1
|
Nojima H, Shimizu H, Murakami T, Shuto K, Koda K. Critical Roles of the Sphingolipid Metabolic Pathway in Liver Regeneration, Hepatocellular Carcinoma Progression and Therapy. Cancers (Basel) 2024; 16:850. [PMID: 38473211 DOI: 10.3390/cancers16050850] [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: 01/08/2024] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
The sphingolipid metabolic pathway, an important signaling pathway, plays a crucial role in various physiological processes including cell proliferation, survival, apoptosis, and immune regulation. The liver has the unique ability to regenerate using bioactive lipid mediators involving multiple sphingolipids, including ceramide and sphingosine 1-phosphate (S1P). Dysregulation of the balance between sphingomyelin, ceramide, and S1P has been implicated in the regulation of liver regeneration and diseases, including liver fibrosis and hepatocellular carcinoma (HCC). Understanding and modulating this balance may have therapeutic implications for tumor proliferation, progression, and metastasis in HCC. For cancer therapy, several inhibitors and activators of sphingolipid signaling, including ABC294640, SKI-II, and FTY720, have been discussed. Here, we elucidate the critical roles of the sphingolipid pathway in the regulation of liver regeneration, fibrosis, and HCC. Regulation of sphingolipids and their corresponding enzymes may considerably influence new insights into therapies for various liver disorders and diseases.
Collapse
Affiliation(s)
- Hiroyuki Nojima
- Department of Surgery, Teikyo University Chiba Medical Center, 3426-3, Anesaki, Ichihara, Chiba 299-0011, Japan
| | - Hiroaki Shimizu
- Department of Surgery, Teikyo University Chiba Medical Center, 3426-3, Anesaki, Ichihara, Chiba 299-0011, Japan
| | - Takashi Murakami
- Department of Surgery, Teikyo University Chiba Medical Center, 3426-3, Anesaki, Ichihara, Chiba 299-0011, Japan
| | - Kiyohiko Shuto
- Department of Surgery, Teikyo University Chiba Medical Center, 3426-3, Anesaki, Ichihara, Chiba 299-0011, Japan
| | - Keiji Koda
- Department of Surgery, Teikyo University Chiba Medical Center, 3426-3, Anesaki, Ichihara, Chiba 299-0011, Japan
| |
Collapse
|
2
|
Mir IH, Thirunavukkarasu C. The relevance of acid sphingomyelinase as a potential target for therapeutic intervention in hepatic disorders: current scenario and anticipated trends. Arch Toxicol 2023; 97:2069-2087. [PMID: 37248308 PMCID: PMC10226719 DOI: 10.1007/s00204-023-03529-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
Acid sphingomyelinase (ASMase) serves as one of the most remarkable enzymes in sphingolipid biology. ASMase facilitates the hydrolysis of sphingomyelin, yielding ceramide and phosphorylcholine via the phospholipase C signal transduction pathway. Owing to its prominent intervention in apoptosis, ASMase, and its product ceramide is now at the bleeding edge of lipid research due to the coalesced efforts of several research institutions over the past 40 years. ASMase-catalyzed ceramide synthesis profoundly alters the physiological properties of membrane structure in response to a broad range of stimulations, orchestrating signaling cascades for endoplasmic reticulum stress, autophagy, and lysosomal membrane permeabilization, which influences the development of hepatic disorders, such as steatohepatitis, hepatic fibrosis, drug-induced liver injury, and hepatocellular carcinoma. As a result, the potential to modulate the ASMase action with appropriate pharmaceutical antagonists has sparked a lot of curiosity. This article emphasizes the fundamental mechanisms of the systems that govern ASMase aberrations in various hepatic pathologies. Furthermore, we present an insight into the potential therapeutic agents used to mitigate ASMase irregularities and the paramountcy of such inhibitors in drug repurposing.
Collapse
Affiliation(s)
- Ishfaq Hassan Mir
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, 605 014, India
| | | |
Collapse
|
3
|
Chen J, Jian X, Li C, Cheng B. Therapeutic potential of amitriptyline for paraquat-induced pulmonary fibrosis: Involvement of caveolin-1-mediated anti-epithelial-mesenchymal transition and inhibition of apoptosis. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 254:114732. [PMID: 36898313 DOI: 10.1016/j.ecoenv.2023.114732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/28/2023] [Accepted: 03/03/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVE Treatment of pulmonary fibrosis caused by paraquat (PQ) poisoning remains problematic. Amitriptyline (AMT) has multiple pharmacological effects. Here we investigated the anti-fibrotic effect of AMT on PQ-induced pulmonary fibrosis and its possible mechanism. METHODS C57BL/6 mice were randomly divided into control, PQ, PQ + AMT and AMT groups. Histopathology of the lungs, blood gas analysis, and levels of hydroxyproline (HYP), transforming growth factor β1 (TGF-β1) and interleukin 17 (IL-17) were measured. The siRNA transfection inhibited caveolin-1 in A549 cells, which induced epithelial-mesenchymal transition (EMT) by PQ and followed intervention with AMT. E-cadherin, N-cadherin, α-smooth muscle actin (α-SMA) and caveolin-1 were studied by immunohistochemistry and western blot analysis. The apoptosis rate was measured by flow cytometry. RESULTS Compared with the PQ group, the PQ + AMT group displayed mild pathological changes in pulmonary fibrosis, lower HYP, IL-17 and TGF- β1 levels in lung, but high TGF- β1 in serum. Levels of N-cadherin and α-SMA in the lungs were significantly decreased, but caveolin-1 was increased, while SaO2 and PaO2 levels were higher. Compared with the PQ group, the apoptosis rate, N-cadherin and α-SMA levels in A549 cells were significantly decreased after PQ treatment and high dose AMT intervention (p < 0.01). The expressions of E-cadherin, N-cadherin and α-SMA in the PQ-induced cells transfected with caveolin-1 siRNA or siControl RNA were significantly different (p < 0.01), but the apoptosis rate was unaltered. CONCLUSION AMT inhibited PQ-induced EMT in A549 cells and improved lung histopathology and oxygenation in mice by up-regulating caveolin-1.
Collapse
Affiliation(s)
- Jianshi Chen
- Department of Intensive Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Xiangdong Jian
- Department of Poisoning and Occupational Diseases, Qilu Hospital of Shandong University, Jinan 250000, China
| | - Chunmei Li
- Department of Digestive Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Bihuang Cheng
- Department of Intensive Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
4
|
Shi L, Jia F. Association between antidepressant use and liver fibrosis in patients with type 2 diabetes: a population based study. Diabetol Metab Syndr 2023; 15:45. [PMID: 36899407 PMCID: PMC10007740 DOI: 10.1186/s13098-023-01016-x] [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] [Received: 01/12/2023] [Accepted: 03/04/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND The prevalence of liver fibrosis among diabetic patients is increasing rapidly. Our study aims at exploring the relationship between antidepressant use and liver fibrosis in diabetic patients. METHODS We conducted this cross-sectional study through the cycle of National Health and Nutrition Examination Survey (NHANES) 2017-2018. The study population were consisted of patients with type 2 diabetes and reliable vibration-controlled transient elastography (VCTE) results. The presence of liver fibrosis and steatosis were assessed by the median values of liver stiffness measurement (LSM) and controlled attenuation parameter (CAP), respectively. Antidepressants included selective serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants (TCAs), serotonin and norepinephrine reuptake inhibitors (SNRIs) and serotonin antagonists and reuptake inhibitors (SARIs). Patients with evidence of viral hepatitis and significant alcohol consumption were excluded. Logistic regression analysis was performed to evaluate the association between antidepressant use and both steatosis and significant (≥ F3) liver fibrosis after adjustment for potential confounders. RESULTS Our study population consisted of 340 women and 414 men, of whom 87 women(61.3%) and 55(38.7%) men received antidepressants. The most commonly used antidepressants were SSNIs(48.6%), SNRIs(22.5%) and TCAs(12.7%), followed by SARIs(10.6%) and other antidepressants(5.6%). 165 participants had significant liver fibrosis by VCTE, with a weighted overall prevalence of 24%(95% CI 19.2-29.5). In addition, 510 patients had evidence of hepatic steatosis by VCTE with a weighted overall prevalence of 75.4%(95% CI 69.2-80.7). After adjusting confounders, no significant association was observed between antidepressant use and significant liver fibrosis or cirrhosis. CONCLUSIONS In conclusion, in this cross-sectional study, we found that antidepressant drugs was not associated with liver fibrosis and cirrhosis in patients with type 2 diabetes in a nationwide population.
Collapse
Affiliation(s)
- Lin Shi
- Department of Gastroenterology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fangyuan Jia
- Department of Gastroenterology, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Hajduch E, Lachkar F, Ferré P, Foufelle F. Roles of Ceramides in Non-Alcoholic Fatty Liver Disease. J Clin Med 2021; 10:jcm10040792. [PMID: 33669443 PMCID: PMC7920467 DOI: 10.3390/jcm10040792] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease is one of the most common chronic liver diseases, ranging from simple steatosis to steatohepatitis, fibrosis, and cirrhosis. Its prevalence is rapidly increasing and presently affects around 25% of the general population of Western countries, due to the obesity epidemic. Liver fat accumulation induces the synthesis of specific lipid species and particularly ceramides, a sphingolipid. In turn, ceramides have deleterious effects on hepatic metabolism, a phenomenon called lipotoxicity. We review here the evidence showing the role of ceramides in non-alcoholic fatty liver disease and the mechanisms underlying their effects.
Collapse
Affiliation(s)
- Eric Hajduch
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (E.H.); (F.L.); (P.F.)
- Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France
| | - Floriane Lachkar
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (E.H.); (F.L.); (P.F.)
- Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France
| | - Pascal Ferré
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (E.H.); (F.L.); (P.F.)
- Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France
| | - Fabienne Foufelle
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France; (E.H.); (F.L.); (P.F.)
- Institute of Cardiometabolism and Nutrition (ICAN), Hôpital Pitié-Salpêtrière, AP-HP, 75013 Paris, France
- Correspondence: ; Tel.: +33-1-44-27-24-25
| |
Collapse
|
6
|
Chung HY, Claus RA. Keep Your Friends Close, but Your Enemies Closer: Role of Acid Sphingomyelinase During Infection and Host Response. Front Med (Lausanne) 2021; 7:616500. [PMID: 33553211 PMCID: PMC7859284 DOI: 10.3389/fmed.2020.616500] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022] Open
Abstract
Breakdown of the inert and constitutive membrane building block sphingomyelin to the highly active lipid mediator ceramide by extracellularly active acid sphingomyelinase is tightly regulated during stress response and opens the gate for invading pathogens, triggering the immune response, development of remote organ failure, and tissue repair following severe infection. How do one enzyme and one mediator manage all of these affairs? Under physiological conditions, the enzyme is located in the lysosomes and takes part in the noiseless metabolism of sphingolipids, but following stress the protein is secreted into circulation. When secreted, acid sphingomyelinase (ASM) is able to hydrolyze sphingomyelin present at the outer leaflet of membranes to ceramide. Its generation troubles the biophysical context of cellular membranes resulting in functional assembly and reorganization of proteins and receptors, also embedded in highly conserved response mechanisms. As a consequence of cellular signaling, not only induction of cell death but also proliferation, differentiation, and fibrogenesis are affected. Here, we discuss the current state of the art on both the impact and function of the enzyme during host response and damage control. Also, the potential role of lysosomotropic agents as functional inhibitors of this upstream alarming cascade is highlighted.
Collapse
Affiliation(s)
- Ha-Yeun Chung
- Section Translational Neuroimmunology, Department of Neurology, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Ralf A Claus
- Department for Anaesthesiology and Intensive Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
7
|
Li L, Wang H, Jones JW. Sphingolipid metabolism as a marker of hepatotoxicity in drug-induced liver injury. Prostaglandins Other Lipid Mediat 2020; 151:106484. [PMID: 33007444 PMCID: PMC7669681 DOI: 10.1016/j.prostaglandins.2020.106484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/09/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
Drug-induced liver injury (DILI) has a substantial impact on human health and is a major monetary burden on the drug development process. Presently, there is a lack of robust and analytically validated markers for predicting and early diagnosis of DILI. Sphingolipid metabolism and subsequent disruption of sphingolipid homeostasis has been documented to play a key role contributing to hepatocellular death and subsequent liver injury. A more comprehensive understanding of sphingolipid metabolism in response to liver toxicity has great potential to gain mechanistic insight into hepatotoxicity and define molecular markers that are responsible for hepatocyte dysfunction. Here, we present an analytical platform that provides multidimensional mass spectrometry-based datasets for comprehensive structure characterization of sphingolipids extracted from human primary hepatocytes (HPH) exposed to toxic levels of acetaminophen (APAP). Sphingolipid metabolism as measured by characterization of individual sphingolipid structure was sensitive to APAP toxicity displaying a concentration-dependent response. A number of sphingolipid structures were differentially expressed across varying APAP exposures highlighting the unique role sphingolipid metabolism has in response to hepatotoxicity and its potential use as a molecular marker in DILI.
Collapse
Affiliation(s)
- Linhao Li
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States
| | - Hongbing Wang
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States
| | - Jace W Jones
- University of Maryland, School of Pharmacy, Department of Pharmaceutical Sciences, Baltimore, MD, United States.
| |
Collapse
|
8
|
Sphingomyelinases and Liver Diseases. Biomolecules 2020; 10:biom10111497. [PMID: 33143193 PMCID: PMC7692672 DOI: 10.3390/biom10111497] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Sphingolipids (SLs) are critical components of membrane bilayers that play a crucial role in their physico-chemical properties. Ceramide is the prototype and most studied SL due to its role as a second messenger in the regulation of multiple signaling pathways and cellular processes. Ceramide is a heterogeneous lipid entity determined by the length of the fatty acyl chain linked to its carbon backbone sphingosine, which can be generated either by de novo synthesis from serine and palmitoyl-CoA in the endoplasmic reticulum or via sphingomyelin (SM) hydrolysis by sphingomyelinases (SMases). Unlike de novo synthesis, SMase-induced SM hydrolysis represents a rapid and transient mechanism of ceramide generation in specific intracellular sites that accounts for the diverse biological effects of ceramide. Several SMases have been described at the molecular level, which exhibit different pH requirements for activity: neutral, acid or alkaline. Among the SMases, the neutral (NSMase) and acid (ASMase) are the best characterized for their contribution to signaling pathways and role in diverse pathologies, including liver diseases. As part of a Special Issue (Phospholipases: From Structure to Biological Function), the present invited review summarizes the physiological functions of NSMase and ASMase and their role in chronic and metabolic liver diseases, of which the most relevant is nonalcoholic steatohepatitis and its progression to hepatocellular carcinoma, due to the association with the obesity and type 2 diabetes epidemic. A better understanding of the regulation and role of SMases in liver pathology may offer the opportunity for novel treatments of liver diseases.
Collapse
|
9
|
Lv Y, Li Y, Liu N, Dong Y, Deng J. Investigation into imbalance of Th1/Th2 cells in cirrhotic, hypersplenic rats. J Int Med Res 2019; 48:300060519889441. [PMID: 31852338 PMCID: PMC7783262 DOI: 10.1177/0300060519889441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objectives To evaluate the Th1/Th2 cell profile in spleens of cirrhotic and hypersplenic
rats by investigating the expression of Th1-associated chemokine receptors
CXCR3, CCR5 and Th2-associated chemokine receptor CCR3. Methods Experimental liver cirrhosis and hypersplenism were induced in rats by the
intragastric administration of carbon tetrachloride (CCl4; 40%
solution [0.3 ml/100g, twice/week for 8 weeks]) and confirmed by pathology
and hemogram. Presence of the three chemokine receptors was investigated by
real-time polymerase chain reaction (RT-PCR), immunohistochemical staining,
and western blot analysis. Results By comparison with control animals (n=10), RT-PCR demonstrated that CXCR3 and
CCR5-mRNA levels were significantly elevated in the hypersplenic rats (n=26)
and CCR3-mRNA levels were lower. Immunohistochemical staining showed that by
comparison with controls, the mean density of the Th1-associated CXCR3 and
CCR5 receptors was significantly increased but there was no difference
between groups in Th2-associated CCR3 receptors. Western blot analysis
showed that by comparison with controls, hypersplenic rats had higher levels
of CXCR3 and CCR5 protein but lower levels of CCR3 protein. Conclusions The abnormal expression of Th1-associated chemokine receptors in spleens of
rats with cirrhosis and hypersplenism induced by CCL4 suggests that a
functional imbalance between Th1/Th2 cells may play a role in the
pathogenesis of hypersplenism.
Collapse
Affiliation(s)
- Yunfu Lv
- Department of General Surgery, Hainan General Hospital, Haikou, China
| | - Yejuan Li
- Reproductive Medical Centre, Hainan Women and Children's Medical Centre, Haikou, China
| | - Ning Liu
- Department of General Surgery, Hainan General Hospital, Haikou, China
| | - Yonghong Dong
- Department of General Surgery, Shanxi Provincial People's Hospital, Haikou 030001, China
| | - Jie Deng
- Department of General Surgery, Hainan General Hospital, Haikou, China
| |
Collapse
|
10
|
Beckmann N, Becker KA, Kadow S, Schumacher F, Kramer M, Kühn C, Schulz-Schaeffer WJ, Edwards MJ, Kleuser B, Gulbins E, Carpinteiro A. Acid Sphingomyelinase Deficiency Ameliorates Farber Disease. Int J Mol Sci 2019; 20:ijms20246253. [PMID: 31835809 PMCID: PMC6941101 DOI: 10.3390/ijms20246253] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/03/2019] [Accepted: 12/07/2019] [Indexed: 12/20/2022] Open
Abstract
Farber disease is a rare lysosomal storage disorder resulting from acid ceramidase deficiency and subsequent ceramide accumulation. No treatments for Farber disease are clinically available, and affected patients have a severely shortened lifespan. We have recently reported a novel acid ceramidase deficiency model that mirrors the human disease closely. Acid sphingomyelinase is the enzyme that generates ceramide upstream of acid ceramidase in the lysosomes. Using our acid ceramidase deficiency model, we tested if acid sphingomyelinase could be a potential novel therapeutic target for the treatment of Farber disease. A number of functional acid sphingomyelinase inhibitors are clinically available and have been used for decades to treat major depression. Using these as a therapeutic for Farber disease, thus, has the potential to improve central nervous symptoms of the disease as well, something all other treatment options for Farber disease can’t achieve so far. As a proof-of-concept study, we first cross-bred acid ceramidase deficient mice with acid sphingomyelinase deficient mice in order to prevent ceramide accumulation. Double-deficient mice had reduced ceramide accumulation, fewer disease manifestations, and prolonged survival. We next targeted acid sphingomyelinase pharmacologically, to test if these findings would translate to a setting with clinical applicability. Surprisingly, the treatment of acid ceramidase deficient mice with the acid sphingomyelinase inhibitor amitriptyline was toxic to acid ceramidase deficient mice and killed them within a few days of treatment. In conclusion, our study provides the first proof-of-concept that acid sphingomyelinase could be a potential new therapeutic target for Farber disease to reduce disease manifestations and prolong survival. However, we also identified previously unknown toxicity of the functional acid sphingomyelinase inhibitor amitriptyline in the context of Farber disease, strongly cautioning against the use of this substance class for Farber disease patients.
Collapse
Affiliation(s)
- Nadine Beckmann
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | - Stephanie Kadow
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | - Fabian Schumacher
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany;
| | - Melanie Kramer
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | - Claudine Kühn
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
| | | | - Michael J. Edwards
- Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45229, USA;
| | - Burkhard Kleuser
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany;
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
- Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45229, USA;
| | - Alexander Carpinteiro
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, 45147 Essen, Germany; (N.B.); (K.A.B.); (S.K.); (F.S.); (M.K.); (C.K.); (E.G.)
- Department of Hematology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
- Correspondence: ; Tel.: +49-201-723-84579; Fax: +49-201-723-5974
| |
Collapse
|
11
|
Beckmann N, Kadow S, Schumacher F, Göthert JR, Kesper S, Draeger A, Schulz-Schaeffer WJ, Wang J, Becker JU, Kramer M, Kühn C, Kleuser B, Becker KA, Gulbins E, Carpinteiro A. Pathological manifestations of Farber disease in a new mouse model. Biol Chem 2019; 399:1183-1202. [PMID: 29908121 DOI: 10.1515/hsz-2018-0170] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/07/2018] [Indexed: 11/15/2022]
Abstract
Farber disease (FD) is a rare lysosomal storage disorder resulting from acid ceramidase deficiency and subsequent ceramide accumulation. No treatments are clinically available and affected patients have a severely shortened lifespan. Due to the low incidence, the pathogenesis of FD is still poorly understood. Here, we report a novel acid ceramidase mutant mouse model that enables the study of pathogenic mechanisms of FD and ceramide accumulation. Asah1tmEx1 mice were generated by deletion of the acid ceramidase signal peptide sequence. The effects on lysosomal targeting and activity of the enzyme were assessed. Ceramide and sphingomyelin levels were quantified by liquid chromatography tandem-mass spectrometry (LC-MS/MS) and disease manifestations in several organ systems were analyzed by histology and biochemistry. We show that deletion of the signal peptide sequence disrupts lysosomal targeting and enzyme activity, resulting in ceramide and sphingomyelin accumulation. The affected mice fail to thrive and die early. Histiocytic infiltrations were observed in many tissues, as well as lung inflammation, liver fibrosis, muscular disease manifestations and mild kidney injury. Our new mouse model mirrors human FD and thus offers further insights into the pathogenesis of this disease. In the future, it may also facilitate the development of urgently needed therapies.
Collapse
Affiliation(s)
- Nadine Beckmann
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Stephanie Kadow
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Fabian Schumacher
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany.,Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | - Joachim R Göthert
- Department of Hematology, University Hospital Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Stefanie Kesper
- Department of Hematology, University Hospital Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Annette Draeger
- Institute of Anatomy, University of Bern, Baltzerstr. 2, CH-3012 Bern, Switzerland
| | - Walter J Schulz-Schaeffer
- Insitute of Neuropathology, University of the Saarland, Kirrberger Str. 100, D-66421 Homburg, Germany
| | - Jiang Wang
- Department of Pathology and Laboratory Medicine, UC Health University Hospital, 234 Goodman Street, Cincinnati, OH 45219, USA
| | - Jan U Becker
- Institute of Pathology, University Hospital Cologne, Kerpener Straße 62, D-50937 Cologne, Germany
| | - Melanie Kramer
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Claudine Kühn
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Burkhard Kleuser
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Arthur-Scheunert-Allee 114-116, D-14558 Nuthetal, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany.,Department of Surgery, University of Cincinnati, 231 Albert Sabin Way, ML 0558, Cincinnati, OH 45229, USA
| | - Alexander Carpinteiro
- Department of Molecular Biology, University of Duisburg-Essen, Hufelandstraße 55, D-45147 Essen, Germany.,Department of Hematology, University Hospital Essen, Hufelandstraße 55, D-45147 Essen, Germany
| |
Collapse
|
12
|
Chen JY, Ren Y, Yan P, Belina ME, Chung RT, Butt AA. Tricyclic antidepressant use and the risk of fibrosis progression in hepatitis C-infected persons: Results from ERCHIVES. J Viral Hepat 2018; 25:825-833. [PMID: 29478294 PMCID: PMC6019114 DOI: 10.1111/jvh.12884] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 01/15/2018] [Indexed: 12/19/2022]
Abstract
Recent preclinical studies have suggested an antifibrotic role for tricyclic antidepressants (TCA). Using the Electronically Retrieved Cohort of hepatitis C virus (HCV) Infected Veterans, we aimed to evaluate the impact of TCA use on fibrosis progression and development of hepatocellular carcinoma (HCC) among HCV-infected persons. Subjects were categorized according to use of TCAs, selective serotonin reuptake inhibitors (SSRI) or no antidepressants. TCAs or selective serotonin uptake inhibitors use was defined according to cumulative defined daily dose (cDDD), and categories were mutually exclusive. Subjects with HIV coinfection, hepatitis B surface antigen (HbsAg) positivity, cirrhosis or HCC at baseline were excluded. Outcomes were liver fibrosis progression measured by APRI scores and incident HCC. We utilized Cox proportional hazards regression to determine predictors of cirrhosis, defined as APRI > 2, and incident hepatocellular carcinoma (iHCC). Among 128 201 eligible HCV+ persons, 4% received TCAs, 43% received selective serotonin uptake inhibitors, and 53% received no antidepressants. Fewer TCAs users had drug abuse (34% and 43%) and alcohol abuse (32% vs 42%) compared to selective serotonin uptake inhibitor users. After adjusting for age, baseline APRI score, diabetes, hypertension, alcohol use, drug abuse and HCV RNA levels, TCAs use was associated with decreased risk of cirrhosis (hazard ratio [HR] = 0.77, 95% CI = 0.60, 0.99) and delayed time to development of cirrhosis, but not with decreased iHCC. In conclusion among a large cohort of HCV-positive Veterans, TCAs use was associated with decreased fibrosis progression and lower risk of developing cirrhosis. These data provide supportive evidence for the beneficial effects of TCAs on progression of liver fibrosis in patients with chronic HCV infection.
Collapse
Affiliation(s)
- Jennifer Y. Chen
- Department of Medicine, University of California, San Francisco, California USA,The Liver Center, University of California, San Francisco, California USA
| | - Yanjie Ren
- Veterans Research Foundation, Pittsburgh, PA USA
| | - Peng Yan
- Veterans Research Foundation, Pittsburgh, PA USA
| | - Morgan E. Belina
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Raymond T. Chung
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Adeel A. Butt
- VA Pittsburgh Healthcare System, Pittsburgh, PA USA,Weill Cornell Medical College, Doha, Qatar and New York, NY USA,Hamad Healthcare Quality Institute and Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
13
|
Konishi T, Schuster RM, Lentsch AB. Proliferation of hepatic stellate cells, mediated by YAP and TAZ, contributes to liver repair and regeneration after liver ischemia-reperfusion injury. Am J Physiol Gastrointest Liver Physiol 2018; 314:G471-G482. [PMID: 29351389 PMCID: PMC5966748 DOI: 10.1152/ajpgi.00153.2017] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are key regulators of cell proliferation and organ size; however, their physiological contribution after liver injury has not been fully understood. In this study, we sought to determine the role of YAP and TAZ during liver recovery after ischemia-reperfusion (I/R). A murine model of partial (70%) I/R was used to induce liver injury and study the reparative and regenerative response. After liver injury, there was marked activation and proliferation of hepatic stellate cells. The Hippo pathway components, large tumor suppressor 1 (LATS1) and its adapter protein, Mps one binder 1 (MOB1), were inactivated during liver repair, and YAP and TAZ were activated selectively in hepatic stellate cells. Concurrently, the expression of connective tissue growth factor and survivin, both of which are YAP and TAZ target genes, were upregulated. Hepatic stellate cell expansion and concomitant activation of YAP and TAZ occurred only in the injured liver and were not observed in the nonischemic liver. Treatment of mice with verteporfin, an inhibitor of YAP and TAZ, decreased hepatic stellate cell proliferation, survivin, and cardiac ankyrin repeat protein expression. These changes were associated with a significant decrease in hepatocyte proliferation. The data suggest that liver repair and regeneration after I/R injury are dependent on hepatic stellate cell proliferation, which is mediated by YAP and TAZ. NEW & NOTEWORTHY This study is the first to assess the proliferation of hepatic stellate cells (HSCs) after ischemia-reperfusion (I/R) injury and their role in the reparative and regenerative process. Here we show that the Hippo pathway is inactivated after I/R and that Yes-associated protein/transcriptional coactivator with PDZ-binding motif (YAP/TAZ) activation is detected in HSC. HSC proliferation and expansion are prominent during liver recovery after I/R injury. Inhibition of YAP/TAZ activation with verteporfin reduces HSC proliferation and target gene expression and attenuates hepatocyte proliferation.
Collapse
Affiliation(s)
- Takanori Konishi
- Department of Surgery, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Rebecca M. Schuster
- Department of Surgery, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Alex B. Lentsch
- Department of Surgery, University of Cincinnati, College of Medicine, Cincinnati, Ohio
| |
Collapse
|
14
|
Chen JY, Newcomb B, Zhou C, Pondick JV, Ghoshal S, York SR, Motola DL, Coant N, Yi JK, Mao C, Tanabe KK, Bronova I, Berdyshev EV, Fuchs BC, Hannun Y, Chung RT, Mullen AC. Tricyclic Antidepressants Promote Ceramide Accumulation to Regulate Collagen Production in Human Hepatic Stellate Cells. Sci Rep 2017; 7:44867. [PMID: 28322247 PMCID: PMC5359599 DOI: 10.1038/srep44867] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 02/15/2017] [Indexed: 12/21/2022] Open
Abstract
Activation of hepatic stellate cells (HSCs) in response to injury is a key step in hepatic fibrosis, and is characterized by trans-differentiation of quiescent HSCs to HSC myofibroblasts, which secrete extracellular matrix proteins responsible for the fibrotic scar. There are currently no therapies to directly inhibit hepatic fibrosis. We developed a small molecule screen to identify compounds that inactivate human HSC myofibroblasts through the quantification of lipid droplets. We screened 1600 compounds and identified 21 small molecules that induce HSC inactivation. Four hits were tricyclic antidepressants (TCAs), and they repressed expression of pro-fibrotic factors Alpha-Actin-2 (ACTA2) and Alpha-1 Type I Collagen (COL1A1) in HSCs. RNA sequencing implicated the sphingolipid pathway as a target of the TCAs. Indeed, TCA treatment of HSCs promoted accumulation of ceramide through inhibition of acid ceramidase (aCDase). Depletion of aCDase also promoted accumulation of ceramide and was associated with reduced COL1A1 expression. Treatment with B13, an inhibitor of aCDase, reproduced the antifibrotic phenotype as did the addition of exogenous ceramide. Our results show that detection of lipid droplets provides a robust readout to screen for regulators of hepatic fibrosis and have identified a novel antifibrotic role for ceramide.
Collapse
Affiliation(s)
- Jennifer Y Chen
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Benjamin Newcomb
- Health Science Center, Stony Brook University, Stony Brook, NY USA
| | - Chan Zhou
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Joshua V Pondick
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Sarani Ghoshal
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA USA
| | - Samuel R York
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Daniel L Motola
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Nicolas Coant
- Health Science Center, Stony Brook University, Stony Brook, NY USA
| | - Jae Kyo Yi
- Health Science Center, Stony Brook University, Stony Brook, NY USA
| | - Cungui Mao
- Health Science Center, Stony Brook University, Stony Brook, NY USA
| | - Kenneth K Tanabe
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA USA
| | | | | | - Bryan C Fuchs
- Division of Surgical Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA USA
| | - Yusuf Hannun
- Health Science Center, Stony Brook University, Stony Brook, NY USA
| | - Raymond T Chung
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Alan C Mullen
- Gastrointestinal Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA.,Harvard Stem Cell Institute, Cambridge, MA 02138 USA
| |
Collapse
|
15
|
Zoicas I, Reichel M, Gulbins E, Kornhuber J. Role of Acid Sphingomyelinase in the Regulation of Social Behavior and Memory. PLoS One 2016; 11:e0162498. [PMID: 27598773 PMCID: PMC5012580 DOI: 10.1371/journal.pone.0162498] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 08/23/2016] [Indexed: 01/23/2023] Open
Abstract
Major depressive disorder is often associated with deficits in social and cognitive functioning. Mice transgenic for acid sphingomyelinase (t-ASM) were previously shown to have a depressive-like phenotype, which could be normalized by antidepressant treatment. Here, we investigated whether t-ASM mice show deficits in social behavior and memory performance, and whether these possible deficits might be normalized by amitriptyline treatment. Our results revealed that ASM overexpression altered the behavior of mice in a sex-dependent manner. As such, t-ASM female, but not male, mice showed an impaired social preference and a depressive- and anxiogenic-like phenotype, which could be normalized by amitriptyline treatment. Both male and female t-ASM mice showed unaltered preference for social novelty, novel object recognition, and social and object discrimination abilities. Amitriptyline treatment impaired novel object recognition and object discrimination abilities in female, but not in male, wild-type mice, while female t-ASM mice showed unaltered novel object recognition and object discrimination abilities. This study suggests that female t-ASM mice represent a model of depression with comorbid anxiety and social deficits, without memory impairments. It further suggests that ASM overexpression has a protective role against the detrimental effects of amitriptyline on female, but not on male, non-social (object) memory.
Collapse
Affiliation(s)
- Iulia Zoicas
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
- * E-mail:
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
16
|
Ilan Y. Compounds of the sphingomyelin-ceramide-glycosphingolipid pathways as secondary messenger molecules: new targets for novel therapies for fatty liver disease and insulin resistance. Am J Physiol Gastrointest Liver Physiol 2016; 310:G1102-17. [PMID: 27173510 DOI: 10.1152/ajpgi.00095.2016] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/04/2016] [Indexed: 01/31/2023]
Abstract
The compounds of sphingomyelin-ceramide-glycosphingolipid pathways have been studied as potential secondary messenger molecules in various systems, along with liver function and insulin resistance. Secondary messenger molecules act directly or indirectly to affect cell organelles and intercellular interactions. Their potential role in the pathogenesis of steatohepatitis and diabetes has been suggested. Data samples collected from patients with Gaucher's disease, who had high levels of glucocerebroside, support a role for compounds from these pathways as a messenger molecules in the pathogenesis of fatty liver disease and diabetes. The present review summarizes some of the recent data on the role of glycosphingolipid molecules as messenger molecules in various physiological and pathological conditions, more specifically including insulin resistance and fatty liver disease.
Collapse
Affiliation(s)
- Yaron Ilan
- Gastroenterology and Liver Units, Department of Medicine, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
17
|
Wilson GC, Freeman CM, Kuethe JW, Quillin RC, Nojima H, Schuster R, Blanchard J, Edwards MJ, Caldwell CC, Lentsch AB. CXC chemokine receptor-4 signaling limits hepatocyte proliferation after hepatic ischemia-reperfusion in mice. Am J Physiol Gastrointest Liver Physiol 2015; 308:G702-9. [PMID: 25721302 PMCID: PMC4398844 DOI: 10.1152/ajpgi.00257.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 02/18/2015] [Indexed: 01/31/2023]
Abstract
The role of stromal cell-derived factor-1 (SDF-1 or CXCL12) and its receptor CXC chemokine receptor-4 (CXCR4) in ischemic liver injury and recovery has not been studied. Some reports suggest that this chemokine may aid in liver regeneration, but others suggest that it may be profibrotic through its activation of hepatic stellate cells. In this study we sought to elucidate the role of SDF-1 and its receptor CXCR4 during liver injury, recovery, and regeneration after ischemia-reperfusion (I/R). A murine model of partial (70%) I/R was used to induce liver injury and study the reparative and regenerative response. CXCR4 was expressed constitutively in the liver, and hepatic levels of SDF-1 peaked 8 h after reperfusion but remained significantly increased for 96 h. Treatment of mice with the CXCR4 antagonist AMD3100 or agonist SDF-1 had no effect on acute liver injury assessed 8 h after I/R. However, treatment with AMD3100 increased hepatocyte proliferation after 72 and 96 h of reperfusion and reduced the amount of liver necrosis. In contrast, treatment with SDF-1 significantly decreased hepatocyte proliferation. These effects appeared to be dependent on the presence of liver injury, as AMD3100 and SDF-1 had no effect on hepatocyte proliferation or liver mass in mice undergoing 70% partial hepatectomy. The data suggest that signaling through CXCR4 is detrimental to liver recovery and regeneration after I/R and that clinical therapy with a CXCR4 antagonist may improve hepatic recovery following acute liver injury.
Collapse
Affiliation(s)
- Gregory C. Wilson
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Joshua W. Kuethe
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ralph C. Quillin
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Hiroyuki Nojima
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Rebecca Schuster
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - John Blanchard
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Michael J. Edwards
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Charles C. Caldwell
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Alex B. Lentsch
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
18
|
Zhang YF, Wang F, Li FH, Sun YF, Zhang Y, Guo PR, He LL, Zhu YN. Influence of Biejiajian pill on PDGF and Ras signaling pathways in hepatic fibrosis in rats. Shijie Huaren Xiaohua Zazhi 2014; 22:4060-4067. [DOI: 10.11569/wcjd.v22.i27.4060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the anti-fibrotic mechanism of Biejiajian pill, a traditional Chinese medicine for activating blood circulation and eliminating stasis, in rats with hepatic fibrosis.
METHODS: Hepatic fibrosis was induced in rats by subcutaneous injection of CCl4. SD rats were randomly divided into five groups: a control group, a model group, low-, medium- and high-dose Biejiajian pill treatment groups. The three treatment groups were given Biejiajian pill 0.55, 1.10, and 2.20 g/(kg•d), respectively, while the normal control group and model group were treated with equal volume of normal saline by gavage. At the 11th week, hepatic pathological changes were observed. The immunohistochemical technique was applied to analyze the expression of extracellular signal regulated kinase 1 (ERK1) in hepatic tissue. RT-PCR was applied to analyze the expression of platelet derivative growth factor (PDGF) and Ras mRNAs in hepatic tissue.
RESULTS: Compared with the control group, the expression levels of ERK1 protein and PDGF and Ras mRNAs increased significantly in the model group (0.19 ± 0.001 vs 0.71 ± 0.018, 0.12 ± 0.000 vs 0.42 ± 0.006, 0.12 ± 0.000 vs 0.44 ± 0.017, P < 0.05). Compared with the model group, Biejiajian pill significantly improved hepatic pathological changes and decreased the expression of ERK1 protein and PDGF and Ras mRNAs, and the efficacy of high-dose Biejiajian pill was the best.
CONCLUSION: Biejiajian pill can improve hepatic pathological changes in rats with hepatic fibrosis possibly via mechanisms associated with activating the Ras/ERK signaling pathway.
Collapse
|
19
|
Beckmann N, Sharma D, Gulbins E, Becker KA, Edelmann B. Inhibition of acid sphingomyelinase by tricyclic antidepressants and analogons. Front Physiol 2014; 5:331. [PMID: 25228885 PMCID: PMC4151525 DOI: 10.3389/fphys.2014.00331] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/12/2014] [Indexed: 11/13/2022] Open
Abstract
Amitriptyline, a tricyclic antidepressant, has been used in the clinic to treat a number of disorders, in particular major depression and neuropathic pain. In the 1970s the ability of tricyclic antidepressants to inhibit acid sphingomyelinase (ASM) was discovered. The enzyme ASM catalyzes the hydrolysis of sphingomyelin to ceramide. ASM and ceramide were shown to play a crucial role in a wide range of diseases, including cancer, cystic fibrosis, diabetes, Alzheimer's disease, and major depression, as well as viral (e.g., measles virus) and bacterial (e.g., Staphylococcus aureus, Pseudomonas aeruginosa) infections. Ceramide molecules may act in these diseases by the alteration of membrane biophysics, the self-association of ceramide molecules within the cell membrane and the ultimate formation of larger ceramide-enriched membrane domains/platforms. These domains were shown to serve the clustering of certain receptors such as CD95 and may also act in the above named diseases. The potential to block the generation of ceramide by inhibiting the ASM has opened up new therapeutic approaches for the treatment of these conditions. Since amitriptyline is one of the longest used clinical drugs and side effects are well studied, it could potentially become a cheap and easily accessible medication for patients suffering from these diseases. In this review, we aim to provide an overview of current in vitro and in vivo studies and clinical trials utilizing amitriptyline to inhibit ASM and contemplate possible future applications of the drug.
Collapse
Affiliation(s)
- Nadine Beckmann
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Deepa Sharma
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Katrin Anne Becker
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| | - Bärbel Edelmann
- Department of Molecular Biology, Institute of Molecular Biology, University of Duisburg-Essen Essen, Germany
| |
Collapse
|