1
|
Clemens MM, McGill MR, Apte U. Mechanisms and biomarkers of liver regeneration after drug-induced liver injury. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 85:241-262. [PMID: 31307589 DOI: 10.1016/bs.apha.2019.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Liver, the major metabolic organ in the body, is known for its remarkable capacity to regenerate. Whereas partial hepatectomy (PHx) is a popular model for the study of liver regeneration, the liver also regenerates after acute injury, but less is known about the mechanisms that drive it. Recent studies have shown that liver regeneration is critical for survival in acute liver failure (ALF), which is usually due to drug-induced liver injury (DILI). It is sometimes assumed that the signaling pathways involved are similar to those that regulate regeneration after PHx, but there are likely to be critical differences. A better understanding of regeneration mechanisms after DILI and hepatotoxicity in general could lead to development of new therapies for ALF patients and new biomarkers to predict patient outcome. Here, we summarize what is known about the mechanisms of liver regeneration and repair after hepatotoxicity. We also review the literature in the emerging field of liver regeneration biomarkers.
Collapse
Affiliation(s)
- Melissa M Clemens
- Interdisciplinary Biomedical Sciences Graduate Program, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Mitchell R McGill
- Department of Environmental and Occupational Health, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
2
|
High-dose BAFF receptor specific mAb-siRNA conjugate generates Fas-expressing B cells in lymph nodes and high-affinity serum autoantibody in a myasthenia mouse model. Clin Immunol 2017; 176:122-130. [DOI: 10.1016/j.clim.2017.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/11/2017] [Indexed: 01/04/2023]
|
3
|
Gonçalves LA, Rodo J, Rodrigues-Duarte L, de Moraes LV, Penha-Gonçalves C. HGF Secreted by Activated Kupffer Cells Induces Apoptosis of Plasmodium-Infected Hepatocytes. Front Immunol 2017; 8:90. [PMID: 28220125 PMCID: PMC5292919 DOI: 10.3389/fimmu.2017.00090] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/19/2017] [Indexed: 02/03/2023] Open
Abstract
Malaria liver stage infection is an obligatory parasite development step and represents a population bottleneck in Plasmodium infections, providing an advantageous target for blocking parasite cycle progression. Parasite development inside hepatocytes implies a gross cellular insult evoking innate host responses to counteract intra-hepatocytic infection. Using primary hepatocyte cultures, we investigated the role of Kupffer cell-derived hepatocyte growth factor (HGF) in malaria liver stage infection. We found that Kupffer cells from Plasmodium-infected livers produced high levels of HGF, which trigger apoptosis of infected hepatocytes through a mitochondrial-independent apoptosis pathway. HGF action in infected hepatocyte primary cultures results in a potent reduction of parasite yield by specifically sensitizing hepatocytes carrying established parasite exo-erythrocytic forms to undergo apoptosis. This apoptosis mechanism is distinct from cell death that is spontaneously induced in infected cultures and is governed by Fas signaling modulation through a mitochondrial-dependent apoptosis pathway. This work indicates that HGF and Fas signaling pathways are part of an orchestrated host apoptosis response that occurs during malaria liver stage infection, decreasing the success of infection of individual hepatocytes. Our results raise the hypothesis that paracrine signals derived from Kupffer cell activation are implicated in directing death of hepatocytes infected with the malaria parasite.
Collapse
Affiliation(s)
| | - Joana Rodo
- Instituto Gulbenkian de Ciência , Oeiras , Portugal
| | | | | | | |
Collapse
|
4
|
Maraldi T, Beretti F, Guida M, Zavatti M, De Pol A. Role of hepatocyte growth factor in the immunomodulation potential of amniotic fluid stem cells. Stem Cells Transl Med 2015; 4:539-47. [PMID: 25873747 DOI: 10.5966/sctm.2014-0266] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/23/2015] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Human amniotic fluid stem cells (hAFSCs) may be useful for regenerative medicine because of their potential to differentiate into all three germ layers and to modulate immune response with different types of secretion molecules. This last issue has not been completely elucidated. The aim of this study was to investigate the secretome profile of the hAFSC, focusing on the role of hepatocyte growth factor (HGF) in immunoregulation through short and long cocultures with human peripheral blood mononuclear cells. We found that HGF produced by hAFSCs exerts a cytoprotective role, inducing an increase in caspase-dependent apoptosis in human immune cells. This study provides evidence supporting the hypothesis that amniotic fluid is an ideal source of stem cells for expansion and banking properties for therapeutic use. hAFSCs not only are less immunogenic but also can secrete immunoregulatory factors that may be useful in autoimmune diseases or allogenic implants. SIGNIFICANCE New information about the secretome pattern is reported in this paper. Human amniotic fluid stem cells (hAFSCs) possess immunomodulatory properties involving hepatocyte growth factor production. hAFSCs could be used in immunotherapies and might be able to avoid allogenic rejection.
Collapse
Affiliation(s)
- Tullia Maraldi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Francesca Beretti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Marianna Guida
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Manuela Zavatti
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| | - Anto De Pol
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Biomedicine, European Academy of Bozen/Bolzano (EURAC) Research, Bolzano, Italy
| |
Collapse
|
5
|
Deficiency of formyl peptide receptor 1 and 2 is associated with increased inflammation and enhanced liver injury after LPS-stimulation. PLoS One 2014; 9:e100522. [PMID: 24956481 PMCID: PMC4067326 DOI: 10.1371/journal.pone.0100522] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 05/28/2014] [Indexed: 12/30/2022] Open
Abstract
Introduction Formyl peptide-receptor 1 and 2 (FPR1 and FPR2) in mice were identified as receptors with contrary affinity for the PAMP fMLF. Formyl-methionyl-leucyl-phenylalanine is either part of the bacterial membrane and is secreted by the mitochondria of eukaryotic ceslls during apoptosis. Furthermore FPR1 and 2 are described as highly relevant factors for the chemotaxis of immune cells. Their role during the acute liver injury has not been investigated yet. Materials and Methods Constitutive knockout mice for FPR1 (mFPR1-/-), FPR2 (mFPR2-/-) and wild type (WT) mice were challenged with LPS i.p. for 3 h and 6 h. Liver and serum were sampled for further analysis. Results Liver transaminases were elevated in all mice 3 h and 6 h post LPS stimulation. Gene expression analysis displayed a reduced expression of the pro-inflammatory cytokines IL-6 and CXCL1 after 3 h in the mFPR1-/- compared to wild type and mFPR2-/- mice. After 6 h, IL-6, TNF-α and CXCL1 were significantly higher in mice lacking mFPR1 or 2. Consistent to these findings the numbers of CD11b+ and Ly6G+ immune cells were altered in the livers. The analysis of TLR2 and TLR4 revealed time and genotype specific changes in theirs gene expression. Additionally, the liver in mFPR1- and mFPR2-deficient mice seem to be more susceptible to apoptosis by showing a significant higher number of TUNEL+-cells in the liver than WT-mice and displayed less Ki67-positive nuclei in the liver. Conclusion The results suggest a prominent role of FPRs in the regulation of the hepatic inflammatory response after LPS induced liver injury. Deletion of mFPR1 or mFPR2 leads to deregulation of the inflammatory response compared to WT mice, associated with more severe liver injury represented by higher levels of transaminases, apoptotic cells and a reduced regenerative capacity.
Collapse
|
6
|
Raychaudhuri S, Raychaudhuri SC. Monte carlo study elucidates the type 1/type 2 choice in apoptotic death signaling in healthy and cancer cells. Cells 2013; 2:361-92. [PMID: 24709706 PMCID: PMC3972686 DOI: 10.3390/cells2020361] [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] [Received: 03/13/2013] [Revised: 05/10/2013] [Accepted: 05/14/2013] [Indexed: 11/16/2022] Open
Abstract
Apoptotic cell death is coordinated through two distinct (type 1 and type 2) intracellular signaling pathways. How the type 1/type 2 choice is made remains a central problem in the biology of apoptosis and has implications for apoptosis related diseases and therapy. We study the problem of type 1/type 2 choice in silico utilizing a kinetic Monte Carlo model of cell death signaling. Our results show that the type 1/type 2 choice is linked to deterministic versus stochastic cell death activation, elucidating a unique regulatory control of the apoptotic pathways. Consistent with previous findings, our results indicate that caspase 8 activation level is a key regulator of the choice between deterministic type 1 and stochastic type 2 pathways, irrespective of cell types. Expression levels of signaling molecules downstream also regulate the type 1/type 2 choice. A simplified model of DISC clustering elucidates the mechanism of increased active caspase 8 generation and type 1 activation in cancer cells having increased sensitivity to death receptor activation. We demonstrate that rapid deterministic activation of the type 1 pathway can selectively target such cancer cells, especially if XIAP is also inhibited; while inherent cell-to-cell variability would allow normal cells stay protected.
Collapse
|
7
|
Zinkel SS, Yin XM, Gross A. Rejuvenating Bi(d)ology. Oncogene 2012; 32:3213-3219. [PMID: 23069655 DOI: 10.1038/onc.2012.454] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 08/14/2012] [Accepted: 08/14/2012] [Indexed: 12/25/2022]
Abstract
The BH3-only Bid protein is a critical sentinel of cellular stress in the liver and the hematopoietic system. Bid's initial 'claim to fame' came from its ability-as a caspase-truncated product-to trigger the mitochondrial apoptotic program following death receptor activation. Today we know that Bid can response to multiple types of proteases, which are activated under different conditions such as T-cell activation, ischemical reperfusion injury and lysosomal injury. Activation of the mitochondrial apoptotic program by Bid-via its recently identified receptor mitochondrial carrier homolog 2-involves multiple mechanisms, including release of cytochrome c and second mitochondria-derived activator of caspase (Smac), alteration of mitochondrial cristae organization, generation of reactive oxygen species and engagement of the permeability transition pore. Bid is also emerging-in its full-length form-as a pivotal sentinel of DNA damage in the bone marrow regulated by the ataxia telangiectasia mutated (ATM)/ataxia telangiectasia and Rad3-related (ATR) kinases. The ATM/ATR-Bid pathway is critically involved in preserving the quiescence and survival of hematopoietic stem cells both in the absence and presence of external stress, and a large part of this review will be dedicated to recent advances in this area of research.
Collapse
Affiliation(s)
- S S Zinkel
- Departments of Medicine, Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - X M Yin
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - A Gross
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
8
|
Soleymaninejadian E, Pramanik K, Samadian E. Immunomodulatory Properties of Mesenchymal Stem Cells: Cytokines and Factors. Am J Reprod Immunol 2011; 67:1-8. [DOI: 10.1111/j.1600-0897.2011.01069.x] [Citation(s) in RCA: 175] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
9
|
Grzelakowska-Sztabert B, Dudkowska M. Paradoxical action of growth factors: antiproliferative and proapoptotic signaling by HGF/c-MET. Growth Factors 2011; 29:105-18. [PMID: 21631393 DOI: 10.3109/08977194.2011.585609] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Hepatocyte growth factor (HGF)/mesenchymal-epithelial transition factor (c-MET) signaling is usually associated with the promotion of cellular growth and often with progression of tumors. Nevertheless, under certain conditions HGF can also act as an antiproliferative and proapoptotic factor and can sensitize various cancer cells, treated with anticancer drugs, to apoptosis. Not only HGF but also its various truncated forms as well as intracellular fragments of its membrane receptor, c-MET, may act as antiproliferative and proapoptotic factors toward various cells. This review focuses on different mechanisms responsible for such paradoxical action of the known typical growth factor. It also points toward the possibilities of usage of this information in anticancer therapy.
Collapse
|
10
|
|
11
|
Walter D, Schmich K, Vogel S, Pick R, Kaufmann T, Hochmuth FC, Haber A, Neubert K, McNelly S, von Weizsäcker F, Merfort I, Maurer U, Strasser A, Borner C. Switch from type II to I Fas/CD95 death signaling on in vitro culturing of primary hepatocytes. Hepatology 2008; 48:1942-53. [PMID: 19003879 PMCID: PMC2993691 DOI: 10.1002/hep.22541] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
UNLABELLED Fas/CD95-induced apoptosis of hepatocytes in vivo proceeds through the so-called type II pathway, requiring the proapoptotic BH3-only Bcl-2 family member Bid for mitochondrial death signaling. Consequently, Bid-deficient mice are protected from anti-Fas antibody injection induced fatal hepatitis. We report the unexpected finding that freshly isolated mouse hepatocytes, cultured on collagen or Matrigel, become independent of Bid for Fas-induced apoptosis, thereby switching death signaling from type II to type I. In such in vitro cultures, Fas ligand (FasL) activates caspase-3 without Bid cleavage, Bax/Bak activation or cytochrome c release, and neither Bid ablation nor Bcl-2 overexpression is protective. The type II to type I switch depends on extracellular matrix adhesion, as primary hepatocytes in suspension die in a Bid-dependent manner. Moreover, the switch is specific for FasL-induced apoptosis as collagen-plated Bid-deficient hepatocytes are protected from tumor necrosis factor alpha/actinomycin D (TNFalpha/ActD)-induced apoptosis. CONCLUSION Our data suggest a selective crosstalk between extracellular matrix and Fas-mediated signaling that favors mitochondria-independent type I apoptosis induction.
Collapse
Affiliation(s)
- Dorothée Walter
- Institute of Molecular Medicine and Cell Research, Centre of Biochemistry and Molecular Research (ZBMZ), Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, D-79104 Freiburg, Germany,Faculty of Biology, Albert Ludwigs University Freiburg
| | - Kathrin Schmich
- Department of Pharmaceutical Biology and Biotechnology, Stefan Meier Str. 19, Albert Ludwigs University Freiburg, D-79104 Freiburg
| | - Sandra Vogel
- Institute of Molecular Medicine and Cell Research, Centre of Biochemistry and Molecular Research (ZBMZ), Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, D-79104 Freiburg, Germany,Faculty of Biology, Albert Ludwigs University Freiburg,Spemann Graduate School of Biology and Medicine (SGBM), Albertstrasse 19a, Albert Ludwigs University Freiburg, D-79104 Freiburg
| | - Robert Pick
- Institute of Molecular Medicine and Cell Research, Centre of Biochemistry and Molecular Research (ZBMZ), Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, D-79104 Freiburg, Germany
| | - Thomas Kaufmann
- The Walter and Eliza Hall Institute Medical Research, Parkville, VIC 3050, Australia
| | - Florian Christoph Hochmuth
- Institute of Molecular Medicine and Cell Research, Centre of Biochemistry and Molecular Research (ZBMZ), Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, D-79104 Freiburg, Germany
| | - Angelika Haber
- Institute of Molecular Medicine and Cell Research, Centre of Biochemistry and Molecular Research (ZBMZ), Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, D-79104 Freiburg, Germany
| | - Karin Neubert
- Institute of Molecular Medicine and Cell Research, Centre of Biochemistry and Molecular Research (ZBMZ), Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, D-79104 Freiburg, Germany
| | - Sabine McNelly
- Internal Medicine, University Clinic Freiburg, Hugstetterstrasse 55, D-79106 Freiburg
| | - Fritz von Weizsäcker
- Internal Medicine, University Clinic Freiburg, Hugstetterstrasse 55, D-79106 Freiburg
| | - Irmgard Merfort
- Department of Pharmaceutical Biology and Biotechnology, Stefan Meier Str. 19, Albert Ludwigs University Freiburg, D-79104 Freiburg
| | - Ulrich Maurer
- Institute of Molecular Medicine and Cell Research, Centre of Biochemistry and Molecular Research (ZBMZ), Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, D-79104 Freiburg, Germany
| | - Andreas Strasser
- The Walter and Eliza Hall Institute Medical Research, Parkville, VIC 3050, Australia
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Centre of Biochemistry and Molecular Research (ZBMZ), Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, D-79104 Freiburg, Germany,Department of Pharmaceutical Biology and Biotechnology, Stefan Meier Str. 19, Albert Ludwigs University Freiburg, D-79104 Freiburg,To whom correspondence should be addressed:
| |
Collapse
|
12
|
Li Y, Fan X, Goodwin CR, Laterra J, Xia S. Hepatocyte growth factor enhances death receptor-induced apoptosis by up-regulating DR5. BMC Cancer 2008; 8:325. [PMID: 18992144 PMCID: PMC2590617 DOI: 10.1186/1471-2407-8-325] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Accepted: 11/07/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hepatocyte growth factor (HGF) and its receptor c-MET are commonly expressed in malignant gliomas and embryonic neuroectodermal tumors including medulloblastoma and appear to play an important role in the growth and dissemination of these malignancies. Dependent on cell context and the involvement of specific downstream effectors, both pro- and anti-apoptotic effects of HGF have been reported. METHODS Human medulloblastoma cells were treated with HGF for 24-72 hours followed by death receptor ligand TRAIL (Tumor necrosis factor-related apoptosis-inducing ligand) for 24 hours. Cell death was measured by MTT and Annexin-V/PI flow cytometric analysis. Changes in expression levels of targets of interest were measured by Northern blot analysis, quantitative reverse transcription-PCR, Western blot analysis as well as immunoprecipitation. RESULTS In this study, we show that HGF promotes medulloblastoma cell death induced by TRAIL. TRAIL alone triggered apoptosis in DAOY cells and death was enhanced by pre-treating the cells with HGF for 24-72 h prior to the addition of TRAIL. HGF (100 ng/ml) enhanced TRAIL (10 ng/ml) induced cell death by 36% (P<0.001). No cell death was associated with HGF alone. Treating cells with PHA-665752, a specific c-Met receptor tyrosine kinase inhibitor, significantly abrogated the enhancement of TRAIL-induced cell death by HGF, indicating that its death promoting effect requires activation of its canonical receptor tyrosine kinase. Cell death induced by TRAIL+HGF was predominately apoptotic involving both extrinsic and intrinsic pathways as evidenced by the increased activation of caspase-3, 8, 9. Promotion of apoptosis by HGF occurred via the increased expression of the death receptor DR5 and enhanced formation of death-inducing signal complexes (DISC). CONCLUSION Taken together, these and previous findings indicate that HGF:c-Met pathway either promotes or inhibits medulloblastoma cell death via pathway and context specific mechanisms.
Collapse
Affiliation(s)
- Yang Li
- Hugo W, Moser Research Institute at Kennedy Krieger, Baltimore, MD, USA.
| | | | | | | | | |
Collapse
|