1
|
Kehar M, Klaassen RJ, Sergi CM. Heterozygous missense mutation of the fibrinogen gene associated with cryptogenic liver disease in a 15-months-old Canadian caucasian child. Ultrastruct Pathol 2025; 49:235-242. [PMID: 39739370 DOI: 10.1080/01913123.2024.2447853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/02/2025]
Abstract
Hepatic fibrinogen storage disease is an uncommon autosomal dominant hereditary illness marked by hypofibrinogenemia and the accumulation of variant fibrinogen in the hepatic endoplasmic reticulum. We present an asymptomatic 15-month-old male with elevated liver enzymes. Test results indicate hypofibrinogenemia. The liver biopsy revealed circular eosinophilic inclusion bodies within the hepatocyte cytoplasm. After diastase pretreatment, the inclusion bodies did not stain using the periodic acid - Schiff procedure. Ultrastructural examination revealed the characteristic fibrinogen storage curvilinear inclusions. Sequence analysis using the Blueprint Genetics (BpG) FLEX Bleeding Disorder/Coagulopathy Panel identified a heterozygous missense variant FGG c.1075 G>C, p. (Gly359Arg). Thus, the patient was diagnosed with hepatic fibrinogen storage disease. Our findings suggest that in patients with asymptomatic elevated liver enzymes presenting with unanticipated hypofibrinogenemia, hepatic fibrinogen storage disorder must be included in the differential diagnosis. Furthermore, our results underscore the significance of molecular diagnosis in patients diagnosed with cryptogenic liver disease.
Collapse
Affiliation(s)
- Mohit Kehar
- Division of Pediatric Gastroenterology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Robert J Klaassen
- Division of Hematology/Oncology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| | - Consolato M Sergi
- Anatomical Pathology, Children's Hospital of Eastern Ontario, Ottawa, ON, Canada
| |
Collapse
|
2
|
Zaidalkilani AT, Al‐kuraishy HM, Fahad EH, Al‐Gareeb AI, Elewa YHA, Zahran MH, Alexiou A, Papadakis M, AL‐Farga A, Batiha GE. Autophagy modulators in type 2 diabetes: A new perspective. J Diabetes 2024; 16:e70010. [PMID: 39676616 PMCID: PMC11647182 DOI: 10.1111/1753-0407.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 05/31/2024] [Accepted: 08/27/2024] [Indexed: 12/17/2024] Open
Abstract
Type 2 diabetes (T2D) is a chronic metabolic disorder caused by defective insulin signaling, insulin resistance, and impairment of insulin secretion. Autophagy is a conserved lysosomal-dependent catabolic cellular pathway involved in the pathogenesis of T2D and its complications. Basal autophagy regulates pancreatic β-cell function by enhancing insulin release and peripheral insulin sensitivity. Therefore, defective autophagy is associated with impairment of pancreatic β-cell function and the development of insulin rersistance (IR). However, over-activated autophagy increases apoptosis of pancreatic β-cells leading to pancreatic β-cell dysfunction. Hence, autophagy plays a double-edged sword role in T2D. Therefore, the use of autophagy modulators including inhibitors and activators may affect the pathogenesis of T2D. Hence, this review aims to clarify the potential role of autophagy inhibitors and activators in T2D.
Collapse
Affiliation(s)
- Ayah Talal Zaidalkilani
- Department of Nutrition, Faculty of Pharmacy and Medical SciencesUniversity of PetraAmmanJordan
| | - Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and MedicineCollege of Medicine, Al‐Mustansiriyah UniversityBaghdadIraq
| | - Esraa H. Fahad
- Department of Pharmacology and ToxicologyCollege of Pharmacy, Mustansiriyah UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and MedicineCollege of Medicine, Al‐Mustansiriyah UniversityBaghdadIraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary MedicineZagazig UniversityZagazigEgypt
- Faculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | | | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh UniversityMohaliPunjabIndia
- Department of Research & DevelopmentFunogenAthensGreece
- Department of Research & DevelopmentAFNP MedWienAustria
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐HerdeckeWuppertalGermany
| | - Ammar AL‐Farga
- Department of BiochemistryCollege of Science University of JeddahJeddahSaudi Arabia
| | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhur UniversityDamanhurAlBeheiraEgypt
| |
Collapse
|
3
|
Song Z, Thepsuwan P, Hur WS, Torres M, Wu SA, Wei X, Tushi NJ, Wei J, Ferraresso F, Paton AW, Paton JC, Zheng Z, Zhang K, Fang D, Kastrup CJ, Jaiman S, Flick MJ, Sun S. Regulation of hepatic inclusions and fibrinogen biogenesis by SEL1L-HRD1 ERAD. Nat Commun 2024; 15:9244. [PMID: 39455574 PMCID: PMC11512042 DOI: 10.1038/s41467-024-53639-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Impaired secretion of an essential blood coagulation factor fibrinogen leads to hepatic fibrinogen storage disease (HFSD), characterized by the presence of fibrinogen-positive inclusion bodies and hypofibrinogenemia. However, the molecular mechanisms underlying the biogenesis of fibrinogen in the endoplasmic reticulum (ER) remain unexplored. Here we uncover a key role of SEL1L-HRD1 complex of ER-associated degradation (ERAD) in the formation of aberrant inclusion bodies, and the biogenesis of nascent fibrinogen protein complex in hepatocytes. Acute or chronic deficiency of SEL1L-HRD1 ERAD in the hepatocytes leads to the formation of hepatocellular inclusion bodies. Proteomics studies followed by biochemical assays reveal fibrinogen as a major component of the inclusion bodies. Mechanistically, we show that the degradation of misfolded endogenous fibrinogen Aα, Bβ, and γ chains by SEL1L-HRD1 ERAD is indispensable for the formation of a functional fibrinogen complex in the ER. Providing clinical relevance of these findings, SEL1L-HRD1 ERAD indeed degrades and thereby attenuates the pathogenicity of two disease-causing fibrinogen γ mutants. Together, this study demonstrates an essential role of SEL1L-HRD1 ERAD in fibrinogen biogenesis and provides insight into the pathogenesis of protein-misfolding diseases.
Collapse
Affiliation(s)
- Zhenfeng Song
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Pattaraporn Thepsuwan
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Woosuk Steve Hur
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Mauricio Torres
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Shuangcheng Alivia Wu
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Xiaoqiong Wei
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Nusrat Jahan Tushi
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Juncheng Wei
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Department of Cardiovascular Sciences and Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Francesca Ferraresso
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
- Departments of Surgery, Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Ze Zheng
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Christian J Kastrup
- Blood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI, 53226, USA
- Departments of Surgery, Biochemistry, Biomedical Engineering, and Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Sunil Jaiman
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Matthew James Flick
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Shengyi Sun
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| |
Collapse
|
4
|
Li Y, Shimizu H, Nakamura R, Lu Y, Sakamoto R, Omori E, Takahashi T, Morimatsu H. The protective effect of carbamazepine on acute lung injury induced by hemorrhagic shock and resuscitation in rats. PLoS One 2024; 19:e0309622. [PMID: 39441839 PMCID: PMC11498730 DOI: 10.1371/journal.pone.0309622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 08/14/2024] [Indexed: 10/25/2024] Open
Abstract
Hemorrhagic shock and resuscitation (HSR) enhances the risk of acute lung injury (ALI). This study investigated the protective effect of carbamazepine (CBZ) on HSR-induced ALI in rats. Male Sprague-Dawley rats were allocated into five distinct groups through randomization: control (SHAM), saline + HSR (HSR), CBZ + HSR (CBZ/HSR), dimethyl sulfoxide (DMSO) + HSR (DMSO/HSR), and CBZ + chloroquine (CQ) + HSR (CBZ/CQ/HSR). Subsequently, HSR models were established. To detect tissue damage, we measured lung histological changes, lung injury scores, and wet/dry weight ratios. We measured neutrophil counts as well as assessed the expression of inflammatory factors using RT-PCR to determine the inflammatory response. We detected autophagy-related proteins LC3II/LC3I, P62, Beclin-1, and Atg12-Atg5 using western blotting. Pretreatment with CBZ improved histopathological changes in the lungs and reduced lung injury scores. The CBZ pretreatment group exhibited significantly reduced lung wet/dry weight ratio, neutrophil aggregation and number, and inflammation factor (TNF-α and iNOS) expression. CBZ changed the expression levels of autophagy-related proteins (LC3II/LC3I, beclin-1, Atg12-Atg5, and P62), suggesting autophagy activation. However, after injecting CQ, an autophagy inhibitor, the beneficial effects of CBZ were reversed. Taken together, CBZ pretreatment improved HSR-induced ALI by suppressing inflammation, at least in part, through activating autophagy. Thus, our study offers a novel perspective for treating HSR-induced ALI.
Collapse
Affiliation(s)
- Yaqiang Li
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Hiroko Shimizu
- Department of Anesthesiology and Resuscitology, Okayama University Medical School, Okayama, Japan
| | - Ryu Nakamura
- Department of Anesthesiology and Resuscitology, Okayama University Medical School, Okayama, Japan
| | - Yifu Lu
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Risa Sakamoto
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Emiko Omori
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | - Hiroshi Morimatsu
- Department of Anesthesiology and Resuscitology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
5
|
Pelland-Marcotte MC, Avram AT, Néron H, Demers C, Castilloux J, Gauthier J, Neerman-Arbez M, Casini A, Rivard GE. A novel FGG missense variant associated with fibrinogen storage disease in a large family from Quebec. Haemophilia 2024; 30:858-861. [PMID: 38561627 DOI: 10.1111/hae.15006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Affiliation(s)
- Marie-Claude Pelland-Marcotte
- Department of Paediatrics, Division of Haematology, Centre Mère Enfant Soleil, Centre Hospitalier de l'Université de Québec, Quebec City, Canada
- Centre de recherche du CHU de Québec, axe Reproduction, santé de la mère et de l'enfant, Québec, Canada
| | - Adelina-Teona Avram
- Department of Hematology, Centre Hospitalier de l'Université de Québec, Quebec City, Canada
| | - Hélène Néron
- Department of Hematology, Centre Hospitalier de l'Université de Québec, Quebec City, Canada
| | - Christine Demers
- Department of Hematology, Centre Hospitalier de l'Université de Québec, Quebec City, Canada
| | - Julie Castilloux
- Department of Pediatrics, Division of Gastro-Enterology, Centre Mère Enfant Soleil, Centre Hospitalier de l'Université de Québec, Quebec City, Canada
| | - Julie Gauthier
- Division of Genetics, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Canada
| | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, University Medical School of Geneva, Geneva, Switzerland
- Faculty of Medicine of Geneva, Geneva, Switzerland
| | - Alessandro Casini
- Angiology and Haemostasis University Hospitals and Faculty of Medicine of Geneva, Geneva, Switzerland
- Faculty of Medicine of Geneva, Geneva, Switzerland
| | - Georges-Etienne Rivard
- Division of Hematology/Oncology, Centre Hospitalier Universitaire Sainte-Justine, Montréal, Canada
| |
Collapse
|
6
|
Zhang Y, Chen Q, Fu X, Zhu S, Huang Q, Li C. Current Advances in the Regulatory Effects of Bioactive Compounds from Dietary Resources on Nonalcoholic Fatty Liver Disease: Role of Autophagy. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:17554-17569. [PMID: 37955247 DOI: 10.1021/acs.jafc.3c04692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease characterized by lipid metabolic disorder primarily due to sedentary lifestyles and excessive food consumption. However, there are currently no approved and effective drugs available to treat NAFLD. In recent years, research has shown that dietary bioactive compounds, such as polysaccharides, polyphenols, flavones, and alkaloids, have the potential to improve NAFLD by regulating autophagy. However, there is no up-to-date review of research progress in this field. This review aims to systematically summarize and discuss the regulatory effects and molecular mechanisms of dietary bioactive compounds on NAFLD through the modulation of autophagy. The existing research has demonstrated that some dietary bioactive compounds can effectively improve various aspects of NAFLD progression, such as lipid metabolism, insulin resistance (IR), endoplasmic reticulum (ER) stress, oxidative stress, mitochondrial homeostasis, and inflammation. Molecular mechanism studies have revealed that they exert their beneficial effects on NAFLD through autophagy-mediated signaling pathways, predominantly involving transcription factor EB (TFEB), mammalian target of rapamycin (mTOR), adenosine monophosphate-activated protein kinase (AMPK), peroxisome proliferator-activated receptors (PPARs), SIRT, and PTEN-induced kinase 1 (PINK1)/parkin. Furthermore, the challenges and prospects of current research in this field are highlighted. Overall, this review provides valuable insights into the potential treatment of NAFLD using dietary bioactive compounds that can modulate autophagy.
Collapse
Affiliation(s)
- Yue Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Qing Chen
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
- School of Food Science and Dietetics, Guangzhou City Polytechnic, Guangzhou 510405, China
| | - Xiong Fu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Siming Zhu
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Qiang Huang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Chao Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
7
|
Kawaguchi D, Kawakami T, Kakuda Y, Yamazaki K. Rare Case of Advanced Gastric Cancer Complicated with Fibrinogen Storage Disease Treated with Chemotherapy plus Immune Checkpoint Inhibitor: A Case Report. Case Rep Oncol 2023; 16:1267-1273. [PMID: 37928864 PMCID: PMC10622163 DOI: 10.1159/000534145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/12/2023] [Indexed: 11/07/2023] Open
Abstract
The administration of chemotherapy to cancer patients with organ dysfunction raises concerns regarding its safety. The safety profile of patients with organ dysfunction due to rare diseases treated with chemotherapy plus immune checkpoint inhibitor is limited. Fibrinogen storage disease (FSD) is a rare disease that causes liver dysfunction through endoplasmic reticulum stress response due to abnormal accumulation of fibrinogen in the endoplasmic reticulum of hepatocytes. Although chemotherapy plus nivolumab is recommended as a standard first-line treatment for patients with advanced gastric cancer (AGC), its safety profile for patients with FSD is rarely available. In this study, an 80-year-old male with gastric cancer with positive lavage cytology was scheduled to receive palliative chemotherapy. This case had liver dysfunction of unknown cause, and a liver biopsy was performed. Histopathological findings revealed a diagnosis of type II/III fibrinogen inclusion based on morphology and immunohistochemistry. Liver function was recovered by administering ursodeoxycholic acid. Therefore, the combination chemotherapy of S-1, oxaliplatin, with nivolumab as palliative chemotherapy was initiated. The case responded well to chemotherapy and achieved conversion surgery without worsening of liver function. We report a case of AGC with fibrinogen inclusion complication where chemotherapy was safely administered with a good outcome. The combination therapy of cytotoxic drugs and immune checkpoint inhibitors may be safely and effectively administered to such patients.
Collapse
Affiliation(s)
- Daiki Kawaguchi
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
- Division of Gastroenterology, Shizuoka City Shizuoka Hospital, Shizuoka, Japan
| | - Takeshi Kawakami
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Yuko Kakuda
- Division of Pathology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| |
Collapse
|
8
|
Ryan JL, Sherman AK, Heble DE, Friesen CA, Daniel JF, Fischer RT, Slowik V. The effect of neuropsychiatric medication on pediatric nonalcoholic fatty liver disease. Clin Transl Sci 2022; 15:2241-2250. [PMID: 35769031 PMCID: PMC9468556 DOI: 10.1111/cts.13358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/13/2022] [Accepted: 06/01/2022] [Indexed: 01/25/2023] Open
Abstract
Obese and overweight children are at risk of developing nonalcoholic fatty liver disease (NAFLD), which can lead to steatohepatitis, cirrhosis, and liver transplantation. Neuropsychiatric conditions affect an increasing proportion of children and often require neuropsychiatric medications (NPMs) that are associated with weight gain and/or drug-induced liver injury. We sought to evaluate the role that the extended use of NPMs play in pediatric NAFLD. Medical chart review was conducted for 260 patients with NAFLD (NPM = 77, non-NPM = 183) seen in the Liver Care Center at Children's Mercy Hospital between 2000 and 2016. Outcome measures included body mass index (BMI) percentile, BMI z-score, aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin, and gamma glutamyltransferase, and were collected at diagnosis, 6-18 month follow-up, and 18-36 months. Controlling for race and metformin, there was a significant increase over time in BMI z-score (p < 0.01) and total bilirubin (p = 0.03), with only initial decreases in ALT (p < 0.01) and AST (p < 0.01). Except for higher total bilirubin in the non-NPM group, no main effect of group or interaction effect was found. Similar patterns remained when subjects were analyzed by NPM drug class. Further study is needed to confirm these findings and to evaluate the effects of NPM dose and duration of exposure, by drug class, on pediatric NAFLD outcomes.
Collapse
Affiliation(s)
- Jamie L. Ryan
- Division of Pediatric Gastroenterology, Hepatology, and NutritionChildren’s Mercy HospitalKansas CityMissouriUSA,Division of Developmental and Behavioral HealthChildren’s Mercy HospitalKansas CityMissouriUSA
| | - Ashley K. Sherman
- Division of Health Services and Outcomes ResearchChildren’s Mercy HospitalKansas CityMissouriUSA
| | - Daniel E. Heble
- Department of PharmacyChildren’s Mercy HospitalKansas CityMissouriUSA
| | - Craig A. Friesen
- Division of Pediatric Gastroenterology, Hepatology, and NutritionChildren’s Mercy HospitalKansas CityMissouriUSA,Department of PediatricsUniversity of Missouri – Kansas City School of MedicineKansas CityMissouriUSA
| | - James F. Daniel
- Division of Pediatric Gastroenterology, Hepatology, and NutritionChildren’s Mercy HospitalKansas CityMissouriUSA,Department of PediatricsUniversity of Missouri – Kansas City School of MedicineKansas CityMissouriUSA
| | - Ryan T. Fischer
- Division of Pediatric Gastroenterology, Hepatology, and NutritionChildren’s Mercy HospitalKansas CityMissouriUSA,Department of PediatricsUniversity of Missouri – Kansas City School of MedicineKansas CityMissouriUSA
| | - Voytek Slowik
- Division of Pediatric Gastroenterology, Hepatology, and NutritionChildren’s Mercy HospitalKansas CityMissouriUSA,Department of PediatricsUniversity of Missouri – Kansas City School of MedicineKansas CityMissouriUSA
| |
Collapse
|
9
|
Song E, Nagarapu A, van Nispen J, Armstrong A, Manithody C, Murali V, Voigt M, Samaddar A, Hutchinson C, Jain S, Roenker J, Krebs J, Jain AK. Carbamazepine mitigates parenteral nutrition-associated liver disease in a novel ambulatory piglet model. JPEN J Parenter Enteral Nutr 2022; 46:1384-1392. [PMID: 35072265 PMCID: PMC9308820 DOI: 10.1002/jpen.2330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/02/2022] [Accepted: 01/05/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Parenteral nutrition (PN) remains a critical therapeutic option in patients who cannot tolerate enteral feeding. However, although lifesaving, PN is associated with significant side effects, including liver injury, the etiology of which is multifactorial. Carbamazepine (CBZ), an antiepileptic medication, is known to modulate hepatic fibrosis and hepatocellular injury in a variety of liver diseases. We hypothesized that CBZ could prevent PN-associated liver disease (PNALD), which we tested by using our novel ambulatory PN piglet model. METHODS Piglets were fitted with jugular catheters and infusion pumps for PN and randomized to enteral nutrition (n = 7), PN (n = 6), or PN with parenteral CBZ (n = 6) for 2 weeks. Serum and liver tissue were analyzed via light microscopy, quantification of serum liver injury markers, Ki67 and cytokeratin-7 indexing, and real-time quantitative polymerase chain reaction. RESULTS PN-fed piglets in our model developed manifestations of PNALD-particularly, increased serum bilirubin, gamma-glutamyltransferase, liver cholestasis, and Ki67 expression compared with that of EN-fed animals (P < 0.03). CBZ therapy in PN-fed animals led to a significant reduction in these markers of injury (P < 0.05). Investigation into the mechanism of these therapeutic effects revealed increased expression of sterol regulatory element-binding protein 1 (SREBP-1), peroxisome proliferator-activated receptor alpha (PPAR-α), and fatty acid binding protein (FABP) in PN-fed animals receiving CBZ (P < 0.03). Further investigation revealed increased LC3 expression and decreased lysosomal-associated membrane protein (LAMP1) expression with CBZ (P < 0.03). CONCLUSION CBZ administration mitigates PNALD severity, suggesting a novel therapeutic strategy targeting PN-associated side effects, and may present a paradigm change to current treatment options.
Collapse
Affiliation(s)
- Eric Song
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Aakash Nagarapu
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Johan van Nispen
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Austin Armstrong
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | | | - Vidul Murali
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Marcus Voigt
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Ashish Samaddar
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Chelsea Hutchinson
- Department of Surgery, St. Louis University School of Medicine, St. Louis, Missouri
| | - Sonali Jain
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Jeremy Roenker
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Joseph Krebs
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| | - Ajay K Jain
- Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri
| |
Collapse
|
10
|
Li H, Sun S. Protein Aggregation in the ER: Calm behind the Storm. Cells 2021; 10:cells10123337. [PMID: 34943844 PMCID: PMC8699410 DOI: 10.3390/cells10123337] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
As one of the largest organelles in eukaryotic cells, the endoplasmic reticulum (ER) plays a vital role in the synthesis, folding, and assembly of secretory and membrane proteins. To maintain its homeostasis, the ER is equipped with an elaborate network of protein folding chaperones and multiple quality control pathways whose cooperative actions safeguard the fidelity of protein biogenesis. However, due to genetic abnormalities, the error-prone nature of protein folding and assembly, and/or defects or limited capacities of the protein quality control systems, nascent proteins may become misfolded and fail to exit the ER. If not cleared efficiently, the progressive accumulation of misfolded proteins within the ER may result in the formation of toxic protein aggregates, leading to the so-called “ER storage diseases”. In this review, we first summarize our current understanding of the protein folding and quality control networks in the ER, including chaperones, unfolded protein response (UPR), ER-associated protein degradation (ERAD), and ER-selective autophagy (ER-phagy). We then survey recent research progress on a few ER storage diseases, with a focus on the role of ER quality control in the disease etiology, followed by a discussion on outstanding questions and emerging concepts in the field.
Collapse
Affiliation(s)
- Haisen Li
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Shengyi Sun
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Correspondence:
| |
Collapse
|
11
|
Padilla-Godínez FJ, Ramos-Acevedo R, Martínez-Becerril HA, Bernal-Conde LD, Garrido-Figueroa JF, Hiriart M, Hernández-López A, Argüero-Sánchez R, Callea F, Guerra-Crespo M. Protein Misfolding and Aggregation: The Relatedness between Parkinson's Disease and Hepatic Endoplasmic Reticulum Storage Disorders. Int J Mol Sci 2021; 22:ijms222212467. [PMID: 34830348 PMCID: PMC8619695 DOI: 10.3390/ijms222212467] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/21/2022] Open
Abstract
Dysfunction of cellular homeostasis can lead to misfolding of proteins thus acquiring conformations prone to polymerization into pathological aggregates. This process is associated with several disorders, including neurodegenerative diseases, such as Parkinson’s disease (PD), and endoplasmic reticulum storage disorders (ERSDs), like alpha-1-antitrypsin deficiency (AATD) and hereditary hypofibrinogenemia with hepatic storage (HHHS). Given the shared pathophysiological mechanisms involved in such conditions, it is necessary to deepen our understanding of the basic principles of misfolding and aggregation akin to these diseases which, although heterogeneous in symptomatology, present similarities that could lead to potential mutual treatments. Here, we review: (i) the pathological bases leading to misfolding and aggregation of proteins involved in PD, AATD, and HHHS: alpha-synuclein, alpha-1-antitrypsin, and fibrinogen, respectively, (ii) the evidence linking each protein aggregation to the stress mechanisms occurring in the endoplasmic reticulum (ER) of each pathology, (iii) a comparison of the mechanisms related to dysfunction of proteostasis and regulation of homeostasis between the diseases (such as the unfolded protein response and/or autophagy), (iv) and clinical perspectives regarding possible common treatments focused on improving the defensive responses to protein aggregation for diseases as different as PD, and ERSDs.
Collapse
Affiliation(s)
- Francisco J. Padilla-Godínez
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.J.P.-G.); (R.R.-A.); (H.A.M.-B.); (L.D.B.-C.); (J.F.G.-F.); (M.H.)
- Regenerative Medicine Laboratory, Department of Surgery, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.H.-L.); (R.A.-S.)
| | - Rodrigo Ramos-Acevedo
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.J.P.-G.); (R.R.-A.); (H.A.M.-B.); (L.D.B.-C.); (J.F.G.-F.); (M.H.)
- Regenerative Medicine Laboratory, Department of Surgery, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.H.-L.); (R.A.-S.)
| | - Hilda Angélica Martínez-Becerril
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.J.P.-G.); (R.R.-A.); (H.A.M.-B.); (L.D.B.-C.); (J.F.G.-F.); (M.H.)
- Regenerative Medicine Laboratory, Department of Surgery, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.H.-L.); (R.A.-S.)
| | - Luis D. Bernal-Conde
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.J.P.-G.); (R.R.-A.); (H.A.M.-B.); (L.D.B.-C.); (J.F.G.-F.); (M.H.)
- Regenerative Medicine Laboratory, Department of Surgery, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.H.-L.); (R.A.-S.)
| | - Jerónimo F. Garrido-Figueroa
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.J.P.-G.); (R.R.-A.); (H.A.M.-B.); (L.D.B.-C.); (J.F.G.-F.); (M.H.)
- Regenerative Medicine Laboratory, Department of Surgery, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.H.-L.); (R.A.-S.)
| | - Marcia Hiriart
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.J.P.-G.); (R.R.-A.); (H.A.M.-B.); (L.D.B.-C.); (J.F.G.-F.); (M.H.)
| | - Adriana Hernández-López
- Regenerative Medicine Laboratory, Department of Surgery, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.H.-L.); (R.A.-S.)
| | - Rubén Argüero-Sánchez
- Regenerative Medicine Laboratory, Department of Surgery, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.H.-L.); (R.A.-S.)
| | - Francesco Callea
- Department of Histopathology, Bugando Medical Centre, Catholic University of Healthy and Allied Sciences, Mwanza 1464, Tanzania;
| | - Magdalena Guerra-Crespo
- Neurosciences Division, Cell Physiology Institute, National Autonomous University of Mexico, Mexico City 04510, Mexico; (F.J.P.-G.); (R.R.-A.); (H.A.M.-B.); (L.D.B.-C.); (J.F.G.-F.); (M.H.)
- Regenerative Medicine Laboratory, Department of Surgery, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico; (A.H.-L.); (R.A.-S.)
- Correspondence:
| |
Collapse
|
12
|
Gunzer S, Kraus A, Buchroth I, Grüneberg M, Westermann C, Biskup S, Reunert J, Grünewald I, Marquardt T. Hypertransaminasemia and liver fibrosis associated with haptoglobin retention and anhaptoglobinemia in a paediatric patient. Liver Int 2021; 41:2427-2432. [PMID: 34358398 DOI: 10.1111/liv.15029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/31/2021] [Accepted: 08/03/2021] [Indexed: 12/13/2022]
Abstract
Cryptogenic elevation of transaminases in childhood can in a few instances be linked to rare hereditary causes. In this paper, a 7-year old girl is reported who was diagnosed with elevated transaminases of unknown origin since infancy. A liver biopsy showed bridging fibrosis, pale eosinophilic intracytoplasmic hepatocellular inclusions and enlarged endoplasmic reticulum cisternae in the hepatocytes. Whole-exome sequencing revealed a homozygous in-frame deletion of 3 base pairs in the haptoglobin gene. The patient is anhaptoglobinemic measured by standard laboratory turbidometry, which was confirmed by Western Blotting and thereby shown to affect both protein chains of haptoglobin. A polyclonal antibody revealed haptoglobin retention in hepatocytes suggesting a defect in haptoglobin secretion. A novel, previously unknown haptoglobin storage disease is suspected to be the reason for the elevated liver enzymes and tissue abnormalities in this patient. The pathophysiology appears to be similar to endoplasmic reticulum storage diseases like alpha-1-antitrypsin-deficiency.
Collapse
Affiliation(s)
- Sophia Gunzer
- Department of General Paediatrics, Metabolic Diseases, University Children's Hospital Muenster, Muenster, Germany
| | - Andreas Kraus
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Inka Buchroth
- Gerhard-Domagk-Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | - Marianne Grüneberg
- Department of General Paediatrics, Metabolic Diseases, University Children's Hospital Muenster, Muenster, Germany
| | - Cordula Westermann
- Gerhard-Domagk-Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | | | - Janine Reunert
- Department of General Paediatrics, Metabolic Diseases, University Children's Hospital Muenster, Muenster, Germany
| | - Inga Grünewald
- Gerhard-Domagk-Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | - Thorsten Marquardt
- Department of General Paediatrics, Metabolic Diseases, University Children's Hospital Muenster, Muenster, Germany
| |
Collapse
|
13
|
Yoda M, Kaido T, Kamijo T, Taira C, Higuchi Y, Arai S, Okumura N. Novel variant fibrinogen γp.C352R produced hypodysfibrinogenemia leading to a bleeding episode and failure of infertility treatment. Int J Hematol 2021; 114:325-333. [PMID: 34117991 DOI: 10.1007/s12185-021-03174-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION We identified a patient with a novel heterozygous variant fibrinogen, γp.C352R (Niigata II; N-II), who had a bleeding episode and failed infertility treatment and was suspected to have hypodysfibrinogenemia based on low and discordant fibrinogen levels (functional assay 0.33 g/L, immunological assay 0.91 g/L). We analyzed the mechanism of this rare phenotype of a congenital fibrinogen disorder. MATERIALS AND METHODS Patient plasma fibrinogen was purified and protein characterization and thrombin-catalyzed fibrin polymerization performed. Recombinant fibrinogen-producing Chinese hamster ovary (CHO) cells were established and the assembly and secretion of variant fibrinogen analyzed by ELISA and western blotting. RESULTS Purified N-II plasma fibrinogen had a small lower molecular weight band below the normal γ-chain and slightly reduced fibrin polymerization. A limited proportion of p.C352R fibrinogen was secreted into the culture medium of established CHO cell lines, but the γ-chain of p.C352R was synthesized and variant fibrinogen was assembled inside the cells. CONCLUSION We demonstrated that fibrinogen N-II, γp.C352R was associated with markedly reduced secretion of variant fibrinogen from CHO cells, that fibrin polymerization of purified plasma fibrinogen was only slightly affected, and that fibrinogen N-II produces hypodysfibrinogenemia in plasma.
Collapse
Affiliation(s)
- Masahiro Yoda
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Takahiro Kaido
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Tomu Kamijo
- Department of Medical Sciences, Graduate School of Medicine, Science and Technology, Shinshu University, Matsumoto, Japan
- Department of Laboratory Medicine, Shinshu University Hospital, Matsumoto, Japan
| | - Chiaki Taira
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Yumiko Higuchi
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
| | - Shinpei Arai
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| | - Nobuo Okumura
- Department of Clinical Laboratory Investigation, Graduate School of Medicine, Shinshu University, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Clinical Laboratory Sciences, School of Health Sciences, Shinshu University, Matsumoto, Japan
| |
Collapse
|
14
|
Kouroumalis E, Voumvouraki A, Augoustaki A, Samonakis DN. Autophagy in liver diseases. World J Hepatol 2021; 13:6-65. [PMID: 33584986 PMCID: PMC7856864 DOI: 10.4254/wjh.v13.i1.6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/10/2020] [Accepted: 12/26/2020] [Indexed: 02/06/2023] Open
Abstract
Autophagy is the liver cell energy recycling system regulating a variety of homeostatic mechanisms. Damaged organelles, lipids and proteins are degraded in the lysosomes and their elements are re-used by the cell. Investigations on autophagy have led to the award of two Nobel Prizes and a health of important reports. In this review we describe the fundamental functions of autophagy in the liver including new data on the regulation of autophagy. Moreover we emphasize the fact that autophagy acts like a two edge sword in many occasions with the most prominent paradigm being its involvement in the initiation and progress of hepatocellular carcinoma. We also focused to the implication of autophagy and its specialized forms of lipophagy and mitophagy in the pathogenesis of various liver diseases. We analyzed autophagy not only in well studied diseases, like alcoholic and nonalcoholic fatty liver and liver fibrosis but also in viral hepatitis, biliary diseases, autoimmune hepatitis and rare diseases including inherited metabolic diseases and also acetaminophene hepatotoxicity. We also stressed the different consequences that activation or impairment of autophagy may have in hepatocytes as opposed to Kupffer cells, sinusoidal endothelial cells or hepatic stellate cells. Finally, we analyzed the limited clinical data compared to the extensive experimental evidence and the possible future therapeutic interventions based on autophagy manipulation.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Liver Research Laboratory, University of Crete Medical School, Heraklion 71110, Greece
| | - Argryro Voumvouraki
- 1 Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54636, Greece
| | - Aikaterini Augoustaki
- Department of Gastroenterology and Hepatology, University Hospital of Crete, Heraklion 71110, Greece
| | - Dimitrios N Samonakis
- Department of Gastroenterology and Hepatology, University Hospital of Crete, Heraklion 71110, Greece.
| |
Collapse
|
15
|
Wang Q, Liu W, Liu G, Li P, Guo X, Zhang C. AMPK-mTOR-ULK1-mediated autophagy protects carbon tetrachloride-induced acute hepatic failure by inhibiting p21 in rats. J Toxicol Pathol 2021; 34:73-82. [PMID: 33627946 PMCID: PMC7890163 DOI: 10.1293/tox.2020-0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/26/2020] [Indexed: 12/25/2022] Open
Abstract
Autophagy is a lysosomal-dependent degradation pathway in eukaryotic cells. Recent
studies have reported that autophagy can facilitate the activation of hepatic stellate
cells (HSCs) and fibrogenesis of the liver during long-term carbon tetrachloride
(CCl4) exposure. However, little is known about the role of autophagy in
CCl4-induced acute hepatic failure (AHF). This study aimed to identify
whether modulation of autophagy can affect CCl4-induced AHF and evaluate the
upstream signaling pathways mediated by CCl4-induced autophagy in rats. The
accumulation of specific punctate distribution of endogenous LC3-II, increased expression
of LC3-II, Atg5, and Atg7 genes/proteins, and decreased expression of p62 gene were
observed after acute liver injury was induced by CCl4 in rats, indicating that
CCl4 resulted in a high level of autophagy. Moreover, loss of autophagic
function by using chloroquine (CQ, an autophagic inhibitor) aggravated liver function,
leading to increased expression of p21 (a cyclin-dependent kinase inhibitor) in
CCl4-treated rats. Furthermore, the AMPK-mTORC1-ULK1 axis was found to serve
a function in CCl4-induced autophagy. These results reveal that
AMPK-mTORC1-ULK1 signaling-induced autophagy has a protective role in
CCl4-induced hepatotoxicity by inhibiting the p21 pathway. This study suggests
a useful strategy aimed at ameliorating CCl4-induced acute hepatotoxicity by
autophagy.
Collapse
Affiliation(s)
- Qiwen Wang
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,College of Life Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Institute of Biomedical Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China
| | - Weixia Liu
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,College of Life Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Institute of Biomedical Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China
| | - Gaopeng Liu
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,College of Life Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Institute of Biomedical Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China
| | - Pan Li
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,College of Life Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Institute of Biomedical Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China
| | - Xueqiang Guo
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,College of Life Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Institute of Biomedical Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China
| | - Chunyan Zhang
- State Key Laboratory Cell Differentiation and Regulation, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Henan Center for Outstanding Overseas Scientists of Pulmonary Fibrosis, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,College of Life Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Institute of Biomedical Science, Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China.,Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis (111 Project), Henan Normal University, #46 East of Construction Road, Xinxiang, 453007 Henan, China
| |
Collapse
|
16
|
Asselta R, Paraboschi EM, Duga S. Hereditary Hypofibrinogenemia with Hepatic Storage. Int J Mol Sci 2020; 21:ijms21217830. [PMID: 33105716 PMCID: PMC7659954 DOI: 10.3390/ijms21217830] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022] Open
Abstract
Fibrinogen is a 340-kDa plasma glycoprotein constituted by two sets of symmetrical trimers, each formed by the Aα, Bβ, and γ chains (respectively coded by the FGA, FGB, and FGG genes). Quantitative fibrinogen deficiencies (hypofibrinogenemia, afibrinogenemia) are rare congenital disorders characterized by low or unmeasurable plasma fibrinogen antigen levels. Their genetic basis is represented by mutations within the fibrinogen genes. To date, only eight mutations, all affecting a small region of the fibrinogen γ chain, have been reported to cause hereditary hypofibrinogenemia with hepatic storage (HHHS), a disorder characterized by protein aggregation in the endoplasmic reticulum, hypofibrinogenemia, and liver disease of variable severity. Here, we will briefly review the clinic characteristics of HHHS patients and the histological feature of their hepatic inclusions, and we will focus on the molecular genetic basis of this peculiar type of coagulopathy.
Collapse
Affiliation(s)
- Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (E.M.P.); (S.D.)
- Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
- Correspondence: ; Tel.: +39-02-8224-5215
| | - Elvezia Maria Paraboschi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (E.M.P.); (S.D.)
- Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Stefano Duga
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20090 Milan, Italy; (E.M.P.); (S.D.)
- Humanitas Clinical and Research Center, IRCCS, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
17
|
Fan J, Shi Y, Peng Y. Autophagy and Liver Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1207:497-528. [PMID: 32671772 DOI: 10.1007/978-981-15-4272-5_37] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autophagy plays an important role in the physiology and pathology of the liver. It is involved in the development of many liver diseases such as α-1-antitrypsin deficiency, chronic hepatitis virus infection, alcoholic liver disease, nonalcoholic fatty liver disease, and liver cancer. Autophagy has thus become a new target for the treatment of liver diseases. How to treat liver diseases by regulating autophagy has been a hot topic.
Collapse
Affiliation(s)
- Jia Fan
- Zhongshan Hospital, Fudan University, 180 FengLin Road, Shanghai, China.
| | - Yinghong Shi
- Zhongshan Hospital, Fudan University, 180 FengLin Road, Shanghai, China
| | - Yuanfei Peng
- Zhongshan Hospital, Fudan University, 180 FengLin Road, Shanghai, China
| |
Collapse
|
18
|
Structural Characteristics in the γ Chain Variants Associated with Fibrinogen Storage Disease Suggest the Underlying Pathogenic Mechanism. Int J Mol Sci 2020; 21:ijms21145139. [PMID: 32698516 PMCID: PMC7404023 DOI: 10.3390/ijms21145139] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 11/24/2022] Open
Abstract
Particular fibrinogen γ chain mutations occurring in the γ-module induce changes that hamper γ-γ dimerization and provoke intracellular aggregation of the mutant fibrinogen, defective export and plasma deficiency. The hepatic storage predisposes to the development of liver disease. This condition has been termed hereditary hypofibrinogenemia with hepatic storage (HHHS). So far, seven of such mutations in the fibrinogen γ chain have been detected. We are reporting on an additional mutation occurring in a 3.5-year-old Turkish child undergoing a needle liver biopsy because of the concomitance of transaminase elevation of unknown origin and low plasma fibrinogen level. The liver biopsy showed an intra-hepatocytic storage of fibrinogen. The molecular analysis of the three fibrinogen genes revealed a mutation (Fibrinogen Trabzon Thr371Ile) at exon 9 of the γ chain in the child and his father, while the mother and the brother were normal. Fibrinogen Trabzon represents a new fibrinogen γ chain mutation fulfilling the criteria for HHHS. Its occurrence in a Turkish child confirms that HHHS can present in early childhood and provides relevant epidemiological information on the worldwide distribution of the fibrinogen γ chain mutations causing this disease. By analyzing fibrinogen crystal structures and calculating the folding free energy change (ΔΔG) to infer how the variants can affect the conformation and function, we propose a mechanism for the intracellular aggregation of Fibrinogen Trabzon and other γ-module mutations causing HHHS.
Collapse
|
19
|
Abstract
Amiodarone is a widely used antiarrhythmic drug that can cause the development of steatohepatitis as well as liver fibrosis and cirrhosis. The molecular mechanisms of amiodarone-mediated liver injury remain largely unknown. We therefore analyzed amiodarone-mediated hepatocellular injury in patients with chronic heart failure, in primary hepatocytes and HepG2 cells. We found that amiodarone-treated patients with chronic heart failure revealed significantly higher serum levels of caspase-cleaved keratin-18, an apoptosis biomarker, compared to healthy individuals or patients not receiving amiodarone. Furthermore, amiodarone treatment of hepatocytes resulted in apoptosis associated with lipid accumulation and ER-stress induction. Liver cell steatosis was accompanied by enhanced de novo lipogenesis which, after reaching peak levels, declined together with decreased activation of ER stress. The decline of amiodarone-mediated lipotoxicity was associated with protective autophagy induction. In contrast, in hepatocytes treated with the autophagy inhibitor chloroquine as well as in autophagy gene (ATG5 or ATG7)-deficient hepatocytes, amiodarone-triggered toxicity was increased. In conclusion, we demonstrate that amiodarone induces lipid accumulation associated with ER stress and apoptosis in hepatocytes, which is mirrored by increased keratin-18 fragment serum levels in amiodarone-treated patients. Autophagy reduces amiodarone-mediated lipotoxicity and could provide a therapeutic strategy for protection from drug-induced liver injury.
Collapse
|
20
|
Gu L, Wang B, Liu L, Gan Q, Liu X, Chen L, Chen L. Hepatic fibrinogen storage disease and hypofibrinogenemia caused by fibrinogen Aguadilla mutation: a case report. J Int Med Res 2020; 48:300060519898033. [PMID: 31965886 PMCID: PMC7169362 DOI: 10.1177/0300060519898033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/10/2019] [Indexed: 12/17/2022] Open
Abstract
Hepatic fibrinogen storage disease is a rare autosomal dominant genetic disorder characterized by hypofibrinogenemia, as well as the retention of variant fibrinogen within the hepatocellular endoplasmic reticulum. Here, we describe an asymptomatic 4-year-old boy with abnormal liver function test results and unexpected hypofibrinogenemia. Liver biopsy showed circular eosinophil inclusion bodies in the hepato-cytoplasm. Immunostaining results of eosinophil inclusion bodies were positive for fibrinogen. Following pretreatment with diastase, the inclusion bodies failed to stain with the periodic acid–Schiff technique; moreover, immunostaining results were positive for fibrinogen, but negative for alpha-1-antitrypsin. Genetic analysis identified a heterozygous missense mutation c.1201C > T (p. Arg401Trp) within the fibrinogen γ-chain (FGG) gene and an additional single nucleotide polymorphism c.-58 A > G within the 5′-untranslated region of the fibrinogen Aα-chain (FGA) gene. Thus, the patient was diagnosed with hepatic fibrinogen storage disease. Our results indicate that, for patients who exhibit chronic liver disease with unexpected hypofibrinogenemia, hepatic fibrinogen storage disease should be considered in the differential diagnosis. Moreover, our findings emphasize the importance of molecular diagnosis in patients with cryptogenic liver disease.
Collapse
Affiliation(s)
- Leilei Gu
- Department of Gastroenterology, Ruijin Hospital North, Shanghai JiaoTong University, School of Medicine, Shanghai, China
| | - Bin Wang
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, China
- Department of Pathology, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, China
| | - Lu Liu
- Department of Hepatology, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Qiaorong Gan
- Department of Hepatology, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaolong Liu
- Department of Hepatology, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Lihong Chen
- Department of Pathology, School of Basic Medical Sciences of Fujian Medical University, Fuzhou, China
- Department of Pathology, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, China
| | - Li Chen
- Department of Gastroenterology, Ruijin Hospital North, Shanghai JiaoTong University, School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Ji CH, Kim HY, Heo AJ, Lee SH, Lee MJ, Kim SB, Srinivasrao G, Mun SR, Cha-Molstad H, Ciechanover A, Choi CY, Lee HG, Kim BY, Kwon YT. The N-Degron Pathway Mediates ER-phagy. Mol Cell 2019; 75:1058-1072.e9. [PMID: 31375263 DOI: 10.1016/j.molcel.2019.06.028] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 04/04/2019] [Accepted: 06/19/2019] [Indexed: 12/29/2022]
Abstract
The endoplasmic reticulum (ER) is susceptible to wear-and-tear and proteotoxic stress, necessitating its turnover. Here, we show that the N-degron pathway mediates ER-phagy. This autophagic degradation initiates when the transmembrane E3 ligase TRIM13 (also known as RFP2) is ubiquitinated via the lysine 63 (K63) linkage. K63-ubiquitinated TRIM13 recruits p62 (also known as sequestosome-1), whose complex undergoes oligomerization. The oligomerization is induced when the ZZ domain of p62 is bound by the N-terminal arginine (Nt-Arg) of arginylated substrates. Upon activation by the Nt-Arg, oligomerized TRIM13-p62 complexes are separated along with the ER compartments and targeted to autophagosomes, leading to lysosomal degradation. When protein aggregates accumulate within the ER lumen, degradation-resistant autophagic cargoes are co-segregated by ER membranes for lysosomal degradation. We developed synthetic ligands to the p62 ZZ domain that enhance ER-phagy for ER protein quality control and alleviate ER stresses. Our results elucidate the biochemical mechanisms and pharmaceutical means that regulate ER homeostasis.
Collapse
Affiliation(s)
- Chang Hoon Ji
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Hee Yeon Kim
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea; AUTOTAC, Changkkyunggung-ro 254, Jongno-gu, Seoul 110-799, Republic of Korea
| | - Ah Jung Heo
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Su Hyun Lee
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Min Ju Lee
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Su Bin Kim
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Ganipisetti Srinivasrao
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea; AUTOTAC, Changkkyunggung-ro 254, Jongno-gu, Seoul 110-799, Republic of Korea
| | - Su Ran Mun
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Hyunjoo Cha-Molstad
- World Class Institute, Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 28116, Republic of Korea
| | - Aaron Ciechanover
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea; Technion Integrated Cancer Center, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Cheol Yong Choi
- Department of Biological Sciences, Sungkyunkwan University, Suwon 440-746, Republic of Korea.
| | - Hee Gu Lee
- Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
| | - Bo Yeon Kim
- World Class Institute, Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongwon 28116, Republic of Korea.
| | - Yong Tae Kwon
- Protein Metabolism Medical Research Center and Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea; Protech, Yongeon 103 Daehangno, Jongno-gu, Seoul 110-799, Republic of Korea; Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea.
| |
Collapse
|
22
|
Panda PK, Fahrner A, Vats S, Seranova E, Sharma V, Chipara M, Desai P, Torresi J, Rosenstock T, Kumar D, Sarkar S. Chemical Screening Approaches Enabling Drug Discovery of Autophagy Modulators for Biomedical Applications in Human Diseases. Front Cell Dev Biol 2019; 7:38. [PMID: 30949479 PMCID: PMC6436197 DOI: 10.3389/fcell.2019.00038] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 03/01/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an intracellular degradation pathway for malfunctioning aggregation-prone proteins, damaged organelles, unwanted macromolecules and invading pathogens. This process is essential for maintaining cellular and tissue homeostasis that contribute to organismal survival. Autophagy dysfunction has been implicated in the pathogenesis of diverse human diseases, and therefore, therapeutic exploitation of autophagy is of potential biomedical relevance. A number of chemical screening approaches have been established for the drug discovery of autophagy modulators based on the perturbations of autophagy reporters or the clearance of autophagy substrates. These readouts can be detected by fluorescence and high-content microscopy, flow cytometry, microplate reader and immunoblotting, and the assays have evolved to enable high-throughput screening and measurement of autophagic flux. Several pharmacological modulators of autophagy have been identified that act either via the classical mechanistic target of rapamycin (mTOR) pathway or independently of mTOR. Many of these autophagy modulators have been demonstrated to exert beneficial effects in transgenic models of neurodegenerative disorders, cancer, infectious diseases, liver diseases, myopathies as well as in lifespan extension. This review describes the commonly used chemical screening approaches in mammalian cells and the key autophagy modulators identified through these methods, and highlights the therapeutic benefits of these compounds in specific disease contexts.
Collapse
Affiliation(s)
- Prashanta Kumar Panda
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Alexandra Fahrner
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Somya Vats
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Elena Seranova
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Vartika Sharma
- Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Miruna Chipara
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Priyal Desai
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Jorge Torresi
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Tatiana Rosenstock
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Department of Physiological Science, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Dhiraj Kumar
- Cellular Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sovan Sarkar
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
23
|
Guillaud O, Dumortier J, Traclet J, Restier L, Joly P, Chapuis-Cellier C, Lachaux A, Mornex JF. Assessment of liver fibrosis by transient elastography (Fibroscan ®) in patients with A1AT deficiency. Clin Res Hepatol Gastroenterol 2019; 43:77-81. [PMID: 30612958 DOI: 10.1016/j.clinre.2018.08.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/17/2018] [Accepted: 08/27/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND Alpha-1-antitrypsin deficiency (A1ATD) is a common genetic condition which predisposes to emphysema and liver disorders. It is estimated that 10-15% of homozygous individuals for the Z allele (PiZZ) may develop liver fibrosis. The optimal modalities to detect liver disease in PiZZ adult patients need to be defined. The aim of this prospective study was to perform a systematic non-invasive evaluation of the liver fibrosis by elastometry using Fibroscan® in a cohort of A1ATD patients with emphysema. METHODS Patients followed in our respiratory unit were enrolled in this prospective study and underwent on the same day a physical examination, a biochemical profiling, an abdominal ultrasound (US) and a Fibroscan®. RESULTS Twenty-nine PiZZ adults (19 male) were included. Median age was 50.4 yrs (21.5-67.2). Median serum A1AT level was 0.20 g/L (0.15-0.33). Liver Function Tests (LFT) were not normal in 2 patients and US was abnormal in 6 patients. Two patients had both abdnormal LFT and US. Fibroscan® was technically feasible in 28/29 (97%) patients. Median liver stiffness was 4.5 kPa (2.8-32.8), and was > 7.2 kPa in 5/28 (18%) and > 14 kPa in 2/28 (7%) patients. Liver stiffness was increased in 2/2 (100%) patients with abnormal LFT and US, in 1/4 (25%) with abnormal LFT or US and in 2/22 (10%) patients with normal LFT and US. CONCLUSIONS Fibroscan® is an easy and repeatable tool which can be used in PiZZ patients to screen for the presence of significant liver fibrosis and to identify patients at higher risk to develop liver complications in the future and who may benefit from a closer surveillance.
Collapse
Affiliation(s)
- Olivier Guillaud
- Service d'hépato-gastro-entérologie, hôpital Édouard-Herriot, hospices civils de Lyon, 69437 Lyon, France.
| | - Jérôme Dumortier
- Service d'hépato-gastro-entérologie, hôpital Édouard-Herriot, hospices civils de Lyon, 69437 Lyon, France; Université Claude-Bernard Lyon 1, 69100 Villeurbanne, France.
| | - Julie Traclet
- Service de pneumologie, groupement hospitalier Est, hospices civils de Lyon, 69677 Bron, France.
| | - Lioara Restier
- Service de gastro-entérologie, hépatologie et nutrition pédiatriques, hôpital Femme-Mère-Enfant, hospices civils de Lyon, 69677 Bron, France.
| | - Philippe Joly
- Unité de pathologie moléculaire du globule rouge, laboratoire de biochimie et de biologie moléculaire, hôpital Édouard-Herriot, hospices civils de Lyon, 69437 Lyon, France.
| | - Colette Chapuis-Cellier
- Université Claude-Bernard Lyon 1, 69100 Villeurbanne, France; Laboratoire d'immunologie, centre de biologie Sud, centre hospitalier Lyon-Sud, hospices civils, 69310 Pierre-Bénite, France.
| | - Alain Lachaux
- Université Claude-Bernard Lyon 1, 69100 Villeurbanne, France; Service de gastro-entérologie, hépatologie et nutrition pédiatriques, hôpital Femme-Mère-Enfant, hospices civils de Lyon, 69677 Bron, France.
| | - Jean François Mornex
- Service de pneumologie, groupement hospitalier Est, hospices civils de Lyon, 69677 Bron, France; Université de Lyon, Inra, UMR754, 69007, Lyon, France.
| |
Collapse
|
24
|
Ke PY. Diverse Functions of Autophagy in Liver Physiology and Liver Diseases. Int J Mol Sci 2019; 20:E300. [PMID: 30642133 PMCID: PMC6358975 DOI: 10.3390/ijms20020300] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/05/2019] [Accepted: 01/08/2019] [Indexed: 01/09/2023] Open
Abstract
Autophagy is a catabolic process by which eukaryotic cells eliminate cytosolic materials through vacuole-mediated sequestration and subsequent delivery to lysosomes for degradation, thus maintaining cellular homeostasis and the integrity of organelles. Autophagy has emerged as playing a critical role in the regulation of liver physiology and the balancing of liver metabolism. Conversely, numerous recent studies have indicated that autophagy may disease-dependently participate in the pathogenesis of liver diseases, such as liver hepatitis, steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma. This review summarizes the current knowledge on the functions of autophagy in hepatic metabolism and the contribution of autophagy to the pathophysiology of liver-related diseases. Moreover, the impacts of autophagy modulation on the amelioration of the development and progression of liver diseases are also discussed.
Collapse
Affiliation(s)
- Po-Yuan Ke
- Department of Biochemistry & Molecular Biology and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
- Division of Allergy, Immunology, and Rheumatology, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan.
| |
Collapse
|
25
|
Zhang L, Yao Z, Ji G. Herbal Extracts and Natural Products in Alleviating Non-alcoholic Fatty Liver Disease via Activating Autophagy. Front Pharmacol 2018; 9:1459. [PMID: 30618753 PMCID: PMC6297257 DOI: 10.3389/fphar.2018.01459] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/29/2018] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease world-wide, and currently therapeutic options for NAFLD are limited. Herbal medicine (HM) may offer an attractive alternative for the treatment of NAFLD. Recent years have witnessed a growing interest in the autophagy-inducing agents, and autophagy activation has been recognized as an efficient strategy in managing NAFLD and related complications. Pharmacological studies have demonstrated certain potential of HM extracts and natural products in inducing autophagy, which might contribute to the efficacy of HM in preventing and treating NAFLD. This review aims to summarize current understanding of mechanisms of HM extracts and natural products in preventing and treating NAFLD. Specially, we focused on mechanisms by which autophagy can target the main pathogenesis events associated with NAFLD, including hepatic steatosis, inflammation, oxidative stress, and apoptosis. It is hoped that this brief review can provide a general understanding of HM extracts and natural products in treating NAFLD, and raise awareness of potential clinical application of HM in general.
Collapse
Affiliation(s)
- Li Zhang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zemin Yao
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Guang Ji
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
26
|
Kuscuoglu D, Janciauskiene S, Hamesch K, Haybaeck J, Trautwein C, Strnad P. Liver - master and servant of serum proteome. J Hepatol 2018; 69:512-524. [PMID: 29709680 DOI: 10.1016/j.jhep.2018.04.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 12/20/2022]
Abstract
Hepatocytes synthesise the majority of serum proteins. This production occurs in the endoplasmic reticulum (ER) and is adjusted by complex local and systemic regulatory mechanisms. Accordingly, serum levels of hepatocyte-made proteins constitute important biomarkers that reflect both systemic processes and the status of the liver. For example, C-reactive protein is an established marker of inflammatory reaction, whereas transferrin emerges as a liver stress marker and an attractive mortality predictor. The high protein flow through the ER poses a continuous challenge that is handled by a complex proteostatic network consisting of ER folding machinery, ER stress response, ER-associated degradation and autophagy. Various disorders disrupt this delicate balance and result in protein accumulation in the ER. These include chronic hepatitis B infection with overproduction of hepatitis B surface antigen or inherited alpha1-antitrypsin deficiency that give rise to ground glass hepatocytes and alpha1-antitrypsin aggregates, respectively. We review these ER storage disorders and their downstream consequences. The interaction between proteotoxic stress and other ER challenges such as lipotoxicity is also discussed. Collectively, this article aims to sharpen our view of liver hepatocytes as the central hubs of protein metabolism.
Collapse
Affiliation(s)
- Deniz Kuscuoglu
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany; The Interdisciplinary Center for Clinical Research (IZKF), University Hospital Aachen, Aachen, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Hannover Medical School, BREATH, German Center for Lung Research (DZL), Hannover, Germany
| | - Karim Hamesch
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Johannes Haybaeck
- Institute of Pathology, Medical University Graz, Graz, Austria; Department of Pathology, Medical Faculty, Otto-von-Guericke University of Magdeburg, Magdeburg, Germany
| | - Christian Trautwein
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
| | - Pavel Strnad
- Medical Clinic III, Gastroenterology, Metabolic Diseases and Intensive Care, University Hospital RWTH Aachen, Aachen, Germany; The Interdisciplinary Center for Clinical Research (IZKF), University Hospital Aachen, Aachen, Germany.
| |
Collapse
|
27
|
Zhang JJ, Zhou QM, Chen S, Le WD. Repurposing carbamazepine for the treatment of amyotrophic lateral sclerosis in SOD1-G93A mouse model. CNS Neurosci Ther 2018; 24:1163-1174. [PMID: 29656576 DOI: 10.1111/cns.12855] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/27/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022] Open
Abstract
AIMS To investigate the effect and mechanisms of carbamazepine (CBZ) on the onset and progression of amyotrophic lateral sclerosis (ALS) in SOD1-G93A mouse model. METHODS Starting from 64 days of age, SOD1-G93A mice were orally administered with CBZ at 200 mg/kg once daily until death. The disease onset and life span of SOD1-G93A mice were recorded. Motor neurons (MNs) in anterior horn of spinal cord were quantified by Nissl staining and SMI-32 immunostaining. Hematoxylin and eosin (H&E), nicotinamide adenine dinucleotide hydrogen (NADH), modified Gomori trichrome (MGT), and α-bungarotoxin-ATTO-488 staining were also performed to evaluate muscle and neuromuscular junction (NMJ) damage. Expressions of aggregated SOD1 protein and autophagy-related proteins were further detected by Western blot and immunofluorescent staining. RESULTS Carbamazepine treatment could delay the disease onset and extend life span of SOD1-G93A mice by about 14.5% and 13.9%, respectively. Furthermore, CBZ treatment reduced MNs loss by about 46.6% and ameliorated the altered muscle morphology and NMJ. Much more interestingly, mechanism study revealed that CBZ treatment activated autophagy via AMPK-ULK1 pathway and promoted the clearance of mutant SOD1 aggregation. CONCLUSION Our findings uncovered the therapeutic effects of CBZ against disease pathogenesis in SOD1-G93A mice, indicating a promising clinical utilization of CBZ in ALS therapy.
Collapse
Affiliation(s)
- Jing-Jing Zhang
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Chifeng Municipal Hospital, Chifeng, China
| | - Qin-Ming Zhou
- Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sheng Chen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Dong Le
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Collaborative Innovation Center for Brain Science, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
28
|
Neerman-Arbez M, Casini A. Clinical Consequences and Molecular Bases of Low Fibrinogen Levels. Int J Mol Sci 2018; 19:E192. [PMID: 29316703 PMCID: PMC5796141 DOI: 10.3390/ijms19010192] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 12/22/2017] [Accepted: 12/22/2017] [Indexed: 12/19/2022] Open
Abstract
The study of inherited fibrinogen disorders, characterized by extensive allelic heterogeneity, allows the association of defined mutations with specific defects providing significant insight into the location of functionally important sites in fibrinogen and fibrin. Since the identification of the first causative mutation for congenital afibrinogenemia, studies have elucidated the underlying molecular pathophysiology of numerous causative mutations leading to fibrinogen deficiency, developed cell-based and animal models to study human fibrinogen disorders, and further explored the clinical consequences of absent, low, or dysfunctional fibrinogen. Since qualitative disorders are addressed by another review in this special issue, this review will focus on quantitative disorders and will discuss their diagnosis, clinical features, molecular bases, and introduce new models to study the phenotypic consequences of fibrinogen deficiency.
Collapse
Affiliation(s)
- Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, 1211 Geneva, Switzerland.
| | - Alessandro Casini
- Division of Angiology and Hemostasis, Faculty of Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland.
| |
Collapse
|
29
|
Callea F, Giovannoni I, Sari S, Guldal E, Dalgic B, Akyol G, Sogo T, Al-Hussaini A, Maggiore G, Bartuli A, Boldrini R, Francalanci P, Bellacchio E. Fibrinogen Gamma Chain Mutations Provoke Fibrinogen and Apolipoprotein B Plasma Deficiency and Liver Storage. Int J Mol Sci 2017; 18:ijms18122717. [PMID: 29244742 PMCID: PMC5751318 DOI: 10.3390/ijms18122717] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 01/12/2023] Open
Abstract
p.R375W (Fibrinogen Aguadilla) is one out of seven identified mutations (Brescia, Aguadilla, Angers, Al du Pont, Pisa, Beograd, and Ankara) causing hepatic storage of the mutant fibrinogen γ. The Aguadilla mutation has been reported in children from the Caribbean, Europe, Japan, Saudi Arabia, Turkey, and China. All reported children presented with a variable degree of histologically proven chronic liver disease and low plasma fibrinogen levels. In addition, one Japanese and one Turkish child had concomitant hypo-APOB-lipoproteinemia of unknown origin. We report here on an additional child from Turkey with hypofibrinogenemia due to the Aguadilla mutation, massive hepatic storage of the mutant protein, and severe hypo-APOB-lipoproteinemia. The liver biopsy of the patient was studied by light microscopy, electron microscopy (EM), and immunohistochemistry. The investigation included the DNA sequencing of the three fibrinogen and APOB-lipoprotein regulatory genes and the analysis of the encoded protein structures. Six additional Fibrinogen Storage Disease (FSD) patients with either the Aguadilla, Ankara, or Brescia mutations were investigated with the same methodology. A molecular analysis revealed the fibrinogen gamma p.R375W mutation (Aguadilla) but no changes in the APOB and MTTP genes. APOB and MTTP genes showed no abnormalities in the other study cases. Light microscopy and EM studies of liver tissue samples from the child led to the demonstration of the simultaneous accumulation of both fibrinogen and APOB in the same inclusions. Interestingly enough, APOB-containing lipid droplets were entrapped within the fibrinogen inclusions in the hepatocytic Endoplasmic Reticulum (ER). Similar histological, immunohistochemical, EM, and molecular genetics findings were found in the other six FSD cases associated with the Aguadilla, as well as with the Ankara and Brescia mutations. The simultaneous retention of fibrinogen and APOB-lipoproteins in FSD can be detected in routinely stained histological sections. The analysis of protein structures unraveled the pathomorphogenesis of this unexpected phenomenon. Fibrinogen gamma chain mutations provoke conformational changes in the region of the globular domain involved in the "end-to-end" interaction, thus impairing the D-dimer formation. Each monomeric fibrinogen gamma chain is left with an abnormal exposure of hydrophobic patches that become available for interactions with APOB and lipids, causing their intracellular retention and impairment of export as a secondary unavoidable phenomenon.
Collapse
Affiliation(s)
- Francesco Callea
- Department Pathology and Molecular Histopathology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy.
| | - Isabella Giovannoni
- Department Pathology and Molecular Histopathology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy.
| | - Sinan Sari
- Department Pediatric Gastroenterology, Gazi University Ankara, 06560 Ankara, Turkey.
| | - Esendagli Guldal
- Department Pathology, Gazi University Ankara, 06560 Ankara, Turkey.
| | - Buket Dalgic
- Department Pediatric Gastroenterology, Gazi University Ankara, 06560 Ankara, Turkey.
| | - Gulen Akyol
- Department Pathology, Gazi University Ankara, 06560 Ankara, Turkey.
| | - Tsuyoshi Sogo
- Department of Pediatric Hepatology and Gastroenterology, Saiseikai Yokohama City Tobu Hospital 3-6-1, Shimosueyoshi, Tsurumi Ward, Yokohama City, Kanagawa, Japan.
| | - Abdulrahman Al-Hussaini
- Division of Pediatric Gastroenterology, Children's Specialized Hospital, King Fahad Medical City, College of Medicine, Alfaisal University Riyadh 11525, Saudi Arabia.
| | - Giuseppe Maggiore
- Section of Pediatrics, Department of Medical Sciences, University of Ferrara, University Hospital Arcispedale Sant'Anna, 44100 Ferrara, Italy.
| | - Andrea Bartuli
- Rare Disease and Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy.
| | - Renata Boldrini
- Department Pathology and Molecular Histopathology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy.
| | - Paola Francalanci
- Department Pathology and Molecular Histopathology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy.
| | - Emanuele Bellacchio
- Genetics and Rare Diseases, Research Division, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy.
| |
Collapse
|
30
|
Drug Repurposing Patent Applications July–September 2017. Assay Drug Dev Technol 2017; 15:378-382. [DOI: 10.1089/adt.2017.29069.pq3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
31
|
The fibrous form of intracellular inclusion bodies in recombinant variant fibrinogen-producing cells is specific to the hepatic fibrinogen storage disease-inducible variant fibrinogen. Int J Hematol 2017; 105:758-768. [PMID: 28161763 DOI: 10.1007/s12185-017-2185-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 10/20/2022]
Abstract
Fibrinogen storage disease (FSD) is a rare disorder that is characterized by the accumulation of fibrinogen in hepatocytes and induces liver injury. Six mutations in the γC domain (γG284R, γT314P, γD316N, the deletion of γG346-Q350, γG366S, and γR375W) have been identified for FSD. Our group previously established γ375W fibrinogen-producing Chinese hamster ovary (CHO) cells and observed aberrant large granular and fibrous forms of intracellular inclusion bodies. The aim of this study was to investigate whether fibrous intracellular inclusion bodies are specific to FSD-inducible variant fibrinogen. Thirteen expression vectors encoding the variant γ-chain were stably or transiently transfected into CHO cells expressing normal fibrinogen Aα- and Bβ-chains or HuH-7 cells, which were then immunofluorescently stained. Six CHO and HuH-7 cell lines that transiently produced FSD-inducible variant fibrinogen presented the fibrous (3.2-22.7 and 2.1-24.5%, respectively) and large granular (5.4-25.5 and 7.7-23.9%) forms of intracellular inclusion bodies. Seven CHO and HuH-7 cell lines that transiently produced FSD-non-inducible variant fibrinogen only exhibit the large granular form. These results demonstrate that transiently transfected variant fibrinogen-producing CHO cells and inclusion bodies of the fibrous form may be useful in non-invasive screening for FSD risk factors for FSD before its onset.
Collapse
|
32
|
Tipping the scales: Lessons from simple model systems on inositol imbalance in neurological disorders. Eur J Cell Biol 2017; 96:154-163. [PMID: 28153412 DOI: 10.1016/j.ejcb.2017.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/18/2017] [Accepted: 01/21/2017] [Indexed: 11/20/2022] Open
Abstract
Inositol and inositol-containing compounds have signalling and regulatory roles in many cellular processes, suggesting that inositol imbalance may lead to wide-ranging changes in cellular functions. Indeed, changes in inositol-dependent signalling have been implicated in various diseases and cellular functions such as autophagy, and these changes have often been proposed as therapeutic targets. However, few studies have highlighted the links between inositol depletion and the downstream effects on inositol phosphates and phosphoinositides in disease states. For this research, many advances have employed simple model systems that include the social amoeba D. discoideum and the yeast S. cerevisiae, since these models enable a range of experimental approaches that are not possible in mammalian models. In this review, we discuss recent findings initiated in simple model systems and translated to higher model organisms where the effect of altered inositol, inositol phosphate and phosphoinositide levels impact on bipolar disorder, Alzheimer disease, epilepsy and autophagy.
Collapse
|
33
|
Zhang MH, Knisely AS, Wang NL, Gong JY, Wang JS. Fibrinogen storage disease in a Chinese boy with de novo fibrinogen Aguadilla mutation: Incomplete response to carbamazepine and ursodeoxycholic acid. BMC Gastroenterol 2016; 16:92. [PMID: 27520927 PMCID: PMC4981954 DOI: 10.1186/s12876-016-0507-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/02/2016] [Indexed: 01/04/2023] Open
Abstract
Background Fibrinogen storage disease (FSD) is a rare autosomal-dominant disorder caused by mutation in FGG, encoding the fibrinogen gamma chain. Here we report the first Han Chinese patient with FSD, caused by de novo fibrinogen Aguadilla mutation, and his response to pharmacologic management. Case presentation Epistaxis and persistent clinical-biochemistry test-result abnormalities prompted liver biopsy in a boy, with molecular study of FGG in him and his parents. He was treated with the autophagy enhancer carbamazepine, reportedly effective in FSD, and with ursodeoxycholic acid thereafter. Inclusion bodies in hepatocellular cytoplasm stained immune-histochemically for fibrinogen. Selective analysis of FGG found the heterozygous mutation c.1201C > T (p.Arg401Trp), absent in both parents. Over more than one year’s follow-up, transaminase and gamma-glutamyl transpeptidase activities have lessened but not normalized. Conclusion This report expands the epidemiology of FSD and demonstrates idiosyncrasy in response to oral carbamazepine and/or ursodeoxycholic acid in FSD. Electronic supplementary material The online version of this article (doi:10.1186/s12876-016-0507-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mei-Hong Zhang
- Department of Pediatrics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - A S Knisely
- Institute of Liver Studies, King's College Hospital, Denmark Hill, London, SE5 9RS, UK.,Present address: Institute of Pathology, Medical University Graz, Auenbruggerplatz 25, 8036, Graz, Austria
| | - Neng-Li Wang
- Department of Pediatrics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Jing-Yu Gong
- Department of Pediatrics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Jian-She Wang
- Department of Pediatrics, Jinshan Hospital, Fudan University, Shanghai, 201508, China. .,The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, Shanghai, 201102, China. .,Department of Pediatrics, Shanghai Medical College, Fudan University, Shanghai, 201102, China.
| |
Collapse
|
34
|
Uggenti C, Briant K, Streit AK, Thomson S, Koay YH, Baines RA, Swanton E, Manson FD. Restoration of mutant bestrophin-1 expression, localisation and function in a polarised epithelial cell model. Dis Model Mech 2016; 9:1317-1328. [PMID: 27519691 PMCID: PMC5117222 DOI: 10.1242/dmm.024216] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 07/21/2016] [Indexed: 01/13/2023] Open
Abstract
Autosomal recessive bestrophinopathy (ARB) is a retinopathy caused by mutations in the bestrophin-1 protein, which is thought to function as a Ca2+-gated Cl− channel in the basolateral surface of the retinal pigment epithelium (RPE). Using a stably transfected polarised epithelial cell model, we show that four ARB mutant bestrophin-1 proteins were mislocalised and subjected to proteasomal degradation. In contrast to the wild-type bestrophin-1, each of the four mutant proteins also failed to conduct Cl− ions in transiently transfected cells as determined by whole-cell patch clamp. We demonstrate that a combination of two clinically approved drugs, bortezomib and 4-phenylbutyrate (4PBA), successfully restored the expression and localisation of all four ARB mutant bestrophin-1 proteins. Importantly, the Cl− conductance function of each of the mutant bestrophin-1 proteins was fully restored to that of wild-type bestrophin-1 by treatment of cells with 4PBA alone. The functional rescue achieved with 4PBA is significant because it suggests that this drug, which is already approved for long-term use in infants and adults, might represent a promising therapy for the treatment of ARB and other bestrophinopathies resulting from missense mutations in BEST1. Summary: Chemical chaperone 4PBA fully restores Cl− conductance activity for mutant bestrophin-1 proteins associated with inherited retinal dystrophy, autosomal recessive bestrophinopathy.
Collapse
Affiliation(s)
- Carolina Uggenti
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Kit Briant
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Anne-Kathrin Streit
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Steven Thomson
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Yee Hui Koay
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Richard A Baines
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Eileithyia Swanton
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Forbes D Manson
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
35
|
Casini A, de Moerloose P. Can the phenotype of inherited fibrinogen disorders be predicted? Haemophilia 2016; 22:667-75. [PMID: 27293018 DOI: 10.1111/hae.12967] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2016] [Indexed: 12/17/2022]
Abstract
Congenital fibrinogen disorders are rare diseases affecting either the quantity (afibrinogenaemia and hypofibrinogenaemia) or the quality (dysfibrinogenaemia) or both (hypodysfibrinogenaemia) of fibrinogen. In addition to bleeding, unexpected thrombosis, spontaneous spleen ruptures, painful bone cysts and intrahepatic inclusions can complicate the clinical course of patients with quantitative fibrinogen disorders. Clinical manifestations of dysfibrinogenaemia include absence of symptoms, major bleeding or thrombosis as well as systemic amyloidosis. Although the diagnosis of any type of congenital fibrinogen disorders is usually not too difficult with the help of conventional laboratory tests completed by genetic studies, the correlation between all available tests and the clinical manifestations is more problematic in many cases. Improving accuracy of diagnosis, performing genotype, analysing function of fibrinogen variants and carefully investigating the personal and familial histories may lead to a better assessment of patients' phenotype and therefore help in identifying patients at increased risk of adverse clinical outcomes. This review provides an update of various tests (conventional and global assays, molecular testing, fibrin clot analysis) and clinical features, which may help to better predict the phenotype of the different types of congenital fibrinogen disorders.
Collapse
Affiliation(s)
- A Casini
- Division of Angiology and Haemostasis, University Hospitals and Faculty of Medicine, Geneva, Switzerland.
| | - P de Moerloose
- Division of Angiology and Haemostasis, University Hospitals and Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
36
|
Sari S, Yilmaz G, Gonul II, Dalgic B, Akyol G, Giovannoni I, Francalanci P, Callea F. Fibrinogen storage disease and cirrhosis associated with hypobetalipoproteinemia owing to fibrinogen Aguadilla in a Turkish child. Liver Int 2015; 35:2501-5. [PMID: 26176881 DOI: 10.1111/liv.12914] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 06/29/2015] [Indexed: 02/13/2023]
Abstract
BACKGROUND AND AIMS Fibrinogen gene mutations can rarely result in hepatic fibrinogen storage disease (HFSD). Herein, we report on the first Turkish family carrying the mutation p.Arg375Trp (fibrinogen Aguadilla) in the γ-chain of the fibrinogen (FGG) gene. METHODS Clinical, laboratory and histopathological findings of the patient were documented. Molecular study of fibrinogen gene was performed in the patient and her family members. RESULTS The proband was 5 years old girl presenting with advanced liver fibrosis of unknown origin. The child had very low plasma levels of fibrinogen and hypobetalipoproteinemia. Immunomorphologic and electron microscopic studies showed selective and exclusive accumulation of fibrinogen within the endoplasmic reticulum in liver biopsy of the patient. Patient, mother, two sisters and one brother carried p.Arg375Trp mutation (fibrinogen Aguadilla) in FGG gene. The patient was treated with ursodeoxycholic acid and carbamazepine. After 3 months, carbamazepine was suspended upon family decision and unresponsiveness of carbamazepine. CONCLUSIONS HFSD is characterized by hypofibrinogenemia and accumulation of abnormal fibrinogen within hepatocytes. In addition, hypofibrinogenemia is associated with hypobetalipoproteinemia in Aguadilla mutation.
Collapse
Affiliation(s)
- Sinan Sari
- Department of Pediatric Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Guldal Yilmaz
- Department of Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Ipek I Gonul
- Department of Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Buket Dalgic
- Department of Pediatric Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Gulen Akyol
- Department of Pathology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Isabella Giovannoni
- Department of Pathology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paola Francalanci
- Department of Pathology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Francesco Callea
- Department of Pathology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
37
|
Hidvegi T, Stolz DB, Alcorn JF, Yousem SA, Wang J, Leme AS, Houghton AM, Hale P, Ewing M, Cai H, Garchar EA, Pastore N, Annunziata P, Kaminski N, Pilewski J, Shapiro SD, Pak SC, Silverman GA, Brunetti-Pierri N, Perlmutter DH. Enhancing Autophagy with Drugs or Lung-directed Gene Therapy Reverses the Pathological Effects of Respiratory Epithelial Cell Proteinopathy. J Biol Chem 2015; 290:29742-57. [PMID: 26494620 DOI: 10.1074/jbc.m115.691253] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Indexed: 11/06/2022] Open
Abstract
Recent studies have shown that autophagy mitigates the pathological effects of proteinopathies in the liver, heart, and skeletal muscle but this has not been investigated for proteinopathies that affect the lung. This may be due at least in part to the lack of an animal model robust enough for spontaneous pathological effects from proteinopathies even though several rare proteinopathies, surfactant protein A and C deficiencies, cause severe pulmonary fibrosis. In this report we show that the PiZ mouse, transgenic for the common misfolded variant α1-antitrypsin Z, is a model of respiratory epithelial cell proteinopathy with spontaneous pulmonary fibrosis. Intracellular accumulation of misfolded α1-antitrypsin Z in respiratory epithelial cells of the PiZ model resulted in activation of autophagy, leukocyte infiltration, and spontaneous pulmonary fibrosis severe enough to elicit functional restrictive deficits. Treatment with autophagy enhancer drugs or lung-directed gene transfer of TFEB, a master transcriptional activator of the autophagolysosomal system, reversed these proteotoxic consequences. We conclude that this mouse is an excellent model of respiratory epithelial proteinopathy with spontaneous pulmonary fibrosis and that autophagy is an important endogenous proteostasis mechanism and an attractive target for therapy.
Collapse
Affiliation(s)
- Tunda Hidvegi
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | | | - John F Alcorn
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | | | | | | | | | - Pamela Hale
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Michael Ewing
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Houming Cai
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Evelyn Akpadock Garchar
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Nunzia Pastore
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138
| | - Patrizia Annunziata
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138
| | | | | | | | - Stephen C Pak
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224
| | - Gary A Silverman
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224, Cell Biology, and
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Federico II University, Naples, Italy, 80138 Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy, 80131, and
| | - David H Perlmutter
- From the Departments of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15224, Cell Biology, and
| |
Collapse
|
38
|
Asselta R, Robusto M, Braidotti P, Peyvandi F, Nastasio S, D'Antiga L, Perisic VN, Maggiore G, Caccia S, Duga S. Hepatic fibrinogen storage disease: identification of two novel mutations (p.Asp316Asn, fibrinogen Pisa and p.Gly366Ser, fibrinogen Beograd) impacting on the fibrinogen γ-module. J Thromb Haemost 2015; 13:1459-67. [PMID: 26039544 DOI: 10.1111/jth.13021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/13/2015] [Indexed: 08/31/2023]
Abstract
BACKGROUND Quantitative fibrinogen deficiencies (hypofibrinogenemia and afibrinogenemia) are rare congenital disorders characterized by low/unmeasurable plasma fibrinogen antigen levels. Their genetic basis is invariably represented by mutations within the fibrinogen genes (FGA, FGB and FGG coding for the Aα, Bβ and γ chains). Currently, only four mutations (p.Gly284Arg, p.Arg375Trp, delGVYYQ 346-350, p.Thr314Pro), all affecting the fibrinogen γ chain, have been reported to cause fibrinogen storage disease (FSD), a disorder characterized by protein aggregation, endoplasmic reticulum retention and hypofibrinogenemia. OBJECTIVES To investigate the genetic basis of FSD in two hypofibrinogenemic patients. METHODS The mutational screening of the fibrinogen genes was performed by direct DNA sequencing. The impact of identified mutations on fibrinogen structure was investigated by in-silico molecular modeling. Liver histology was evaluated by light microscopy, electron microscopy and immunocytochemistry. RESULTS Here, we describe two hypofibrinogenemic children with persistent abnormal liver function parameters. Direct sequencing of the coding portion of fibrinogen genes disclosed two novel FGG missense variants (p.Asp316Asn, fibrinogen Pisa; p.Gly366Ser, fibrinogen Beograd), both present in the heterozygous state and affecting residues located in the fibrinogen C-terminal γ-module. Liver sections derived from biopsies of the two patients were examined by immunocytochemical analyses, revealing hepatocyte cytoplasmic inclusions immunoreactive to anti-fibrinogen antibodies. CONCLUSIONS Our work strongly confirms the clustering of mutations causing FSD in the fibrinogen γ chain between residues 284 and 375. Based on an in-depth structural analysis of all FSD-causing mutations and on their resemblance to mutations leading to serpinopathies, we also comment on a possible mechanism explaining fibrinogen polymerization within hepatocytes.
Collapse
Affiliation(s)
- R Asselta
- Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Italy
| | - M Robusto
- Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Italy
| | - P Braidotti
- Pathology Department, S. Paolo Hospital, Milan, Italy
| | - F Peyvandi
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Angelo Bianchi Bonomi Haemophilia and Thrombosis Centre, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico and Luigi Villa Foundation, Milan, Italy
| | - S Nastasio
- Department of Clinical and Experimental Medicine, University of Pisa, Pediatric Gastroenterology, University Hospital Santa Chiara, Pisa, Italy
| | - L D'Antiga
- Paediatric Liver, GI and Transplantation, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - V N Perisic
- Department of Gastroenterology and Hepatology, University Children's Hospital, Belgrade, Serbia
| | - G Maggiore
- Department of Clinical and Experimental Medicine, University of Pisa, Pediatric Gastroenterology, University Hospital Santa Chiara, Pisa, Italy
| | - S Caccia
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università degli Studi di Milano, Segrate, Milan, Italy
| | - S Duga
- Department of Biomedical Sciences, Humanitas University, Rozzano, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Italy
| |
Collapse
|
39
|
Casini A, Sokollik C, Lukowski SW, Lurz E, Rieubland C, de Moerloose P, Neerman-Arbez M. Hypofibrinogenemia and liver disease: a new case of Aguadilla fibrinogen and review of the literature. Haemophilia 2015; 21:820-7. [PMID: 25990487 DOI: 10.1111/hae.12719] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2015] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Fibrinogen storage disease (FSD) is characterized by hypofibrinogenemia and hepatic inclusions due to impaired release of mutant fibrinogen which accumulates and aggregates in the hepatocellular endoplasmic reticulum. Liver disease is variable. AIM We studied a new Swiss family with fibrinogen Aguadilla. In order to understand the molecular peculiarity of FSD mutations, fibrinogen Aguadilla and the three other causative mutations, all located in the γD domain, were modelled. METHOD The proband is a Swiss girl aged 4 investigated because of fatigue and elevated liver enzymes. Protein structure models were prepared using the Swiss-PdbViewer and POV-Ray software. RESULTS The proband was found to be heterozygous for fibrinogen Aguadilla: FGG Arg375Trp. Familial screening revealed that her mother and maternal grandmother were also affected and, in addition, respectively heterozygous and homozygous for the hereditary haemochromatosis mutation HFE C282Y. Models of backbone and side-chain interactions for fibrinogen Aguadilla in a 10-angstrom region revealed the loss of five H-bonds and the gain of one H-bond between structurally important amino acids. The structure predicted for fibrinogen Angers showed a novel helical structure in place of hole 'a' on the outer edge of γD likely to have a negative impact on fibrinogen assembly and secretion. CONCLUSION The mechanism by which FSD mutations generate hepatic intracellular inclusions is still not clearly established although the promotion of aberrant intermolecular strand insertions is emerging as a likely cause. Reporting new cases is essential in the light of novel opportunities of treatment offered by increasing knowledge of the degradation pathway and autophagy.
Collapse
Affiliation(s)
- A Casini
- Angiology and Haemostasis, University Hospitals and Faculty of Medicine of Geneva, Geneva, Switzerland
| | - C Sokollik
- Paediatric Gastroenterology, Hepatology and Nutrition, University Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - S W Lukowski
- Department of Genetic Medicine and Development, University Medical School of Geneva, Geneva, Switzerland
| | - E Lurz
- Paediatric Gastroenterology, Hepatology and Nutrition, University Children's Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - C Rieubland
- Division of Human Genetics, Department of Paediatrics, Inselspital, Bern, Switzerland
| | - P de Moerloose
- Angiology and Haemostasis, University Hospitals and Faculty of Medicine of Geneva, Geneva, Switzerland
| | - M Neerman-Arbez
- Angiology and Haemostasis, University Hospitals and Faculty of Medicine of Geneva, Geneva, Switzerland.,Department of Genetic Medicine and Development, University Medical School of Geneva, Geneva, Switzerland
| |
Collapse
|
40
|
Hauff P, Gottwald U, Ocker M. Early to Phase II drugs currently under investigation for the treatment of liver fibrosis. Expert Opin Investig Drugs 2015; 24:309-327. [PMID: 25547844 DOI: 10.1517/13543784.2015.997874] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Chronic liver diseases represent a high unmet medical need and are characterized by persistent inflammation, parenchymal damage and fibrotic remodeling, leading eventually to cirrhosis and hepatic failure. Besides the persisting high prevalence of chronic viral hepatitis B and C, the dramatic increase in nonalcoholic steatohepatitis is now considered to be a major pathophysiologic driver for fibrosis development and subsequently cirrhosis. Increasing evidence suggests that also liver cirrhosis can regress when treated adequately. AREAS COVERED Herein, the authors review the underlying pathophysiologic mechanisms leading to fibrotic remodeling in the liver. They also highlight the options for novel treatment strategies by using molecular targeted agents. EXPERT OPINION New in vitro and preclinical animal models, and the careful selection of patients with high disease dynamics for clinical studies, provide a sound basis for the clinical development of antifibrotic agents in humans. Surrogate parameters of liver function, inflammation, tissue remodeling and damage, as well as noninvasive imaging techniques, can be applied in clinical trials to provide fast readouts and novel and reliable endpoints for trial design, and provide an attractive regulatory environment for this emerging disease area.
Collapse
|
41
|
Asselta R, Platè M, Robusto M, Borhany M, Guella I, Soldà G, Afrasiabi A, Menegatti M, Shamsi T, Peyvandi F, Duga S. Clinical and molecular characterisation of 21 patients affected by quantitative fibrinogen deficiency. Thromb Haemost 2014; 113:567-76. [PMID: 25427968 DOI: 10.1160/th14-07-0629] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 09/30/2014] [Indexed: 12/14/2022]
Abstract
Fibrinogen is a plasma glycoprotein mainly synthesised by hepatocytes and circulating as a 340-kDa hexamer consisting of two sets of three different polypeptide chains (Aα, Bβ, and γ, encoded by the FGA, FGB, and FGG gene, respectively). Congenital afibrinogenaemia and hypofibrinogenaemia are rare bleeding disorders characterised by abnormally low levels of functional and immunoreactive fibrinogen in plasma, associated with haemorrhagic manifestations of variable severity. While afibrinogenaemia is caused by mutations in the homozygous or compound heterozygous state in one of the three fibrinogen genes, hypofibrinogenaemia is generally due to heterozygous mutations, and is usually characterised by a milder phenotype. The mutational spectrum of these quantitative fibrinogen disorders includes large deletions, point mutations causing premature termination codons, and missense mutations often affecting fibrinogen assembly and/or secretion. Here we report the clinical and molecular characterisation of 13 unrelated afibrinogenaemic and eight hypofibrinogenaemic patients, leading to the identification of 17 different mutations (10 hitherto unknown). All the newly-identified missense and splicing mutations werein vitro expressed to verify their pathogenic role. Our data increase the number of mutations causing quantitative fibrinogen deficiencies by about 7 %. The high number of private mutations identified in the analysed probands indicates that the full mutational screening of the three fibrinogen genes is still required for molecular diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Stefano Duga
- Prof. Stefano Duga, PhD, Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Viotti 3/5, 20133 Milan, Italy, Tel.: +39 02 50315823, Fax: +39 02 50315864, E-mail:
| |
Collapse
|
42
|
Capitalizing on the autophagic response for treatment of liver disease caused by alpha-1-antitrypsin deficiency and other genetic diseases. BIOMED RESEARCH INTERNATIONAL 2014; 2014:459823. [PMID: 25025052 PMCID: PMC4065733 DOI: 10.1155/2014/459823] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/23/2014] [Indexed: 11/29/2022]
Abstract
Alpha-1-antitrypsin deficiency (ATD) is one of the most common genetic causes of liver disease and is a prototype of liver diseases caused by the pathologic accumulation of aggregated mutant alpha-1-antitrypsin Z (ATZ) within liver cells. In the case of ATD-associated liver disease, the resulting “gain-of-function” toxicity can lead to serious clinical manifestations, including cirrhosis and hepatocellular carcinoma. Currently, the only definitive therapy for ATD-associated liver disease is liver transplantation, but recent efforts have demonstrated the exciting potential for novel therapies that target disposal of the mutant protein aggregates by harnessing a cellular homeostasis mechanism called autophagy. In this review, we will summarize research advances on autophagy and genetic liver diseases. We will discuss autophagy enhancer strategies for liver disease due to ATD and another genetic liver disease, inherited hypofibrinogenemia, caused by the proteotoxic effects of a misfolded protein. On the basis of recent evidence that autophagy plays a role in cellular lipid degradation, we also speculate about autophagy enhancer strategies for treatment of hepatic lipid storage diseases such as cholesterol ester storage disease.
Collapse
|
43
|
Gracia-Sancho J, Guixé-Muntet S, Hide D, Bosch J. Modulation of autophagy for the treatment of liver diseases. Expert Opin Investig Drugs 2014; 23:965-77. [DOI: 10.1517/13543784.2014.912274] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Jordi Gracia-Sancho
- Barcelona Hepatic Hemodynamic Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Hospital Clínic de Barcelona – CIBEREHD,
Barcelona, Spain ;
| | - Sergi Guixé-Muntet
- Barcelona Hepatic Hemodynamic Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Hospital Clínic de Barcelona – CIBEREHD,
Barcelona, Spain ;
| | - Diana Hide
- Barcelona Hepatic Hemodynamic Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Hospital Clínic de Barcelona – CIBEREHD,
Barcelona, Spain ;
| | | |
Collapse
|
44
|
Yang L, Rozenfeld R, Wu D, Devi LA, Zhang Z, Cederbaum A. Cannabidiol protects liver from binge alcohol-induced steatosis by mechanisms including inhibition of oxidative stress and increase in autophagy. Free Radic Biol Med 2014; 68:260-7. [PMID: 24398069 PMCID: PMC4112960 DOI: 10.1016/j.freeradbiomed.2013.12.026] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 12/23/2013] [Accepted: 12/27/2013] [Indexed: 12/31/2022]
Abstract
Acute alcohol drinking induces steatosis, and effective prevention of steatosis can protect liver from progressive damage caused by alcohol. Increased oxidative stress has been reported as one mechanism underlying alcohol-induced steatosis. We evaluated whether cannabidiol, which has been reported to function as an antioxidant, can protect the liver from alcohol-generated oxidative stress-induced steatosis. Cannabidiol can prevent acute alcohol-induced liver steatosis in mice, possibly by preventing the increase in oxidative stress and the activation of the JNK MAPK pathway. Cannabidiol per se can increase autophagy both in CYP2E1-expressing HepG2 cells and in mouse liver. Importantly, cannabidiol can prevent the decrease in autophagy induced by alcohol. In conclusion, these results show that cannabidiol protects mouse liver from acute alcohol-induced steatosis through multiple mechanisms including attenuation of alcohol-mediated oxidative stress, prevention of JNK MAPK activation, and increasing autophagy.
Collapse
Affiliation(s)
- Lili Yang
- School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | | | - Defeng Wu
- Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | - Zhenfeng Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China.
| | | |
Collapse
|
45
|
Puri P, Chandra A. Autophagy modulation as a potential therapeutic target for liver diseases. J Clin Exp Hepatol 2014; 4:51-9. [PMID: 25755534 PMCID: PMC4017203 DOI: 10.1016/j.jceh.2014.04.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/01/2014] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a critical intracellular pathway which maintains cellular function by lysosomal degradation of damaged proteins and organelles besides elimination of invading pathogens. Its primary function is to prevent cell death. Autophagy has diverse physiological functions namely; starvation adaptation, prevention of tumorigenesis, energy homeostasis, intracellular quality control and degradation of abnormal intracellular protein aggregates. Understanding the molecular mechanisms of autophagy has given key insights into the pathogenesis of various diseases like Non Alcoholic Steato-Hepatitis, Hepatitis B and C infections, Alpha-1 antitrypsin deficiency and hepatocellular carcinoma. Pharmacological modulation of autophagy may have a therapeutic potential in management of these liver diseases.
Collapse
Key Words
- AMPk, adenosine monophosphate-activated protein linase
- AT, antitrypsin
- ER, endoplasmic reticulum
- HBV, hepatitis B virus
- HCC, hepatocellular carcinoma
- HCV, hepatitis C virus
- HSC, hepatic stellate cells
- NASH, Non Alcoholic Steato-Hepatitis
- STEBPs, sterol regulatory element-binding proteins
- TG, triglyceride
- ULK1, Uncoordinated 51-like kinase 1
- autophagosome
- autophagy
- liver diseases
- mTORC1, mTOR complex 1
- mTORC2, mTOR complex 2
Collapse
Affiliation(s)
- Pankaj Puri
- Department of Gastroenterology, Army Hospital (Research and Referral), Dhaula Kuan, New Delhi 110010, India,Address for correspondence: Pankaj Puri, Department of Gastroenterology, Army Hospital (Research and Referral), Dhaula Kuan, New Delhi 110010, India.
| | - Alok Chandra
- Department of Gastroenterology, Command Hospital (EC), Alipore Road, Kolkata 700027, West Bengal, India
| |
Collapse
|
46
|
Lei YC, Luo P, Li W. Carbamazepine protects the liver against ischemia/reperfusion injury in mice. Shijie Huaren Xiaohua Zazhi 2013; 21:3617-3622. [DOI: 10.11569/wcjd.v21.i33.3617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the effects of carbamazepine (CBZ) on hepatic ischemia/reperfusion (I/R) injury in mice.
METHODS: Hepatic ischemia in male Balb/c mice was induced by occluding the portal triad for 1 h, and reperfusion was initiated by removing a microvascular clamp. Mice were randomly assigned to three groups (n = 6 for each group): I/R group as control, CBZ treatment group, and CBZ plus chloroquine (CQ) group. Serum ALT/AST levels at different time points were measured using biochemical methods. Hepatic morphological changes at 6 h after I/R were assessed by HE staining, and hepatocyte high mobility group box 1 (HMGB1) cytoplasmic translocation was detected by immunohistochemistry. Expression of Caspase3, Atg7, Beclin-1 and light chain 3 Ⅱ (LC3Ⅱ) in liver tissue was analyzed by Western blot.
RESULTS: CBZ blocked the depletion of Atg7 and Beclin-1 and LC3II expression after reperfusion. CBZ treatment decreased ALT/AST levels significantly 2, 6 and 12 h after I/R compared with the I/R group (all P < 0.01). Expression of Caspase3 in liver tissue and hepatocyte HMGB1 cytoplasmic translocation at 6 h after I/R were also decreased significantly in the CBZ group (both P < 0.01). CQ antagonized the effect of CBZ in decreasing ALT/AST levels, Caspase3 expression and hepatocyte HMGB1 cytoplasmic translocation.
CONCLUSION: CBZ protects the liver against I/R injury in mice.
Collapse
|