1
|
Gadd VL, Ferreira-Gonzalez S, Man TY, Kilpatrick AM, Aird RE, Smith IP, Rodrigo-Torres D, Kurian D, Hallett JM, Ashmore-Harris C, Esser H, Ferreira MF, Macmillan MT, Lu WY, Forbes SJ. Host hepatocyte senescence determines the success of hepatocyte transplantation in a mouse model of liver injury. J Hepatol 2025:S0168-8278(24)02830-7. [PMID: 39755157 DOI: 10.1016/j.jhep.2024.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/06/2025]
Abstract
BACKGROUND & AIMS Hepatocyte transplantation has shown promise for genetic diseases of the hepatocytes but to date has shown limited efficacy for non-genetic forms of severe liver injury. Limited cell engraftment and poor function of donor hepatocytes in recipient livers impacts the clinical utility of hepatocyte cell therapy. The mechanisms underpinning this are poorly understood. We explored this in a liver injury model, where predictable levels of injury and hepatocyte senescence were induced in AhCreMdm2fl/fl mice through genetic excision of hepatocyte Mdm2. METHODS Freshly isolated mouse or human cryopreserved hepatocytes were delivered via intrasplenic injection into AhCreMdm2fl/fl (immune competent and deficient strains) mice. Engraftment kinetics, donor cell engraftment and host liver function were assessed. Paired transcriptomic and proteomic analyses were performed on healthy vs. senescent mouse hepatocytes. RESULTS We found inhibition of host hepatocyte proliferation and liver injury is a requirement for donor hepatocyte engraftment and long-term repopulation, improving liver repair and function, but excessive senescence inhibited this process, causing a decline in graft function due to transmission of senescence from host to donor cells. Paired proteomic and transcriptomic analyses of healthy vs. senescent hepatocytes reveal a unique senescent signature associated with paracrine senescence. Modification of the host niche prior to transplantation with the senotherapeutic drug ABT737 improved donor cell proliferative capacity. CONCLUSIONS The host niche impacts the initial engraftment and long-term function of transplanted hepatocytes. Targeting paracrine senescence may be a way to improve donor hepatocyte function, optimise therapy and guide translation into the clinic. IMPACT AND IMPLICATIONS Hepatocyte transplantation has shown promise for genetic diseases but has limited efficacy for acute and severe liver injury. Poor engraftment and functionality have prevented large-scale clinical application. We show that host senescence provides the required non-competitive niche for donor hepatocytes to repopulate the recipient liver, but can, paradoxically, negatively impact donor function. These findings demonstrate a requirement for a clear understanding of the host niche prior to cell transfusion. This has significant implications not only for hepatocellular therapies, but also when developing and optimising any preclinical and clinical cell therapies.
Collapse
Affiliation(s)
- Victoria L Gadd
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Sofia Ferreira-Gonzalez
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom; Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Tak Yung Man
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Rhona E Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Ian P Smith
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Daniel Rodrigo-Torres
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Dominic Kurian
- Proteomic and Metabolomics Unit, Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, United Kingdom
| | - John M Hallett
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Candice Ashmore-Harris
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Hannah Esser
- Department of Visceral, Transplant and Thoracic Surgery, Centre of Operative Medicine, Medical University of Innsbruck, Innsbruck, 6020, Austria
| | - Marisa F Ferreira
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Mark T Macmillan
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Wei-Yu Lu
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom; Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, EH16 4UU, United Kingdom.
| |
Collapse
|
2
|
Sun Z, Yuan X, Wu J, Wang C, Zhang K, Zhang L, Hui L. Hepatocyte transplantation: The progress and the challenges. Hepatol Commun 2023; 7:e0266. [PMID: 37695736 PMCID: PMC10497249 DOI: 10.1097/hc9.0000000000000266] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/26/2023] [Indexed: 09/13/2023] Open
Abstract
Numerous studies have shown that hepatocyte transplantation is a promising approach for liver diseases, such as liver-based metabolic diseases and acute liver failure. However, it lacks strong evidence to support the long-term therapeutic effects of hepatocyte transplantation in clinical practice. Currently, major hurdles include availability of quality-assured hepatocytes, efficient engraftment and repopulation, and effective immunosuppressive regimens. Notably, cell sources have been advanced recently by expanding primary human hepatocytes by means of dedifferentiation in vitro. Moreover, the transplantation efficiency was remarkably improved by the established preparative hepatic irradiation in combination with hepatic mitogenic stimuli regimens. Finally, immunosuppression drugs, including glucocorticoid and inhibitors for co-stimulating signals of T cell activation, were proposed to prevent innate and adaptive immune rejection of allografted hepatocytes. Despite remarkable progress, further studies are required to improve in vitro cell expansion technology, develop clinically feasible preconditioning regimens, and further optimize immunosuppression regimens or establish ex vivo gene correction-based autologous hepatocyte transplantation.
Collapse
Affiliation(s)
- Zhen Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Xiang Yuan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jingqi Wu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Chenhua Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Kun Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ludi Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
3
|
Ma H, Wang C, Liang S, Yu X, Yuan Y, Lv Z, Zhang J, Jin C, Zhu J, Wang C, Sun P, Li W. ROCK inhibition enhanced hepatocyte liver engraftment by retaining membrane CD59 and attenuating complement activation. Mol Ther 2023; 31:1846-1856. [PMID: 36860134 PMCID: PMC10277888 DOI: 10.1016/j.ymthe.2023.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/19/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Hepatocyte transplantation can be an effective treatment for patients with certain liver-based metabolic disorders and liver injuries. Hepatocytes are usually infused into the portal vein, from which hepatocytes migrate into the liver and integrate into the liver parenchyma. However, early cell loss and poor liver engraftment represent major hurdles to sustaining the recovery of diseased livers after transplantation. In the present study, we found that ROCK (Rho-associated kinase) inhibitors significantly enhanced in vivo hepatocyte engraftment. Mechanistic studies suggested that the isolation of hepatocytes caused substantial degradation of cell membrane proteins, including the complement inhibitor CD59, probably due to shear stress-induced endocytosis. ROCK inhibition by ripasudil, a clinically used ROCK inhibitor, can protect transplanted hepatocytes by retaining cell membrane CD59 and blocking the formation of the membrane attack complex. Knockdown of CD59 in hepatocytes eliminates ROCK inhibition-enhanced hepatocyte engraftment. Ripasudil can accelerate liver repopulation of fumarylacetoacetate hydrolase-deficient mice. Our work reveals a mechanism underlying hepatocyte loss after transplantation and provides immediate strategies to enhance hepatocyte engraftment by inhibiting ROCK.
Collapse
Affiliation(s)
- Haoxin Ma
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
| | - Chao Wang
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
| | - Shulong Liang
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
| | - Xinlu Yu
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
| | - Yuan Yuan
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
| | - Zhuanman Lv
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
| | - Jiqianzhu Zhang
- Department of Health Toxicology, Naval Medical University, Shanghai 200433, China
| | - Caixia Jin
- Department of Regenerative Medicine, College of Medicine, Tongji University, Shanghai 200433, China
| | - Jiangbo Zhu
- Department of Health Toxicology, Naval Medical University, Shanghai 200433, China
| | - Chao Wang
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
| | - Pingxin Sun
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
| | - Wenlin Li
- Department of Cell Biology, Naval Medical University, Shanghai 200433, China
- Shanghai Key Laboratory of Cell Engineering, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
4
|
Angelica sinensis Polysaccharide and Astragalus membranaceus Polysaccharide Accelerate Liver Regeneration by Enhanced Glycolysis via Activation of JAK2/STAT3/HK2 Pathway. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227890. [PMID: 36431990 PMCID: PMC9695464 DOI: 10.3390/molecules27227890] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/05/2022] [Accepted: 11/09/2022] [Indexed: 11/17/2022]
Abstract
The promotion of liver regeneration is crucial to avoid liver failure after hepatectomy. Angelica sinensis polysaccharide (ASP) and Astragalus membranaceus polysaccharide (AMP) have been identified as being associated with hepatoprotective effects. However, their roles and specific mechanisms in liver regeneration remain to be elucidated. In the present study, it suggested that the respective use of ASP or AMP strikingly promoted hepatocyte proliferation in vitro with a wide range of concentrations (from 12.5 μg/mL to 3200 μg/mL), and a stronger promoting effect was observed in combined interventions. A significantly enhanced liver/body weight ratio (4.20%) on day 7 and reduced serum transaminase (ALT 243.53 IU/L and AST 423.74 IU/L) and total bilirubin (52.61 IU/L) levels on day 3 were achieved by means of ASP-AMP administration after partial hepatectomy in mice. Metabonomics showed that differential metabolites were enriched in glycolysis with high expression of beta-d-fructose 6-phosphate and lactate, followed by significantly strengthened lactate secretion in the supernatant (0.54) and serum (0.43) normalized to control. Upon ASP-AMP treatment, the knockdown of hexokinase 2 (HK2) or inhibited glycolysis caused by 2-deoxy-d-glucose decreased hepatocyte proliferation in vitro and in vivo. Furthermore, pathway analysis predicted the role of JAK2/STAT3 pathway in ASP-AMP-regulated liver regeneration, and phosphorylation of JAK2 and STAT3 was proven to be elevated in this promoting process. Finally, downregulated expression of HK2, an attenuated level of lactate secretion, and reduced hepatocyte proliferation were displayed when STAT3 was knocked out in vitro. Therefore, it can be concluded that ASP-AMP accelerated liver regeneration and exerted a hepatoprotective effect after hepatectomy, in which the JAK2/STAT3/HK2 pathway was actively involved in activating glycolysis.
Collapse
|
5
|
Hung YT, Chang YR, Wang HN, Lee WC, Lee CF, Chen CB. Diagnostic and therapeutic dilemma in Stevens–Johnson syndrome-like acute graft-versus-host disease after liver transplantation: A case report. Front Immunol 2022; 13:917782. [PMID: 36059444 PMCID: PMC9433559 DOI: 10.3389/fimmu.2022.917782] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022] Open
Abstract
Background Acute graft-versus-host disease (aGVHD) is a severe and fatal complication after orthotopic liver transplantation (OLT). Clinical manifestations of severe aGVHD can resemble drug-induced Stevens–Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN), and there are also various medications, such as antibiotics and immunosuppressants, used after transplantation, causing a diagnostic dilemma. Furthermore, there have been no standardized diagnostic and therapeutic strategies for OLT-aGVHD due to its rarity. Case summary A 52-year-old man presented with generalized maculopapular eruptions, fever, and pancytopenia 1 month after OLT and 4 days after taking sulfamethoxazole/trimethoprim. After assessment of the scoring criteria for drug causality of drug allergy, histopathological findings of skin biopsy, lymphocyte activation test of the potential offending drug, and microchimerism study, the diagnosis was in favor of aGVHD mimicking SJS/TEN. Considering severe sepsis, the anti-tumor necrosis factor alpha (TNF-α) agent, etanercept, was used to replace tacrolimus and corticosteroid. Skin lesions resolved gradually after anti-TNF-α biologics rescue; tacrolimus and corticosteroid therapy were re-administrated after controlling sepsis. Pancytopenia recovered and the patient was discharged in a stable condition. Conclusion We demonstrated a diagnostic strategy for OLT-aGVHD. Targeting therapy with anti-TNF-α blockade and a temporary withdrawal of traditional immunosuppressants may be among effective and safe therapeutic options of OLT-aGVHD for those with severe sepsis.
Collapse
Affiliation(s)
- Yi-Teng Hung
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei and Keelung, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Yau-Ren Chang
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Hsuan-Ning Wang
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei and Keelung, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Dermatology, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
| | - Wei-Chen Lee
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chen-Fang Lee
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Liver and Transplantation Surgery, Chang Gung Memorial Hospital, Linkou, Taiwan
- *Correspondence: Chen-Fang Lee, ; Chun-Bing Chen,
| | - Chun-Bing Chen
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei and Keelung, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou, Taiwan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan City, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- *Correspondence: Chen-Fang Lee, ; Chun-Bing Chen,
| |
Collapse
|
6
|
Liu Y, Yu M, Cui J, Du Y, Teng X, Zhang Z. Heat shock proteins took part in oxidative stress-mediated inflammatory injury via NF-κB pathway in excess manganese-treated chicken livers. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 226:112833. [PMID: 34600291 DOI: 10.1016/j.ecoenv.2021.112833] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 06/13/2023]
Abstract
Manganese (Mn) is an essential metal in humans and animals. However, excess Mn entered environment due to the wide application of Mn in industry and agriculture, and became an environmental pollutant. Exposure to high doses of Mn is toxic to humans and animals (including chickens). Liver is a target organ of Mn poisoning. Nevertheless, there were few studies on whether Mn poisoning damages chicken livers and poisoning mechanism of Mn in chicken livers. Herein, the aim of this study was to explore if oxidative stress, heat shock proteins (HSPs), and inflammatory response were involved in the mechanism of Mn poisoning-caused damage in chicken livers. A chicken Mn poisoning model was established. One hundred and eighty chickens were randomly divided into one control group (containing 127.88 mg Mn kg-1) and three Mn-treated groups (containing 600, 900, and 1800 mg Mn kg-1, respectively). Histomorphological structure was observed via microstructure and ultrastructure. Spectrophotometry was used to detect total antioxidant capacity (T-AOC) and inducible nitric oxide synthase (iNOS) activity, as well as nitric oxide (NO) content. And qRT-PCR was performed to measure mRNA expression of inflammatory genes (nuclear factor kappa B (NF-κB), tumor necrosis factor α (TNF-α), cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), and iNOS) and heat shock protein (HSP) genes (HSP27, HSP40, HSP60, HSP70, and HSP90). Multivariate correlation analysis, principal component analysis, and cluster analysis were used to demonstrate the reliability of mechanism of Mn poisoning in our experiment. The results indicated that excess Mn led to inflammatory injury at three contents and three time points. Meanwhile, we found that NO content, iNOS activity, and NF-κB, TNF-α, COX-2, PGE2, and iNOS mRNA expression increased after Mn treatment, meaning that exposure to Mn induced inflammatory response via NF-κB pathway in chicken livers. Moreover, excess Mn decreased T-AOC activity, indicating that Mn exposure caused oxidative stress. Furthermore, mRNA expression of above five HSP genes was up-regulated during Mn exposure. Oxidative stress triggered the increase of HSPs and the increase of HSPs mediated inflammatory response induced by Mn. In addition, there were time- and dose-dependent effects on Mn-caused chicken liver inflammatory injury. Taken together, HSPs participated in oxidative stress-mediated inflammatory damage caused by excess Mn in chicken livers via NF-κB pathway. For the first time, we found that oxidative stress can trigger HSP70 and HSPs can trigger poisoning-caused inflammatory damage, which needs to be further explored. This study provided a new insight into environmental pollutants and a reference for further study on molecular mechanisms of poisoning.
Collapse
Affiliation(s)
- Yuhao Liu
- School of Animal Science, Inner Mongolia Agricultural University, Hohhot City 010018, PR China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Meijin Yu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Jiawen Cui
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Ye Du
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China; Electrical and Information Engineering College, Jilin Agricultural Science and Technology University, Jilin 132101, PR China.
| | - Zuozhong Zhang
- School of Animal Science, Inner Mongolia Agricultural University, Hohhot City 010018, PR China.
| |
Collapse
|
7
|
Viswanathan P, Sharma Y, Jaber FL, Tchaikovskaya T, Gupta S. Transplanted hepatocytes rescue mice in acetaminophen-induced acute liver failure through paracrine signals for hepatic ATM and STAT3 pathways. FASEB J 2021; 35:e21471. [PMID: 33683737 DOI: 10.1096/fj.202002421r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/27/2021] [Accepted: 02/08/2021] [Indexed: 12/29/2022]
Abstract
Acute liver failure constitutes a devastating condition that needs novel cell and molecular therapies. To elicit synergisms in cell types of therapeutic interest, we studied hepatocytes and liver sinusoidal endothelial in mice with acetaminophen-induced acute liver failure. The context of regenerative signals was examined by transplants in peritoneal cavity because it possesses considerable capacity and allows soluble signals to enter the systemic circulation. Whereas transplanted hepatocytes and liver sinusoidal endothelial cells engrafted in peritoneal cavity, only the former could rescue mice in liver failure by improving injury outcomes, activating hepatic DNA damage repair, and inducing liver regeneration. The cytokines secreted by donor hepatocytes or liver sinusoidal endothelial cells differed and in hepatocytes from mice undergoing acetaminophen toxicity major cytokines were even rendered deficient (eg, G-CSF, VEGF, and others). Significantly, recapitulating hepatotoxicity-related DNA damage response in cultured cells identified impairments in ATM and JAK/STAT3 intersections since replacing cytokines produced less from injured hepatocytes restored these pathways to avoid acetaminophen hepatotoxicity. Similarly, hepatocyte transplantation in acute liver failure restored ATM and JAK/STAT3 pathways to advance DNA damage/repair and liver regeneration. The unexpected identification of novel hepatic G-CSF receptor expression following injury allowed paradigmatic studies of G-CSF supplementation to confirm the centrality of this paracrine ATM and STAT3 intersection. Remarkably, DNA damage/repair and hepatic regeneration directed by G-CSF concerned rebalancing of regulatory gene networks overseeing inflammation, metabolism, and cell viability. We conclude that healthy donor hepatocytes offer templates for generating specialized cell types to replace metabolic functions and regenerative factors in liver failure.
Collapse
Affiliation(s)
- Preeti Viswanathan
- Division of Pediatric Gastroenterology, Department of Pediatrics, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA.,Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yogeshwar Sharma
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Fadi-Luc Jaber
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Tatyana Tchaikovskaya
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sanjeev Gupta
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA.,Diabetes Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, USA.,Irwin S. and Sylvia Chanin Institute for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA.,Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
8
|
Jaber FL, Sharma Y, Mui BG, Kapoor S, Gupta S. Tumor Necrosis Factor Directs Allograft-Related Innate Responses and Its Neutralization Improves Hepatocyte Engraftment in Rats. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:79-89. [PMID: 33127336 PMCID: PMC7768347 DOI: 10.1016/j.ajpath.2020.09.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/09/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
The innate immune system plays a critical role in allograft rejection. Alloresponses involve numerous cytokines, chemokines, and receptors that cause tissue injury during rejection. To dissect these inflammatory mechanisms, we developed cell transplantation models in dipeptidylpeptidase-deficient F344 rats using mycophenolate mofetil and tacrolimus for partial lymphocyte-directed immunosuppression. Syngeneic hepatocytes engrafted in liver, whereas allogeneic hepatocytes were rejected but engrafted after immunosuppression. These transplants induced mRNAs for >40 to 50 cytokines, chemokines, and receptors. In allografts, innate cell type-related regulatory networks extended to granulocytes, monocytes, and macrophages. Activation of Tnfa and its receptors or major chemokine receptor-ligand subsets persisted in the long term. An examination of the contribution of Tnfa in allograft response revealed that it was prospectively antagonized by etanercept or thalidomide, which resolved cytokine, chemokine, and receptor cascades. In bioinformatics analysis of upstream regulator networks, the Cxcl8 pathway exhibited dominance despite immunosuppression. Significantly, Tnfa antagonism silenced the Cxcl8 pathway and decreased neutrophil and Kupffer cell recruitment, resulting in multifold greater engraftment of allogeneic hepatocytes and substantially increased liver repopulation in retrorsine/partial hepatectomy model. We conclude that Tnfa is a major driver for persistent innate immune responses after allogeneic cells. Neutralizing Tnfa should help in avoiding rejection and associated tissue injury in the allograft setting.
Collapse
Affiliation(s)
- Fadi Luc Jaber
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Yogeshwar Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Brandon G Mui
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Sorabh Kapoor
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York
| | - Sanjeev Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York; Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York; Department of Pathology, Albert Einstein College of Medicine, Bronx, New York; Diabetes Center, Albert Einstein College of Medicine, Bronx, New York; Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, New York; Irwin S. and Sylvia Chanin Institute for Cancer Research, Albert Einstein College of Medicine, Bronx, New York; Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
9
|
Yadav N, Jaber FL, Sharma Y, Gupta P, Viswanathan P, Gupta S. Efficient Reconstitution of Hepatic Microvasculature by Endothelin Receptor Antagonism in Liver Sinusoidal Endothelial Cells. Hum Gene Ther 2018; 30:365-377. [PMID: 30266073 DOI: 10.1089/hum.2018.166] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Reconstitution of healthy endothelial cells in vascular beds offers opportunities for mechanisms in tissue homeostasis, organ regeneration, and correction of deficient functions. Liver sinusoidal endothelial cells express unique functions, and their transplantation is relevant for disease models and for cell therapy. As molecular targets for improving transplanted cell engraftment and proliferation will be highly significant, this study determined whether ETA/B receptor antagonism by the drug bosentan could overcome cell losses due to cell transplantation-induced cytotoxicity. Cell engraftment and proliferation assays were performed with healthy wild-type liver sinusoidal endothelial cells transplanted into the liver of dipeptidylpeptidase IV knockout mice. Transplanted cells were identified in tissues by enzyme histochemistry. Cells with prospective ETA/B antagonism engrafted significantly better in hepatic sinusoids. Moreover, these cells underwent multiple rounds of division under liver repopulation conditions. The gains of ETA/B antagonism resulted from benefits in cell viability and membrane integrity. Also, in bosentan-treated cells, mitochondrial homeostasis was better maintained with less oxidative stress and DNA damage after injuries. Intracellular effects of ETA/B antagonism were transduced by conservation of ataxia telangiectasia mutated protein, which directs DNA damage response. Therefore, ETA/B antagonism in donor cells will advance vascular reconstitution. Extensive experience with ETA/B antagonists will facilitate translation in people.
Collapse
Affiliation(s)
- Neelam Yadav
- 1 Department of Medicine, Albert Einstein College of Medicine, Bronx, New York.,2 Department of Biochemistry, Dr. RML Avadh University, Faizabad, India
| | - Fadi Luc Jaber
- 1 Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Yogeshwar Sharma
- 1 Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Priya Gupta
- 1 Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| | - Preeti Viswanathan
- 3 Department of Pediatrics, Albert Einstein College of Medicine and Children's Hospital at Montefiore, Bronx, New York
| | - Sanjeev Gupta
- 1 Department of Medicine, Albert Einstein College of Medicine, Bronx, New York.,4 Department of Pathology, Albert Einstein College of Medicine, Bronx, New York.,5 Marion Bessin Liver Research Center, Diabetes Center, Irwin S. and Sylvia Chanin Institute for Cancer Research, and Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
10
|
Sharma Y, Liu J, Kristian KE, Follenzi A, Gupta S. In Atp7b-/- Mice Modeling Wilson's Disease Liver Repopulation With Bone Marrow-Derived Myofibroblasts or Inflammatory Cells and Not Hepatocytes Is Deleterious. Gene Expr 2018; 19:15-24. [PMID: 30029699 PMCID: PMC6290324 DOI: 10.3727/105221618x15320123457380] [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] [Indexed: 11/24/2022]
Abstract
In Wilson's disease, Atp7b mutations impair copper excretion with liver or brain damage. Healthy transplanted hepatocytes repopulate the liver, excrete copper, and reverse hepatic damage in animal models of Wilson's disease. In Fah-/- mice with tyrosinemia and α-1 antitrypsin mutant mice, liver disease is resolved by expansions of healthy hepatocytes derived from transplanted healthy bone marrow stem cells. This potential of stem cells has not been defined for Wilson's disease. In diseased Atp7b-/- mice, we reconstituted bone marrow with donor cells expressing green fluorescent protein reporter from healthy transgenic mice. Mature hepatocytes originating from donor bone marrow were identified by immunostaining for green fluorescence protein and bile canalicular marker, dipeptidylpeptidase-4. Mesenchymal and inflammatory cell markers were used for other cells from donor bone marrow cells. Gene expression, liver tests, and tissues were analyzed for outcomes in Atp7b-/- mice. After bone marrow transplantation in Atp7b-/- mice, donor-derived hepatocytes containing bile canaliculi appeared within weeks. Despite this maturity, donor-derived hepatocytes neither divided nor expanded. The liver of Atp7b-/- mice was not repopulated by donor-derived hepatocytes: Atp7b mRNA remained undetectable; liver tests, copper content, and fibrosis actually worsened. Restriction of proliferation in hepatocytes accompanied oxidative DNA damage. By contrast, donor-derived mesenchymal and inflammatory cells extensively proliferated. These contributed to fibrogenesis through greater expression of inflammatory cytokines. In Wilson's disease, donor bone marrow-derived cells underwent different fates: hepatocytes failed to proliferate; inflammatory cells proliferated to worsen disease outcomes. This will help guide stem cell therapies for conditions with proinflammatory or profibrogenic microenvironments.
Collapse
Affiliation(s)
- Yogeshwar Sharma
- *Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jinghua Liu
- †Department of Obstetrics and Gynecology, Shanghai Public Health Clinical Center, Shanghai, P.R. China
| | | | - Antonia Follenzi
- §Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
- ¶Department of Health Sciences, Università del Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Sanjeev Gupta
- *Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- §Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
- #Marion Bessin Liver Research Center, Diabetes Center, Cancer Center, and Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
11
|
Kakabadze Z, Kakabadze A, Chakhunashvili D, Karalashvili L, Berishvili E, Sharma Y, Gupta S. Decellularized human placenta supports hepatic tissue and allows rescue in acute liver failure. Hepatology 2018; 67:1956-1969. [PMID: 29211918 PMCID: PMC5906146 DOI: 10.1002/hep.29713] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/29/2017] [Accepted: 12/04/2017] [Indexed: 12/29/2022]
Abstract
UNLABELLED Tissue engineering with scaffolds to form transplantable organs is of wide interest. Decellularized tissues have been tested for this purpose, although supplies of healthy donor tissues, vascular recellularization for perfusion, and tissue homeostasis in engineered organs pose challenges. We hypothesized that decellularized human placenta will be suitable for tissue engineering. The universal availability and unique structures of placenta for accommodating tissue, including presence of embedded vessels, were major attractions. We found decellularized placental vessels were reendothelialized by adjacent native cells and bridged vessel defects in rats. In addition, implantation of liver fragments containing all cell types successfully hepatized placenta with maintenance of albumin and urea synthesis, as well as hepatobiliary transport of 99m Tc-mebrofenin, up to 3 days in vitro. After hepatized placenta containing autologous liver was transplanted into sheep, tissue units were well-perfused and self-assembled. Histological examination indicated transplanted tissue retained hepatic cord structures with characteristic hepatic organelles, such as gap junctions, and hepatic sinusoids lined by endothelial cells, Kupffer cells, and other cell types. Hepatocytes in this neo-organ expressed albumin and contained glycogen. Moreover, transplantation of hepatized placenta containing autologous tissue rescued sheep in extended partial hepatectomy-induced acute liver failure. This rescue concerned amelioration of injury and induction of regeneration in native liver. The grafted hepatized placenta was intact with healthy tissue that neither proliferated nor was otherwise altered. CONCLUSION The unique anatomic structure and matrix of human placenta were effective for hepatic tissue engineering. This will advance applications ranging from biological studies, drug development, and toxicology to patient therapies. (Hepatology 2018;67:1956-1969).
Collapse
Affiliation(s)
- Zurab Kakabadze
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - Ann Kakabadze
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - David Chakhunashvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - Lia Karalashvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - Ekaterine Berishvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | | | - Sanjeev Gupta
- Department of Medicine, Bronx, New York, USA,Department of Pathology, Marion Bessin Liver Research Center, Diabetes Center, The Irwin S. and Sylvia Chanin Institute for Cancer Research, Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
12
|
Kakabadze Z, Kakabadze A, Chakhunashvili D, Karalashvili L, Berishvili E, Sharma Y, Gupta S. Decellularized human placenta supports hepatic tissue and allows rescue in acute liver failure. Hepatology 2018. [PMID: 29211918 DOI: 10.1002/hep.v67.5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
UNLABELLED Tissue engineering with scaffolds to form transplantable organs is of wide interest. Decellularized tissues have been tested for this purpose, although supplies of healthy donor tissues, vascular recellularization for perfusion, and tissue homeostasis in engineered organs pose challenges. We hypothesized that decellularized human placenta will be suitable for tissue engineering. The universal availability and unique structures of placenta for accommodating tissue, including presence of embedded vessels, were major attractions. We found decellularized placental vessels were reendothelialized by adjacent native cells and bridged vessel defects in rats. In addition, implantation of liver fragments containing all cell types successfully hepatized placenta with maintenance of albumin and urea synthesis, as well as hepatobiliary transport of 99m Tc-mebrofenin, up to 3 days in vitro. After hepatized placenta containing autologous liver was transplanted into sheep, tissue units were well-perfused and self-assembled. Histological examination indicated transplanted tissue retained hepatic cord structures with characteristic hepatic organelles, such as gap junctions, and hepatic sinusoids lined by endothelial cells, Kupffer cells, and other cell types. Hepatocytes in this neo-organ expressed albumin and contained glycogen. Moreover, transplantation of hepatized placenta containing autologous tissue rescued sheep in extended partial hepatectomy-induced acute liver failure. This rescue concerned amelioration of injury and induction of regeneration in native liver. The grafted hepatized placenta was intact with healthy tissue that neither proliferated nor was otherwise altered. CONCLUSION The unique anatomic structure and matrix of human placenta were effective for hepatic tissue engineering. This will advance applications ranging from biological studies, drug development, and toxicology to patient therapies. (Hepatology 2018;67:1956-1969).
Collapse
Affiliation(s)
- Zurab Kakabadze
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - Ann Kakabadze
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - David Chakhunashvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - Lia Karalashvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - Ekaterine Berishvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | - Yogeshwar Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Sanjeev Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY
- Diabetes Center, Albert Einstein College of Medicine, Bronx, NY
- The Irwin S. and Sylvia Chanin Institute for Cancer Research, Albert Einstein College of Medicine, Bronx, NY
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY
| |
Collapse
|
13
|
Viswanathan P, Sharma Y, Gupta P, Gupta S. Replicative stress and alterations in cell cycle checkpoint controls following acetaminophen hepatotoxicity restrict liver regeneration. Cell Prolif 2018; 51:e12445. [PMID: 29504225 DOI: 10.1111/cpr.12445] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/16/2018] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Acetaminophen hepatotoxicity is a leading cause of hepatic failure with impairments in liver regeneration producing significant mortality. Multiple intracellular events, including oxidative stress, mitochondrial damage, inflammation, etc., signify acetaminophen toxicity, although how these may alter cell cycle controls has been unknown and was studied for its significance in liver regeneration. MATERIALS AND METHODS Assays were performed in HuH-7 human hepatocellular carcinoma cells, primary human hepatocytes and tissue samples from people with acetaminophen-induced acute liver failure. Cellular oxidative stress, DNA damage and cell proliferation events were investigated by mitochondrial membrane potential assays, flow cytometry, fluorescence staining, comet assays and spotted arrays for protein expression after acetaminophen exposures. RESULTS In experimental groups with acetaminophen toxicity, impaired mitochondrial viability and substantial DNA damage were observed with rapid loss of cells in S and G2/M and cell cycle restrictions or even exit in the remainder. This resulted from altered expression of the DNA damage regulator, ATM and downstream transducers, which imposed G1/S checkpoint arrest, delayed entry into S and restricted G2 transit. Tissues from people with acute liver failure confirmed hepatic DNA damage and cell cycle-related lesions, including restrictions of hepatocytes in aneuploid states. Remarkably, treatment of cells with a cytoprotective cytokine reversed acetaminophen-induced restrictions to restore cycling. CONCLUSIONS Cell cycle lesions following mitochondrial and DNA damage led to failure of hepatic regeneration in acetaminophen toxicity but their reversibility offers molecular targets for treating acute liver failure.
Collapse
Affiliation(s)
- Preeti Viswanathan
- Division of Pediatric Gastroenterology and Hepatology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yogeshwar Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Priya Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sanjeev Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA.,Marion Bessin Liver Research Center, Diabetes Center, Irwin S. and Sylvia Chanin Institute for Cancer Research, Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
14
|
Jaber FL, Sharma Y, Gupta S. Demonstrating Potential of Cell Therapy for Wilson's Disease with the Long-Evans Cinnamon Rat Model. Methods Mol Biol 2017; 1506:161-178. [PMID: 27830552 DOI: 10.1007/978-1-4939-6506-9_11] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Wilson's disease (WD) is characterized by the inability to excrete copper (Cu) from the body with progressive tissue injury, especially in liver and brain. The molecular defect in WD concerns mutations in ATP7B gene leading to loss of Cu transport from the hepatocyte to the bile canaliculus. While drugs, e.g., Cu chelators, have been available for several decades, these must be taken lifelong, which can be difficult due to issues of compliance or side effects. Many individuals may require liver transplantation, which can also be difficult due to donor organ shortages. Therefore, achieving permanent cures via cell or gene therapy are of great interest for WD. Cell therapy is feasible because transplanted hepatocytes can integrate in liver parenchyma and restore deficient functions, including transport of Cu into bile. The availability of authentic animal models that recapitulate hepatic WD, especially the Long-Evans Cinnamon (LEC) rat, has advanced cell transplantation research in WD. We describe requirements for cell therapy in animal models with several standardized methods for studies to test or refine cell therapy strategies in WD.
Collapse
Affiliation(s)
- Fadi Luc Jaber
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yogeshwar Sharma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sanjeev Gupta
- Departments of Medicine and Pathology, Marion Bessin Liver Research Center, Diabetes Center, Cancer Center, Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Ullmann Building, Room 625, 1300 Morris Park Avenue, Bronx, NY, 10461, USA.
| |
Collapse
|
15
|
Moreno D, Neri L, Vicente E, Vales A, Aldabe R. Use of Thymidine Kinase Recombinant Adenovirus and Ganciclovir Mediated Mouse Liver Preconditioning for Hepatocyte Xenotransplantation. Methods Mol Biol 2017; 1506:179-192. [PMID: 27830553 DOI: 10.1007/978-1-4939-6506-9_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Hepatocyte transplantation is the best approach to maintain and propagate differentiated hepatocytes from different species. Host liver has to be adapted for transplanted hepatocytes productive engraftment and proliferation being required a chronic liver injury to eliminate host hepatocytes and provide a proliferative advantage to the transplanted hepatocytes. Most valuable mouse models for xenograft hepatocyte transplantation are based on genetically modified animals to cause a chronic liver damage and to limit host hepatocyte regeneration potential. We present a methodology that generates a chronic liver damage and can be applied to any host mouse strain and animal species based on the inoculation of a recombinant adenovirus to express herpes simplex thymidine kinase in host hepatocytes sensitizing them to ganciclovir treatment. This causes a prolonged liver damage that allows hepatocyte transplantation and generation of regenerative nodules in recipient mouse liver integrated by transplanted cells and host sinusoidal. Obtained chimeric animals maintain functional chimeric nodules for several weeks, ready to be used in any study.
Collapse
Affiliation(s)
- Daniel Moreno
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Leire Neri
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Eva Vicente
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Africa Vales
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Rafael Aldabe
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain.
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
16
|
Viswanathan P, Gupta P, Kapoor S, Gupta S. Thalidomide promotes transplanted cell engraftment in the rat liver by modulating inflammation and endothelial integrity. J Hepatol 2016; 65:1171-1178. [PMID: 27422749 PMCID: PMC5116265 DOI: 10.1016/j.jhep.2016.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 05/26/2016] [Accepted: 07/04/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND & AIMS For liver-directed cell therapy, efficient engraftment of transplanted cells is critical. This study delineated whether anti-inflammatory and endothelial disrupting properties of thalidomide could promote transplanted cell engraftment and proliferation in liver. METHODS We used dipeptidyl peptidase IV-deficient rats for cell transplantation studies, including gene expression analysis, morphological tissue analysis, serological assays, cell culture assays, and assays of transplanted cell engraftment and proliferation. RESULTS Thalidomide-pretreatment increased engraftment and proliferation of transplanted hepatocytes due to decreased inflammation. Moreover, thalidomide exacerbated cell transplantation-induced endothelial injury. This combined anti-inflammatory and endothelial injury effect of thalidomide was superior to the anti-inflammatory effect alone of repertaxin or etanercept, which block cytokines/chemokines/receptor-dependent inflammation. In thalidomide-pretreated animals, liver repopulation accelerated, including when cells were primed with bosentan to block endothelin-1 receptors. CONCLUSIONS Thalidomide improved transplanted cell engraftment and liver repopulation. Therefore, this class of drugs will advance applications of liver cell therapy in people. LAY SUMMARY This work aimed to develop effective drug treatments for improving engraftment of transplanted cells because that constitutes a critical step in rebuilding liver with healthy cells. Studies in animal models of cell transplantation led to identification of an old drug, thalidomide, which blocked inflammation and altered the liver microenvironment to yield superior engraftment and proliferation of transplanted cells. This will be appropriate for liver cell therapy in people.
Collapse
Affiliation(s)
- Preeti Viswanathan
- Division of Pediatric Gastroenterology, Department of Pediatrics, Children’s Hospital at Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY
| | - Priya Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Sorabh Kapoor
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY
| | - Sanjeev Gupta
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States; Departments of Medicine and Pathology, Marion Bessin Liver Research Center, Diabetes Center, Cancer Center, Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
17
|
Infliximab and Dexamethasone Attenuate the Ductular Reaction in Mice. Sci Rep 2016; 6:36586. [PMID: 27824131 PMCID: PMC5100545 DOI: 10.1038/srep36586] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 10/17/2016] [Indexed: 12/16/2022] Open
Abstract
Chronic hepatic injury is accompanied by a ductular response that is strongly correlated with disease severity and progression of fibrosis. To investigate whether anti-inflammatory drugs can modulate the ductular response, we treated mice suffering from a steatotic or cholestatic injury with anti-TNF-α antibodies (Infliximab) or glucocorticoids (Dexamethasone). We discovered that Dexamethasone and Infliximab can both modulate the adaptive remodeling of the biliary architecture that occurs upon liver injury and limit extracellular matrix deposition. Infliximab treatment, at least in these steatotic and cholestatic mouse models, is the safer approach since it does not increase liver injury, allows inflammation to take place but inhibits efficiently the ductular response and extracellular matrix deposition. Infliximab-based therapy could, thus, still be of importance in multiple chronic liver disorders that display a ductular response such as alcoholic liver disease or sclerosing cholangitis.
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW To date, the only curative treatment for end-stage liver disease is liver transplantation, which is limited by the shortage of available organs. Cell therapy, in the form of cell transplantation or cell-based extracorporeal support devices, may in the future offer an alternative to transplantation, or at least provide liver function support as a bridging therapy until surgery may be performed. The purpose of this review is to highlight the most recent advances made in the field of cell therapy and regenerative medicine for the treatment of chronic liver disease. RECENT FINDINGS After hepatocyte transplantation, long-term engraftment in the liver and spleen may be achieved, which can be stimulated through preconditioning, multiple infusions, and inflammatory response blockade. Mesenchymal stem cells are promising candidates for cell transplantation, as they have been shown to reduce liver fibrosis and support endogenous regeneration. Adipose tissue-derived stem cells are also being tested in this setting, because of their ready availability. Bioartificial liver devices are being built that allow for effective preservation of hepatocytes, and one such device has recently demonstrated survival benefit in a porcine model of liver failure. SUMMARY Cell transplantation of primary hepatocytes or stem cell-derived hepatocyte-like cells for the treatment of chronic liver disease holds promise. Bioartificial liver systems may in the future be able to bridge acute-on-chronic liver failure patients to liver transplantation.
Collapse
|
19
|
Ding G, Li Z, Zhang L, Ma H, Li H. Circulating full-length cytokeratin 18 for predicting prognosis of hepatitis B virus-related acute-on-chronic liver failure patients with lamivudine treatment. Hepatol Res 2016; 46:329-34. [PMID: 26138382 DOI: 10.1111/hepr.12549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 06/27/2015] [Accepted: 06/30/2015] [Indexed: 02/08/2023]
Abstract
AIM The predictive value of circulating full-length cytokeratin 18 (M65) on prognosis of hepatitis B virus-related liver failure is still not well investigated. The aim of this study was to reveal the association between serum M65 levels and outcomes of hepatitis B virus-related acute-on-chronic liver failure, and to further clarify clinical significance of M65 in lamivudine treatment. METHODS Ninety-six patients with hepatitis B virus-related acute-on-chronic liver failure were recruited between January 2011 and December 2013, and 52 of them received lamivudine treatment. Blood were obtained from participants at admission, and M65 were determined by enzyme-linked immunosorbent assay. Other necessary data were collected from medical records. RESULTS The liver failure patients were divided into high and low level groups according to the serum M65 levels. After 24-week follow up, survival incidences were significantly higher in the low level group than that in the high level group (odds ratio [OR] = 0.3, 95% confidence interval = 0.1-0.7). In addition, the survival incidences and albumin levels improved in the patients with lamivudine treatment compared with the patients without lamivudine treatment in the low level group (OR = 5.4, 95% CI = 1.1-26.0 and OR = 4.2, 95% CI = 1.3-13.2). A similar improvement was not observed in the high level group. CONCLUSION The study suggested that the circulating M65 may serve as a relatively independent prognostic parameter for liver failure. If prospectively validated in further studies, M65 measurement may also be a useful method for optimizing antiviral therapy.
Collapse
Affiliation(s)
| | - Zhi Li
- Intensive Care Unit, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | | | - Hui Ma
- Departments of Gerontology, Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Hui Li
- Department of Infectious Disease
| |
Collapse
|
20
|
Oldhafer F, Bock M, Falk CS, Vondran FWR. Immunological aspects of liver cell transplantation. World J Transplant 2016; 6:42-53. [PMID: 27011904 PMCID: PMC4801804 DOI: 10.5500/wjt.v6.i1.42] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 10/21/2015] [Accepted: 12/08/2015] [Indexed: 02/05/2023] Open
Abstract
Within the field of regenerative medicine, the liver is of major interest for adoption of regenerative strategies due to its well-known and unique regenerative capacity. Whereas therapeutic strategies such as liver resection and orthotopic liver transplantation (OLT) can be considered standards of care for the treatment of a variety of liver diseases, the concept of liver cell transplantation (LCTx) still awaits clinical breakthrough. Success of LCTx is hampered by insufficient engraftment/long-term acceptance of cellular allografts mainly due to rejection of transplanted cells. This is in contrast to the results achieved for OLT where long-term graft survival is observed on a regular basis and, hence, the liver has been deemed an immune-privileged organ. Immune responses induced by isolated hepatocytes apparently differ considerably from those observed following transplantation of solid organs and, thus, LCTx requires refined immunological strategies to improve its clinical outcome. In addition, clinical usage of LCTx but also related basic research efforts are hindered by the limited availability of high quality liver cells, strongly emphasizing the need for alternative cell sources. This review focuses on the various immunological aspects of LCTx summarizing data available not only for hepatocyte transplantation but also for transplantation of non-parenchymal liver cells and liver stem cells.
Collapse
|
21
|
Merlin S, Bhargava KK, Ranaldo G, Zanolini D, Palestro CJ, Santambrogio L, Prat M, Follenzi A, Gupta S. Kupffer Cell Transplantation in Mice for Elucidating Monocyte/Macrophage Biology and for Potential in Cell or Gene Therapy. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:539-51. [PMID: 26773351 DOI: 10.1016/j.ajpath.2015.11.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 10/16/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
Kupffer cells (KC) play major roles in immunity and tissue injury or repair. Because recapitulation of KC biology and function within liver will allow superior insights into their functional repertoire, we studied the efficacy of the cell transplantation approach for this purpose. Mouse KC were isolated from donor livers, characterized, and transplanted into syngeneic recipients. To promote cell engraftment through impairments in native KC, recipients were preconditioned with gadolinium chloride. The targeting, fate, and functionality of transplanted cells were evaluated. The findings indicated that transplanted KC engrafted and survived in recipient livers throughout the study period of 3 months. Transplanted KC expressed macrophage functions, including phagocytosis and cytokine expression, with or without genetic modifications using lentiviral vectors. This permitted studies of whether transplanted KC could affect outcomes in the context of acetaminophen hepatotoxicity or hepatic ischemia-reperfusion injury. Transplanted KC exerted beneficial effects in these injury settings. The benefits resulted from cytoprotective factors including vascular endothelial growth factor. In conclusion, transplanted adult KC were successfully targeted and engrafted in the liver with retention of innate immune and tissue repair functions over the long term. This will provide excellent opportunities to address critical aspects in the biogenesis, fate, and function of KC within their native liver microenvironment and to develop the cell and gene therapy potential of KC transplantation.
Collapse
Affiliation(s)
- Simone Merlin
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Kuldeep K Bhargava
- Division of Nuclear Medicine and Molecular Imaging, North Shore - Long Island Jewish Health System, New Hyde Park, New York
| | - Gabriella Ranaldo
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Diego Zanolini
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Christopher J Palestro
- Division of Nuclear Medicine and Molecular Imaging, North Shore - Long Island Jewish Health System, New Hyde Park, New York
| | - Laura Santambrogio
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Maria Prat
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Antonia Follenzi
- Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy; Department of Pathology, Albert Einstein College of Medicine, Bronx, New York.
| | - Sanjeev Gupta
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York; Department of Medicine, Marion Bessin Liver Research Center, Cancer Research Center, Diabetes Center, Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, and Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
22
|
Aronson SJ, Beuers U, Bosma PJ. Progress and challenges in gene therapy for Crigler–Najjar syndrome. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1100991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
23
|
Wangensteen KJ, Zhang S, Greenbaum LE, Kaestner KH. A genetic screen reveals Foxa3 and TNFR1 as key regulators of liver repopulation. Genes Dev 2015; 29:904-9. [PMID: 25934503 PMCID: PMC4421979 DOI: 10.1101/gad.258855.115] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Wangensteen et al. employed a parallel screen to test the impact of 43 selected genes on liver repopulation in the Fah−/− mouse model of hereditary tyrosinemia. The transcription factor Foxa3 was a strong promoter of liver regeneration, while tumor necrosis factor receptor 1 (TNFR1) was the most significant suppressor of repopulation among all of the genes tested. The fundamental question of which genes are most important in controlling liver regeneration remains unanswered. We employed a parallel screen to test the impact of 43 selected genes on liver repopulation in the Fah−/− mouse model of hereditary tyrosinemia. We discovered that the transcription factor Foxa3 was a strong promoter of liver regeneration, while tumor necrosis factor receptor 1 (TNFR1) was the most significant suppressor of repopulation among all of the genes tested. Our approach enabled the identification of these factors as important regulators of liver repopulation and potential drug targets for the promotion of liver repopulation.
Collapse
Affiliation(s)
- Kirk J Wangensteen
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; Center for Molecular Studies in Digestive and Liver Diseases, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Sophia Zhang
- Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Linda E Greenbaum
- Janssen Research and Development, Spring House, Pennsylvania 19477, USA
| | - Klaus H Kaestner
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA; Center for Molecular Studies in Digestive and Liver Diseases, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
24
|
Forbes SJ, Gupta S, Dhawan A. Cell therapy for liver disease: From liver transplantation to cell factory. J Hepatol 2015; 62:S157-69. [PMID: 25920085 DOI: 10.1016/j.jhep.2015.02.040] [Citation(s) in RCA: 218] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/20/2015] [Accepted: 02/27/2015] [Indexed: 02/08/2023]
Abstract
Work over several decades has laid solid foundations for the advancement of liver cell therapy. To date liver cell therapy in people has taken the form of hepatocyte transplantation for metabolic disorders with a hepatic basis, and for acute or chronic liver failure. Although clinical trials using various types of autologous cells have been implemented to promote liver regeneration or reduce liver fibrosis, clear evidence of therapeutic benefits have so far been lacking. Cell types that have shown efficacy in preclinical models include hepatocytes, liver sinusoidal endothelial cells, mesenchymal stem cells, endothelial progenitor cells, and macrophages. However, positive results in animal models have not always translated through to successful clinical therapies and more realistic preclinical models need to be developed. Studies defining the optimal repopulation by transplanted cells, including routes of cell transplantation, superior engraftment and proliferation of transplanted cells, as well as optimal immunosuppression regimens are required. Tissue engineering approaches to transplant cells in extrahepatic locations have also been proposed. The derivation of hepatocytes from pluripotent or reprogrammed cells raises hope that donor organ and cell shortages could be overcome in the future. Critical hurdles to be overcome include the production of hepatocytes from pluripotent cells with equal functional capacity to primary hepatocytes and long-term phenotypic stability in vivo.
Collapse
Affiliation(s)
- Stuart J Forbes
- MRC Centre for Regenerative Medicine, Scottish Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh EH16 4UU, United Kingdom.
| | - Sanjeev Gupta
- Departments of Medicine and Pathology, Albert Einstein College of Medicine, Jack and Pearl Resnick Campus, 1300 Morris Park Avenue, Ullmann Building, Room 625, Bronx, NY 10461, United States
| | - Anil Dhawan
- Paediatric Liver GI and Nutrition Center and NIHR/Wellcome Cell Therapy Unit, King's College Hospital at King's College, London SE59RS, United Kingdom
| |
Collapse
|