1
|
Ogino N, Leite MF, Guerra MT, Kruglov E, Asashima H, Hafler DA, Ito T, Pereira JP, Peiffer BJ, Sun Z, Ehrlich BE, Nathanson MH. Neutrophils insert elastase into hepatocytes to regulate calcium signaling in alcohol-associated hepatitis. J Clin Invest 2024; 134:e171691. [PMID: 38916955 PMCID: PMC11324315 DOI: 10.1172/jci171691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/20/2024] [Indexed: 06/27/2024] Open
Abstract
Neutrophil infiltration occurs in a variety of liver diseases, but it is unclear how neutrophils and hepatocytes interact. Neutrophils generally use granule proteases to digest phagocytosed bacteria and foreign substances or neutralize them in neutrophil extracellular traps. In certain pathological states, granule proteases play a destructive role against the host as well. More recently, nondestructive actions of neutrophil granule proteins have been reported, such as modulation of tissue remodeling and metabolism. Here, we report a completely different mechanism by which neutrophils act nondestructively, by inserting granules directly into hepatocytes. Specifically, elastase-containing granules were transferred to hepatocytes where elastase selectively degraded intracellular calcium channels to reduce cell proliferation without cytotoxicity. In response, hepatocytes increased expression of Serpin E2 and A3, which inhibited elastase activity. Elastase insertion was seen in patient specimens of alcohol-associated hepatitis, and the relationship between elastase-mediated ITPR2 degradation and reduced cell proliferation was confirmed in mouse models. Moreover, neutrophils from patients with alcohol-associated hepatitis were more prone to degranulation and more potent in reducing calcium channel expression than neutrophils from healthy individuals. This nondestructive and reversible action on hepatocytes defines a previously unrecognized role for neutrophils in the transient regulation of epithelial calcium signaling mechanisms.
Collapse
Affiliation(s)
- Noriyoshi Ogino
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - M. Fatima Leite
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
- INCT - NanoBiofar – Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mateus T. Guerra
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emma Kruglov
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | - Takeshi Ito
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - João P. Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Brandon J. Peiffer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Barbara E. Ehrlich
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, New York University School of Medicine, New York, New York, USA
| | - Michael H. Nathanson
- Yale Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
2
|
Albalawi SS, Aljabri A, Alshibani M, Al-Gayyar MM. The Involvement of Calcium Channels in the Endoplasmic Reticulum Membrane in Nonalcoholic Fatty Liver Disease Pathogenesis. Cureus 2023; 15:e49150. [PMID: 38024063 PMCID: PMC10663096 DOI: 10.7759/cureus.49150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 12/01/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent and complex condition that affects millions of people globally. It occurs when fat, primarily triglycerides, accumulates in liver cells, leading to inflammation and damage. Calcium, an essential mineral, is involved in various physiological processes, including the regeneration process following liver injury. The endoplasmic reticulum (ER), a complex organelle involved in protein synthesis and lipid metabolism, regulates intracellular calcium levels. Dysregulation of this process can lead to calcium overload, oxidative stress, and cellular damage, all of which are hallmarks of NAFLD. Inositol 1,4,5-trisphosphate receptor (IP3R), a type of calcium ion channel, is found throughout the body, including the liver. IP3R is classified into three subtypes: IP3R1, IP3R2, and IP3R3, and it plays a critical role in regulating intracellular calcium levels. However, excessive calcium accumulation in the mitochondria due to an overload of calcium ions or increased IP3R activity can lead to NAFLD. Therefore, targeting calcium channels in the ER membrane may represent a promising therapeutic strategy for preventing and treating this increasingly prevalent metabolic disorder. It may help prevent mitochondrial calcium accumulation and reduce the risk of hepatic damage. This review article aimed to review the relationship between IP3R modulation and the pathogenicity of NAFLD, providing valuable insights to help researchers develop more effective treatments for the condition.
Collapse
Affiliation(s)
- Sarah S Albalawi
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
| | - Ahmed Aljabri
- Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, SAU
- Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
| | - Mohannad Alshibani
- Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, SAU
| | - Mohammed M Al-Gayyar
- Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, SAU
- Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, EGY
| |
Collapse
|
3
|
Rodrigues MA, Gomes DA, Fiorotto R, Guerra MT, Weerachayaphorn J, Bo T, Sessa WC, Strazzabosco M, Nathanson MH. Molecular determinants of peri-apical targeting of inositol 1,4,5-trisphosphate receptor type 3 in cholangiocytes. Hepatol Commun 2022; 6:2748-2764. [PMID: 35852334 PMCID: PMC9512452 DOI: 10.1002/hep4.2042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/03/2022] [Accepted: 06/20/2022] [Indexed: 11/19/2022] Open
Abstract
Fluid and bicarbonate secretion is a principal function of cholangiocytes, and impaired secretion results in cholestasis. Cholangiocyte secretion depends on peri-apical expression of the type 3 inositol trisphosphate receptor (ITPR3), and loss of this intracellular Ca2+ release channel is a final common event in most cholangiopathies. Here we investigated the mechanism by which ITPR3 localizes to the apical region to regulate secretion. Isolated bile duct units, primary mouse cholangiocytes, and polarized Madin-Darby canine kidney (MDCK) cells were examined using a combination of biochemical and fluorescence microscopy techniques to investigate the mechanism of ITPR3 targeting to the apical region. Apical localization of ITPR3 depended on the presence of intact lipid rafts as well as interactions with both caveolin 1 (CAV1) and myosin heavy chain 9 (MYH9). Chemical disruption of lipid rafts or knockdown of CAV1 or MYH9 redistributed ITPR3 away from the apical region. MYH9 interacted with the five c-terminal amino acids of the ITPR3 peptide. Disruption of lipid rafts impaired Ca2+ signaling, and absence of CAV1 impaired both Ca2+ signaling and fluid secretion. Conclusion: A cooperative mechanism involving MYH9, CAV1, and apical lipid rafts localize ITPR3 to the apical region to regulate Ca2+ signaling and secretion in cholangiocytes.
Collapse
Affiliation(s)
- Michele A. Rodrigues
- Section of Digestive Diseases, Internal MedicineYale UniversityNew HavenConnecticutUSA
- Department of Biochemistry and ImmunologyFederal University of Minas Gerais (UFMG)Belo HorizonteMGBrazil
| | - Dawidson A. Gomes
- Section of Digestive Diseases, Internal MedicineYale UniversityNew HavenConnecticutUSA
- Department of Biochemistry and ImmunologyFederal University of Minas Gerais (UFMG)Belo HorizonteMGBrazil
| | - Romina Fiorotto
- Section of Digestive Diseases, Internal MedicineYale UniversityNew HavenConnecticutUSA
| | - Mateus T. Guerra
- Section of Digestive Diseases, Internal MedicineYale UniversityNew HavenConnecticutUSA
| | | | - Tao Bo
- Department of Pharmacology and Program in Vascular Cell Signaling and TherapeuticsYale University School of MedicineNew HavenConnecticutUSA
| | - William C. Sessa
- Department of Pharmacology and Program in Vascular Cell Signaling and TherapeuticsYale University School of MedicineNew HavenConnecticutUSA
| | - Mario Strazzabosco
- Section of Digestive Diseases, Internal MedicineYale UniversityNew HavenConnecticutUSA
| | - Michael H. Nathanson
- Section of Digestive Diseases, Internal MedicineYale UniversityNew HavenConnecticutUSA
| |
Collapse
|
4
|
Han B, Zhen F, Zheng XS, Hu J, Chen XS. Systematic analysis of the expression and prognostic value of ITPR1 and correlation with tumor infiltrating immune cells in breast cancer. BMC Cancer 2022; 22:297. [PMID: 35313846 PMCID: PMC8939201 DOI: 10.1186/s12885-022-09410-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND ITPR1 is a key gene for autophagy, but its biological function is still unclear, and there are few studies on the correlation between ITPR1 gene expression and the occurrence and development of breast cancer. METHODS Analyze the expression of ITPR1 through online databases such as Oncomine and TIMER. Kaplan-Meier plotter and other databases were used to evaluate the impact of ITPR1 on clinical prognosis. The expression of ITPR1 in analysis of 145 cases of breast cancer and 30 cases of adjacent normal tissue was detected by Immunohistochemistry. Statistical analysis was used to evaluate the clinical relevance and prognostic significance of abnormally expressed proteins. And the Western Blot was used to detect the expression of ITPR1 between breast cancer tissues and cells. The TIMER database studied the relationship between ITPR1 and cancer immune infiltration. And used the ROC plotter database to predict the response of ITPR1 to chemotherapy, endocrine therapy and anti-HER2 therapy in patients with breast cancer. RESULTS Compared with normal breast samples, ITPR1 was significantly lower in patients with breast cancer. And the increased expression of ITPR1 mRNA was closely related to longer overall survival (OS), distant metastasis free survival (DMFS), disease specific survival (DSS) and relapse free survival (RFS) in breast cancer. And the expression level of ITPR1 was higher in patients treated with chemotherapy than untreated patients. In addition, the expression of ITPR1 was positively correlated with related gene markers of immune cells in different types of breast cancer, especially with BRCA basal tissue breast cancer. CONCLUSION ITPR1 was lower expressed in breast cancer. The higher expression of ITPR1 suggested favorable prognosis for patients. ITPR1 was related to the level of immune infiltration, especially in BRCA-Basal patients. All research results indicated that ITPR1 might affect breast cancer prognosis and participate in immune regulation. In short, ITPR1 might be a potential target for breast cancer therapy.
Collapse
Affiliation(s)
- Bing Han
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Fang Zhen
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China
| | - Xiu-Shuang Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, 150001, China
| | - Jing Hu
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| | - Xue-Song Chen
- Department of Breast Medical Oncology, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150040, China.
| |
Collapse
|
5
|
Verma A, Manchel A, Narayanan R, Hoek JB, Ogunnaike BA, Vadigepalli R. A Spatial Model of Hepatic Calcium Signaling and Glucose Metabolism Under Autonomic Control Reveals Functional Consequences of Varying Liver Innervation Patterns Across Species. Front Physiol 2021; 12:748962. [PMID: 34899380 PMCID: PMC8662697 DOI: 10.3389/fphys.2021.748962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/11/2021] [Indexed: 11/25/2022] Open
Abstract
Rapid breakdown of hepatic glycogen stores into glucose plays an important role during intense physical exercise to maintain systemic euglycemia. Hepatic glycogenolysis is governed by several different liver-intrinsic and systemic factors such as hepatic zonation, circulating catecholamines, hepatocellular calcium signaling, hepatic neuroanatomy, and the central nervous system (CNS). Of the factors regulating hepatic glycogenolysis, the extent of lobular innervation varies significantly between humans and rodents. While rodents display very few autonomic nerve terminals in the liver, nearly every hepatic layer in the human liver receives neural input. In the present study, we developed a multi-scale, multi-organ model of hepatic metabolism incorporating liver zonation, lobular scale calcium signaling, hepatic innervation, and direct and peripheral organ-mediated communication between the liver and the CNS. We evaluated the effect of each of these governing factors on the total hepatic glucose output and zonal glycogenolytic patterns within liver lobules during simulated physical exercise. Our simulations revealed that direct neuronal stimulation of the liver and an increase in circulating catecholamines increases hepatic glucose output mediated by mobilization of intracellular calcium stores and lobular scale calcium waves. Comparing simulated glycogenolysis between human-like and rodent-like hepatic innervation patterns (extensive vs. minimal) suggested that propagation of calcium transients across liver lobules acts as a compensatory mechanism to improve hepatic glucose output in sparsely innervated livers. Interestingly, our simulations suggested that catecholamine-driven glycogenolysis is reduced under portal hypertension. However, increased innervation coupled with strong intercellular communication can improve the total hepatic glucose output under portal hypertension. In summary, our modeling and simulation study reveals a complex interplay of intercellular and multi-organ interactions that can lead to differing calcium dynamics and spatial distributions of glycogenolysis at the lobular scale in the liver.
Collapse
Affiliation(s)
- Aalap Verma
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States.,Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexandra Manchel
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Rahul Narayanan
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jan B Hoek
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Babatunde A Ogunnaike
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics and Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
6
|
Ren T, Pang L, Dai W, Wu S, Kong J. Regulatory mechanisms of the bile salt export pump (BSEP/ABCB11) and its role in related diseases. Clin Res Hepatol Gastroenterol 2021; 45:101641. [PMID: 33581308 DOI: 10.1016/j.clinre.2021.101641] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/03/2021] [Accepted: 01/21/2021] [Indexed: 02/04/2023]
Abstract
The bile salt export pump (BSEP/ABCB11) is located on the apical membrane and mediates the secretion of bile salts from hepatocytes into the bile. BSEP-mediated bile salt efflux is the rate-limiting step of bile salt secretion and the main driving force of bile flow. BSEP drives and maintains the enterohepatic circulation of bile salts. In recent years, research efforts have been focused on understanding the physiological and pathological functions and regulatory mechanisms of BSEP. These studies elucidated the roles of farnesoid X receptor (FXR), AMP-activated protein kinase (AMPK), liver receptor homolog-1(LRH-1) and nuclear factor erythroid 2-related factor 2 (Nrf-2) in BSEP expression and discovered some regulatory factors which participate in its post-transcriptional regulation. A series of liver diseases have also been shown to be related to BSEP expression and dysfunction, such as cholestasis, drug-induced liver injury, and gallstones. Here, we systematically review and summarize recent literature on BSEP structure, physiological functions, regulatory mechanisms, and related diseases.
Collapse
Affiliation(s)
- Tengqi Ren
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Liwei Pang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wanlin Dai
- Innovation Institute of China Medical University, Shenyang, Liaoning, China
| | - Shuodong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Kong
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
7
|
Jin C, Kumar P, Gracia-Sancho J, Dufour JF. Calcium transfer between endoplasmic reticulum and mitochondria in liver diseases. FEBS Lett 2021; 595:1411-1421. [PMID: 33752262 DOI: 10.1002/1873-3468.14078] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/21/2021] [Accepted: 02/25/2021] [Indexed: 01/07/2023]
Abstract
Calcium (Ca2+ ) is a second messenger essential for cellular homeostasis. Inside the cell, Ca2+ is compartmentalized and exchanged among organelles in response to both external and internal stimuli. Mitochondria-associated membranes (MAMs) provide a platform for proteins and channels involved in Ca2+ transfer between the endoplasmic reticulum (ER) and mitochondria. Deregulated Ca2+ signaling and proteins regulating ER-mitochondria interactions have been linked to liver diseases and intensively investigated in recent years. In this review, we summarize the role of MAM-resident proteins in Ca2+ transfer and their association with different liver diseases.
Collapse
Affiliation(s)
- Chaonan Jin
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland
| | - Pavitra Kumar
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland
| | - Jordi Gracia-Sancho
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland.,Liver Vascular Biology Research Group, CIBEREHD, IDIBAPS Research Institute, Barcelona, Spain
| | - Jean-François Dufour
- Hepatology, Department for BioMedical Research, University of Bern, Switzerland.,University Clinic of Visceral Surgery and Medicine, Inselspital, Bern, Switzerland
| |
Collapse
|
8
|
Liang L, Zhu K, Tao J, Lu S. ORN: Inferring patient-specific dysregulation status of pathway modules in cancer with OR-gate Network. PLoS Comput Biol 2021; 17:e1008792. [PMID: 33819263 PMCID: PMC8049496 DOI: 10.1371/journal.pcbi.1008792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 04/15/2021] [Accepted: 02/15/2021] [Indexed: 01/26/2023] Open
Abstract
Pathway level understanding of cancer plays a key role in precision oncology. However, the current amount of high-throughput data cannot support the elucidation of full pathway topology. In this study, instead of directly learning the pathway network, we adapted the probabilistic OR gate to model the modular structure of pathways and regulon. The resulting model, OR-gate Network (ORN), can simultaneously infer pathway modules of somatic alterations, patient-specific pathway dysregulation status, and downstream regulon. In a trained ORN, the differentially expressed genes (DEGs) in each tumour can be explained by somatic mutations perturbing a pathway module. Furthermore, the ORN handles one of the most important properties of pathway perturbation in tumours, the mutual exclusivity. We have applied the ORN to lower-grade glioma (LGG) samples and liver hepatocellular carcinoma (LIHC) samples in TCGA and breast cancer samples from METABRIC. Both datasets have shown abnormal pathway activities related to immune response and cell cycles. In LGG samples, ORN identified pathway modules closely related to glioma development and revealed two pathways closely related to patient survival. We had similar results with LIHC samples. Additional results from the METABRIC datasets showed that ORN could characterize critical mechanisms of cancer and connect them to less studied somatic mutations (e.g., BAP1, MIR604, MICAL3, and telomere activities), which may generate novel hypothesis for targeted therapy.
Collapse
Affiliation(s)
- Lifan Liang
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Kunju Zhu
- Clinical Medicine Research Institute, Jinan University, Guangzhou, Guangdong, China
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Songjian Lu
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
9
|
Gaspers LD, Thomas AP, Hoek JB, Bartlett PJ. Ethanol Disrupts Hormone-Induced Calcium Signaling in Liver. FUNCTION (OXFORD, ENGLAND) 2021; 2:zqab002. [PMID: 33604575 PMCID: PMC7875097 DOI: 10.1093/function/zqab002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/22/2020] [Accepted: 12/30/2020] [Indexed: 01/06/2023]
Abstract
Receptor-coupled phospholipase C (PLC) is an important target for the actions of ethanol. In the ex vivo perfused rat liver, concentrations of ethanol >100 mM were required to induce a rise in cytosolic calcium (Ca2+) suggesting that these responses may only occur after binge ethanol consumption. Conversely, pharmacologically achievable concentrations of ethanol (≤30 mM) decreased the frequency and magnitude of hormone-stimulated cytosolic and nuclear Ca2+ oscillations and the parallel translocation of protein kinase C-β to the membrane. Ethanol also inhibited gap junction communication resulting in the loss of coordinated and spatially organized intercellular Ca2+ waves in hepatic lobules. Increasing the hormone concentration overcame the effects of ethanol on the frequency of Ca2+ oscillations and amplitude of the individual Ca2+ transients; however, the Ca2+ responses in the intact liver remained disorganized at the intercellular level, suggesting that gap junctions were still inhibited. Pretreating hepatocytes with an alcohol dehydrogenase inhibitor suppressed the effects of ethanol on hormone-induced Ca2+ increases, whereas inhibiting aldehyde dehydrogenase potentiated the inhibitory actions of ethanol, suggesting that acetaldehyde is the underlying mediator. Acute ethanol intoxication inhibited the rate of rise and the magnitude of hormone-stimulated production of inositol 1,4,5-trisphosphate (IP3), but had no effect on the size of Ca2+ spikes induced by photolysis of caged IP3. These findings suggest that ethanol inhibits PLC activity, but does not affect IP3 receptor function. We propose that by suppressing hormone-stimulated PLC activity, ethanol interferes with the dynamic modulation of [IP3] that is required to generate large, amplitude Ca2+ oscillations.
Collapse
Affiliation(s)
- Lawrence D Gaspers
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA,Address correspondence to L.D.G. (e-mail: )
| | - Andrew P Thomas
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Jan B Hoek
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Paula J Bartlett
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
10
|
Lemos FO, Guerra MT, Leite MDF. Inositol 1,4,5 trisphosphate receptors in secretory epithelial cells of the gastrointestinal tract. CURRENT OPINION IN PHYSIOLOGY 2020. [DOI: 10.1016/j.cophys.2020.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Lima Filho ACM, França A, Florentino RM, Dos Santos ML, de Oliveira Lemos F, Missiaggia DG, Fonseca RC, Gustavo Oliveira A, Ananthanarayanan M, Guerra MT, de Castro Fonseca M, Vidigal PVT, Lima CX, Nathanson MH, Fatima Leite M. Inositol 1,4,5-trisphosphate receptor type 3 plays a protective role in hepatocytes during hepatic ischemia-reperfusion injury. Cell Calcium 2020; 91:102264. [PMID: 32957029 DOI: 10.1016/j.ceca.2020.102264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 01/02/2023]
Abstract
Hepatic ischemia-reperfusion injury is seen in a variety of clinical conditions, including hepatic thrombosis, systemic hypotension, and liver transplantation. Calcium (Ca2+) signaling mediates several pathophysiological processes in the liver, but it is not known whether and how intracellular Ca2+ channels are involved in the hepatocellular events secondary to ischemia-reperfusion. Using an animal model of hepatic ischemia-reperfusion injury, we observed a progressive increase in expression of the type 3 isoform of the inositol trisphosphate receptor (ITPR3), an intracellular Ca2+ channel that is not normally expressed in healthy hepatocytes. ITPR3 expression was upregulated, at least in part, by a combination of demethylation of the ITPR3 promoter region and the increased transcriptional activity of the nuclear factor of activated T-cells (NFAT). Additionally, expression of pro-inflammatory interleukins and necrotic surface area were less pronounced in livers of control animals compared to liver-specific ITPR3 KO mice subjected to hepatic damage. Corroborating these findings, ITPR3 expression and activation of NFAT were observed in hepatocytes of liver biopsies from patients who underwent liver ischemia caused by thrombosis after organ transplant. Together, these results are consistent with the idea that ITPR3 expression in hepatocytes plays a protective role during hepatic injury induced by ischemia-reperfusion.
Collapse
Affiliation(s)
| | - Andressa França
- Department of Molecular Medicine, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | - Rodrigo M Florentino
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | | | | | | | | | - André Gustavo Oliveira
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| | | | - Mateus T Guerra
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, CT, United States.
| | - Matheus de Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials, SP, Brazil.
| | | | - Cristiano Xavier Lima
- Department of Surgery, Medicine School of Federal University of Minas Gerais (UFMG), MG, United States.
| | - Michael H Nathanson
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, CT, United States.
| | - M Fatima Leite
- Department of Biophysics and Physiology, Federal University of Minas Gerais (UFMG), MG, Brazil.
| |
Collapse
|
12
|
Lemos FDO, França A, Lima Filho ACM, Florentino RM, Santos ML, Missiaggia DG, Rodrigues GOL, Dias FF, Souza Passos IB, Teixeira MM, Andrade AMDF, Lima CX, Vidigal PVT, Costa VV, Fonseca MC, Nathanson MH, Leite MF. Molecular Mechanism for Protection Against Liver Failure in Human Yellow Fever Infection. Hepatol Commun 2020; 4:657-669. [PMID: 32363317 PMCID: PMC7193135 DOI: 10.1002/hep4.1504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/18/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Yellow fever (YF) is a viral hemorrhagic fever that typically involves the liver. Brazil recently experienced its largest recorded YF outbreak, and the disease was fatal in more than a third of affected individuals, mostly because of acute liver failure. Affected individuals are generally treated only supportively, but during the recent Brazilian outbreak, selected patients were treated with liver transplant. We took advantage of this clinical experience to better characterize the clinical and pathological features of YF-induced liver failure and to examine the mechanism of hepatocellular injury in YF, to identify targets that would be amenable to therapeutic intervention in preventing progression to liver failure and death. Patients with YF liver failure rapidly developed massive transaminase elevations, with jaundice, coagulopathy, thrombocytopenia, and usually hepatic encephalopathy, along with pathological findings that included microvesicular steatosis and lytic necrosis. Hepatocytes began to express the type 3 isoform of the inositol trisphosphate receptor (ITPR3), an intracellular calcium (Ca2+) channel that is not normally expressed in hepatocytes. Experiments in an animal model, isolated hepatocytes, and liver-derived cell lines showed that this new expression of ITPR3 was associated with increased nuclear Ca2+ signaling and hepatocyte proliferation, and reduced steatosis and cell death induced by the YF virus. Conclusion: Yellow fever often induces liver failure characterized by massive hepatocellular damage plus steatosis. New expression of ITPR3 also occurs in YF-infected hepatocytes, which may represent an endogenous protective mechanism that could suggest approaches to treat affected individuals before they progress to liver failure, thereby decreasing the mortality of this disease in a way that does not rely on the costly and limited resource of liver transplantation.
Collapse
Affiliation(s)
| | - Andressa França
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Rodrigo M. Florentino
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Marcone Loiola Santos
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | - Dabny G. Missiaggia
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Felipe Ferraz Dias
- Center of MicroscopyUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Mauro M. Teixeira
- Department of Biochemistry and ImmunologyUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | - Cristiano Xavier Lima
- Hepatic Transplant ServiceHospital Felício RochoBelo HorizonteBrazil
- SurgeryUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| | | | | | - Matheus Castro Fonseca
- Brazilian Biosciences National Laboratory (LNBio)Brazilian Center for Research in Energy and MaterialsRua Giuseppe Máximo ScolfaroCampinasBrazil
| | - Michael H. Nathanson
- Section of Digestive DiseasesDepartment of Internal MedicineYale University School of MedicineNew HavenCT
| | - M. Fatima Leite
- Department of Physiology and BiophysicsUniversidade Federal de Minas GeraisBelo HorizonteBrazil
| |
Collapse
|
13
|
Alogaili F, Chinnarasu S, Jaeschke A, Kranias EG, Hui DY. Hepatic HAX-1 inactivation prevents metabolic diseases by enhancing mitochondrial activity and bile salt export. J Biol Chem 2020; 295:4631-4646. [PMID: 32079675 DOI: 10.1074/jbc.ra119.012361] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/04/2020] [Indexed: 12/26/2022] Open
Abstract
Increasing hepatic mitochondrial activity through pyruvate dehydrogenase and elevating enterohepatic bile acid recirculation are promising new approaches for metabolic disease therapy, but neither approach alone can completely ameliorate disease phenotype in high-fat diet-fed mice. This study showed that diet-induced hepatosteatosis, hyperlipidemia, and insulin resistance can be completely prevented in mice with liver-specific HCLS1-associated protein X-1 (HAX-1) inactivation. Mechanistically, we showed that HAX-1 interacts with inositol 1,4,5-trisphosphate receptor-1 (InsP3R1) in the liver, and its absence reduces InsP3R1 levels, thereby improving endoplasmic reticulum-mitochondria calcium homeostasis to prevent excess calcium overload and mitochondrial dysfunction. As a result, HAX-1 ablation activates pyruvate dehydrogenase and increases mitochondria utilization of glucose and fatty acids to prevent hepatosteatosis, hyperlipidemia, and insulin resistance. In contrast to the reduction of InsP3R1 levels, hepatic HAX-1 deficiency increases bile salt exporter protein levels, thereby promoting enterohepatic bile acid recirculation, leading to activation of bile acid-responsive genes in the intestinal ileum to augment insulin sensitivity and of cholesterol transport genes in the liver to suppress hyperlipidemia. The dual mechanisms of increased mitochondrial respiration and enterohepatic bile acid recirculation due to improvement of endoplasmic reticulum-mitochondria calcium homeostasis with hepatic HAX-1 inactivation suggest that this may be a potential therapeutic target for metabolic disease intervention.
Collapse
Affiliation(s)
- Fawzi Alogaili
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237.,Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Sivaprakasam Chinnarasu
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Anja Jaeschke
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - David Y Hui
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Research Center, University of Cincinnati College of Medicine, Cincinnati, Ohio 45237
| |
Collapse
|
14
|
Ueasilamongkol P, Khamphaya T, Guerra MT, Rodrigues M, Gomes DA, Kong Y, Wei W, Jain D, Trampert DC, Ananthanarayanan M, Banales JM, Roberts LR, Farshidfar F, Nathanson MH, Weerachayaphorn J. Type 3 Inositol 1,4,5-Trisphosphate Receptor Is Increased and Enhances Malignant Properties in Cholangiocarcinoma. Hepatology 2020; 71:583-599. [PMID: 31251815 PMCID: PMC6934938 DOI: 10.1002/hep.30839] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
Cholangiocarcinoma (CCA) is the second most common malignancy arising in the liver. It carries a poor prognosis, in part because its pathogenesis is not well understood. The type 3 inositol 1,4,5-trisphosphate receptor (ITPR3) is the principal intracellular calcium ion (Ca2+ ) release channel in cholangiocytes, and its increased expression has been related to the pathogenesis of malignancies in other types of tissues, so we investigated its role in CCA. ITPR3 expression was increased in both hilar and intrahepatic CCA samples as well as in CCA cell lines. Deletion of ITPR3 from CCA cells impaired proliferation and cell migration. A bioinformatic analysis suggested that overexpression of ITPR3 in CCA would have a mitochondrial phenotype, so this was also examined. ITPR3 normally is concentrated in a subapical region of endoplasmic reticulum (ER) in cholangiocytes, but both immunogold electron microscopy and super-resolution microscopy showed that ITPR3 in CCA cells was also in regions of ER in close association with mitochondria. Deletion of ITPR3 from these cells impaired mitochondrial Ca2+ signaling and led to cell death. Conclusion: ITPR3 expression in cholangiocytes becomes enhanced in CCA. This contributes to malignant features, including cell proliferation and migration and enhanced mitochondrial Ca2+ signaling.
Collapse
Affiliation(s)
| | - Tanaporn Khamphaya
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Mateus T. Guerra
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michele Rodrigues
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dawidson A. Gomes
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yong Kong
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut, USA
| | - Wei Wei
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut, USA
| | - Dhanpat Jain
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - David C. Trampert
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBERehd, Ikerbasque, San Sebastian, Spain
| | - Lewis R. Roberts
- Divisions of Gastroenterology and Hepatology and Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Farshad Farshidfar
- Department of Oncology, Cumming School of Medicine, University of Calgary, Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Canada
| | - Michael H. Nathanson
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jittima Weerachayaphorn
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
15
|
Lemos FDO, Florentino RM, Lima Filho ACM, Santos MLD, Leite MF. Inositol 1,4,5-trisphosphate receptor in the liver: Expression and function. World J Gastroenterol 2019; 25:6483-6494. [PMID: 31802829 PMCID: PMC6886013 DOI: 10.3748/wjg.v25.i44.6483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/22/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The liver is a complex organ that performs several functions to maintain homeostasis. These functions are modulated by calcium, a second messenger that regulates several intracellular events. In hepatocytes and cholangiocytes, which are the epithelial cell types in the liver, inositol 1,4,5-trisphosphate (InsP3) receptors (ITPR) are the only intracellular calcium release channels. Three isoforms of the ITPR have been described, named type 1, type 2 and type 3. These ITPR isoforms are differentially expressed in liver cells where they regulate distinct physiological functions. Changes in the expression level of these receptors correlate with several liver diseases and hepatic dysfunctions. In this review, we highlight how the expression level, modulation, and localization of ITPR isoforms in hepatocytes and cholangiocytes play a role in hepatic homeostasis and liver pathology.
Collapse
Affiliation(s)
- Fernanda de Oliveira Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Rodrigo M Florentino
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Antônio Carlos Melo Lima Filho
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Marcone Loiola dos Santos
- Department of Cell Biology, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - M Fatima Leite
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| |
Collapse
|
16
|
Type 3 inositol 1,4,5-trisphosphate receptor: A calcium channel for all seasons. Cell Calcium 2019; 85:102132. [PMID: 31790953 DOI: 10.1016/j.ceca.2019.102132] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022]
Abstract
Inositol 1,4,5 trisphosphate receptors (ITPRs) are a family of endoplasmic reticulum Ca2+ channels essential for the control of intracellular Ca2+ levels in virtually every mammalian cell type. The three isoforms (ITPR1, ITPR2 and ITPR3) are highly homologous in amino acid sequence, but they differ considerably in terms of biophysical properties, subcellular localization, and tissue distribution. Such differences underscore the variety of cellular responses triggered by each isoform and suggest that the expression/activity of specific isoforms might be linked to particular pathophysiological states. Indeed, recent findings demonstrate that changes in expression of ITPR isoforms are associated with a number of human diseases ranging from fatty liver disease to cancer. ITPR3 is emerging as the isoform that is particularly important in the pathogenesis of various human diseases. Here we review the physiological and pathophysiological roles of ITPR3 in various tissues and the mechanisms by which the expression of this isoform is modulated in health and disease.
Collapse
|
17
|
Deferm N, De Vocht T, Qi B, Van Brantegem P, Gijbels E, Vinken M, de Witte P, Bouillon T, Annaert P. Current insights in the complexities underlying drug-induced cholestasis. Crit Rev Toxicol 2019; 49:520-548. [PMID: 31589080 DOI: 10.1080/10408444.2019.1635081] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Drug-induced cholestasis (DIC) poses a major challenge to the pharmaceutical industry and regulatory agencies. It causes both drug attrition and post-approval withdrawal of drugs. DIC represents itself as an impaired secretion and flow of bile, leading to the pathological hepatic and/or systemic accumulation of bile acids (BAs) and their conjugate bile salts. Due to the high number of mechanisms underlying DIC, predicting a compound's cholestatic potential during early stages of drug development remains elusive. A profound understanding of the different molecular mechanisms of DIC is, therefore, of utmost importance. Although many knowledge gaps and caveats still exist, it is generally accepted that alterations of certain hepatobiliary membrane transporters and changes in hepatocellular morphology may cause DIC. Consequently, liver models, which represent most of these mechanisms, are valuable tools to predict human DIC. Some of these models, such as membrane-based in vitro models, are exceptionally well-suited to investigate specific mechanisms (i.e. transporter inhibition) of DIC, while others, such as liver slices, encompass all relevant biological processes and, therefore, offer a better representation of the in vivo situation. In the current review, we highlight the principal molecular mechanisms associated with DIC and offer an overview and critical appraisal of the different liver models that are currently being used to predict the cholestatic potential of drugs.
Collapse
Affiliation(s)
- Neel Deferm
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Tom De Vocht
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Bing Qi
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Pieter Van Brantegem
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Eva Gijbels
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mathieu Vinken
- Entity of In Vitro Toxicology and Dermato-Cosmetology, Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Peter de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Thomas Bouillon
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Pieter Annaert
- Department of Pharmaceutical and Pharmacological Sciences, Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Guerra MT, Florentino RM, Franca A, Filho ACL, dos Santos ML, Fonseca RC, Lemos FO, Fonseca MC, Kruglov E, Mennone A, Njei B, Gibson J, Guan F, Cheng YC, Ananthanarayanam M, Gu J, Jiang J, Zhao H, Lima CX, Vidigal PT, Oliveira AG, Nathanson MH, Leite MF. Expression of the type 3 InsP 3 receptor is a final common event in the development of hepatocellular carcinoma. Gut 2019; 68:1676-1687. [PMID: 31315892 PMCID: PMC7087395 DOI: 10.1136/gutjnl-2018-317811] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 06/25/2019] [Accepted: 06/30/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND & OBJECTIVES Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide. Several types of chronic liver disease predispose to HCC, and several different signalling pathways have been implicated in its pathogenesis, but no common molecular event has been identified. Ca2+ signalling regulates the proliferation of both normal hepatocytes and liver cancer cells, so we investigated the role of intracellular Ca2+ release channels in HCC. DESIGN Expression analyses of the type 3 isoform of the inositol 1, 4, 5-trisphosphate receptor (ITPR3) in human liver samples, liver cancer cells and mouse liver were combined with an evaluation of DNA methylation profiles of ITPR3 promoter in HCC and characterisation of the effects of ITPR3 expression on cellular proliferation and apoptosis. The effects of de novo ITPR3 expression on hepatocyte calcium signalling and liver growth were evaluated in mice. RESULTS ITPR3 was absent or expressed in low amounts in hepatocytes from normal liver, but was expressed in HCC specimens from three independent patient cohorts, regardless of the underlying cause of chronic liver disease, and its increased expression level was associated with poorer survival. The ITPR3 gene was heavily methylated in control liver specimens but was demethylated at multiple sites in specimens of patient with HCC. Administration of a demethylating agent in a mouse model resulted in ITPR3 expression in discrete areas of the liver, and Ca2+ signalling was enhanced in these regions. In addition, cell proliferation and liver regeneration were enhanced in the mouse model, and deletion of ITPR3 from human HCC cells enhanced apoptosis. CONCLUSIONS These results provide evidence that de novo expression of ITPR3 typically occurs in HCC and may play a role in its pathogenesis.
Collapse
MESH Headings
- Adult
- Animals
- Apoptosis/physiology
- Calcium Signaling/physiology
- Carcinogenesis/genetics
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Proliferation/physiology
- Cells, Cultured
- DNA Methylation
- Female
- Gene Expression Regulation, Neoplastic/physiology
- Hepatocytes/metabolism
- Humans
- Inositol 1,4,5-Trisphosphate Receptors/deficiency
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Liver/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Regeneration/physiology
- Male
- Mice, Knockout
- Middle Aged
- Survival Analysis
Collapse
Affiliation(s)
- Mateus T Guerra
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Rodrigo M Florentino
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andressa Franca
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antonio C Lima Filho
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcone L dos Santos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Roberta C Fonseca
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda O Lemos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus C Fonseca
- Centro Nacional de Pesquisa em Energia e Materiais, Campinas, Brazil
| | - Emma Kruglov
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Albert Mennone
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Basile Njei
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Joanna Gibson
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Fulan Guan
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yung-Chi Cheng
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jianlei Gu
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Bioinformatics and Biostatistics, SJTU-Yale Joint Center for Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, China
| | - Jianping Jiang
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Bioinformatics and Biostatistics, SJTU-Yale Joint Center for Biostatistics, School of Life Science and Biotechnology, Shanghai Jiao Tong University, China
| | - Hongyu Zhao
- Department of Biostatistics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Cristiano X Lima
- Department of Surgery, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paula T Vidigal
- Department of Pathological Anatomy and Forensic Medicine, School of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Andre G Oliveira
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michael H Nathanson
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Maria Fatima Leite
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
19
|
Franca A, Filho ACML, Guerra MT, Weerachayaphorn J, dos Santos ML, Njei B, Robert M, Lima CX, Vidigal PVT, Banales JM, Ananthanarayanam M, Leite MF, Nathanson MH. Effects of Endotoxin on Type 3 Inositol 1,4,5-Trisphosphate Receptor in Human Cholangiocytes. Hepatology 2019; 69:817-830. [PMID: 30141207 PMCID: PMC6351171 DOI: 10.1002/hep.30228] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022]
Abstract
Clinical conditions that result in endotoxemia, such as sepsis and alcoholic hepatitis (AH), often are accompanied by cholestasis. Although hepatocellular changes in response to lipopolysaccharide (LPS) have been well characterized, less is known about whether and how cholangiocytes contribute to this form of cholestasis. We examined effects of endotoxin on expression and function of the type 3 inositol trisphosphate receptor (ITPR3), because this is the main intracellular Ca2+ release channel in cholangiocytes, and loss of it impairs ductular bicarbonate secretion. Bile duct cells expressed the LPS receptor, Toll-like receptor 4 (TLR4), which links to activation of nuclear factor-κB (NF-κB). Analysis of the human ITPR3 promoter revealed five putative response elements to NF-κB, and promoter activity was inhibited by p65/p50. Nested 0.5- and 1.0-kilobase (kb) deletion fragments of the ITPR3 promoter were inhibited by NF-κB subunits. Chromatin immunoprecipitation (ChIP) assay showed that NF-κB interacts with the ITPR3 promoter, with an associated increase in H3K9 methylation. LPS decreased ITPR3 mRNA and protein expression and also decreased sensitivity of bile duct cells to calcium agonist stimuli. This reduction was reversed by inhibition of TLR4. ITPR3 expression was decreased or absent in cholangiocytes from patients with cholestasis of sepsis and from those with severe AH. Conclusion: Stimulation of TLR4 by LPS activates NF-κB to down-regulate ITPR3 expression in human cholangiocytes. This may contribute to the cholestasis that can be observed in conditions such as sepsis or AH.
Collapse
Affiliation(s)
- Andressa Franca
- Federal University of Minas Gerais (UFMG), Belo Horizonte, MG
| | | | - Mateus T. Guerra
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Jittima Weerachayaphorn
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Basile Njei
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| | - Marie Robert
- Department of Pathology, Yale University School of Medicine, New Haven, CT
| | | | | | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | | | - M. Fatima Leite
- Federal University of Minas Gerais (UFMG), Belo Horizonte, MG
| | - Michael H. Nathanson
- Section of Digestive Disease, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
20
|
Verma A, Antony AN, Ogunnaike BA, Hoek JB, Vadigepalli R. Causality Analysis and Cell Network Modeling of Spatial Calcium Signaling Patterns in Liver Lobules. Front Physiol 2018; 9:1377. [PMID: 30337879 PMCID: PMC6180170 DOI: 10.3389/fphys.2018.01377] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 09/11/2018] [Indexed: 01/21/2023] Open
Abstract
Dynamics as well as localization of Ca2+ transients plays a vital role in liver function under homeostatic conditions, repair, and disease. In response to circulating hormonal stimuli, hepatocytes exhibit intracellular Ca2+ responses that propagate through liver lobules in a wave-like fashion. Although intracellular processes that control cell autonomous Ca2+ spiking behavior have been studied extensively, the intra- and inter-cellular signaling factors that regulate lobular scale spatial patterns and wave-like propagation of Ca2+ remain to be determined. To address this need, we acquired images of cytosolic Ca2+ transients in 1300 hepatocytes situated across several mouse liver lobules over a period of 1600 s. We analyzed this time series data using correlation network analysis, causal network analysis, and computational modeling, to characterize the spatial distribution of heterogeneity in intracellular Ca2+ signaling components as well as intercellular interactions that control lobular scale Ca2+ waves. Our causal network analysis revealed that hepatocytes are causally linked to multiple other co-localized hepatocytes, but these influences are not necessarily aligned uni-directionally along the sinusoids. Our computational model-based analysis showed that spatial gradients of intracellular Ca2+ signaling components as well as intercellular molecular exchange are required for lobular scale propagation of Ca2+ waves. Additionally, our analysis suggested that causal influences of hepatocytes on Ca2+ responses of multiple neighbors lead to robustness of Ca2+ wave propagation through liver lobules.
Collapse
Affiliation(s)
- Aalap Verma
- Department of Biomedical Engineering, University of Delaware, Newark, DE, United States.,Department of Pathology, Anatomy and Cell Biology, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Anil Noronha Antony
- Department of Pathology, Anatomy and Cell Biology, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Babatunde A Ogunnaike
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, United States
| | - Jan B Hoek
- Department of Pathology, Anatomy and Cell Biology, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Rajanikanth Vadigepalli
- Department of Pathology, Anatomy and Cell Biology, Daniel Baugh Institute for Functional Genomics and Computational Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
21
|
Oliva-Vilarnau N, Hankeova S, Vorrink SU, Mkrtchian S, Andersson ER, Lauschke VM. Calcium Signaling in Liver Injury and Regeneration. Front Med (Lausanne) 2018; 5:192. [PMID: 30023358 PMCID: PMC6039545 DOI: 10.3389/fmed.2018.00192] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/11/2018] [Indexed: 12/12/2022] Open
Abstract
The liver fulfills central roles in metabolic control and detoxification and, as such, is continuously exposed to a plethora of insults. Importantly, the liver has a unique ability to regenerate and can completely recoup from most acute, non-iterative insults. However, multiple conditions, including viral hepatitis, non-alcoholic fatty liver disease (NAFLD), long-term alcohol abuse and chronic use of certain medications, can cause persistent injury in which the regenerative capacity eventually becomes dysfunctional, resulting in hepatic scaring and cirrhosis. Calcium is a versatile secondary messenger that regulates multiple hepatic functions, including lipid and carbohydrate metabolism, as well as bile secretion and choleresis. Accordingly, dysregulation of calcium signaling is a hallmark of both acute and chronic liver diseases. In addition, recent research implicates calcium transients as essential components of liver regeneration. In this review, we provide a comprehensive overview of the role of calcium signaling in liver health and disease and discuss the importance of calcium in the orchestration of the ensuing regenerative response. Furthermore, we highlight similarities and differences in spatiotemporal calcium regulation between liver insults of different etiologies. Finally, we discuss intracellular calcium control as an emerging therapeutic target for liver injury and summarize recent clinical findings of calcium modulation for the treatment of ischemic-reperfusion injury, cholestasis and NAFLD.
Collapse
Affiliation(s)
- Nuria Oliva-Vilarnau
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Simona Hankeova
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,Faculty of Science, Institute of Experimental Biology, Masaryk University, Brno, Czechia
| | - Sabine U Vorrink
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Souren Mkrtchian
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Emma R Andersson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Volker M Lauschke
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Trampert DC, Nathanson MH. Regulation of bile secretion by calcium signaling in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1761-1770. [PMID: 29787781 DOI: 10.1016/j.bbamcr.2018.05.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/12/2018] [Accepted: 05/16/2018] [Indexed: 12/15/2022]
Abstract
Calcium (Ca2+) signaling controls secretion in many types of cells and tissues. In the liver, Ca2+ regulates secretion in both hepatocytes, which are responsible for primary formation of bile, and cholangiocytes, which line the biliary tree and further condition the bile before it is secreted. Cholestatic liver diseases, which are characterized by impaired bile secretion, may result from impaired Ca2+ signaling mechanisms in either hepatocytes or cholangiocytes. This review will discuss the Ca2+ signaling machinery and mechanisms responsible for regulation of secretion in both hepatocytes and cholangiocytes, and the pathophysiological changes in Ca2+ signaling that can occur in each of these cell types to result in cholestasis.
Collapse
Affiliation(s)
- David C Trampert
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA
| | - Michael H Nathanson
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA.
| |
Collapse
|
23
|
Ryan J, Morgan RE, Chen Y, Volak LP, Dunn RT, Dunn KW. Intravital Multiphoton Microscopy with Fluorescent Bile Salts in Rats as an In Vivo Biomarker for Hepatobiliary Transport Inhibition. Drug Metab Dispos 2018; 46:704-718. [PMID: 29467212 DOI: 10.1124/dmd.117.079277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/15/2018] [Indexed: 12/27/2022] Open
Abstract
The bile salt export pump (BSEP) is expressed at the canalicular domain of hepatocytes, where it mediates the elimination of monovalent bile salts into the bile. Inhibition of BSEP is considered a susceptibility factor for drug-induced liver injury that often goes undetected during nonclinical testing. Although in vitro assays exist for screening BSEP inhibition, a reliable and specific method for confirming Bsep inhibition in vivo would be a valuable follow up to a BSEP screening strategy, helping to put a translatable context around in vitro inhibition data, incorporating processes such as metabolism, protein binding, and other exposure properties that are lacking in most in vitro BSEP models. Here, we describe studies in which methods of quantitative intravital microscopy were used to identify dose-dependent effects of two known BSEP/Bsep inhibitors, 2-[4-[4-(butylcarbamoyl)-2-[(2,4-dichlorophenyl)sulfonylamino]phenoxy]-3-methoxyphenyl]acetic acid (AMG-009) and bosentan, on hepatocellular transport of the fluorescent bile salts cholylglycyl amidofluorescein and cholyl-lysyl-fluorescein in rats. Results of these studies demonstrate that the intravital microscopy approach is capable of detecting Bsep inhibition at drug doses well below those found to increase serum bile acid levels, and also indicate that basolateral efflux transporters play a significant role in preventing cytosolic accumulation of bile acids under conditions of Bsep inhibition in rats. Studies of this kind can both improve our understanding of exposures needed to inhibit Bsep in vivo and provide unique insights into drug effects in ways that can improve our ability interpret animal studies for the prediction of human drug hepatotoxicity.
Collapse
Affiliation(s)
- Jennifer Ryan
- Division of Nephrology, Department of Medicine, Indiana University Medical Center, Indianapolis, Indiana (J.R., K.W.D.); Department of Comparative Biology and Safety Sciences, Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (R.E.M., Y.C., L.P.V., R.T.D.)
| | - Ryan E Morgan
- Division of Nephrology, Department of Medicine, Indiana University Medical Center, Indianapolis, Indiana (J.R., K.W.D.); Department of Comparative Biology and Safety Sciences, Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (R.E.M., Y.C., L.P.V., R.T.D.)
| | - Yuan Chen
- Division of Nephrology, Department of Medicine, Indiana University Medical Center, Indianapolis, Indiana (J.R., K.W.D.); Department of Comparative Biology and Safety Sciences, Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (R.E.M., Y.C., L.P.V., R.T.D.)
| | - Laurie P Volak
- Division of Nephrology, Department of Medicine, Indiana University Medical Center, Indianapolis, Indiana (J.R., K.W.D.); Department of Comparative Biology and Safety Sciences, Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (R.E.M., Y.C., L.P.V., R.T.D.)
| | - Robert T Dunn
- Division of Nephrology, Department of Medicine, Indiana University Medical Center, Indianapolis, Indiana (J.R., K.W.D.); Department of Comparative Biology and Safety Sciences, Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (R.E.M., Y.C., L.P.V., R.T.D.)
| | - Kenneth W Dunn
- Division of Nephrology, Department of Medicine, Indiana University Medical Center, Indianapolis, Indiana (J.R., K.W.D.); Department of Comparative Biology and Safety Sciences, Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (R.E.M., Y.C., L.P.V., R.T.D.)
| |
Collapse
|
24
|
Khamphaya T, Chukijrungroat N, Saengsirisuwan V, Mitchell-Richards KA, Robert ME, Mennone A, Nathanson MH, Weerachayaphorn J, Weerachayaphorn J. Nonalcoholic fatty liver disease impairs expression of the type II inositol 1,4,5-trisphosphate receptor. Hepatology 2018; 67:560-574. [PMID: 29023819 PMCID: PMC5893412 DOI: 10.1002/hep.29588] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/07/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disease worldwide. It may result in several types of liver problems, including impaired liver regeneration (LR), but the mechanism for this is unknown. Because LR depends on calcium signaling, we examined the effects of NAFLD on expression of the type II inositol 1,4,5-trisphosphate receptor (ITPR2), the principle calcium release channel in hepatocytes. ITPR2 promoter activity was measured in Huh7 and HepG2 cells. ITPR2 and c-Jun protein levels were evaluated in Huh7 cells, in liver tissue from a rat model of NAFLD, and in liver biopsy specimens of patients with simple steatosis and nonalcoholic steatohepatitis (NASH). LR was assessed in wild-type and Itpr2 knockout (Itpr2-/- ) mice following 67% hepatectomy. Cell proliferation was examined in ITPR2-knockout HepG2 cells generated by the CRISPR/Cas9 system. c-Jun dose dependently decreased activity of the human ITPR2 promoter. c-Jun expression was increased and ITPR2 was decreased in fat-loaded Huh7 cells and in livers of rats fed a high-fat, high-fructose diet. Overexpression of c-Jun reduced protein and mRNA expression of ITPR2 in Huh7 cells, whereas knockdown of c-Jun prevented the decrease of ITPR2 in fat-loaded Huh7 cells. ITPR2 expression was decreased and c-Jun was increased in liver biopsies of patients with steatosis and NASH compared to controls. ITPR2-knockout cells exhibited less nuclear calcium signaling and cell proliferation than control cells. LR assessed by Ki-67 and proliferating cell nuclear antigen was markedly decreased in Itpr2-/- mice. Conclusion: Fatty liver induces a c-Jun-mediated decrease in ITPR2 in hepatocytes. This may account for the impaired LR that occurs in NAFLD. (Hepatology 2018;67:560-574).
Collapse
Affiliation(s)
- Tanaporn Khamphaya
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Natsasi Chukijrungroat
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Vitoon Saengsirisuwan
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Marie E. Robert
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut 06519, USA
| | - Albert Mennone
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven 06519, Connecticut, USA
| | - Michael H. Nathanson
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven 06519, Connecticut, USA,Corresponding Authors: Michael H. Nathanson, M.D., Ph.D., Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06519, USA. Phone: (+1) 203-785-7312; Fax: (+1) 203-785-7273, ; Jittima Weerachayaphorn, Ph.D., Department of Physiology, Faculty of Science, Mahidol University, Rama 6 Road, Ratchathewi, Bangkok 10400, Thailand. Phone: (+66) 2201-5514; Fax: (+66) 2354-7154, ,
| | - Jittima Weerachayaphorn
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand,Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven 06519, Connecticut, USA,Corresponding Authors: Michael H. Nathanson, M.D., Ph.D., Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06519, USA. Phone: (+1) 203-785-7312; Fax: (+1) 203-785-7273, ; Jittima Weerachayaphorn, Ph.D., Department of Physiology, Faculty of Science, Mahidol University, Rama 6 Road, Ratchathewi, Bangkok 10400, Thailand. Phone: (+66) 2201-5514; Fax: (+66) 2354-7154, ,
| | - Jittima Weerachayaphorn
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
25
|
Dunn KW, Ryan JC. Using quantitative intravital multiphoton microscopy to dissect hepatic transport in rats. Methods 2017; 128:40-51. [PMID: 28434905 DOI: 10.1016/j.ymeth.2017.04.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 12/24/2022] Open
Abstract
Hepatic solute transport is a complex process whose disruption is associated with liver disease and drug-induced liver injury. Intravital multiphoton fluorescence excitation microscopy provides the spatial and temporal resolution necessary to characterize hepatic transport at the level of individual hepatocytes in vivo and thus to identify the mechanisms and cellular consequences of cholestasis. Here we present an overview of the use of fluorescence microscopy for studies of hepatic transport in living animals, and describe how we have combined methods of intravital microscopy and digital image analysis to dissect the effects of drugs and pathological conditions on the function of hepatic transporters in vivo.
Collapse
Affiliation(s)
- Kenneth W Dunn
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, United States.
| | - Jennifer C Ryan
- Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|
26
|
Kruglov E, Ananthanarayanan M, Sousa P, Weerachayaphorn J, Guerra MT, Nathanson MH. Type 2 inositol trisphosphate receptor gene expression in hepatocytes is regulated by cyclic AMP. Biochem Biophys Res Commun 2017; 486:659-664. [PMID: 28327356 DOI: 10.1016/j.bbrc.2017.03.086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 03/17/2017] [Indexed: 01/19/2023]
Abstract
The type 2 inositol 1,4,5-trisphosphate receptor (IP3R2) is the principal intracellular Ca2+ release channel in hepatocytes, and so is important for bile secretion and other functions. IP3R2 activity is regulated in part by post-translational modifications but little is known about transcriptional regulation of its expression. We found that both IP3R2 mRNA and protein levels in liver were increased during fasting. Treatment of hepatocytes with forskolin or 8-CPT-cAMP also increased IP3R2, and this was reduced by actinomycin D. Analysis of the IP3R2 promoter revealed five CREs, and CREB potently increased promoter activity. Mutation of CRE4 or CRE5 decreased induction by CREB, and ChIP assay showed recruitment of CREB to these sites. Adenylyl cyclase (AC) 6 and 9 were the principal AC isoforms detected in rat hepatocytes, and silencing either one decreased organic anion secretion, which depends on IP3R2. Secretion furthermore was increased by overnight but not acute treatment with forskolin or 8-CPT-cAMP. These findings provide evidence that IP3R2 expression is transcriptionally regulated by cAMP via CREB binding to CRE elements in its promoter. The findings furthermore suggest that this mechanism is relevant for hormonal regulation of bile secretion.
Collapse
Affiliation(s)
- Emma Kruglov
- Digestive Diseases Section, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA
| | | | - Pedro Sousa
- Digestive Diseases Section, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA
| | - Jittima Weerachayaphorn
- Digestive Diseases Section, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA
| | - Mateus T Guerra
- Digestive Diseases Section, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA
| | - Michael H Nathanson
- Digestive Diseases Section, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520-8019, USA.
| |
Collapse
|
27
|
Sandwich-Cultured Hepatocytes as a Tool to Study Drug Disposition and Drug-Induced Liver Injury. J Pharm Sci 2016; 105:443-459. [PMID: 26869411 DOI: 10.1016/j.xphs.2015.11.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/06/2015] [Accepted: 11/09/2015] [Indexed: 12/21/2022]
Abstract
Sandwich-cultured hepatocytes (SCH) are metabolically competent and have proper localization of basolateral and canalicular transporters with functional bile networks. Therefore, this cellular model is a unique tool that can be used to estimate biliary excretion of compounds. SCH have been used widely to assess hepatobiliary disposition of endogenous and exogenous compounds and metabolites. Mechanistic modeling based on SCH data enables estimation of metabolic and transporter-mediated clearances, which can be used to construct physiologically based pharmacokinetic models for prediction of drug disposition and drug-drug interactions in humans. In addition to pharmacokinetic studies, SCH also have been used to study cytotoxicity and perturbation of biological processes by drugs and hepatically generated metabolites. Human SCH can provide mechanistic insights underlying clinical drug-induced liver injury (DILI). In addition, data generated in SCH can be integrated into systems pharmacology models to predict potential DILI in humans. In this review, applications of SCH in studying hepatobiliary drug disposition and bile acid-mediated DILI are discussed. An example is presented to show how data generated in the SCH model were used to establish a quantitative relationship between intracellular bile acids and cytotoxicity, and how this information was incorporated into a systems pharmacology model for DILI prediction.
Collapse
|
28
|
Feriod CN, Oliveira AG, Guerra MT, Nguyen L, Richards KM, Jurczak MJ, Ruan HB, Camporez JP, Yang X, Shulman GI, Bennett AM, Nathanson MH, Ehrlich BE. Hepatic Inositol 1,4,5 Trisphosphate Receptor Type 1 Mediates Fatty Liver. Hepatol Commun 2016; 1:23-35. [PMID: 28966992 PMCID: PMC5613674 DOI: 10.1002/hep4.1012] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Fatty liver is the most common type of liver disease, affecting nearly one third of the US population and more than half a billion people worldwide. Abnormalities in ER calcium handling and mitochondrial function each have been implicated in abnormal lipid droplet formation. Here we show that the type 1 isoform of the inositol 1,4,5-trisphosphate receptor (InsP3R1) specifically links ER calcium release to mitochondrial calcium signaling and lipid droplet formation in hepatocytes. Moreover, liver-specific InsP3R1 knockout mice have impaired mitochondrial calcium signaling, decreased hepatic triglycerides, reduced lipid droplet formation and are resistant to development of fatty liver. Patients with non-alcoholic steatohepatitis, the most malignant form of fatty liver, have increased hepatic expression of InsP3R1 and the extent of ER-mitochondrial co-localization correlates with the degree of steatosis in human liver biopsies. CONCLUSION InsP3R1 plays a central role in lipid droplet formation in hepatocytes and the data suggest that it is involved in the development of human fatty liver disease.
Collapse
Affiliation(s)
- Colleen N Feriod
- Department of Cellular and Molecular Physiology, Yale University School of Medicine New Haven, CT 06520.,Department of Pharmacology, Yale University School of Medicine New Haven, CT 06520
| | - Andre Gustavo Oliveira
- Section of Digestive Diseases, Internal Medicine, Yale University School of Medicine New Haven, CT 06520
| | - Mateus T Guerra
- Section of Digestive Diseases, Internal Medicine, Yale University School of Medicine New Haven, CT 06520
| | - Lily Nguyen
- Department of Pharmacology, Yale University School of Medicine New Haven, CT 06520
| | | | - Michael J Jurczak
- Department of Internal Medicine, Yale University School of Medicine New Haven, CT 06520
| | - Hai-Bin Ruan
- Department of Comparative Medicine, Yale University School of Medicine New Haven, CT 06520.,Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine New Haven, CT 06520
| | - Joao Paulo Camporez
- Department of Internal Medicine, Yale University School of Medicine New Haven, CT 06520
| | - Xiaoyong Yang
- Department of Cellular and Molecular Physiology, Yale University School of Medicine New Haven, CT 06520.,Department of Comparative Medicine, Yale University School of Medicine New Haven, CT 06520.,Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine New Haven, CT 06520
| | - Gerald I Shulman
- Department of Cellular and Molecular Physiology, Yale University School of Medicine New Haven, CT 06520.,Department of Internal Medicine, Yale University School of Medicine New Haven, CT 06520.,Howard Hughes Medical Institute, Yale University School of Medicine New Haven, CT 06520
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine New Haven, CT 06520.,Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine New Haven, CT 06520
| | - Michael H Nathanson
- Section of Digestive Diseases, Internal Medicine, Yale University School of Medicine New Haven, CT 06520
| | - Barbara E Ehrlich
- Department of Cellular and Molecular Physiology, Yale University School of Medicine New Haven, CT 06520.,Department of Pharmacology, Yale University School of Medicine New Haven, CT 06520
| |
Collapse
|
29
|
Jemnitz K, Bátai-Konczos A, Szabó M, Ioja E, Kolacsek O, Orbán TI, Török G, Homolya L, Kovács E, Jablonkai I, Veres Z. A transgenic rat hepatocyte - Kupffer cell co-culture model for evaluation of direct and macrophage-related effect of poly(amidoamine) dendrimers. Toxicol In Vitro 2016; 38:159-169. [PMID: 27717685 DOI: 10.1016/j.tiv.2016.09.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/07/2016] [Accepted: 09/16/2016] [Indexed: 12/12/2022]
Abstract
Increasing number of papers demonstrate that Kupffer cells (KCs) play a role in the development of drug induced liver injury (DILI). Furthermore, elevated intracellular Ca2+ level of hepatocytes is considered as a common marker of DILI. Here we applied an in vitro model based on hepatocyte mono- and hepatocyte/KC co-cultures (H/KC) isolated from transgenic rats stably expressing the GCaMP2 fluorescent Ca2+ sensor protein to investigate the effects of polycationic (G5), polyanionic (G4.5) and polyethylene-glycol coated neutral (G5 Peg) dendrimers known to accumulate in the liver, primarily in KCs. Following dendrimer exposure, hepatocyte homeostasis was measured by MTT cytotoxicity assay and by Ca2+ imaging, while hepatocyte functions were studied by CYP2B1/2 inducibility, and bilirubin and taurocholate transport. G5 was significantly more cytotoxic than G4.5 for hepatocytes and induced Ca2+ oscillation and sustained Ca2+ signals at 1μM and10 μM, respectively both in hepatocytes and KCs. Dendrimer-induced Ca2+ signals in hepatocytes were attenuated by macrophages. Activation of KCs by lipopolysaccharide and G5 decreased the inducibility of CYP2B1/2, which was restored by depleting the KCs with gadolinium-chloride and pentoxyphylline, suggesting a role of macrophages in the hindrance of CYP2B1/2 induction by G5 and lipopolysaccharide. In the H/KC, but not in the hepatocyte mono-culture, G5 reduced the canalicular efflux of bilirubin and stimulated the uptake and canalicular efflux of taurocholate. In conclusion, H/KC provides a good model for the prediction of hepatotoxic potential of drugs, especially of nanomaterials known to be trapped by macrophages, activation of which presumably contributes to DILI.
Collapse
Affiliation(s)
- Katalin Jemnitz
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.
| | - Attila Bátai-Konczos
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Mónika Szabó
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Enikő Ioja
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Orsolya Kolacsek
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Tamás I Orbán
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - György Török
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - László Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | | | - István Jablonkai
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Zsuzsa Veres
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| |
Collapse
|
30
|
Nakamura T, Kamishikiryo J, Morita T. Prazosin-stimulated release of hepatic triacylglyceride lipase from primary cultured rat hepatocytes is involved in the regulation of cAMP-dependent protein kinase through activation of the Ca2+/calmodulin-dependent protein kinase-II. Pharmacol Rep 2016; 68:649-53. [DOI: 10.1016/j.pharep.2016.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/06/2016] [Accepted: 02/08/2016] [Indexed: 10/22/2022]
|
31
|
Verma A, Makadia H, Hoek JB, Ogunnaike BA, Vadigepalli R. Computational Modeling of Spatiotemporal Ca(2+) Signal Propagation Along Hepatocyte Cords. IEEE Trans Biomed Eng 2016; 63:2047-55. [PMID: 27076052 DOI: 10.1109/tbme.2016.2550045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE The purpose of this study is to model the dynamics of lobular Ca(2+) wave propagation induced by an extracellular stimulus, and to analyze the effect of spatially systematic variations in cell-intrinsic signaling parameters on sinusoidal Ca(2+) response. METHODS We developed a computational model of lobular scale Ca(2+) signaling that accounts for receptor- mediated initiation of cell-intrinsic Ca(2+) signal in hepatocytes and its propagation to neighboring hepatocytes through gap junction-mediated molecular exchange. RESULTS Analysis of the simulations showed that a pericentral-to-periportal spatial gradient in hormone sensitivity and/or rates of IP3 synthesis underlies the Ca(2+) wave propagation. We simulated specific cases corresponding to localized disruptions in the graded pattern of these parameters along a hepatic sinusoid. Simulations incorporating locally altered parameters exhibited Ca(2+) waves that do not propagate throughout the hepatic plate. Increased gap junction coupling restored normal Ca(2+) wave propagation when hepatocytes with low Ca(2+) signaling ability were localized in the midlobular or the pericentral region. CONCLUSION Multiple spatial patterns in intracellular signaling parameters can lead to Ca(2+) wave propagation that is consistent with the experimentally observed spatial patterns of Ca(2+) dynamics. Based on simulations and analysis, we predict that increased gap junction-mediated intercellular coupling can induce robust Ca(2+) signals in otherwise poorly responsive hepatocytes, at least partly restoring the sinusoidally oriented Ca (2+) waves. SIGNIFICANCE Our bottom-up model of agonist-evoked spatial Ca(2+) patterns can be integrated with detailed descriptions of liver histology to study Ca(2+) regulation at the tissue level.
Collapse
|
32
|
Chandrasekhar R, Alzayady KJ, Wagner LE, Yule DI. Unique Regulatory Properties of Heterotetrameric Inositol 1,4,5-Trisphosphate Receptors Revealed by Studying Concatenated Receptor Constructs. J Biol Chem 2016; 291:4846-60. [PMID: 26755721 DOI: 10.1074/jbc.m115.705301] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Indexed: 02/02/2023] Open
Abstract
The ability of inositol 1,4,5-trisphosphate receptors (IP3R) to precisely initiate and generate a diverse variety of intracellular Ca(2+) signals is in part mediated by the differential regulation of the three subtypes (R1, R2, and R3) by key functional modulators (IP3, Ca(2+), and ATP). However, the contribution of IP3R heterotetramerization to Ca(2+) signal diversity has largely been unexplored. In this report, we provide the first definitive biochemical evidence of endogenous heterotetramer formation. Additionally, we examine the contribution of individual subtypes within defined concatenated heterotetramers to the shaping of Ca(2+) signals. Under conditions where key regulators of IP3R function are optimal for Ca(2+) release, we demonstrate that individual monomers within heteromeric IP3Rs contributed equally toward generating a distinct 'blended' sensitivity to IP3 that is likely dictated by the unique IP3 binding affinity of the heteromers. However, under suboptimal conditions where [ATP] were varied, we found that one subtype dictated the ATP regulatory properties of heteromers. We show that R2 monomers within a heterotetramer were both necessary and sufficient to dictate the ATP regulatory properties. Finally, the ATP-binding site B in R2 critical for ATP regulation was mutated and rendered non-functional to address questions relating to the stoichiometry of IP3R regulation. Two intact R2 monomers were sufficient to maintain ATP regulation in R2 homotetramers. In summary, we demonstrate that heterotetrameric IP3R do not necessarily behave as the sum of the constituent subunits, and these properties likely extend the versatility of IP3-induced Ca(2+) signaling in cells expressing multiple IP3R isoforms.
Collapse
Affiliation(s)
- Rahul Chandrasekhar
- From the Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642
| | - Kamil J Alzayady
- From the Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642
| | - Larry E Wagner
- From the Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642
| | - David I Yule
- From the Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642
| |
Collapse
|
33
|
Delgado-Coello B, Mas-Oliva J. Relevance of the plasma membrane calcium-ATPase in the homeostasis of calcium in the fetal liver. Organogenesis 2015; 10:333-9. [PMID: 25836032 PMCID: PMC4594366 DOI: 10.1080/15476278.2015.1011918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
During the early stages of development, the embryo depends on the placenta as provider of oxygen and calcium, among other essential compounds. Although fetal liver accomplishes a well-known haematopoietic function, its contribution to calcium homeostasis upon development is poorly understood. The homeostasis of cell calcium contributes to diverse signaling pathways across developmental stages of most tissues and the calcium-ATPase located at the plasma membrane (PMCA) helps pumping excess calcium into the extracellular space. To date, the understanding of the equilibrium shift between PMCA isoforms during liver development is still missing. This review focuses on the characterization of the hepatic PMCA along the early stages of development, followed by a description of modern approaches to study calcium homeostasis involving several types of pluripotent cells. The application of interdisciplinary techniques to improve our understanding of liver development and the role calcium homeostasis plays in the definition of pathogenesis is also discussed.
Collapse
Affiliation(s)
- Blanca Delgado-Coello
- a Departamento de Bioquímica y Biología Estructural ; Instituto de Fisiología Celular ; Universidad Nacional Autónoma de México ; México D.F. , México
| | | |
Collapse
|
34
|
Chandrasekhar R, Alzayady KJ, Yule DI. Using concatenated subunits to investigate the functional consequences of heterotetrameric inositol 1,4,5-trisphosphate receptors. Biochem Soc Trans 2015; 43:364-70. [PMID: 26009177 PMCID: PMC4677331 DOI: 10.1042/bst20140287] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Inositol 1,4,5-trisphosphate receptors (IP3Rs) are a family of ubiquitous, ER localized, tetrameric Ca2+ release channels. There are three subtypes of the IP3Rs (R1, R2, R3), encoded by three distinct genes, that share ∼60-70% sequence identity. The diversity of Ca2+ signals generated by IP3Rs is thought to be largely the result of differential tissue expression, intracellular localization and subtype-specific regulation of the three subtypes by various cellular factors, most significantly InsP3, Ca2+ and ATP. However, largely unexplored is the notion of additional signal diversity arising from the assembly of both homo and heterotetrameric InsP3Rs. In the present article, we review the biochemical and functional evidence supporting the existence of homo and heterotetrameric populations of InsP3Rs. In addition, we consider a strategy that utilizes genetically concatenated InsP3Rs to study the functional characteristics of heterotetramers with unequivocally defined composition. This approach reveals that the overall properties of IP3R are not necessarily simply a blend of the constituent monomers but that specific subtypes appear to dominate the overall characteristics of the tetramer. It is envisioned that the ability to generate tetramers with defined wild type and mutant subunits will be useful in probing fundamental questions relating to IP3R structure and function.
Collapse
MESH Headings
- Adenosine Triphosphate/chemistry
- Adenosine Triphosphate/metabolism
- Calcium Signaling/genetics
- Humans
- Inositol 1,4,5-Trisphosphate/chemistry
- Inositol 1,4,5-Trisphosphate/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/chemistry
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Protein Multimerization
- Protein Structure, Tertiary
- Structure-Activity Relationship
Collapse
|
35
|
Vervloessem T, Yule DI, Bultynck G, Parys JB. The type 2 inositol 1,4,5-trisphosphate receptor, emerging functions for an intriguing Ca²⁺-release channel. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:1992-2005. [PMID: 25499268 DOI: 10.1016/j.bbamcr.2014.12.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 12/02/2014] [Accepted: 12/03/2014] [Indexed: 12/19/2022]
Abstract
The inositol 1,4,5-trisphosphate (IP3) receptor (IP3R) type 2 (IP3R2) is an intracellular Ca²⁺-release channel located on the endoplasmic reticulum (ER). IP3R2 is characterized by a high sensitivity to both IP3 and ATP and is biphasically regulated by Ca²⁺. Furthermore, IP3R2 is modulated by various protein kinases. In addition to its regulation by protein kinase A, IP3R2 forms a complex with adenylate cyclase 6 and is directly regulated by cAMP. Finally, in the ER, IP3R2 is less mobile than the other IP3R isoforms, while its functional properties appear dominant in heterotetramers. These properties make the IP3R2 a Ca²⁺ channel with exquisite properties for setting up intracellular Ca²⁺ signals with unique characteristics. IP3R2 plays a crucial role in the function of secretory cell types (e.g. pancreatic acinar cells, hepatocytes, salivary gland, eccrine sweat gland). In cardiac myocytes, the role of IP3R2 appears more complex, because, together with IP3R1, it is needed for normal cardiogenesis, while its aberrant activity is implicated in cardiac hypertrophy and arrhythmias. Most importantly, its high sensitivity to IP3 makes IP3R2 a target for anti-apoptotic proteins (e.g. Bcl-2) in B-cell cancers. Disrupting IP3R/Bcl-2 interaction therefore leads in those cells to increased Ca²⁺ release and apoptosis. Intriguingly, IP3R2 is not only implicated in apoptosis but also in the induction of senescence, another tumour-suppressive mechanism. These results were the first to unravel the physiological and pathophysiological role of IP3R2 and we anticipate that further progress will soon be made in understanding the function of IP3R2 in various tissues and organs.
Collapse
Affiliation(s)
- Tamara Vervloessem
- KU Leuven, Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven, Belgium
| | - David I Yule
- University of Rochester, Department of Pharmacology and Physiology, Rochester, NY, USA
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, Leuven, Belgium.
| |
Collapse
|
36
|
Feriod CN, Nguyen L, Jurczak MJ, Kruglov EA, Nathanson MH, Shulman GI, Bennett AM, Ehrlich BE. Inositol 1,4,5-trisphosphate receptor type II (InsP3R-II) is reduced in obese mice, but metabolic homeostasis is preserved in mice lacking InsP3R-II. Am J Physiol Endocrinol Metab 2014; 307:E1057-64. [PMID: 25315698 PMCID: PMC4254986 DOI: 10.1152/ajpendo.00236.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Inositol 1,4,5-trisphosphate receptor type II (InsP3R-II) is the most prevalent isoform of the InsP3R in hepatocytes and is concentrated under the canalicular membrane, where it plays an important role in bile secretion. We hypothesized that altered calcium (Ca(2+)) signaling may be involved in metabolic dysfunction, as InsP3R-mediated Ca(2+) signals have been implicated in the regulation of hepatic glucose homeostasis. Here, we find that InsP3R-II, but not InsP3R-I, is reduced in the livers of obese mice. In our investigation of the functional consequences of InsP3R-II deficiency, we found that organic anion secretion at the canalicular membrane and Ca(2+) signals were impaired. However, mice lacking InsP3R-II showed no deficits in energy balance, glucose production, glucose tolerance, or susceptibility to hepatic steatosis. Thus, our results suggest that reduced InsP3R-II expression is not sufficient to account for any disruptions in metabolic homeostasis that are observed in mouse models of obesity. We conclude that metabolic homeostasis is maintained independently of InsP3R-II. Loss of InsP3R-II does impair secretion of bile components; therefore, we suggest that conditions of obesity would lead to a decrease in this Ca(2+)-sensitive process.
Collapse
Affiliation(s)
- Colleen N Feriod
- Department of Cellular and Molecular Physiology, Yale University School of Medicine New Haven, Connecticut
| | - Lily Nguyen
- Department of Pharmacology, Yale University School of Medicine New Haven, Connecticut
| | - Michael J Jurczak
- Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut
| | - Emma A Kruglov
- Section of Digestive Diseases, Yale University School of Medicine New Haven, Connecticut
| | - Michael H Nathanson
- Section of Digestive Diseases, Yale University School of Medicine New Haven, Connecticut
| | - Gerald I Shulman
- Department of Cellular and Molecular Physiology, Yale University School of Medicine New Haven, Connecticut; Department of Internal Medicine, Yale University School of Medicine New Haven, Connecticut; Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Anton M Bennett
- Department of Pharmacology, Yale University School of Medicine New Haven, Connecticut; Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine New Haven, Connecticut; and
| | - Barbara E Ehrlich
- Department of Cellular and Molecular Physiology, Yale University School of Medicine New Haven, Connecticut; Department of Pharmacology, Yale University School of Medicine New Haven, Connecticut;
| |
Collapse
|
37
|
Apical localization of inositol 1,4,5-trisphosphate receptors is independent of extended synaptotagmins in hepatocytes. PLoS One 2014; 9:e114043. [PMID: 25437447 PMCID: PMC4250053 DOI: 10.1371/journal.pone.0114043] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 11/03/2014] [Indexed: 12/12/2022] Open
Abstract
Extended synaptotagmins (E-Syts) are a recently identified family of proteins that tether the endoplasmic reticulum (ER) to the plasma membrane (PM) in part by conferring regulation of cytosolic calcium (Ca2+) at these contact sites (Cell, 2013). However, the mechanism by which E-Syts link this tethering to Ca2+ signaling is unknown. Ca2+ waves in polarized epithelia are initiated by inositol 1,4,5-trisphosphate receptors (InsP3Rs), and these waves begin in the apical region because InsP3Rs are targeted to the ER adjacent to the apical membrane. In this study we investigated whether E-Syts are responsible for this targeting. Primary rat hepatocytes were used as a model system, because a single InsP3R isoform (InsP3R-II) is tethered to the peri-apical ER in these cells. Additionally, it has been established in hepatocytes that the apical localization of InsP3Rs is responsible for Ca2+ waves and secretion and is disrupted in disease states in which secretion is impaired. We found that rat hepatocytes express two of the three identified E-Syts (E-Syt1 and E-Syt2). Individual or simultaneous siRNA knockdown of these proteins did not alter InsP3R-II expression levels, apical localization or average InsP3R-II cluster size. Moreover, apical secretion of the organic anion 5-chloromethylfluorescein diacetate (CMFDA) was not changed in cells lacking E-Syts but was reduced in cells in which cytosolic Ca2+ was buffered. These data provide evidence that E-Syts do not participate in the targeting of InsP3Rs to the apical region. Identifying tethers that bring InsP3Rs to the apical region remains an important question, since mis-targeting of InsP3Rs leads to impaired secretory activity.
Collapse
|
38
|
Ananthanarayanan M, Banales JM, Guerra MT, Spirli C, Munoz-Garrido P, Mitchell-Richards K, Tafur D, Saez E, Nathanson MH. Post-translational regulation of the type III inositol 1,4,5-trisphosphate receptor by miRNA-506. J Biol Chem 2014; 290:184-96. [PMID: 25378392 DOI: 10.1074/jbc.m114.587030] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The type III isoform of the inositol 1,4,5-trisphosphate receptor (InsP3R3) is apically localized and triggers Ca(2+) waves and secretion in a number of polarized epithelia. However, nothing is known about epigenetic regulation of this InsP3R isoform. We investigated miRNA regulation of InsP3R3 in primary bile duct epithelia (cholangiocytes) and in the H69 cholangiocyte cell line, because the role of InsP3R3 in cholangiocyte Ca(2+) signaling and secretion is well established and because loss of InsP3R3 from cholangiocytes is responsible for the impairment in bile secretion that occurs in a number of liver diseases. Analysis of the 3'-UTR of human InsP3R3 mRNA revealed two highly conserved binding sites for miR-506. Transfection of miR-506 mimics into cell lines expressing InsP3R3-3'UTR-luciferase led to decreased reporter activity, whereas co-transfection with miR-506 inhibitors led to enhanced activity. Reporter activity was abrogated in isolated mutant proximal or distal miR-506 constructs in miR-506-transfected HEK293 cells. InsP3R3 protein levels were decreased by miR-506 mimics and increased by inhibitors, and InsP3R3 expression was markedly decreased in H69 cells stably transfected with miR-506 relative to control cells. miR-506-H69 cells exhibited a fibrotic signature. In situ hybridization revealed elevated miR-506 expression in vivo in human-diseased cholangiocytes. Histamine-induced, InsP3-mediated Ca(2+) signals were decreased by 50% in stable miR-506 cells compared with controls. Finally, InsP3R3-mediated fluid secretion was significantly decreased in isolated bile duct units transfected with miR-506, relative to control IBDU. Together, these data identify miR-506 as a regulator of InsP3R3 expression and InsP3R3-mediated Ca(2+) signaling and secretion.
Collapse
Affiliation(s)
| | - Jesus M Banales
- the Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of Basque Country (UPV/EHU), CIBERehd, IKERBASQUE, AECC, 20014 San Sebastian, Spain, and the Division of Gene Therapy and Hepatology, CIMA of the University of Navarra, Ciberehd, 31009 Pamplona, Spain
| | | | | | - Patricia Munoz-Garrido
- the Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of Basque Country (UPV/EHU), CIBERehd, IKERBASQUE, AECC, 20014 San Sebastian, Spain, and
| | - Kisha Mitchell-Richards
- Pathology, Section of Digestive Diseases and the Liver Center, Yale University School of Medicine, New Haven, Connecticut 06520
| | | | - Elena Saez
- the Division of Gene Therapy and Hepatology, CIMA of the University of Navarra, Ciberehd, 31009 Pamplona, Spain
| | | |
Collapse
|
39
|
Abstract
Mutations in polycystin 1 and 2 (PC1 and PC2) cause the common genetic kidney disorder autosomal dominant polycystic kidney disease (ADPKD). It is unknown how these mutations result in renal cysts, but dysregulation of calcium (Ca(2+)) signaling is a known consequence of PC2 mutations. PC2 functions as a Ca(2+)-activated Ca(2+) channel of the endoplasmic reticulum. We hypothesize that Ca(2+) signaling through PC2, or other intracellular Ca(2+) channels such as the inositol 1,4,5-trisphosphate receptor (InsP3R), is necessary to maintain renal epithelial cell function and that disruption of the Ca(2+) signaling leads to renal cyst development. The cell line LLC-PK1 has traditionally been used for studying PKD-causing mutations and Ca(2+) signaling in 2D culture systems. We demonstrate that this cell line can be used in long-term (8 wk) 3D tissue culture systems. In 2D systems, knockdown of InsP3R results in decreased Ca(2+) transient signals that are rescued by overexpression of PC2. In 3D systems, knockdown of either PC2 or InsP3R leads to cyst formation, but knockdown of InsP3R type 1 (InsP3R1) generated the largest cysts. InsP3R1 and InsP3R3 are differentially localized in both mouse and human kidney, suggesting that regional disruption of Ca(2+) signaling contributes to cystogenesis. All cysts had intact cilia 2 wk after starting 3D culture, but the cells with InsP3R1 knockdown lost cilia as the cysts grew. Studies combining 2D and 3D cell culture systems will assist in understanding how mutations in PC2 that confer altered Ca(2+) signaling lead to ADPKD cysts.
Collapse
|
40
|
Miszczuk GS, Barosso IR, Zucchetti AE, Boaglio AC, Pellegrino JM, Sánchez Pozzi EJ, Roma MG, Crocenzi FA. Sandwich-cultured rat hepatocytes as an in vitro model to study canalicular transport alterations in cholestasis. Arch Toxicol 2014; 89:979-90. [PMID: 24912783 DOI: 10.1007/s00204-014-1283-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 05/20/2014] [Indexed: 12/28/2022]
Abstract
At present, it has not been systematically evaluated whether the functional alterations induced by cholestatic compounds in canalicular transporters involved in bile formation can be reproduced in sandwich-cultured rat hepatocytes (SCRHs). Here, we focused on two clinically relevant cholestatic agents, such as estradiol 17β-D-glucuronide (E17G) and taurolithocholate (TLC), also testing the ability of dibutyryl cyclic AMP (DBcAMP) to prevent their effects. SCRHs were incubated with E17G (200 µM) or TLC (2.5 µM) for 30 min, with or without pre-incubation with DBcAMP (10 µM) for 15 min. Then, the increase in glutathione methyl fluorescein (GS-MF)-associated fluorescence inside the canaliculi was monitored by quantitative time-lapse imaging, and Mrp2 transport activity was calculated by measuring the slope of the time-course fluorescence curves during the initial linear phase, which was considered to be the Mrp2-mediated initial transport rate (ITR). E17G and TLC impaired canalicular bile formation, as evidenced by a decrease in both the bile canaliculus volume and the bile canaliculus width, estimated from 3D and 2D confocal images, respectively. These compounds decreased ITR and induced retrieval of Mrp2, a main pathomechanism involved in their cholestatic effects. Finally, DBcAMP prevented these effects, and its well-known choleretic effect was evident from the increase in the canalicular volume/width values; this choleretic effect is associated in part with its capability to increase Mrp2 activity, evidenced here by the increase in ITR of GS-MF. Our study supports the use of SCRHs as an in vitro model useful to quantify canalicular transport function under conditions of cholestasis and choleresis.
Collapse
Affiliation(s)
- Gisel S Miszczuk
- Instituto de Fisiología Experimental (IFISE) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Ciencias Bioquímicas y Farmacéuticas - Universidad Nacional de Rosario (UNR), Suipacha 570, S2002LRL, Rosario, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Soroka CJ, Boyer JL. Biosynthesis and trafficking of the bile salt export pump, BSEP: therapeutic implications of BSEP mutations. Mol Aspects Med 2014; 37:3-14. [PMID: 23685087 PMCID: PMC3784619 DOI: 10.1016/j.mam.2013.05.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 04/25/2013] [Accepted: 05/07/2013] [Indexed: 12/17/2022]
Abstract
The bile salt export pump (BSEP, ABCB11) is the primary transporter of bile acids from the hepatocyte to the biliary system. This rate-limiting step in bile formation is essential to the formation of bile salt dependent bile flow, the enterohepatic circulation of bile acids, and the digestion of dietary fats. Mutations in BSEP are associated with cholestatic diseases such as progressive familial intrahepatic cholestasis type 2 (PFIC2), benign recurrent intrahepatic cholestasis type 2 (BRIC2), drug-induced cholestasis, and intrahepatic cholestasis of pregnancy. Development of clinical therapies for these conditions necessitates a clear understanding of the cell biology of biosynthesis, trafficking, and transcriptional and translational regulation of BSEP. This chapter will focus on the molecular and cell biological aspects of this critical hepatic membrane transporter.
Collapse
Affiliation(s)
- Carol J Soroka
- Yale University School of Medicine, Department of Internal Medicine, New Haven, CT 06520, United States.
| | - James L Boyer
- Yale University School of Medicine, Department of Internal Medicine, New Haven, CT 06520, United States.
| |
Collapse
|
42
|
Kagawa T, Orii R, Hirose S, Arase Y, Shiraishi K, Mizutani A, Tsukamoto H, Mine T. Ursodeoxycholic acid stabilizes the bile salt export pump in the apical membrane in MDCK II cells. J Gastroenterol 2014; 49:890-9. [PMID: 23722250 DOI: 10.1007/s00535-013-0833-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 05/03/2013] [Indexed: 02/04/2023]
Abstract
BACKGROUND Ursodeoxycholic acid (UDCA) partly exerts choleretic effects by modifying the function of the bile salt export pump (Bsep, ABCB11). UDCA induces insertion of Bsep into the canalicular membrane of hepatocytes; however, underlying mechanisms remain unknown. We aimed to elucidate molecular mechanisms behind UDCA-induced Bsep activation. METHODS We established MDCK II cells stably expressing both Bsep and Na(+)-taurocholate cotransporting polypeptide, and investigated the effect of UDCA on activity and protein expression of Bsep using these cells. We performed inhibitor study to know the molecules involved in UDCA-induced Bsep activation, and also tested the influence of UDCA on Bsep having a disease-associated mutation. RESULTS UDCA activated Bsep in a dose-dependent manner. UDCA did not affect Bsep protein expression in whole cell lysates but increased its apical surface expression by extending the half-life from 2.4 to 5.0 h. This effect was specific to Bsep because UDCA did not affect other apical and basolateral proteins, and was independent of protein kinase A, adenylate cyclase, p38(MAPK), phosphatidylinositide 3-kinase, Ca(2+), and microtubules. NorUDCA activated Bsep similar to UDCA; however, cholic acid, taurocholic acid, and tauroUDCA had no effect. UDCA significantly increased the activity of Bsep with a benign recurrent intrahepatic cholestasis 2 mutation (A570T) but did not affect Bsep with a progressive familial intrahepatic cholestasis 2 mutation (G982R or D482G). CONCLUSIONS We demonstrated that UDCA stabilizes Bsep protein in the apical membrane and increases its activity in MDCK II cells, presumably by retarding the endocytotic process.
Collapse
Affiliation(s)
- Tatehiro Kagawa
- Division of Gastroenterology, Department of Internal Medicine, Tokai University School of Medicine, Shimokasuya 143, Isehara, Kanagawa, 259-1193, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Amaya MJ, Nathanson MH. Calcium signaling and the secretory activity of bile duct epithelia. Cell Calcium 2014; 55:317-24. [PMID: 24612866 DOI: 10.1016/j.ceca.2014.02.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 02/03/2014] [Accepted: 02/04/2014] [Indexed: 12/20/2022]
Abstract
Cytosolic calcium (Cai(2+)) is a second messenger that is important for the regulation of secretion in many types of tissues. Bile duct epithelial cells, or cholangiocytes, are polarized epithelia that line the biliary tree in liver and are responsible for secretion of bicarbonate and other solutes into bile. Cai(2+) signaling plays an important role in the regulation of secretion by cholangiocytes, and this review discusses the machinery involved in the formation of Ca(2+) signals in cholangiocytes, along with the evidence that these signals regulate ductular secretion. Finally, this review discusses the evidence that impairments in cholangiocyte Ca(2+) signaling play a primary role in the pathogenesis of cholestatic disorders, in which hepatic bile secretion is impaired.
Collapse
Affiliation(s)
- Maria Jimena Amaya
- Section of Digestive Diseases, Department of Internal Medicine, Yale University, 333 Cedar Street, PO Box 208019, New Haven, CT 06520-8019, USA
| | - Michael H Nathanson
- Section of Digestive Diseases, Department of Internal Medicine, Yale University, 333 Cedar Street, PO Box 208019, New Haven, CT 06520-8019, USA.
| |
Collapse
|
44
|
Abstract
Bile is a unique and vital aqueous secretion of the liver that is formed by the hepatocyte and modified down stream by absorptive and secretory properties of the bile duct epithelium. Approximately 5% of bile consists of organic and inorganic solutes of considerable complexity. The bile-secretory unit consists of a canalicular network which is formed by the apical membrane of adjacent hepatocytes and sealed by tight junctions. The bile canaliculi (∼1 μm in diameter) conduct the flow of bile countercurrent to the direction of portal blood flow and connect with the canal of Hering and bile ducts which progressively increase in diameter and complexity prior to the entry of bile into the gallbladder, common bile duct, and intestine. Canalicular bile secretion is determined by both bile salt-dependent and independent transport systems which are localized at the apical membrane of the hepatocyte and largely consist of a series of adenosine triphosphate-binding cassette transport proteins that function as export pumps for bile salts and other organic solutes. These transporters create osmotic gradients within the bile canalicular lumen that provide the driving force for movement of fluid into the lumen via aquaporins. Species vary with respect to the relative amounts of bile salt-dependent and independent canalicular flow and cholangiocyte secretion which is highly regulated by hormones, second messengers, and signal transduction pathways. Most determinants of bile secretion are now characterized at the molecular level in animal models and in man. Genetic mutations serve to illuminate many of their functions.
Collapse
Affiliation(s)
- James L Boyer
- Department of Medicine and Liver Center, Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
45
|
Abstract
Intracellular free Ca(2+) ([Ca(2+)]i) is a highly versatile second messenger that regulates a wide range of functions in every type of cell and tissue. To achieve this versatility, the Ca(2+) signaling system operates in a variety of ways to regulate cellular processes that function over a wide dynamic range. This is particularly well exemplified for Ca(2+) signals in the liver, which modulate diverse and specialized functions such as bile secretion, glucose metabolism, cell proliferation, and apoptosis. These Ca(2+) signals are organized to control distinct cellular processes through tight spatial and temporal coordination of [Ca(2+)]i signals, both within and between cells. This article will review the machinery responsible for the formation of Ca(2+) signals in the liver, the types of subcellular, cellular, and intercellular signals that occur, the physiological role of Ca(2+) signaling in the liver, and the role of Ca(2+) signaling in liver disease.
Collapse
Affiliation(s)
- Maria Jimena Amaya
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
46
|
Báez-Ruiz A, Cázares-Gómez K, Vázquez-Martínez O, Aguilar-Roblero R, Díaz-Muñoz M. Diurnal and nutritional adjustments of intracellular Ca2+ release channels and Ca2+ ATPases associated with restricted feeding schedules in the rat liver. J Circadian Rhythms 2013; 11:8. [PMID: 23962056 PMCID: PMC3850936 DOI: 10.1186/1740-3391-11-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 08/07/2013] [Indexed: 01/18/2023] Open
Abstract
Background Intracellular calcium is a biochemical messenger that regulates part of the metabolic adaptations in the daily fed-fast cycle. The aim of this study was to characterize the 24-h variations of the liver ryanodine and IP3 receptors (RyR and IP3R) as well as of the endoplasmic-reticulum and plasma-membrane Ca2+-ATPases (SERCA and PMCA) in daytime restricted feeding protocol. Methods A biochemical and immunohistochemical approach was implemented in this study: specific ligand-binding for RyR and IP3R, enzymatic activity (SERCA and PMCA), and protein levels and zonational hepatic-distribution were determined by immunoblot and immunohistochemistry respectively under conditions of fasting, feeding, and temporal food-restriction. Results Binding assays and immunoblots for IP3R1 and 2 showed a peak at the light/dark transition in the ad-libitum (AL) group, whereas in the restricted-feeding (RF) group the peak shifted towards the food-access time. In the case of RyR binding experiments, both AL and RF groups showed a modest elevation during the dark period, with the RF rats exhibiting increased binding in response to feeding. The AL group showed 24-h rhythmicity in SERCA level; in contrast, RF group showed a pronounced amplitude elevation and a peak phase-shift during the light-period in SERCA level and activity. The activity of PMCA was constant along day in both groups; PMCA1 levels showed a 24-h rhythmicity in the RF rats (with a peak in the light period), meanwhile PMCA4 protein levels showed rhythmicity in both groups. The fasted condition promoted an increase in IP3R binding and protein level; re-feeding increased the amount of RyR; neither the activity nor expression of SERCA and PMCA protein was affected by fasting–re-feeding conditions. Histochemical experiments showed that the distribution of the Ca2+-handling proteins, between periportal and pericentral zones of the liver, varied with the time of day and the feeding protocol. Conclusions Our findings show that RF influences mainly the phase and amplitude of hepatic IP3R and SERCA rhythms as well as discrete zonational distribution for RyR, IP3Rs, SERCA, and PMCA within the liver acinus, suggesting that intracellular calcium dynamics could be part of the rheostatic adaptation of the liver due to diurnal meal entrainment/food entrained oscillator expression.
Collapse
Affiliation(s)
- Adrián Báez-Ruiz
- Departamento de Neurobiología Molecular y Celular, Instituto de Neurobiología, UNAM-Juriquilla, Boulevard Juriquilla #3001, Apdo, Postal 1-1141, Querétaro, QRO 76230, México.
| | | | | | | | | |
Collapse
|
47
|
Distribution of voltage-dependent and intracellular Ca2+ channels in submucosal neurons from rat distal colon. Cell Tissue Res 2013; 353:355-66. [DOI: 10.1007/s00441-013-1643-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 04/18/2013] [Indexed: 10/26/2022]
|
48
|
The bile salt export pump (BSEP) in health and disease. Clin Res Hepatol Gastroenterol 2012; 36:536-53. [PMID: 22795478 DOI: 10.1016/j.clinre.2012.06.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 05/29/2012] [Accepted: 06/06/2012] [Indexed: 02/04/2023]
Abstract
The bile salt export pump (BSEP) is the major transporter for the secretion of bile acids from hepatocytes into bile in humans. Mutations of BSEP are associated with cholestatic liver diseases of varying severity including progressive familial intrahepatic cholestasis type 2 (PFIC-2), benign recurrent intrahepatic cholestasis type 2 (BRIC-2) and genetic polymorphisms are linked to intrahepatic cholestasis of pregnancy (ICP) and drug-induced liver injury (DILI). Detailed analysis of these diseases has considerably increased our knowledge about physiology and pathophysiology of bile secretion in humans. This review focuses on expression, localization, and function, short- and long-term regulation of BSEP as well as diseases association and treatment options for BSEP-associated diseases.
Collapse
|