101
|
Van Vaisberg V, Tannuri ACA, Lima FR, Tannuri U. Ileal exclusion for pruritus treatment in children with progressive familial intrahepatic cholestasis and other cholestatic diseases. J Pediatr Surg 2020; 55:1385-1391. [PMID: 31708211 DOI: 10.1016/j.jpedsurg.2019.09.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pruritus is a major health-related quality-of-life burden in progressive familial intrahepatic cholestasis (PFIC) and other childhood cholestatic liver diseases. Several nontransplant surgical techniques were developed in an attempt to ameliorate symptoms and slow disease progression. Very few case-series have been published on a particular intervention, ileal exclusion (IE), which has been considered to be inferior to the other approaches. METHODS We conducted a single-center retrospective chart-review case-series of patients submitted to IE as the first-line surgical treatment at our institution from 1995 to 2018. The primary goal was pruritus relief, followed by survival with the native liver and improvement in biochemical parameters. RESULTS Eleven patients were submitted to IE, with a mean follow-up of 60 months. Complete resolution or significant reduction of pruritus was obtained in 72.7% (n = 8) of patients. One patient (9.1%) had a major postoperative complication that required surgery. No other morbidities were reported. Two cases progressed to end-stage liver disease (ESLD) within the short-term and one year after surgery. CONCLUSIONS This case series study shows that IE provided excellent results in pruritus control and permitted survival with the native liver. We believe IE is a safe procedure, with few associated morbidities, and should be considered more often as primary surgical treatment for PFIC and other cholestasis. LEVEL OF EVIDENCE IV.
Collapse
Affiliation(s)
- Victor Van Vaisberg
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Ana Cristina Aoun Tannuri
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Fabiana Roberto Lima
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Uenis Tannuri
- Pediatric Surgery Division, Pediatric Liver Transplantation Unit and Laboratory of Research in Pediatric Surgery (LIM 30), University of Sao Paulo Medical School, Sao Paulo, Brazil.
| |
Collapse
|
102
|
Slavetinsky C, Sturm E. Odevixibat and partial external biliary diversion showed equal improvement of cholestasis in a patient with progressive familial intrahepatic cholestasis. BMJ Case Rep 2020; 13:13/6/e234185. [PMID: 32601135 PMCID: PMC7326258 DOI: 10.1136/bcr-2019-234185] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Untreated progressive familial intrahepatic cholestasis (PFIC) type 2, or bile salt exporter protein deficiency, frequently leads to severe pruritus, impaired growth and progressive liver fibrosis with risk of organ failure. We describe a 15-month-old male patient with severe pruritus diagnosed with PFIC type 2 enrolled in an open-label phase 2 study who received 4 weeks of treatment with odevixibat, an ileal bile acid transporter inhibitor under development for cholestatic liver disease treatment. The patient experienced reductions in serum bile acids and improvement in itching and sleep scores, and odevixibat was well tolerated. After the odevixibat study, symptoms returned and the patient underwent partial external biliary diversion (PEBD). Odevixibat treatment and PEBD produced similar normalisation of serum bile acid levels and improvements in pruritus and sleep disruptions. Thus, odevixibat appeared to be as effective as invasive PEBD in treating serum bile acids and cholestatic pruritus in this patient.
Collapse
Affiliation(s)
- Christoph Slavetinsky
- Paediatric Gastroenterology and Hepatology, University Children's Hospital, Eberhard Karls University Tubingen, Tubingen, Germany
| | - Ekkehard Sturm
- Paediatric Gastroenterology and Hepatology, University Children's Hospital, Eberhard Karls University Tubingen, Tubingen, Germany
| |
Collapse
|
103
|
Sura SR, Germain-Lee EL. Treatment of rickets and dyslipidemia in twins with progressive familial intrahepatic cholestasis type 2. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2020; 2020:9. [PMID: 32508937 PMCID: PMC7249403 DOI: 10.1186/s13633-020-00079-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 04/24/2020] [Indexed: 12/01/2022]
Abstract
Background Progressive Familial Intrahepatic Cholestasis Type 2 (PFIC2) is a rare congenital cholestatic liver disease that progresses to end stage liver disease. It is associated with fat soluble vitamin D deficiency rickets and severe dyslipidemia; however, treatment of these secondary effects remains a challenge. Case presentation One year old twin males born to a mother with intrahepatic cholestasis during pregnancy presented with jaundice, pruritus and failure to thrive. Lab evaluation revealed significant transaminitis, direct hyperbilirubinemia and normal gamma glutamyl transferase (GGT). Genetic studies confirmed PFIC2. Further evaluation for fat soluble vitamin deficiencies revealed severe vitamin D deficiency rickets. High dose vitamin D replacement therapy using Ergocalciferol (Vitamin D2) 50,000 IU three times a week over 10 weeks led to the improvement of Vitamin D, 25-Hydroxy (25-OH) serum levels and resolution of rickets. Dyslipidemia with very low high density lipoprotein-cholesterol (HDL-C) and high triglycerides was more profound in our patients compared to what has been described in the literature thus far. The dyslipidemia improved 2 months after internal biliary diversion. Conclusions Higher doses of Vitamin D therapy are needed for treatment of rickets secondary to cholestasis. Extremely low HDL-C levels are characteristic of PFIC and improve with treatment of underlying cholestasis. Maternal intrahepatic cholestasis during pregnancy can be an early warning sign.
Collapse
Affiliation(s)
- Sunitha R Sura
- Connecticut Children's Medical Center, 505 Farmington Avenue, Farmington, CT 06032 USA.,Department of Pediatrics, University of Connecticut School of Medicine, 505 Farmington Avenue, Farmington, CT 06032 USA
| | - Emily L Germain-Lee
- Connecticut Children's Medical Center, 505 Farmington Avenue, Farmington, CT 06032 USA.,Department of Pediatrics, University of Connecticut School of Medicine, 505 Farmington Avenue, Farmington, CT 06032 USA
| |
Collapse
|
104
|
Foroutan HR, Bahador A, Ghanim SM, Dehghani SM, Anbardar MH, Fattahi MR, Forooghi M, Azh O, Tadayon A, Sherafat A, Yaghoobi AA, Ashraf MA. Effects of partial internal biliary diversion on long-term outcomes in patients with progressive familial intrahepatic cholestasis: experience in 44 patients. Pediatr Surg Int 2020; 36:603-610. [PMID: 32206891 DOI: 10.1007/s00383-020-04641-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE Progressive familial intrahepatic cholestasis (PFIC) is a hereditary disease characterized by cholestasis, which may cause jaundice, severe pruritus, and cirrhosis in the later stages. By the invention of biliary diversion methods, these patients were prevented from undergoing liver transplant. Using biliary diversion techniques, the entero-hepatic cycle was interrupted. This lowers the bile acid pool and resolves the pruritus. Herein, we report 44 cases of PFIC who underwent partial internal biliary diversion (PIBD) and long-term follow-up of these children. This comprises the largest case series of PIBD. METHODS All patients were diagnosed by liver biopsy as PFIC before the operation. All underwent cholecysto colic bypass by jejunal interposition due to severe pruritus unresponsive to medication. Laboratory blood tests, sonography, and physical exam were done before and after the operation once every 3 months. Besides, a questionnaire was designed to ask the patients about the symptoms after the operation, and a pruritus score was measured using the 5D-itch scale. RESULTS 44 children (25 boys, 19 girls), between 1.75 and 27.5 years (at the time of this study) were followed for a median period of 54 months. Age at operation ranged from 2 months to 18 years, with a median of 29 months. Of these children, 14 were lost to follow up. Results showed a significant decrease in pruritus and sleep disturbance after the surgery (p < 0.001). Also, jaundice decreased from 82.1 before to 7.1% following the surgery. 50% of the patients became medication-free at follow-up. CONCLUSION PIBD is a safe procedure which helps non-cirrhotic children preserve their liver function. Therefore, PIBD prevents them from undergoing liver transplant. Effective results were achieved in terms of severe pruritus and jaundice, and children were able to regain their sleep patterns. It also avoided external stoma, which is more convenient from the patient's point of view.
Collapse
Affiliation(s)
- Hamid Reza Foroutan
- Laparoscopy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pediatric Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran
| | - Ali Bahador
- Department of Pediatric Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran
| | - Sultan Mohsin Ghanim
- Department of Pediatric Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran
| | - Seyed Mohsen Dehghani
- Gastroenterohepatology Research Center, Department of Pediatric Gastroenterology, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Reza Fattahi
- Department of Gastroenterology, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Forooghi
- Department of Pediatric Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran
| | - Omidreza Azh
- Department of Pediatric Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran
| | - Ali Tadayon
- Department of Pediatric Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran
| | - Alireza Sherafat
- School of Medicine, University of Central Lancashire, Preston, England
| | - Amir Arsalan Yaghoobi
- Department of Pediatric Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran
| | - Mohammad Ali Ashraf
- Department of Pediatric Surgery, Nemazee Hospital, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran.
- Student Research Committee, Shiraz University of Medical Sciences, Zand St., 71 32326645, Shiraz, Iran.
| |
Collapse
|
105
|
Novel ABCB4 mutation in a Chinese female patient with progressive familial intrahepatic cholestasis type 3: a case report. Diagn Pathol 2020; 15:39. [PMID: 32321542 PMCID: PMC7175503 DOI: 10.1186/s13000-020-00955-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/08/2020] [Indexed: 02/02/2023] Open
Abstract
Background Progressive familial intrahepatic cholestasis (PFIC) is a rare group of autosomal recessive hereditary hepatic diseases. There are three types of PFIC, classified according to the mutated gene. For example, PFIC type 3 (PFIC3) is due to mutations in the ABCB4 gene (encoding multidrug-resistant protein 3 [MDR3]). Case presentation We present a 19-year-old Chinese female patient who had a 2-year history of recurrent liver dysfunction, with mainly elevated alkaline phosphatase and γ-glutamyl transpeptidase(γ-GT) levels. After excluding other causes of abnormal liver function and cholestasis, the final diagnosis of PFIC3 was confirmed by histopathological examination and gene detection. The immunohistochemical results showed no MDR3 protein expression in the bile duct membrane. Genetic sequencing analysis revealed a novel heterozygous 2137G > A; p. V713M mutation (Exon 17) and a synonymous 504C > T; p. N168N mutation (Exon 6) in ABCB4. Conclusions Our patient with long-term liver dysfunction demonstrated that elevated alkaline phosphatase and γ-GT levels should be associated with the diagnosis of PFIC3, and gene detection is the key to diagnosis. From our in silico analysis, the novel mutation p. V713M in Exon 17 was predicted to affect protein function, with a SIFT (Sorting Intolerant from Tolerant) score of 0.02, indicating a deleterious effect. Further studies are necessary to investigate the impact of the novel heterozygous 2137G > A; p. V713M mutation (Exon 17) on functional defects of MDR3 and PFIC3.
Collapse
|
106
|
Mitra S, Das A, Thapa B, Kumar Vasishta R. Phenotype-Genotype Correlation of North Indian Progressive Familial Intrahepatic Cholestasis type2 Children Shows p.Val444Ala and p.Asn591Ser Variants and Retained BSEP Expression. Fetal Pediatr Pathol 2020; 39:107-123. [PMID: 31335238 DOI: 10.1080/15513815.2019.1641860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Backgrounds and Aims: Progressive familial intrahepatic cholestasis type 2 (PFIC2) is caused by a defect or deficiency of bile salt export protein (BSEP) due to mutation in the ABCB11 gene. We intend to evaluate the phenotype-genotype correlation in 10 diagnosed cases of PFIC2 in a single tertiary care center in North India. Methods: The clinical, biochemical, histopathological, immunohistochemical, ultrastructural and genetic data of the 10 diagnosed cases of PFIC2 were recorded. Results: Icterus, pruritus and bleeding manifestations were the commonest clinical symptoms. Giant cell transformation was seen in 50% of the patients. Two predominant genetic variants were ABCB11 missense p.Val444Ala (c. 1331 T > C) and ABCB11 missense p.Asn591Ser (c. 1772 A > G) in their homozygous or compound heterozygous states and were associated with retained BSEP immunopositivity and reduced but retained BSEP immunopositivity respectively. Conclusion: Retention of BSEP is common in North Indian children of PFIC2 with no phenotype-genotype correlation.
Collapse
Affiliation(s)
| | - Ashim Das
- PGIMER, Histopathology, Chandigarh, India
| | - Baburam Thapa
- Post Graduate Institute of Medical Education and Research, Pediatric Gastroenterology, Nehru Hospital, Chandigarh, India
| | | |
Collapse
|
107
|
Mechanism-based integrated assay systems for the prediction of drug-induced liver injury. Toxicol Appl Pharmacol 2020; 394:114958. [PMID: 32198022 DOI: 10.1016/j.taap.2020.114958] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 02/29/2020] [Accepted: 03/13/2020] [Indexed: 12/18/2022]
Abstract
Drug-induced liver injury (DILI) can cause hepatic failure and result in drug withdrawal from the market. It has host-related and compound-dependent mechanisms. Preclinical prediction of DILI risk is very challenging and safety assessments based on animals inadequately forecast human DILI risk. In contrast, human-derived in vitro cell culture-based models could improve DILI risk prediction accuracy. Here, we developed and validated an innovative method to assess DILI risk associated with various compounds. Fifty-four marketed and withdrawn drugs classified as DILI risks of "most concern", "less concern", and "no concern" were tested using a combination of four assays addressing mitochondrial injury, intrahepatic lipid accumulation, inhibition of bile canalicular network formation, and bile acid accumulation. Using the inhibitory potencies of the drugs evaluated in these in vitro tests, an algorithm with the highest available DILI risk prediction power was built by artificial neural network (ANN) analysis. It had an overall forecasting accuracy of 73%. We excluded the intrahepatic lipid accumulation assay to avoid overfitting. The accuracy of the algorithm in terms of predicting DILI risks was 62% when it was constructed by ANN but only 49% when it was built by the point-added scoring method. The final algorithm based on three assays made no DILI risk prediction errors such as "most concern " instead of "no concern" and vice-versa. Our mechanistic approach may accurately predict DILI risks associated with numerous candidate drugs.
Collapse
|
108
|
Adult onset of genetic disorders in bile acid transport in the liver. Hum Pathol 2020; 96:2-7. [DOI: 10.1016/j.humpath.2019.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/09/2019] [Indexed: 12/27/2022]
|
109
|
Bekdas M, Can G, Eroz R, Duzcu SE. Combination of Novel c.3484G> T/p.Glu162Ter Variant in ABCB11 and c.208G> A/p.Asp70Asn Variant in ATP8B1 Are Associated with Severe Symptoms in Progressive Family Intrahepatic Cholestasis. J Pediatr Genet 2020; 9:285-288. [PMID: 32765934 DOI: 10.1055/s-0039-1700971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/25/2019] [Indexed: 10/25/2022]
Abstract
Progressive family intrahepatic cholestasis (PFIC) is an autosomal recessive disease that causes chronic cholestasis. It is associated with pathogenic variants in genes that encode proteins involved in bile secretion to canaliculus from hepatocytes. In this study, we present a 16-year-old boy who presented with severe pruritus and cholestatic jaundice. All possible infectious etiologies were negative. A liver biopsy was consistent with intrahepatic cholestasis and portal fibrosis. DNA was isolated from a peripheral blood sample, and whole exome sequencing was performed. A novel c.3484G > T/p.Glu162Ter variant in the ABCB11 gene and a c.208G> A/p.Asp70Asn variant in the ATP8B1 gene were detected. Despite traditional treatment, the patient's recurrent severe symptoms did not improve. The patient was referred for a liver transplantation. This novel c.3484G > T/p.Glu162Ter variant is associated with a severe and recurrent presentation, and the two compound variants could explain the severity of PFIC.
Collapse
Affiliation(s)
- Mervan Bekdas
- Department of Pediatrics, Bolu Abant Izzet Baysal University Medical Faculty, Bolu, Turkey
| | - Guray Can
- Department of Gastroenterology, Bolu Abant Izzet Baysal University Medical Faculty, Bolu, Turkey
| | - Recep Eroz
- Department of Medical Genetics, Duzce University Medical Faculty, Duzce, Turkey
| | - Selma Erdogan Duzcu
- Department of Pathology, Bolu Abant Izzet Baysal University Medical Faculty, Bolu, Turkey
| |
Collapse
|
110
|
Ticho AL, Malhotra P, Dudeja PK, Gill RK, Alrefai WA. Intestinal Absorption of Bile Acids in Health and Disease. Compr Physiol 2019; 10:21-56. [PMID: 31853951 PMCID: PMC7171925 DOI: 10.1002/cphy.c190007] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The intestinal reclamation of bile acids is crucial for the maintenance of their enterohepatic circulation. The majority of bile acids are actively absorbed via specific transport proteins that are highly expressed in the distal ileum. The uptake of bile acids by intestinal epithelial cells modulates the activation of cytosolic and membrane receptors such as the farnesoid X receptor (FXR) and G protein-coupled bile acid receptor 1 (GPBAR1), which has a profound effect on hepatic synthesis of bile acids as well as glucose and lipid metabolism. Extensive research has focused on delineating the processes of bile acid absorption and determining the contribution of dysregulated ileal signaling in the development of intestinal and hepatic disorders. For example, a decrease in the levels of the bile acid-induced ileal hormone FGF15/19 is implicated in bile acid-induced diarrhea (BAD). Conversely, the increase in bile acid absorption with subsequent overload of bile acids could be involved in the pathophysiology of liver and metabolic disorders such as fatty liver diseases and type 2 diabetes mellitus. This review article will attempt to provide a comprehensive overview of the mechanisms involved in the intestinal handling of bile acids, the pathological implications of disrupted intestinal bile acid homeostasis, and the potential therapeutic targets for the treatment of bile acid-related disorders. Published 2020. Compr Physiol 10:21-56, 2020.
Collapse
Affiliation(s)
- Alexander L. Ticho
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Pooja Malhotra
- Division of Gastroenterology & Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Pradeep K. Dudeja
- Division of Gastroenterology & Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Ravinder K. Gill
- Division of Gastroenterology & Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Waddah A. Alrefai
- Division of Gastroenterology & Hepatology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
- jesse Brown VA Medical Center, Chicago, Illinois, USA
| |
Collapse
|
111
|
Panasiti I, Briuglia S, Costa S, Caminiti L. Comorbidity between progressive familial intrahepatic cholestasis and atopic dermatitis in a 19-month-old child. BMJ Case Rep 2019; 12:12/10/e230152. [DOI: 10.1136/bcr-2019-230152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Atopic dermatitis (AD) is the most common chronic skin disease in children, with an increasing prevalence in the past three decades. Adequate treatment is prescribed for individual patient based on symptoms and disease severity. However, further underlying diagnosis should be researched when therapeutic strategies for symptoms fail and skin lesions and pruritus persist. We reported herein the case of a 19-month-old infant with a history of AD unresponsive to treatment due to the type 2 progressive familial intrahepatic cholestasis (PFIC). A new homozygous mutation of the ABCB11 gene was found. The severe pruritus, the early onset jaundice, poor growth and raised transaminase levels with normal gamma glutamyl transpeptidase have led to the suspicion of PFIC. The presence of severe AD and intrahepatic chronic cholestasis, both pruritus associated, could delay a proper diagnosis. To our knowledge, for the first time, a case of comorbidity between type 2 PFIC and AD-like disease had been described.
Collapse
|
112
|
Baier V, Cordes H, Thiel C, Castell JV, Neumann UP, Blank LM, Kuepfer L. A Physiology-Based Model of Human Bile Acid Metabolism for Predicting Bile Acid Tissue Levels After Drug Administration in Healthy Subjects and BRIC Type 2 Patients. Front Physiol 2019; 10:1192. [PMID: 31611804 PMCID: PMC6777137 DOI: 10.3389/fphys.2019.01192] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 09/03/2019] [Indexed: 12/23/2022] Open
Abstract
Drug-induced liver injury (DILI) is a matter of concern in the course of drug development and patient safety, often leading to discontinuation of drug-development programs or early withdrawal of drugs from market. Hepatocellular toxicity or impairment of bile acid (BA) metabolism, known as cholestasis, are the two clinical forms of DILI. Whole-body physiology-based modelling allows a mechanistic investigation of the physiological processes leading to cholestasis in man. Objectives of the present study were: (1) the development of a physiology-based model of the human BA metabolism, (2) population-based model validation and characterisation, and (3) the prediction and quantification of altered BA levels in special genotype subgroups and after drug administration. The developed physiology-based bile acid (PBBA) model describes the systemic BA circulation in humans and includes mechanistically relevant active and passive processes such as the hepatic synthesis, gallbladder emptying, transition through the gastrointestinal tract, reabsorption into the liver, distribution within the whole body, and excretion via urine and faeces. The kinetics of active processes were determined for the exemplary BA glycochenodeoxycholic acid (GCDCA) based on blood plasma concentration-time profiles. The robustness of our PBBA model was verified with population simulations of healthy individuals. In addition to plasma levels, the possibility to estimate BA concentrations in relevant tissues like the intracellular space of the liver enhance the mechanistic understanding of cholestasis. We analysed BA levels in various tissues of Benign Recurrent Intrahepatic Cholestasis type 2 (BRIC2) patients and our simulations suggest a higher susceptibility of BRIC2 patients toward cholestatic DILI due to BA accumulation in the liver. The effect of drugs on systemic BA levels were simulated for cyclosporine A (CsA). Our results confirmed the higher risk of DILI after CsA administration in healthy and BRIC2 patients. The presented PBBA model enhances our mechanistic understanding underlying cholestasis and drug-induced alterations of BA levels in blood and organs. The developed PBBA model might be applied in the future to anticipate potential risk of cholestasis in patients.
Collapse
Affiliation(s)
- Vanessa Baier
- Institute of Applied Microbiology (iAMB), Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany.,Department of Surgery, University Hospital Aachen, Aachen, Germany
| | - Henrik Cordes
- Institute of Applied Microbiology (iAMB), Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - Christoph Thiel
- Institute of Applied Microbiology (iAMB), Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - José V Castell
- Unit of Experimental Hepatology, IIS Hospital La Fe, Faculty of Medicine, University of Valencia and CIBEREHD, Valencia, Spain
| | - Ulf P Neumann
- Department of Surgery, University Hospital Aachen, Aachen, Germany
| | - Lars M Blank
- Institute of Applied Microbiology (iAMB), Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| | - Lars Kuepfer
- Institute of Applied Microbiology (iAMB), Aachen Biology and Biotechnology (ABBt), RWTH Aachen University, Aachen, Germany
| |
Collapse
|
113
|
Rhee ES, Kim YB, Lee S, Oh SH, Lee BH, Kim KM, Yoo HW. Novel ATP8B1 Gene Mutations in a Child with Progressive Familial Intrahepatic Cholestasis Type 1. Pediatr Gastroenterol Hepatol Nutr 2019; 22:479-486. [PMID: 31555573 PMCID: PMC6751104 DOI: 10.5223/pghn.2019.22.5.479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 08/31/2018] [Indexed: 01/26/2023] Open
Abstract
Progressive familial intrahepatic cholestasis (PFIC) is a group of severe genetic disorders, inherited in an autosomal recessive manner, causing cholestasis of hepatocellular origin, later progressing to biliary cirrhosis and liver failure. This is the first report of PFIC type 1 with novel compound heterozygous mutations in Korea. The patient was presented with intrahepatic cholestasis, a normal level of serum γ-glutamyl transferase, steatorrhea, and growth failure. Genetic testing of this patient revealed novel compound heterozygous mutations (p.Glu585Ter and p.Leu749Pro) in the ATP8B1 gene. After a liver transplantation at age 19 months, the patient developed severe post-transplant steatohepatitis.
Collapse
Affiliation(s)
- Eun Sang Rhee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Yu Bin Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Sunghee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Seak Hee Oh
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Kyung Mo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| | - Han-Wook Yoo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
114
|
Sissung TM, Huang PA, Hauke RJ, McCrea EM, Peer CJ, Barbier RH, Strope JD, Ley AM, Zhang M, Hong JA, Venzon D, Jackson JP, Brouwer KR, Grohar P, Glod J, Widemann BC, Heller T, Schrump DS, Figg WD. Severe Hepatotoxicity of Mithramycin Therapy Caused by Altered Expression of Hepatocellular Bile Transporters. Mol Pharmacol 2019; 96:158-167. [PMID: 31175181 PMCID: PMC6608607 DOI: 10.1124/mol.118.114827] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Mithramycin demonstrates preclinical anticancer activity, but its therapeutic dose is limited by the development of hepatotoxicity that remains poorly characterized. A pharmacogenomics characterization of mithramycin-induced transaminitis revealed that hepatotoxicity is associated with germline variants in genes involved in bile disposition: ABCB4 (multidrug resistance 3) rs2302387 and ABCB11 [bile salt export pump (BSEP)] rs4668115 reduce transporter expression (P < 0.05) and were associated with ≥grade 3 transaminitis developing 24 hours after the third infusion of mithramycin (25 mcg/kg, 6 hours/infusion, every day ×7, every 28 days; P < 0.0040). A similar relationship was observed in a pediatric cohort. We therefore undertook to characterize the mechanism of mithramycin-induced acute transaminitis. As mithramycin affects cellular response to bile acid treatment by altering the expression of multiple bile transporters (e.g., ABCB4, ABCB11, sodium/taurocholate cotransporting polypeptide, organic solute transporter α/β) in several cell lines [Huh7, HepaRG, HepaRG BSEP (-/-)] and primary human hepatocytes, we hypothesized that mithramycin inhibited bile-mediated activation of the farnesoid X receptor (FXR). FXR was downregulated in all hepatocyte cell lines and primary human hepatocytes (P < 0.0001), and mithramycin inhibited chenodeoxycholic acid- and GW4046-induced FXR-galactose-induced gene 4 luciferase reporter activity (P < 0.001). Mithramycin promoted glycochenodeoxycholic acid-induced cytotoxicity in ABCB11 (-/-) cells and increased the overall intracellular concentration of bile acids in primary human hepatocytes grown in sandwich culture (P < 0.01). Mithramycin is a FXR expression and FXR transactivation inhibitor that inhibits bile flow and potentiates bile-induced cellular toxicity, particularly in cells with low ABCB11 function. These results suggest that mithramycin causes hepatotoxicity through derangement of bile acid disposition; results also suggest that pharmacogenomic markers may be useful to identify patients who may tolerate higher mithramycin doses. SIGNIFICANCE STATEMENT: The present study characterizes a novel mechanism of drug-induced hepatotoxicity in which mithramycin not only alters farnesoid X receptor (FXR) and small heterodimer partner gene expression but also inhibits bile acid binding to FXR, resulting in deregulation of cellular bile homeostasis. Two novel single-nucleotide polymorphisms in bile flow transporters are associated with mithramycin-induced liver function test elevations, and the present results are the rationale for a genotype-directed clinical trial using mithramycin in patients with thoracic malignancies.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism
- Adult
- Aged
- Antibiotics, Antineoplastic/adverse effects
- Cell Line, Tumor
- Chemical and Drug Induced Liver Injury/genetics
- Chemical and Drug Induced Liver Injury/metabolism
- Clinical Trials, Phase II as Topic
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Male
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/metabolism
- Middle Aged
- Plicamycin/adverse effects
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Thoracic Neoplasms/drug therapy
- Thoracic Neoplasms/genetics
- Thoracic Neoplasms/metabolism
Collapse
Affiliation(s)
- Tristan M Sissung
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Phoebe A Huang
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Ralph J Hauke
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Edel M McCrea
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Cody J Peer
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Roberto H Barbier
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Jonathan D Strope
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Ariel M Ley
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Mary Zhang
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Julie A Hong
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - David Venzon
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Jonathan P Jackson
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Kenneth R Brouwer
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Patrick Grohar
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Jon Glod
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Brigitte C Widemann
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - Theo Heller
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - David S Schrump
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| | - William D Figg
- Clinical Pharmacology Program (T.M.S., C.J.P., W.D.F.), Molecular Pharmacology Section (P.A.H., R.J.H., E.M.M., R.H.B., J.D.S., A.M.L., W.D.F.), Biostatistics and Data Management Section (M.Z., J.A.H., D.V.), Pediatric Oncology Branch (P.G., J.G., B.C.W.), Thoracic Surgery Branch, Center for Cancer Research, National Cancer Institute (D.S.S.), and Translational Hepatology Section (T.H.), Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland; and BioIVT, ADME-Tox Division, Durham, North Carolina (J.P.J., K.R.B.)
| |
Collapse
|
115
|
Fabris L, Fiorotto R, Spirli C, Cadamuro M, Mariotti V, Perugorria MJ, Banales JM, Strazzabosco M. Pathobiology of inherited biliary diseases: a roadmap to understand acquired liver diseases. Nat Rev Gastroenterol Hepatol 2019; 16:497-511. [PMID: 31165788 PMCID: PMC6661007 DOI: 10.1038/s41575-019-0156-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bile duct epithelial cells, also known as cholangiocytes, regulate the composition of bile and its flow. Acquired, congenital and genetic dysfunctions in these cells give rise to a set of diverse and complex diseases, often of unknown aetiology, called cholangiopathies. New knowledge has been steadily acquired about genetic and congenital cholangiopathies, and this has led to a better understanding of the mechanisms of acquired cholangiopathies. This Review focuses on findings from studies on Alagille syndrome, polycystic liver diseases, fibropolycystic liver diseases (Caroli disease and congenital hepatic fibrosis) and cystic fibrosis-related liver disease. In particular, knowledge on the role of Notch signalling in biliary repair and tubulogenesis has been advanced by work on Alagille syndrome, and investigations in polycystic liver diseases have highlighted the role of primary cilia in biliary pathophysiology and the concept of biliary angiogenic signalling and its role in cyst growth and biliary repair. In fibropolycystic liver disease, research has shown that loss of fibrocystin generates a signalling cascade that increases β-catenin signalling, activates the NOD-, LRR- and pyrin domain-containing 3 inflammasome, and promotes production of IL-1β and other chemokines that attract macrophages and orchestrate the process of pericystic and portal fibrosis, which are the main mechanisms of progression in cholangiopathies. In cystic fibrosis-related liver disease, lack of cystic fibrosis transmembrane conductance regulator increases the sensitivity of epithelial Toll-like receptor 4 that sustains the secretion of nuclear factor-κB-dependent cytokines and peribiliary inflammation in response to gut-derived products, providing a model for primary sclerosing cholangitis. These signalling mechanisms may be targeted therapeutically and they offer a possibility for the development of novel treatments for acquired cholangiopathies.
Collapse
Affiliation(s)
- Luca Fabris
- Liver Center, Department of Medicine, Yale University, New Haven, CT, USA
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Romina Fiorotto
- Liver Center, Department of Medicine, Yale University, New Haven, CT, USA
| | - Carlo Spirli
- Liver Center, Department of Medicine, Yale University, New Haven, CT, USA
| | | | - Valeria Mariotti
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
- National Institute for the Study of Liver and Gastrointestinal Diseases (CIBERehd, Instituto de Salud Carlos III), Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Mario Strazzabosco
- Liver Center, Department of Medicine, Yale University, New Haven, CT, USA.
- Department of Molecular Medicine, University of Padova, Padova, Italy.
| |
Collapse
|
116
|
Takemura A, Gong S, Sekine S, Ito K. Inhibition of biliary network reconstruction by benzbromarone delays recovery from pre-existing liver injury. Toxicology 2019; 423:32-41. [DOI: 10.1016/j.tox.2019.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 04/09/2019] [Accepted: 05/13/2019] [Indexed: 01/23/2023]
|
117
|
Vitale G, Gitto S, Vukotic R, Raimondi F, Andreone P. Familial intrahepatic cholestasis: New and wide perspectives. Dig Liver Dis 2019; 51:922-933. [PMID: 31105019 DOI: 10.1016/j.dld.2019.04.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/01/2019] [Accepted: 04/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Progressive familial intrahepatic cholestasis (PFIC) includes autosomal recessive cholestatic rare diseases of childhood. AIMS To update the panel of single genes mutations involved in familial cholestasis. METHODS PubMed search for "familial intrahepatic cholestasis" alone as well as in combination with other key words was performed considering primarily original studies and meta-analyses. RESULTS PFIC1 involves ATP8B1 gene encoding for aminophospholipid flippase FIC1. PFIC2 includes ABCB11 gene, encoding for protein functioning as bile salt export pump. PFIC3 is due to mutations of ABCB4 gene responsible for the synthesis of class III multidrug resistance P-glycoprotein flippase. PFIC4 and PFIC5 involve tight junction protein-2 gene and NR1H4 gene encoding for farnesoid X receptor. Benign Intrahepatic Cholestasis, Intrahepatic Cholestasis of Pregnancy and Low-phospholipid-associated cholelithiasis involve the same genes and are characterized by intermittent attacks of cholestasis, no progression to cirrhosis, reversible pregnancy-specific cholestasis and cholelithiasis in young people. Blood and liver tissue levels of bile-excreted drugs can be influenced by the presence of mutations in PFIC genes, causing drug-induced cholestasis. Mutations in PFIC genes might increase the risk of liver cancer. CONCLUSION There is a high proportion of unexplained cholestasis potentially caused by specific genetic pathophysiologic pathways. The use of next generation sequencing and whole-exome sequencing could improve the diagnostic process in this setting.
Collapse
Affiliation(s)
- Giovanni Vitale
- ITEC Unit, Department of Medical and Surgical Sciences, University of Bologna, Italy; Research Centre for the Study of Hepatitis, University of Bologna, Italy; End-stage Liver Disease Unit, Department of Medical and Surgical Sciences, Sant'Orsola-Malpighi Hospital, University of Bologna, Italy
| | - Stefano Gitto
- ITEC Unit, Department of Medical and Surgical Sciences, University of Bologna, Italy; Research Centre for the Study of Hepatitis, University of Bologna, Italy
| | - Ranka Vukotic
- ITEC Unit, Department of Medical and Surgical Sciences, University of Bologna, Italy; Research Centre for the Study of Hepatitis, University of Bologna, Italy
| | - Francesco Raimondi
- Bioquant Institute, Heidelberg University, Germany; Heidelberg University Biochemistry Center (BZH), Germany
| | - Pietro Andreone
- ITEC Unit, Department of Medical and Surgical Sciences, University of Bologna, Italy; Research Centre for the Study of Hepatitis, University of Bologna, Italy.
| |
Collapse
|
118
|
Henkel SAF, Squires JH, Ayers M, Ganoza A, Mckiernan P, Squires JE. Expanding etiology of progressive familial intrahepatic cholestasis. World J Hepatol 2019; 11:450-463. [PMID: 31183005 PMCID: PMC6547292 DOI: 10.4254/wjh.v11.i5.450] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 04/19/2019] [Accepted: 04/27/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Progressive familial intrahepatic cholestasis (PFIC) refers to a disparate group of autosomal recessive disorders that are linked by the inability to appropriately form and excrete bile from hepatocytes, resulting in a hepatocellular form of cholestasis. While the diagnosis of such disorders had historically been based on pattern recognition of unremitting cholestasis without other identified molecular or anatomic cause, recent scientific advancements have uncovered multiple specific responsible proteins. The variety of identified defects has resulted in an ever-broadening phenotypic spectrum, ranging from traditional benign recurrent jaundice to progressive cholestasis and end-stage liver disease.
AIM To review current data on defects in bile acid homeostasis, explore the expanding knowledge base of genetic based diseases in this field, and report disease characteristics and management.
METHODS We conducted a systemic review according to PRISMA guidelines. We performed a Medline/PubMed search in February-March 2019 for relevant articles relating to the understanding, diagnosis, and management of bile acid homeostasis with a focus on the family of diseases collectively known as PFIC. English only articles were accessed in full. The manual search included references of retrieved articles. We extracted data on disease characteristics, associations with other diseases, and treatment. Data was summarized and presented in text, figure, and table format.
RESULTS Genetic-based liver disease resulting in the inability to properly form and secrete bile constitute an important cause of morbidity and mortality in children and increasingly in adults. A growing number of PFIC have been described based on an expanded understanding of biliary transport mechanism defects and the development of a common phenotype.
CONCLUSION We present a summary of current advances made in a number of areas relevant to both the classically described FIC1 (ATP8B1), BSEP (ABCB11), and MDR3 (ABCB4) transporter deficiencies, as well as more recently described gene mutations -- TJP2 (TJP2), FXR (NR1H4), MYO5B (MYO5B), and others which expand the etiology and understanding of PFIC-related cholestatic diseases and bile transport.
Collapse
Affiliation(s)
- Sarah AF Henkel
- Division of Gastroenterology, Hepatology, and Nutrition, Emory School of Medicine, Atlanta, GA 30322, United States
| | - Judy H Squires
- Department of Radiology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, United States
| | - Mary Ayers
- Division of Gastroenterology, Hepatology, and Nutrition, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, United States
| | - Armando Ganoza
- Division of Pediatric Transplantation, Department of Surgery, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, United States
| | - Patrick Mckiernan
- Division of Gastroenterology, Hepatology, and Nutrition, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, United States
| | - James E Squires
- Division of Gastroenterology, Hepatology, and Nutrition, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, United States
| |
Collapse
|
119
|
Predisposing Conditions to Pediatric Hepatocellular Carcinoma and Association With Outcomes: Single-center Experience. J Pediatr Gastroenterol Nutr 2019; 68:695-699. [PMID: 30676520 DOI: 10.1097/mpg.0000000000002285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Hepatocellular carcinoma (HCC) has been linked to chronic viral or metabolic liver disease and other conditions. The characteristics of children with HCC have not been fully elucidated and outcomes in children with predisposing liver disease are not well defined. METHODS Patients ≤21 years old with HCC managed at our institution and through external consultation between 1996 and 2016 were included. Demographics, clinical history, and pathology were tabulated. Fisher exact test and Wilcoxon test were employed for subgroup comparison, and survival differences were evaluated by Kaplan-Meier method. RESULTS Sixty-one cases of HCC were identified. Seven of 16 patients (44%) at our institution and 18 of 45 consult patients (40%) had a predisposing condition: cryptogenic cirrhosis/steatosis (9), genetic (7), biliary pathology (4), viral hepatitis (1), and other (4). Thirteen of 27 patients with de novo HCC had fibrolamellar HCC. Clinical characteristics were grouped by presence or absence of predisposing conditions: age at diagnosis (7.2 vs 10.2 years, P < 0.05), metastatic disease at presentation (15% vs 44%, P = n.s), and tumor size >4 cm (20% vs 100%, P < 0.05). In patients treated at our institution, 5 of 7 with predisposing conditions received liver transplant and achieved complete remission, whereas only 3 of 9 patients with de novo HCC received curative surgery and this group had decreased median overall survival (P < 0.05). CONCLUSIONS The majority of children with HCC did not have predisposing liver or associated disease. These patients were diagnosed later with more advanced stage disease and had significantly decreased overall survival.
Collapse
|
120
|
Zhang BP, Huang ZH, Dong C. Biliary atresia combined with progressive familial intrahepatic cholestasis type 3: A case report and review of the literature. Medicine (Baltimore) 2019; 98:e15593. [PMID: 31083246 PMCID: PMC6531222 DOI: 10.1097/md.0000000000015593] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/04/2019] [Accepted: 04/16/2019] [Indexed: 01/26/2023] Open
Abstract
RATIONALE Neonatal cholestasis is one of the most serious diseases in infancy. Progressive familial intrahepatic cholestasis (PFIC) is a disease that leads to intrahepatic cholestasis. It is one of the common causes of neonatal cholestasis in addition to biliary atresia (BA). The differential diagnosis of neonatal cholestasis is clinically challenging for pediatricians. PATIENT CONCERNS A 4-month-old female presented with severe jaundice, pruritus, and pale stool for 20 days. Abnormally strong echoes near the portal area, an abnormally small gallbladder with an irregularly stiff wall, and splenomegaly were identified on abdominal ultrasound. Blood tests showed elevated alanine aminotransferase, total bilirubin, conjugated bilirubin, gamma-glutamyltranspeptidase, and total bile acid levels. DIAGNOSIS Intraoperative cholangiography showed BA. ABCB4 gene mutation IVS13+6G>A/G was confirmed by genetic testing. The patient was diagnosed with BA combined with PFIC3. INTERVENTIONS Kasai portoenterostomy and ursodeoxycholic acid were used for treatment. OUTCOMES Her clinical symptoms and blood tests improved gradually. No recurrence was noted during 1 year of follow-up. LESSONS Additional examinations, such as genetic testing, should be considered in patients with BA who had refractory jaundice after Kasai portoenterostomy in order to exclude intrahepatic cholestasis.
Collapse
Affiliation(s)
- Ben-Ping Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | |
Collapse
|
121
|
Abstract
Bile acids facilitate nutrient absorption and are endogenous ligands for nuclear receptors that regulate lipid and energy metabolism. The brain-gut-liver axis plays an essential role in maintaining overall glucose, bile acid, and immune homeostasis. Fasting and feeding transitions alter nutrient content in the gut, which influences bile acid composition and pool size. In turn, bile acid signaling controls lipid and glucose use and protection against inflammation. Altered bile acid metabolism resulting from gene mutations, high-fat diets, alcohol, or circadian disruption can contribute to cholestatic and inflammatory diseases, diabetes, and obesity. Bile acids and their derivatives are valuable therapeutic agents for treating these inflammatory metabolic diseases.
Collapse
Affiliation(s)
- John Y L Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio 44272;
| | - Jessica M Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio 44272;
| |
Collapse
|
122
|
Baker A, Kerkar N, Todorova L, Kamath BM, Houwen RHJ. Systematic review of progressive familial intrahepatic cholestasis. Clin Res Hepatol Gastroenterol 2019; 43:20-36. [PMID: 30236549 DOI: 10.1016/j.clinre.2018.07.010] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 06/29/2018] [Accepted: 07/23/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS Progressive familial intrahepatic cholestasis (PFIC) is a heterogeneous group of rare genetic disorders associated with bile acid secretion or transport defects. This is the first systematic review of the epidemiology, natural history and burden of PFIC. METHODS MEDLINE and Embase were searched for publications on PFIC prevalence, incidence or natural history, and the economic burden or health-related quality of life (HRQoL) of patients with PFIC. Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed. RESULTS Of 1269 records screened, 20 were eligible (epidemiology, 17; humanistic burden, 5; both, 2). Incidence of intrahepatic cholestasis, including but not limited to PFIC, was 1/18 000 live births in one study that did not use genetic testing. In two studies of infants and children (2-18 years) with cholestasis, 12-13% had genetically diagnosed PFIC. Of the three main PFIC subtypes, PFIC2 was the most common (21-91% of patients). Common symptoms (e.g. pruritus, jaundice, hepatomegaly, splenomegaly) generally appeared at about 3 months of age and tended to emerge earliest in patients with PFIC2. Patients reported that pruritus was often severe and led to dermal damage and reduced HRQoL. Disease progression led to complications including liver failure and hepatocellular carcinoma, with 20-83% of patients requiring liver transplantation. Mortality was 0-87% across 10 studies (treatment varied among studies), with a median age at death of ~4 years in one study. CONCLUSIONS Patients with PFIC face debilitating symptoms and poor prognosis. Further research is needed to inform patient management and clinical trial design. Published data on the epidemiology and socioeconomic burden of PFIC is limited.
Collapse
Affiliation(s)
- Alastair Baker
- Paediatric Liver Centre, King's College Hospital, London, UK
| | - Nanda Kerkar
- Division of Gastroenterology, Hepatology and Nutrition, Golisano Children's Hospital, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Binita M Kamath
- Division of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Roderick H J Houwen
- Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center, Utrecht, Netherlands.
| |
Collapse
|
123
|
M. Z, S.M. D, F. E, M.R. F, M. M, S.M.B. T. In-silico Evaluation of Rare Codons and their Positions in the Structure of ATP8b1 Gene. J Biomed Phys Eng 2019; 9:105-120. [PMID: 30881940 PMCID: PMC6409373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/18/2016] [Indexed: 11/30/2022]
Abstract
BACKGROUND Progressive familial intrahepatic cholestases (PFIC) are a spectrum of autosomal progressive liver diseases developing to end-stage liver disease. ATP8B1 deficiency caused by mutations in ATP8B1 gene encoding a P-type ATPase leads to PFIC1. The gene for PFIC1 has been mapped on a 19-cM region of 18q21-q22, and a gene defect in ATP8B1 can cause deregulations in bile salt transporters through decreased expression and/or activity of FXR. Point mutations are the most common, with the majority being missense or nonsense mutations. In addition, approximately 15% of disease-causing ATP8B1 mutations are annotated as splicing disrupting alteration given that they are located at exon-intron borders. OBJECTIVE Here, we describe the hidden layer of computational biology information of rare codons in ATP8B1, which can help us for drug design. METHODS Some rare codons in different locations of ATP8b1 gene were identified using several web servers and by in-silico modelling of ATP8b1 in Phyre2 and I-TASSER server, some rare codons were evaluated. RESULTS Some of these rare codons were located at special positions which seem to have a critical role in proper folding of ATP8b1 protein. Structural analysis showed that some of rare codons are related to mutations in ATP8B1 that are responsible for PFIC1 disease, which may have a critical role in ensuring the correct folding. CONCLUSION Investigation of such hidden information can enhance our understanding of ATP8b1 folding. Moreover, studies of these rare codons help us to clarify their role in rational design of new and effective drugs.
Collapse
Affiliation(s)
- Zarenezhad M.
- MD, PhD, Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
,MD, PhD, legal medicine research center, legal medicine organization, Tehran , iran
| | - Dehghani S.M.
- MD, Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ejtehadi F.
- MD, Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fattahi M.R.
- MD, Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mortazavi M.
- PhD, Department of Biotechnology, Institute of Science and High Technology and Environmental Science, Graduate University of Advanced Technology, Kerman, Iran
| | - Tabei S.M.B.
- MD, Genetic Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
124
|
Zhou J, Yao Y, Huang X, Chen D, Zhang T, Zhang Y. Association between bile salt export pump polymorphisms and intrahepatic cholestasis of pregnancy susceptibility: a meta-analysis of case-control studies. Gynecol Endocrinol 2019; 35:179-183. [PMID: 30614300 DOI: 10.1080/09513590.2018.1512570] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The purpose of this meta-analysis was to investigate the relationship between bile salt export pump (BSEP) polymorphisms and intrahepatic cholestasis of pregnancy (ICP) susceptibility. Retrieved studies from Pubmed, EMBASE, Web of Science, Cochrane Library and CBM databases about BSEP polymorphisms and ICP susceptibility were included. Odds ratio (OR) and 95% confidence interval (CI) and publication bias were calculated. Ten related case-control studies on BSEP polymorphisms and ICP susceptibility were included. The pooled results showed a significant association between BSEP rs2287622 polymorphism and ICP risk in Asian population (OR >1, p < .01 for A vs. a and AA vs. Aa/aa) and general population (OR >1, p < .05 for A vs. a, Aa vs. aa, AA/Aa vs. aa), and a borderline statistical significance was found between BSEP rs473351 polymorphism and ICP susceptibility (OR = 1.66, p < .05), and no statistical significance was found in D482G or rs853782 polymorphisms and ICP risk (all p > .05). Additionally, no publication bias was found in these studies (all p > .05). Our current meta-analysis indicated that BSEP rs2287622 polymorphism could increase the susceptibility of ICP in Asians and in general populations, while rs473351, D482G, and rs853782 polymorphisms were not obviously associated with ICP risk, but it needs further larger study with ethnicity and various etiologies.
Collapse
Affiliation(s)
- Jie Zhou
- a Department of Pharmacy , Wuxi Maternal and Child Health Hospital , Wuxi , Jiangsu Province , China
| | - Ying Yao
- a Department of Pharmacy , Wuxi Maternal and Child Health Hospital , Wuxi , Jiangsu Province , China
| | - Xiaoping Huang
- a Department of Pharmacy , Wuxi Maternal and Child Health Hospital , Wuxi , Jiangsu Province , China
| | - Daozhen Chen
- a Department of Pharmacy , Wuxi Maternal and Child Health Hospital , Wuxi , Jiangsu Province , China
| | - Ting Zhang
- a Department of Pharmacy , Wuxi Maternal and Child Health Hospital , Wuxi , Jiangsu Province , China
| | - Yan Zhang
- a Department of Pharmacy , Wuxi Maternal and Child Health Hospital , Wuxi , Jiangsu Province , China
| |
Collapse
|
125
|
Ge T, Zhang X, Xiao Y, Wang Y, Zhang T. Novel compound heterozygote mutations of TJP2 in a Chinese child with progressive cholestatic liver disease. BMC MEDICAL GENETICS 2019; 20:18. [PMID: 30658709 PMCID: PMC6339326 DOI: 10.1186/s12881-019-0753-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/11/2019] [Indexed: 12/14/2022]
Abstract
Background Progressive familial intrahepatic cholestasis (PFIC) is a group of genetic autosomal recessive disorders that predominantly affects young children and results in early-onset progressive liver damage. Several types of PFIC were defined based on different genetic aetiologies in last decades. Case presentation Here, we report a Chinese young child diagnosed as PFIC with variants in tight junction protein 2 (TJP2). The patient was affected by a long history of jaundice, pruritus, and failure to thrive. Highly elevated level of serum total bile acid (TBA) and normal levels of gamma-glutamyltransferase (GGT) were observed at hospitalization. The patient’s clinical symptoms could be alleviated by administration of ursodeoxycholic acid. Genetic testing by next generation sequencing (NGS) found novel compound heterozygote mutations c.2448 + 1G > C/c.2639delC (p.T880Sfs*12) in TJP2, which were inherited from her mother and father, respectively. Both mutations were predicted to abolish TJP2 protein translation, and neither has previously been identified. Conclusion We report a Chinese female PFIC child with novel compound heterozygous mutations of TJP2. Genetic testing by NGS is valuable in the clinical diagnosis of hereditary liver disease. Electronic supplementary material The online version of this article (10.1186/s12881-019-0753-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ting Ge
- Department of Gastroenterology, Hepatology, and Nutrition, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Xinyue Zhang
- Department of Gastroenterology, Hepatology, and Nutrition, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yongmei Xiao
- Department of Gastroenterology, Hepatology, and Nutrition, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yizhong Wang
- Department of Gastroenterology, Hepatology, and Nutrition, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China.
| | - Ting Zhang
- Department of Gastroenterology, Hepatology, and Nutrition, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200062, China.
| |
Collapse
|
126
|
Lipiński P, Jankowska I. [Progressive familial intrahepatic cholestasis type 3]. DEVELOPMENTAL PERIOD MEDICINE 2019; 22. [PMID: 30636238 PMCID: PMC8522827 DOI: 10.34763/devperiodmed.20182204.385389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Progressive familial intrahepatic cholestasis is caused by mutations in the ABCB4 gene and belongs to the family of familial intrahepatic cholestais disorders inherited in an autosomal recessive pattern. To date, about 200 patients with various hepatobiliary disorders associated with ABCB4 gene mutations have been described in the literature. The aim of this manuscript was to describe the pathogenesis, clinical presentation, diagnostic process and treatment of progressive familial intrahepatic cholestais type 3, based on the literature review.
Collapse
Affiliation(s)
- Patryk Lipiński
- Klinika Gastroenterologii, Hepatologii, Zaburzeń Odżywiania i Pediatrii, Instytut „Pomnik – Centrum Zdrowia Dziecka”, Warszawa, Polska,Patryk Lipiński Klinika Gastroenterologii, Hepatologii, Zaburzeń Odżywiania i Pediatrii, Instytut „Pomnik − Centrum Zdrowia Dziecka” Al. Dzieci Polskich 20, 04-730 Warszawa
| | - Irena Jankowska
- Klinika Gastroenterologii, Hepatologii, Zaburzeń Odżywiania i Pediatrii, Instytut „Pomnik – Centrum Zdrowia Dziecka”, Warszawa, Polska
| |
Collapse
|
127
|
Chandler AM, Center SA, Randolph JF, Davignon DL, McDonough SP, Warner KL. Reference limits for hepatic bile duct-to-arteriole and bile duct-to-portal tract ratios in healthy cats. Am J Vet Res 2019; 80:15-23. [PMID: 30605040 DOI: 10.2460/ajvr.80.1.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To establish reference limits for hepatic bile duct-to-arteriole ratio (BD:A) and bile duct-to-portal tract ratio (BD:PT) in healthy cats and assess whether these parameters could be used to support a diagnosis of biliary ductopenia in cats. SAMPLE Hepatic biopsy samples from healthy cats (n = 20) and cats with ductopenia (2). PROCEDURES Hepatic biopsy samples from healthy cats were used to count the number of bile ducts and hepatic arterioles in 20 portal tracts for each cat. Mean BD:A and mean BD:PT for each cat were calculated, and these values were used to determine reference limits for mean BD:A and mean BD:PT. Results of histologic evaluation, including immunohistochemical staining in some instances, were compared for healthy cats versus cats with ductopenia. RESULTS Of the 400 portal tracts from healthy cats, 382 (95.5%) and 396 (99.0%) had BD:A and BD:PT, respectively, ≥ 1.0, with less variability in BD:A. Mean BD:A and BD:PT were markedly lower in both cats with ductopenia, compared with values for healthy cats. However, only mean BD:A for cats with ductopenia was below the reference limit of 0.59. CONCLUSIONS AND CLINICAL RELEVANCE Results indicated that systematic evaluation of BD:A, with a lower reference limit of 0.59 to define biliary ductopenia in cats, may be a discrete and easily applied morphometric tool to enhance detection of ductopenia in cats. However, application of this ratio required evaluation of ≥ 20 portal tracts with cross-sectioned portal elements to determine a mean BD:A value.
Collapse
|
128
|
Milyukov VE, Sharifova KM. [Multiple organ manifestations of liver failure in acute small bowel obstruction]. Khirurgiia (Mosk) 2019:73-79. [PMID: 31532170 DOI: 10.17116/hirurgia201909173] [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] [Indexed: 06/10/2023]
Abstract
Modern surgical and intensive care achievements did not result decrease of mortality in patients with acute small bowel obstruction (ASBO). Direct cause of mortality in ASBO patients is peritonitis followed by multiple organ failure syndrome including acute failure of liver, kidneys, lungs and other organs. Publications devoted to the problem of acute liver failure and its complicationsare reviewed in the article. Mechanisms of pathogenesis and clinical manifestations of multiple organ failure syndrome in patients with acute small bowel obstruction are analyzed.
Collapse
Affiliation(s)
- V E Milyukov
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russia, Moscow, Russia
| | - Kh M Sharifova
- Sechenov First Moscow State Medical University of the Ministry of Health of the Russia, Moscow, Russia
| |
Collapse
|
129
|
Saha H, Tapanjyoti G, Biswas S, Mishra PK, Basu KS, Chatterjee U. Two Cases of Progressive Familial Intrahepatic Cholestasis Type 2: Role of Surgery with Brief Review of Literature. J Indian Assoc Pediatr Surg 2019; 24:75-77. [PMID: 30686894 PMCID: PMC6322173 DOI: 10.4103/jiaps.jiaps_235_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Progressive familial intrahepatic cholestasis (PFIC) is a rare bile acid transporter defect and autosomal recessive disorder with type 2 being the most common type. Partial internal or external biliary diversion delays its progression to end-stage liver disease. Here, we discuss two cases of type 2 PFIC.
Collapse
Affiliation(s)
- Hinglaj Saha
- Department of Paediatric Surgery, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Ghosh Tapanjyoti
- Department of Paediatric Surgery, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Somak Biswas
- Department of Paediatric Surgery, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Prafulla Kumar Mishra
- Department of Paediatric Surgery, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Kalyani Saha Basu
- Department of Paediatric Surgery, Nil Ratan Sircar Medical College and Hospital, Kolkata, West Bengal, India
| | - Uttara Chatterjee
- Department of Pathology, IPGMER Medical College and Hospital, Kolkata, West Bengal, India
| |
Collapse
|
130
|
Kim KH, Choi JM, Li F, Dong B, Wooton-Kee CR, Arizpe A, Anakk S, Jung SY, Hartig SM, Moore DD. Constitutive Androstane Receptor Differentially Regulates Bile Acid Homeostasis in Mouse Models of Intrahepatic Cholestasis. Hepatol Commun 2018; 3:147-159. [PMID: 30620001 PMCID: PMC6312660 DOI: 10.1002/hep4.1274] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022] Open
Abstract
Bile acid (BA) homeostasis is tightly regulated by multiple transcription factors, including farnesoid X receptor (FXR) and small heterodimer partner (SHP). We previously reported that loss of the FXR/SHP axis causes severe intrahepatic cholestasis, similar to human progressive familial intrahepatic cholestasis type 5 (PFIC5). In this study, we found that constitutive androstane receptor (CAR) is endogenously activated in Fxr:Shp double knockout (DKO) mice. To test the hypothesis that CAR activation protects DKO mice from further liver damage, we generated Fxr;Shp;Car triple knockout (TKO) mice. In TKO mice, residual adenosine triphosphate (ATP) binding cassette, subfamily B member 11 (ABCB11; alias bile salt export pump [BSEP]) function and fecal BA excretion are completely impaired, resulting in severe hepatic and biliary damage due to excess BA overload. In addition, we discovered that pharmacologic CAR activation has different effects on intrahepatic cholestasis of different etiologies. In DKO mice, CAR agonist 1,4‐bis[2‐(3,5‐dichloropyridyloxy)]benzene (TCPOBOP; here on TC) treatment attenuated cholestatic liver injury, as expected. However, in the PFIC2 model Bsep knockout (BKO) mice, TC treatment exhibited opposite effects that reflect increased BA accumulation and liver injury. These contrasting results may be linked to differential regulation of systemic cholesterol homeostasis in DKO and BKO livers. TC treatment selectively up‐regulated hepatic cholesterol levels in BKO mice, supporting de novo BA synthesis. Conclusion: CAR activation in DKO mice is generally protective against cholestatic liver injury in these mice, which model PFIC5, but not in the PFIC2 model BKO mice. Our results emphasize the importance of the genetic and physiologic background when implementing targeted therapies to treat intrahepatic cholestasis.
Collapse
Affiliation(s)
- Kang Ho Kim
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX
| | - Jong Min Choi
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX
| | - Feng Li
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX.,Center for Drug Discovery Baylor College of Medicine Houston TX
| | - Bingning Dong
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX
| | | | - Armando Arizpe
- School of Natural Science University of Texas Austin Austin TX
| | - Sayeepriyadarshini Anakk
- Department of Molecular and Integrative Physiology University of Illinois at Urbana-Champaign Urbana IL
| | - Sung Yun Jung
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX.,Verna and Marrs McLean Department of Biochemistry and Molecular Biology Baylor College of Medicine Houston TX
| | - Sean M Hartig
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine Baylor College of Medicine Houston TX
| | - David D Moore
- Department of Molecular and Cellular Biology Baylor College of Medicine Houston TX
| |
Collapse
|
131
|
Chen L, Xiao H, Ren XH, Li L. Long-term outcomes after cholecystocolostomy for progressive familial intrahepatic cholestasis. Hepatol Res 2018; 48:1163-1171. [PMID: 29934967 DOI: 10.1111/hepr.13222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/26/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022]
Abstract
AIM To evaluate the long-term efficacy of cholecystocolostomy surgery for progressive familial intrahepatic cholestasis (PFIC). METHODS From August 2003 to November 2014, 34 clinically diagnosed children, including 11 with familial intrahepatic cholestasis-1 (FIC1), 13 with bile salt export pump (BSEP) disease, five with low γ-glutamyl transpeptidase (GGT) disease (levels <100 U/L), and five with multidrug resistance class III (MDR3) disease with high GGT (>100 U/L), were identified in our center. Data were collected retrospectively from individuals who collectively had 36 surgical operations and two orthotopic liver transplantations (OLT). RESULTS Serum total bilirubin (0 = 163.54 ± 106.02, 36 months = 23.38 ± 17.66 μmol/L) and bile acid (0 = 325.83 ± 153.09, 36 months = 48.36 ± 79.71 μmol/L) decreased after cholecystocolostomy in PFIC patients (P < 0.001). All patients experienced decreased severity of pruritus (88.2% vs. 16.1%, P < 0.001) and a greater freedom from growth retardation after cholecystocolostomy (-3.35 vs. -1.03, P < 0.001). Defecation frequency increased in PFIC patients after cholecystocolostomy (P = 0.002). Four patients (three with FIC1 and one with BSEP) experienced recurrence of cholestasis and two underwent reoperation. Two BSEP patients underwent OLT. One patient with BSEP and one patient with MDR3 died due to severe diarrhea and dehydration; one BSEP patient died of intractable constipation. CONCLUSIONS This is the first long-term, large-scale analysis of cholecystocolostomy approaches for PFIC. Approaches single and well tolerated, and generally result in improvement of pruritus and cholestasis.
Collapse
Affiliation(s)
- Long Chen
- Department of Pediatric Surgery, Capital Institute of Pediatrics, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Hui Xiao
- Department of Pediatric Surgery, Capital Institute of Pediatrics, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiang-Hai Ren
- Department of Pediatric Surgery, Capital Institute of Pediatrics, Beijing, China.,Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Long Li
- Department of Pediatric Surgery, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
132
|
Abstract
Pediatric cholestasis often results from mechanical obstruction of the biliary tract or dysfunction in the processes of forming and excreting bile. Various genetic defects with resulting molecular inaccuracies are increasingly being recognized, often with specific clinical characteristics. Identifying of the molecular abnormality can enable implementation of timely, appropriate treatment in some affected individuals and provide prognostic indicators for both families and care teams.
Collapse
Affiliation(s)
- James E Squires
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Pittsburgh, One Children's Hospital Drive, 6th Floor FP, 4401 Penn Avenue, Pittsburgh, PA 15224, USA.
| | - Patrick McKiernan
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Pittsburgh, One Children's Hospital Drive, 6th Floor FP, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| |
Collapse
|
133
|
18β-Glycyrrhetinic acid protects against alpha-naphthylisothiocyanate-induced cholestasis through activation of the Sirt1/FXR signaling pathway. Acta Pharmacol Sin 2018; 39:1865-1873. [PMID: 30061734 DOI: 10.1038/s41401-018-0110-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/08/2018] [Indexed: 02/06/2023]
Abstract
Cholestasis is a common feature of liver injury, which manifests as bile acid excretion and/or enterohepatic circulation disorders. However, very few effective therapies exist for cholestasis. Recently, 18β-Glycyrrhetinic acid (18b-GA), a major metabolic component of glycyrrhizin, which is the main ingredient of licorice, was reported to protect against alpha-naphthylisothiocyanate (ANIT)-induced cholestasis. However, its protective mechanism remains unclear. We hypothesized that 18b-GA may stimulate the signaling pathway of bile acid (BA) transportation in hepatocytes, resulting its hepatoprotective effect. According to the results, 18b-GA markedly attenuated ANIT-induced liver injury as indicated the hepatic plasma chemistry index and histopathology examination. In addition, the expression levels of nuclear factors, including Sirt1, FXR and Nrf2, and their target efflux transporters in the liver, which mainly mediate bile acid homeostasis in hepatocytes, significantly increased. Furthermore, we first revealed that 18b-GA treatment significantly activated FXR, and which can be significantly reduced by EX-527 (a potent and selective Sirt1 inhibitor), indicating that 18b-GA activates FXR through Sirt1. Taken together, 18b-GA confers hepatoprotection against ANIT-induced cholestasis by activating FXR through Sirt1, which promotes gene expression of the efflux transporter, and consequently attenuates dysregulation of bile acid homeostasis in hepatocyte compartments.
Collapse
|
134
|
Development of a Novel Tool to Assess the Impact of Itching in Pediatric Cholestasis. PATIENT-PATIENT CENTERED OUTCOMES RESEARCH 2018; 11:69-82. [PMID: 28710680 PMCID: PMC5766715 DOI: 10.1007/s40271-017-0266-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
OBJECTIVES The aim was to develop a clinical outcome assessment (COA) for itching in children with cholestatic pruritus. METHODS This prospective study aimed to enroll patients aged 4-30 years with Alagille syndrome (ALGS) or progressive familial intrahepatic cholestasis type 1 and caregivers of patients aged 5 months to 14 years. Eligible patients experienced itching during ≥3 of the 7 days before enrollment and had not undergone liver transplant or surgical interruption of the enterohepatic circulation. Open-ended qualitative interviews confirmed that itching was a primary concern for patients and caregivers. Diaries were modified and then evaluated by participants during cognitive debriefing. Interview results were reviewed by clinical, COA and statistical experts. Diary questions were revised following an interim analysis before finalizing the Itch Reported Outcome (ItchRO). RESULTS Thirty-six interviews were analyzed, representing 25 families of patients with ALGS. Itching was reported spontaneously (without prompting by the interviewer) by ten of 12 patients with ALGS and 19 of 20 caregivers. Consequences of itching included skin damage (78%), mood changes (59%), and difficulties staying asleep (59%) or falling asleep (53%). Two versions of the ItchRO were developed: ItchRO(Patient) for self-completion by patients and ItchRO(Observer) for caregivers. The ItchRO diaries comprise a single scorable item to assess itch and are to be completed twice daily (morning and evening). CONCLUSIONS Itching was the most bothersome ALGS symptom reported by study participants. We have developed the ItchRO(Patient) and ItchRO(Observer) to assess itching in children with ALGS and other cholestatic liver diseases. These diaries are being validated for use in clinical trials.
Collapse
|
135
|
Abstract
Progressive familial intrahepatic cholestasis (PFIC) is a group of autosomal recessive cholestatic liver diseases which are subgrouped according to the genetic defect, clinical presentation, laboratory findings and liver histology. Progressive liver fibrosis, cirrhosis, and end stage liver disease (ESLD) may eventually develop. PFIC was first described in Amish descendants of Jacob Byler, therefore it was originally called Byler disease. But it can be seen anywhere on the globe. This review summarizes the main features of the subtypes of the disease and discusses the current available diagnosis, conservative and surgical therapeutic options.
Collapse
Affiliation(s)
- Mithat Gunaydin
- Avicenna Hospital, Department of Pediatric Surgery, Istanbul, Turkey,
| | | |
Collapse
|
136
|
Min J, Chen H, Gong Z, Liu X, Wu T, Li W, Fang J, Huang T, Zhang Y, Zhao W, Zhu C, Wang Q, Mi S, Wang N. Pharmacokinetic and Pharmacodynamic Properties of Rosmarinic Acid in Rat Cholestatic Liver Injury. Molecules 2018; 23:E2287. [PMID: 30205454 PMCID: PMC6225135 DOI: 10.3390/molecules23092287] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/18/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022] Open
Abstract
The objective of this study was to evaluate the hepatoprotective and metabolic effects of rosmarinic acid (RA) in rats. RA [100 mg/kg body weight (BW)] was intragastrically (i.g.) administered to Sprague-Dawley (SD) rats once a day for seven consecutive days. The rats were then i.g. administered α-naphthylisothiocyanate (ANIT) (80 mg/kg once on the 5th day) to induce acute intrahepatic cholestasis after the last administration of RA. Blood samples were collected at different time points (0.083 h, 0.17 h, 0.33 h, 0.5 h, 0.75 h, 1 h, 1.5 h, 3 h, 4 h, 6 h, 8 h, 12 h, 20 h) after administration, and the levels of RA were estimated by HPLC. Plasma and bile biochemical analysis, bile flow rate, and liver histopathology were measured to evaluate the hepatoprotective effect of RA. The PK-PD curves showed obviously clockwise (AST and ALT) or anticlockwise (TBA, TBIL). Pretreatment with RA at different doses significantly restrained ANIT-induced pathological changes in bile rate, TBA, TBIL, ALT, AST (p < 0.05 or p < 0.01). The relationship between RA concentration and its hepatoprotective effects on acute cholestasis responses was assessed by PK-PD modeling.
Collapse
Affiliation(s)
- Jianbin Min
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Hao Chen
- College of Food and Drug, Anhui Science and Technology of University, Fengyang 233100, Anhui, China.
| | - Zipeng Gong
- Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Beijing Road, Guiyang 550004, China.
| | - Xian Liu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Tian Wu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Weirong Li
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Jiansong Fang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Tianlai Huang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Yingfeng Zhang
- College of Chinese Medicine, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Wei Zhao
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Chenchen Zhu
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Suiqing Mi
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| | - Ningsheng Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Jichang Road 12, Guangzhou 510405, China.
| |
Collapse
|
137
|
Lin GZ, Qiu JW, Cheng Y, Lin WX, Song YZ. [Clinical and genetic analysis of an infant with progressive familial intrahepatic cholestasis type II]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:758-764. [PMID: 30210030 PMCID: PMC7389172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/16/2018] [Indexed: 11/12/2023]
Abstract
Progressive familial intrahepatic cholestasis type II (PFIC-2) is an autosomal recessive disorder caused by biallelic variants of ABCB11 gene. This paper reports the clinical and laboratory features of a pediatric patient with PFIC-2. The patient was a 2.4-month-old male infant with jaundice and hepatomegaly as the main clinical manifestations. The serum levels of total bilirubin, direct bilirubin and total bile acids were increased, while the serum γ-glutamyl transpeptidase (GGT) level was normal. Next generation sequencing revealed two missense variants, c.1493T>C(p.Ile498Thr) and c.1502T>G(p.Val501Gly), in the ABCB11 gene of the patient, which were inherited from his father and mother, respectively. The latter was a novel variant which was predicted to be pathogenic by using a variety of bioinformatic tools, and the affected p.Val501 residue was highly conserved in 112 homologous peptides.
Collapse
Affiliation(s)
- Gui-Zhi Lin
- Department of Pediatrics, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | | | | | | | | |
Collapse
|
138
|
Lin GZ, Qiu JW, Cheng Y, Lin WX, Song YZ. [Clinical and genetic analysis of an infant with progressive familial intrahepatic cholestasis type II]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2018; 20:758-764. [PMID: 30210030 PMCID: PMC7389172 DOI: 10.7499/j.issn.1008-8830.2018.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/16/2018] [Indexed: 06/08/2023]
Abstract
Progressive familial intrahepatic cholestasis type II (PFIC-2) is an autosomal recessive disorder caused by biallelic variants of ABCB11 gene. This paper reports the clinical and laboratory features of a pediatric patient with PFIC-2. The patient was a 2.4-month-old male infant with jaundice and hepatomegaly as the main clinical manifestations. The serum levels of total bilirubin, direct bilirubin and total bile acids were increased, while the serum γ-glutamyl transpeptidase (GGT) level was normal. Next generation sequencing revealed two missense variants, c.1493T>C(p.Ile498Thr) and c.1502T>G(p.Val501Gly), in the ABCB11 gene of the patient, which were inherited from his father and mother, respectively. The latter was a novel variant which was predicted to be pathogenic by using a variety of bioinformatic tools, and the affected p.Val501 residue was highly conserved in 112 homologous peptides.
Collapse
Affiliation(s)
- Gui-Zhi Lin
- Department of Pediatrics, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China.
| | | | | | | | | |
Collapse
|
139
|
Weiskirchen R, Weiskirchen S, Tacke F. Recent advances in understanding liver fibrosis: bridging basic science and individualized treatment concepts. F1000Res 2018; 7:F1000 Faculty Rev-921. [PMID: 30002817 PMCID: PMC6024236 DOI: 10.12688/f1000research.14841.1] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2018] [Indexed: 02/06/2023] Open
Abstract
Hepatic fibrosis is characterized by the formation and deposition of excess fibrous connective tissue, leading to progressive architectural tissue remodeling. Irrespective of the underlying noxious trigger, tissue damage induces an inflammatory response involving the local vascular system and the immune system and a systemic mobilization of endocrine and neurological mediators, ultimately leading to the activation of matrix-producing cell populations. Genetic disorders, chronic viral infection, alcohol abuse, autoimmune attacks, metabolic disorders, cholestasis, alterations in bile acid composition or concentration, venous obstruction, and parasite infections are well-established factors that predispose one to hepatic fibrosis. In addition, excess fat and other lipotoxic mediators provoking endoplasmic reticulum stress, alteration of mitochondrial function, oxidative stress, and modifications in the microbiota are associated with non-alcoholic fatty liver disease and, subsequently, the initiation and progression of hepatic fibrosis. Multidisciplinary panels of experts have developed practice guidelines, including recommendations of preferred therapeutic approaches to a specific cause of hepatic disease, stage of fibrosis, or occurring co-morbidities associated with ongoing loss of hepatic function. Here, we summarize the factors leading to liver fibrosis and the current concepts in anti-fibrotic therapies.
Collapse
Affiliation(s)
- Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Pauwelsstraße 30, Germany
| | - Sabine Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, Pauwelsstraße 30, Germany
| | - Frank Tacke
- Department of Medicine III, RWTH University Hospital Aachen, D-52074 Aachen, Pauwelsstraße 30, Germany
| |
Collapse
|
140
|
Abstract
The inherited diseases causing conjugated hyperbilirubinemia are diverse, with variability in clinical severity, histologic appearance, and time of onset. The liver biopsy appearances can also vary depending on whether the initial presentation is in the neonatal period or later. Although many of the disorders have specific histologic features in fully developed and classic cases, biopsies taken early in the disease course may be nonspecific, showing either cholestatic hepatitis or an obstructive pattern of injury requiring close correlation with the laboratory and clinical findings to reach the correct diagnosis. Additionally, increased understanding of the range of hepatic changes occurring in mild deficiencies of bile canalicular transporter proteins suggest that these disorders, particularly ABCB4 deficiency, may be more common than previously recognized; improved awareness should prompt further investigation.
Collapse
Affiliation(s)
- Andrew D Clouston
- Faculty of Medicine, University of Queensland, Herston Road, Brisbane, Queensland 4006, Australia.
| |
Collapse
|
141
|
Erginel B, Soysal FG, Durmaz O, Celik A, Salman T. Long-term outcomes of six patients after partial internal biliary diversion for progressive familial intrahepatic cholestasis. J Pediatr Surg 2018; 53:468-471. [PMID: 29174177 DOI: 10.1016/j.jpedsurg.2017.10.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Partial internal biliary diversion (PIBD) is an alternative approach for the treatment of devastating pruritus in patients with progressive familial intrahepatic cholestasis (PFIC). In these patients quality of life can be improved and progression of liver disease can be delayed while waiting for liver transplantation. The aim of our study was to evaluate six patients with PFIC who have undergone PIBD in long-term follow-up. METHODS Retrospective review of the records of six patients who underwent PIBD for PFIC between 2008 and 2010 was conducted to evaluate age, growth, clinical and laboratory studies for long-term outcome. RESULTS Serum postoperative bile acid levels were reduced from a mean 340.1μmol/L (range 851-105) preoperatively to a mean of 96.3μmol/L at postoperative fifth year. The difference between pre- and postoperative bile acid levels was statistically significant (p=0.018). AST decreased from 79.1U/L (range 43-150U/L) to 64.6U/L (range 18-172U/L), ALT decreased from 102.8U/L (range 35-270U/L) to 84.6U/L and total bilirubin decreased from 2.9μmol/L (range 0.35-6.4μmol/L) to 1.53μmol/L (range 0.3-2.4). Again, the decrease in total bilirubin levels was significant (p=0.043). Pruritus was diminished from a mean of +4 (range 4-4) preoperatively to a mean of +2 (4-0). One patient who underwent liver transplantation owing to relapsing pruritus died from postoperative sepsis in the early postoperative period at the fifth year after PBID. Five symptom-free patients have not required liver transplantation at a mean period of 6.1±0.83years (5.1-7.0years) follow-up. CONCLUSION PBID is an effective surgical procedure in the long-term and can delay the need for liver transplantation in children with PFIC by reducing jaundice and pruritus.
Collapse
Affiliation(s)
- Basak Erginel
- Istanbul University, Istanbul Medical Faculty, Department of Pediatric Surgery.
| | - Feryal Gun Soysal
- Istanbul University, Istanbul Medical Faculty, Department of Pediatric Surgery
| | - Ozlem Durmaz
- Istanbul University, Istanbul Medical Faculty, Department of Pediatric Gastroenterology
| | - Alaattin Celik
- Istanbul University, Istanbul Medical Faculty, Department of Pediatric Surgery
| | - Tansu Salman
- Istanbul University, Istanbul Medical Faculty, Department of Pediatric Surgery
| |
Collapse
|
142
|
Access to care in rare liver diseases: New challenges and new opportunities. J Hepatol 2018; 68:577-585. [PMID: 29113911 DOI: 10.1016/j.jhep.2017.11.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 10/12/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022]
Abstract
Patients with rare diseases are often disadvantaged, particularly those with rare liver diseases. Reasons for disadvantage include delayed or overlooked diagnosis, lack of local expertise and high-quality care, poor scientific understanding of the disease process and limited therapeutic options. In adult liver disease this can be compounded by prejudices towards patients with liver disease in general, because of a perception (incorrect for all rare liver diseases) that liver disease is lifestyle related and thus "self-inflicted". In paediatric rare liver diseases, such as biliary atresia, optimising outcomes requires a particularly timely diagnosis. Irrespective of patient age, the scientific and medical community must rise to the challenge of advancing our understanding of rare liver disease, searching for more effective and specific therapies, and providing the infrastructure to provide the best care for all patients, infants, children, young and older adults. The European Reference Network for Rare Liver Diseases is an important step in this direction.
Collapse
|
143
|
Badalyan V, Conklin LS, Snyder JD. Acute Hepatitis. PRINCIPLES AND PRACTICE OF PEDIATRIC INFECTIOUS DISEASES 2018:409-413.e1. [DOI: 10.1016/b978-0-323-40181-4.00060-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
144
|
|
145
|
Wu JS, Li YF, Li YY, Dai Y, Li WK, Zheng M, Shi ZC, Shi R, Wang TM, Ma BL, Liu P, Ma YM. Huangqi Decoction Alleviates Alpha-Naphthylisothiocyanate Induced Intrahepatic Cholestasis by Reversing Disordered Bile Acid and Glutathione Homeostasis in Mice. Front Pharmacol 2017; 8:938. [PMID: 29311939 PMCID: PMC5742571 DOI: 10.3389/fphar.2017.00938] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 12/11/2017] [Indexed: 12/13/2022] Open
Abstract
Intrahepatic cholestasis is a serious symptom of liver disorders with limited therapies. In this study, we investigated the efficacy of Huangqi decoction (HQD), a two-herb classic traditional Chinese medicine (TCM), in the treatment of alpha-naphthylisothiocyanate (ANIT)-induced intrahepatic cholestasis in mice. HQD treatment ameliorated impaired hepatic function and tissue damage. A metabolomics study revealed that the endogenous metabolites significantly affected by HQD were related to bile acid (BA) biosynthesis and glutathione metabolism pathways. HQD treatment decreased the intrahepatic accumulation of cytotoxic BAs, normalized serum BA levels, and increased biliary and urinary BA excretion. Additionally, HQD restored the hepatic glutathione content and suppressed reactive oxygen species (ROS) in cholestatic mice. Protein and gene analysis revealed that HQD increased the expression of the hepatic metabolizing enzymes cytochrome P450 (CYP) 2B10 and UDP glucuronosyltransferase family 1 member A1 (UGT1A1), as well as multidrug resistance-associated protein 2 (Mrp2), Mrp3, and Mrp4, which play crucial roles in BA homeostasis. Further, HQD increased the protein expression of glutamate-cysteine ligase, which is involved in the synthesis of glutathione. Importantly, HQD increased the nuclear expression of nuclear factor-E2-related factor-2 (Nrf2). In conclusion, HQD protects against intrahepatic cholestasis by reversing the disordered homeostasis of BAs and glutathione.
Collapse
Affiliation(s)
- Jia-Sheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-Fei Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan-Yuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Dai
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Kai Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min Zheng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zheng-Chun Shi
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Shi
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Ming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bing-Liang Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue-Ming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai Key Laboratory of Compound Chinese Medicines, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
146
|
Tully DC, Rucker PV, Chianelli D, Williams J, Vidal A, Alper PB, Mutnick D, Bursulaya B, Schmeits J, Wu X, Bao D, Zoll J, Kim Y, Groessl T, McNamara P, Seidel HM, Molteni V, Liu B, Phimister A, Joseph SB, Laffitte B. Discovery of Tropifexor (LJN452), a Highly Potent Non-bile Acid FXR Agonist for the Treatment of Cholestatic Liver Diseases and Nonalcoholic Steatohepatitis (NASH). J Med Chem 2017; 60:9960-9973. [PMID: 29148806 DOI: 10.1021/acs.jmedchem.7b00907] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The farnesoid X receptor (FXR) is a nuclear receptor that acts as a master regulator of bile acid metabolism and signaling. Activation of FXR inhibits bile acid synthesis and increases bile acid conjugation, transport, and excretion, thereby protecting the liver from the harmful effects of bile accumulation, leading to considerable interest in FXR as a therapeutic target for the treatment of cholestasis and nonalcoholic steatohepatitis. We identified a novel series of highly potent non-bile acid FXR agonists that introduce a bicyclic nortropine-substituted benzothiazole carboxylic acid moiety onto a trisubstituted isoxazole scaffold. Herein, we report the discovery of 1 (tropifexor, LJN452), a novel and highly potent agonist of FXR. Potent in vivo activity was demonstrated in rodent PD models by measuring the induction of FXR target genes in various tissues. Tropifexor has advanced into phase 2 human clinical trials in patients with NASH and PBC.
Collapse
Affiliation(s)
- David C Tully
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States.,Novartis Institutes for Biomedical Research , Emeryville, California 94608, United States
| | - Paul V Rucker
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Donatella Chianelli
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Jennifer Williams
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Agnès Vidal
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Phil B Alper
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Daniel Mutnick
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Badry Bursulaya
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - James Schmeits
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Xiangdong Wu
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Dingjiu Bao
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Jocelyn Zoll
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Young Kim
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Todd Groessl
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Peter McNamara
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - H Martin Seidel
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Valentina Molteni
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Bo Liu
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Andrew Phimister
- Novartis Institutes for Biomedical Research , Emeryville, California 94608, United States
| | - Sean B Joseph
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| | - Bryan Laffitte
- Genomics Institute of the Novartis Research Foundation , San Diego, California 92121, United States
| |
Collapse
|
147
|
Shamir SB, Kurian J, Kogan-Liberman D, Taragin BH. Hepatic Imaging in Neonates and Young Infants: State of the Art. Radiology 2017; 285:763-777. [DOI: 10.1148/radiol.2017170305] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Stephanie B. Shamir
- From the Department of Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, 111 E 210 St, Bronx, NY 10467
| | - Jessica Kurian
- From the Department of Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, 111 E 210 St, Bronx, NY 10467
| | - Debora Kogan-Liberman
- From the Department of Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, 111 E 210 St, Bronx, NY 10467
| | - Benjamin H. Taragin
- From the Department of Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, 111 E 210 St, Bronx, NY 10467
| |
Collapse
|
148
|
Pearson HJ, Mosser JL, Jacks SK. The triad of pruritus, xanthomas, and cholestasis: Two cases and a brief review of the literature. Pediatr Dermatol 2017; 34:e305-e308. [PMID: 29144045 DOI: 10.1111/pde.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
When encountered in children, xanthomas are most commonly associated with a group of disorders known as familial hyperlipidemias. Aside from cosmetic concerns, these xanthomas are typically asymptomatic, but when generalized pruritus is a prominent associated symptom, clinicians should consider a different set of diagnoses that includes cholestasis of the liver. In this article we present two illustrative cases of children with cholestatic disease, pruritus, and xanthomas and discuss other disorders that may include this triad.
Collapse
Affiliation(s)
| | - Joy L Mosser
- Nationwide Children's Hospital, Columbus, OH, USA
| | | |
Collapse
|
149
|
The Identification of Two New ABCB11 Gene Mutations and the Treatment Outcome in a Young Adult with Benign Recurrent Intrahepatic Cholestasis: A Case Report. HEPATITIS MONTHLY 2017. [DOI: 10.5812/hepatmon.55087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
|
150
|
Chiang JYL. Linking long noncoding RNA to control bile acid signaling and cholestatic liver fibrosis. Hepatology 2017; 66:1032-1035. [PMID: 28543964 PMCID: PMC5605425 DOI: 10.1002/hep.29289] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/19/2017] [Accepted: 05/19/2017] [Indexed: 01/05/2023]
|