We identify adenine-thymine-rich interactive domain-3a (Arid3a), a factor essential for FL and B lymphopoiesis, as a key transcriptional co-regulator of IFN-α/Stat1 signaling. Arid3a occupies the genomic loci of Stat1 as well as several IFN-α effector genes, acting to regulate their expression. Accordingly, Arid3a(-/-) AGM HSCs had significantly reduced transplant potential, which was rescued by IFN-α treatment. Our results implicate the inflammatory IFN-α/Jak-Stat pathway in the developmental maturation of embryonic HSCs, whose manipulation may lead to increased potency of reprogrammed HSCs for transplantation.


Subject(s)
DNA-Binding Proteins/metabolism , Embryo, Mammalian/metabolism , Hematopoietic Stem Cells/metabolism , Interferon-alpha/metabolism , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Embryo, Mammalian/cytology , Hematopoietic Stem Cells/cytology , Interferon-alpha/genetics , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Transcription Factors/genetics
10.
Mol Cell ; 36(3): 487-99, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19917256

ABSTRACT

While activation of BAX/BAK by BH3-only molecules (BH3s) is essential for mitochondrial apoptosis, the underlying mechanisms remain unsettled. Here we demonstrate that BAX undergoes stepwise structural reorganization leading to mitochondrial targeting and homo-oligomerization. The alpha1 helix of BAX keeps the alpha9 helix engaged in the dimerization pocket, rendering BAX as a monomer in cytosol. The activator BH3s, tBID/BIM/PUMA, attack and expose the alpha1 helix of BAX, resulting in secondary disengagement of the alpha9 helix and thereby mitochondrial insertion. Activator BH3s remain associated with the N-terminally exposed BAX through the BH1 domain to drive homo-oligomerization. BAK, an integral mitochondrial membrane protein, has bypassed the first activation step, explaining why its killing kinetics are faster than those of BAX. Furthermore, death signals initiated at ER induce BIM and PUMA to activate mitochondrial apoptosis. Accordingly, deficiency of Bim/Puma impedes ER stress-induced BAX/BAK activation and apoptosis. Our study provides mechanistic insights regarding the spatiotemporal execution of BAX/BAK-governed cell death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Bcl-2-Like Protein 11 , Cells, Cultured , Etoposide/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoblotting , Immunoprecipitation , Membrane Proteins/genetics , Mice , Mice, Knockout , Microscopy, Fluorescence , Mitochondria/metabolism , Models, Biological , Mutation , Protein Binding/drug effects , Protein Multimerization , Proto-Oncogene Proteins/genetics , Staurosporine/pharmacology , Thapsigargin/pharmacology , Tumor Suppressor Proteins/genetics , Tunicamycin/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/chemistry , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/chemistry , bcl-2-Associated X Protein/genetics
12.
Nature ; 455(7216): 1076-81, 2008 Oct 23.
Article in English | MEDLINE | ID: mdl-18948948

ABSTRACT

BAX is a pro-apoptotic protein of the BCL-2 family that is stationed in the cytosol until activated by a diversity of stress stimuli to induce cell death. Anti-apoptotic proteins such as BCL-2 counteract BAX-mediated cell death. Although an interaction site that confers survival functionality has been defined for anti-apoptotic proteins, an activation site has not been identified for BAX, rendering its explicit trigger mechanism unknown. We previously developed stabilized alpha-helix of BCL-2 domains (SAHBs) that directly initiate BAX-mediated mitochondrial apoptosis. Here we demonstrate by NMR analysis that BIM SAHB binds BAX at an interaction site that is distinct from the canonical binding groove characterized for anti-apoptotic proteins. The specificity of the human BIM-SAHB-BAX interaction is highlighted by point mutagenesis that disrupts functional activity, confirming that BAX activation is initiated at this novel structural location. Thus, we have now defined a BAX interaction site for direct activation, establishing a new target for therapeutic modulation of apoptosis.


Subject(s)
Gene Expression Regulation , bcl-2-Associated X Protein/metabolism , Amino Acid Sequence , Animals , Apoptosis , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Bcl-2-Like Protein 11 , Cell Line , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mutagenesis, Site-Directed , Mutation/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Sequence Alignment , bcl-2-Associated X Protein/chemistry
13.
Hypertension ; 81(7): 1491-1499, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38690653

ABSTRACT

BACKGROUND: Small-interfering RNA (siRNA) targeting hepatic AGT (angiotensinogen) mRNA depletes AGT, lowering blood pressure for up to 6 months. However, certain situations may require a rapid angiotensin increase. The REVERSIR (RVR) - reverse siRNA silencing technology a potential approach to counteract siRNA effects. METHODS: Spontaneously hypertensive rats received 10 mg/kg AGT siRNA, and 3 weeks later were given AGT-RVR (1, 10, or 20 mg/kg). One week after AGT-RVR dosing, a redose of AGT siRNA assessed its post-AGT-RVR effectiveness for 2 weeks. Additionally, the impact of AGT-RVR after an equihypotensive dose of valsartan (4 mg/kg per day) was examined. RESULTS: Baseline mean arterial pressure (MAP) was 144±1 mm Hg. AGT siRNA reduced MAP by ≈16 mm Hg and AGT by >95%, while renin increased 25-fold. All AGT-RVR doses restored MAP to baseline within 4 to 7 days. Notably, 10 and 20 mg/kg restored AGT and renin to baseline, while 1 mg/kg allowed ≈50% AGT restoration, with renin remaining above baseline. A second AGT siRNA treatment, following 1 mg/kg AGT-RVR, reduced MAP to the same degree as the initial dose, while following 10 mg/kg AGT-RVR, it resulted in ≈50% of the first dose's MAP effect at 2 weeks. The valsartan-induced MAP reduction was unaffected by AGT-RVR. CONCLUSIONS: In spontaneously hypertensive rats, angiotensinogen-RVR dose-dependently reversed AGT siRNA-induced AGT reduction, normalizing MAP. MAP normalization persisted even with 50% recovered AGT levels, likely due to upregulated renin maintaining adequate angiotensin generation. Post-AGT-RVR dosing, a second AGT siRNA dose lowered MAP again.


Subject(s)
Angiotensinogen , Antihypertensive Agents , Hypertension , RNA, Small Interfering , Rats, Inbred SHR , Animals , Angiotensinogen/genetics , Angiotensinogen/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/pharmacology , RNA, Small Interfering/genetics , Rats , Hypertension/drug therapy , Hypertension/genetics , Hypertension/metabolism , Antihypertensive Agents/pharmacology , Male , Blood Pressure/drug effects , Disease Models, Animal , Valsartan/pharmacology , Renin-Angiotensin System/drug effects
14.
Nat Cell Biol ; 8(12): 1348-58, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17115033

ABSTRACT

Although the BCL-2 family constitutes a crucial checkpoint in apoptosis, the intricate interplay between these family members remains elusive. Here, we demonstrate that BIM and PUMA, similar to truncated BID (tBID), directly activate BAX-BAK to release cytochrome c. Conversely, anti-apoptotic BCL-2-BCL-X(L)-MCL-1 sequesters these 'activator' BH3-only molecules into stable complexes, thus preventing the activation of BAX-BAK. Extensive mutagenesis of BAX-BAK indicates that their activity is not kept in check by BCL-2-BCL-X(L)-MCL-1. Anti-apoptotic BCL-2 members are differentially inactivated by the remaining 'inactivator' BH3-only molecules including BAD, NOXA, BMF, BIK/BLK and HRK/DP5. BAD displaces tBID, BIM or PUMA from BCL-2-BCL-X(L) to activate BAX-BAK, whereas NOXA specifically antagonizes MCL-1. Coexpression of BAD and NOXA killed wild-type but not Bax, Bak doubly deficient cells or Puma deficient cells with Bim knockdown, indicating that activator BH3-only molecules function downstream of inactivator BH3-only molecules to activate BAX-BAK. Our data establish a hierarchical regulation of mitochondrion-dependent apoptosis by various BCL-2 subfamilies.


Subject(s)
Apoptosis , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Bcl-2-Like Protein 11 , Cytochromes c/metabolism , Membrane Proteins/metabolism , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
15.
bioRxiv ; 2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36747758

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a liver manifestation of metabolic syndrome, and is estimated to affect one billion individuals worldwide. An increased intake of a high-fat diet (HFD) and sugar-sweetened beverages are risk-factors for NAFLD development, but how their combined intake promotes progression to a more severe form of liver injury is unknown. Here we show that fructose metabolism via ketohexokinase (KHK) C isoform increases endoplasmic reticulum (ER) stress in a dose dependent fashion, so when fructose is coupled with a HFD intake it leads to unresolved ER stress. Conversely, a liver-specific knockdown of KHK in C57BL/6J male mice consuming fructose on a HFD is adequate to improve the NAFLD activity score and exert a profound effect on the hepatic transcriptome. Overexpression of KHK-C in cultured hepatocytes is sufficient to induce ER stress in fructose free media. Upregulation of KHK-C is also observed in genetically obesity ob/ob, db/db and lipodystrophic FIRKO male mice, whereas KHK knockdown in these mice improves metabolic function. Additionally, in over 100 inbred strains of male or female mice hepatic KHK expression correlates positively with adiposity, insulin resistance, and liver triglycerides. Similarly, in 241 human subjects and their controls, hepatic Khk expression is upregulated in early, but not late stages of NAFLD. In summary, we describe a novel role of KHK-C in triggering ER stress, which offers a mechanistic understanding of how the combined intake of fructose and a HFD propagates the development of metabolic complications.

16.
Metabolism ; 145: 155591, 2023 08.
Article in English | MEDLINE | ID: mdl-37230214

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is a liver manifestation of metabolic syndrome, and is estimated to affect one billion individuals worldwide. An increased intake of a high-fat diet (HFD) and sugar-sweetened beverages are risk-factors for NAFLD development, but how their combined intake promotes progression to a more severe form of liver injury is unknown. Here we show that fructose metabolism via ketohexokinase (KHK) C isoform leads to unresolved endoplasmic reticulum (ER) stress when coupled with a HFD intake. Conversely, a liver-specific knockdown of KHK in mice consuming fructose on a HFD is adequate to improve the NAFLD activity score and exert a profound effect on the hepatic transcriptome. Overexpression of KHK-C in cultured hepatocytes is sufficient to induce ER stress in fructose free media. Upregulation of KHK-C is also observed in mice with genetically induced obesity or metabolic dysfunction, whereas KHK knockdown in these mice improves metabolic function. Additionally, in over 100 inbred strains of male or female mice hepatic KHK expression correlates positively with adiposity, insulin resistance, and liver triglycerides. Similarly, in 241 human subjects and their controls, hepatic Khk expression is upregulated in early, but not late stages of NAFLD. In summary, we describe a novel role of KHK-C in triggering ER stress, which offers a mechanistic understanding of how the combined intake of fructose and a HFD propagates the development of metabolic complications.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Female , Humans , Male , Mice , Diet, High-Fat/adverse effects , Fructokinases/genetics , Fructokinases/metabolism , Fructose/pharmacology , Lipogenesis/physiology , Liver/metabolism , Models, Genetic , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism
17.
Proc Natl Acad Sci U S A ; 106(4): 1093-8, 2009 Jan 27.
Article in English | MEDLINE | ID: mdl-19144918

ABSTRACT

Three forms of cell death have been described: apoptosis, autophagic cell death, and necrosis. Although genetic and biochemical studies have formulated a detailed blueprint concerning the apoptotic network, necrosis is generally perceived as a passive cellular demise resulted from unmanageable physical damages. Here, we conclude an active de novo genetic program underlying DNA damage-induced necrosis, thus assigning necrotic cell death as a form of "programmed cell death." Cells deficient of the essential mitochondrial apoptotic effectors, BAX and BAK, ultimately succumbed to DNA damage, exhibiting signature necrotic characteristics. Importantly, this genotoxic stress-triggered necrosis was abrogated when either transcription or translation was inhibited. We pinpointed the p53-cathepsin axis as the quintessential framework underlying necrotic cell death. p53 induces cathepsin Q that cooperates with reactive oxygen species (ROS) to execute necrosis. Moreover, we presented the in vivo evidence of p53-activated necrosis in tumor allografts. Current study lays the foundation for future experimental and therapeutic discoveries aimed at "programmed necrotic death."


Subject(s)
Cathepsins/metabolism , Cysteine Endopeptidases/metabolism , DNA Damage , Necrosis/pathology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cathepsins/genetics , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Fibroblasts/pathology , Fibroblasts/ultrastructure , Humans , Mice , Neoplasm Transplantation , Neoplasms/pathology , Neoplasms/ultrastructure , Transcriptional Activation/genetics , Transplantation, Homologous , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/metabolism
18.
HGG Adv ; 3(2): 100079, 2022 Apr 14.
Article in English | MEDLINE | ID: mdl-35493704

ABSTRACT

The age of menopause is associated with fertility and disease risk, and its genetic control is of great interest. We use whole-exome sequences from 132,370 women in the UK Biobank to test for associations between rare damaging variants and age at natural menopause. Rare damaging variants in five genes are significantly associated with menopause: CHEK2 (p = 3.3 × 10-51), DCLRE1A (p = 8.4 × 10-13), and HELB (p = 5.7 × 10-7) with later menopause and TOP3A (p = 7.6 × 10-8) and CLPB (p = 8.1 × 10-7) with earlier menopause. Two additional genes are suggestive: RAD54L (p = 2.4 × 10-6) with later menopause and HROB (p = 2.9 × 10-6) with earlier menopause. In a follow-up analysis of repeated questionnaires in women who were initially premenopausal, CHEK2, TOP3A, and RAD54L genotypes are associated with subsequent menopause. Consistent with previous genome-wide association studies (GWASs), six of the seven genes are involved in the DNA damage repair pathway. Phenome-wide scans across 398,569 men and women revealed that in addition to known associations with cancers and blood cell counts, rare variants in CHEK2 are also associated with increased risk for uterine fibroids, polycystic ovary syndrome, and prostate hypertrophy; these associations are not shared with higher-penetrance breast cancer genes. Causal mediation analysis suggests that approximately 8% of the breast cancer risk conferred by CHEK2 pathogenic variants after menopause is mediated through delayed menopause.

19.
STAR Protoc ; 2(3): 100731, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34409309

ABSTRACT

Ketohexokinase (KHK) catalyzes the first step of fructose metabolism. Inhibitors of KHK enzymatic activity are being evaluated in clinical trials for the treatment of non-alcoholic fatty liver disease (NAFLD) and diabetes. Here, we present a luminescence-based protocol to quantify KHK activity. The accuracy of this technique has been validated using knockdown and overexpression of KHK in vivo and in vitro. The specificity of the assay has been verified using 3-O-methyl-D-fructose, a non-metabolizable analog of fructose, heat inactivation of hexokinases, and depletion of potassium. For complete details on the use of this protocol, please refer to Damen et al. (2021).


Subject(s)
Enzyme Assays/methods , Fructokinases/metabolism , Fructose/metabolism , Luminescent Measurements/methods , Animals , Carbohydrate Metabolism , Fructokinases/antagonists & inhibitors , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Luminescence , Mice , Non-alcoholic Fatty Liver Disease
20.
Nat Metab ; 3(4): 513-522, 2021 04.
Article in English | MEDLINE | ID: mdl-33846641

ABSTRACT

Colchicine has served as a traditional medicine for millennia and remains widely used to treat inflammatory and other disorders. Colchicine binds tubulin and depolymerizes microtubules, but it remains unclear how this mechanism blocks myeloid cell recruitment to inflamed tissues. Here we show that colchicine inhibits myeloid cell activation via an indirect mechanism involving the release of hepatokines. We find that a safe dose of colchicine depolymerizes microtubules selectively in hepatocytes but not in circulating myeloid cells. Mechanistically, colchicine triggers Nrf2 activation in hepatocytes, leading to secretion of anti-inflammatory hepatokines, including growth differentiation factor 15 (GDF15). Nrf2 and GDF15 are required for the anti-inflammatory action of colchicine in vivo. Plasma from colchicine-treated mice inhibits inflammatory signalling in myeloid cells in a GDF15-dependent manner, by positive regulation of SHP-1 (PTPN6) phosphatase, although the precise molecular identities of colchicine-induced GDF15 and its receptor require further characterization. Our work shows that the efficacy and safety of colchicine depend on its selective action on hepatocytes, and reveals a new axis of liver-myeloid cell communication. Plasma GDF15 levels and myeloid cell SHP-1 activity may be useful pharmacodynamic biomarkers of colchicine action.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colchicine/pharmacology , Cytokines/physiology , Liver/drug effects , Liver/metabolism , Myeloid Cells/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antioxidants/pharmacology , Colchicine/pharmacokinetics , Computer Simulation , Cytokines/biosynthesis , Growth Differentiation Factor 15/genetics , Hepatocytes/drug effects , Humans , Mice , Mice, Inbred C57BL , Microtubules/drug effects , Microtubules/metabolism , NF-E2-Related Factor 2/metabolism , Peritonitis/chemically induced , Peritonitis/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL