1
|
Brown GE, Bodke VV, Ware BR, Khetani SR. Liver portal fibroblasts induce the functions of primary human hepatocytes in vitro. Commun Biol 2025; 8:721. [PMID: 40346200 PMCID: PMC12064700 DOI: 10.1038/s42003-025-08135-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
In vitro human liver models are critical to mitigate species-specific differences observed for toxicology, disease modeling, and regenerative medicine. Interactions with mesenchyme (i.e., fibroblasts) can promote phenotypic functions of primary human hepatocytes (PHHs) in culture; however, using liver-derived fibroblasts remains elusive. Portal fibroblasts (PFs) around the portal triad influence bile duct formation during development, but their role in regulating homeostatic hepatic functions remains unknown. Here, we show that human liver PFs induce long-term phenotypic functions in PHHs at higher levels than activated hepatic stellate cells across 2-dimensional and 3-dimensional culture formats. While PF-conditioned media induces some hepatic functions, partly via insulin-like growth factor binding protein-5 signaling, direct contact is necessary to induce optimal functional levels. Inhibiting Notch signaling reduces progenitor-like characteristics of PHHs and further enhances functionality. Overall, this work demonstrates a unique role for PFs in modulating hepatic functions and provides all-human and all-liver coculture strategies for downstream applications.
Collapse
Affiliation(s)
- Grace E Brown
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Vedant V Bodke
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Brenton R Ware
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Salman R Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Choi MA, Rose S, Langouët S. Per- and polyfluoroalkyl substances as potentiators of hepatotoxicity in an exposome framework: Current challenges of environmental toxicology. Toxicology 2025; 515:154167. [PMID: 40300710 DOI: 10.1016/j.tox.2025.154167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/17/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Chronic liver diseases, including metabolic dysfunction-associated steatosic liver disease (MASLD) and hepatocellular carcinoma (HCC), are on the rise, potentially due to daily exposure to complex mixtures of chemical compounds forming part of the exposome. Understanding the mechanisms involved in hepatotoxicity of these mixtures is essential to identify common molecular targets that may highlight potential interactions at the molecular level. We illustrated this issue with two families of environmental contaminants, per- and polyfluoroalkyl substances (PFAS) and heterocyclic aromatic amines (HAAs), both of which could be involved in the progression of chronic liver diseases, and whose toxicity may be potentiated by interactions at the level of xenobiotic metabolism. In the study of exposome effects on chronic liver disease, New Approach Methodologies (NAMs) including omics analyses and data from various in vitro, in vivo and in silico approaches, are crucial for improving predictivity of toxicological studies in humans while reducing animal experimentation. Additionally, the development of complex in vitro human liver cell models, such as organoids, is essential to avoid interspecies differences that minimize the risk for humans. All together, these approaches will contribute to construct Adverse Outcome Pathways (AOPs) and could be applied not only to PFAS mixtures but also to other chemical families, providing valuable insights into mixture hepatotoxicity prediction in the study of the exposome. A better understanding of toxicological mechanisms will clarify the role of environmental contaminant mixtures in the development of MASLD and HCC, supporting risk assessment for better treatment, monitoring and prevention strategies.
Collapse
Affiliation(s)
- Minna A Choi
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France
| | - Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes 35000, France.
| |
Collapse
|
3
|
Hori A, Hirata M, Nonogaki R, Utoh R, Yamada M. Designing Porosity-Tailored Hydrogel Sponges with Controlled Cell Positioning Using Dispersible, Autofragmented Sacrificial Microfibers. ACS OMEGA 2025; 10:11900-11910. [PMID: 40191313 PMCID: PMC11966255 DOI: 10.1021/acsomega.4c08536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 04/09/2025]
Abstract
Hydrogel encapsulation is a rational approach that facilitates three-dimensional inoculation, arrangement, and culture of living mammalian cells for biomedical applications. However, strategies to form capillary-like conduits in hydrogels remain challenging due to low spatial resolution and difficulty in controlling the location of multiple cell types. Herein, we propose a highly unique process of constructing hydrogel sponges with tailored pore densities using finely fragmented microfibers as sacrificial porogens. A facile production process for automatically fragmented hydrogel microfibers (AF fibers) was developed through micronozzle-assisted hydrodynamic spinning and shear force application during gelation. Hydrogel sponges were prepared using photo-cross-linkable gelatin as the matrix and AF fibers dispersed in the precursor solution. We cultured liver cells in the sponges and evaluated the morphology and pore connectivity of the sponges and cellular functions. Furthermore, to create tissue models highly mimicking the cellular assembly in vivo, coculture of two types of cells was demonstrated in a position-controlled manner using cell-encapsulating AF fibers. The proposed approach of rationally designing hydrogel sponges is highly versatile in 3D cell culture for cell-based drug evaluation and regenerative medicine because of the simplicity of preparation and its impact on cellular functions.
Collapse
Affiliation(s)
- Aruto Hori
- Department of Applied Chemistry
and Biotechnology, Graduate School of Science and Engineering, Chiba University, Chiba 263-8522, Japan
| | - Mizuki Hirata
- Department of Applied Chemistry
and Biotechnology, Graduate School of Science and Engineering, Chiba University, Chiba 263-8522, Japan
| | - Rina Nonogaki
- Department of Applied Chemistry
and Biotechnology, Graduate School of Science and Engineering, Chiba University, Chiba 263-8522, Japan
| | - Rie Utoh
- Department of Applied Chemistry
and Biotechnology, Graduate School of Science and Engineering, Chiba University, Chiba 263-8522, Japan
| | - Masumi Yamada
- Department of Applied Chemistry
and Biotechnology, Graduate School of Science and Engineering, Chiba University, Chiba 263-8522, Japan
| |
Collapse
|
4
|
Tang XH, Pesola G, Chen Q, Miller D, Nagy LE, McMullen MR, Schwartz RE, Tsoy S, Lim C, Chikara S, Gross SS, Trasino SE, Gudas LJ, Melis M. Ethanol causes rapid decreases in the hepatic retinoid levels shaping the early steps of alcohol-associated liver disease. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:754-770. [PMID: 40016864 PMCID: PMC12014373 DOI: 10.1111/acer.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Chronic alcohol drinking causes hepatic vitamin A (retinoids and derivatives) decreases, which correlate with the progression and severity of alcohol-associated liver disease (ALD). However, the effects of short-term ethanol (EtOH) intake on liver retinoids and ALD are still undefined. METHODS Using high-performance liquid chromatography and high-performance liquid chromatography coupled with tandem mass spectrometry (HPLC, HPLC-MS/MS), and molecular biology techniques in mice and cultured human hepatocytes, we investigated the temporal EtOH effects on retinoids and ALD. RESULTS In female and male mice, acute EtOH intake caused hepatic retinol (ROL) and retinyl palmitate (RP) decreases within hours, whereas it did not significantly change the retinoic acid (RA) levels, and those of the RA catabolism metabolite, 4-oxo-RA. After EtOH withdrawal, the liver recovered the ROL and RP levels within 48 h, whereas RA and 4-oxo-RA levels remained almost undetectable by this time point. Compared with control diet-fed mice, hepatic ROL and RP levels remained decreased in the 10-day and 3-week-long EtOH treatments, while retinyl oleate and linoleate increased. Interestingly, some of the RA signaling receptors, Rarβ, along with Cyp26a1, revealed dramatic transcript increases during the 10-day-long experiments that attenuated over time (up to 8 weeks), reflecting impaired RA signaling. Our work also showed that primary human hepatocytes serve as a model to better define the role of EtOH in retinoid biology. CONCLUSIONS This work reveals that acute and short-term exposures to EtOH disrupt retinoid homeostasis, identifying key events in the early pathogenesis of ALD.
Collapse
Affiliation(s)
- Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Glen Pesola
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dawson Miller
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Laura E. Nagy
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | - Megan R. McMullen
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | - Robert E. Schwartz
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Sergey Tsoy
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Christine Lim
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Shireen Chikara
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Steven S. Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Steven E. Trasino
- Department of Nutrition and Public Health, Hunter College, City University of New York, New York, NY, USA
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Marta Melis
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
5
|
Adeyemi O, Osuntoki A, Magbagbeola O, Abdel Hamid MM, Elaagip A, Mueller AK, Ibrahim M. Evidence for infectious merozoites of Plasmodium falciparum from natural isolates of cultured hepatoma cells infected with sporozoites. PLoS One 2025; 20:e0319901. [PMID: 40106488 PMCID: PMC11922282 DOI: 10.1371/journal.pone.0319901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Previous cell culture systems using various human hepatoma cell lines established that the intra-hepatic stages of Plasmodium falciparum could be studied ex vivo. However, only one of these culture systems yielded infective merozoites that subsequently completed the parasite's life cycle outside a human host. We hypothesized that a major limitation is the use of laboratory-adapted P. falciparum blood stages for sporozoites generation. Plasmodium falciparum sporozoites were generated by membrane-feeding of gametocyte-infected blood samples from hospital patients to Anopheles arabiensis. Subsequently, cultured HepG2 cells were infected with the sporozoites. From 6 days post-sporozoite inoculation, liver merozoites could be harvested from the cell supernatants. When co-cultured with O + erythrocytes, these merozoites established a blood infection and yielded erythrocytic stage parasites that re-infected erythrocytes. To confirm that the erythrocytic parasites generated were P. falciparum, RNA expressed by the erythrocytic parasites was isolated and used as control in microarray analysis against RNA expressed by irradiated erythrocytic parasites; subsequently, P. falciparum genes were identified. The cultured HepG2 cells permitted the full intra-hepatic maturation of P. falciparum parasites from natural isolates. Infective merozoites were yielded which gave rise to the erythrocytic stage P. falciparum post-infection into O + erythrocytes. The full intra-hepatic maturation of the naturally isolated P. falciparum parasites in a HepG2 cell culture system is possible. This finding has important implications for malaria research and vaccine development.
Collapse
Affiliation(s)
- Olumide Adeyemi
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Akinniyi Osuntoki
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Olubunmi Magbagbeola
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Muzamil Mahdi Abdel Hamid
- Department of Parasitology and Medical Entomology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | - Arwa Elaagip
- Department of Parasitology and Medical Entomology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| | - Ann-Kristin Mueller
- Heidelberg University Hospital, Department of Infectious Diseases, Heidelberg, Germany
| | - Muntaser Ibrahim
- Department of Molecular Biology, Institute of Endemic Diseases, University of Khartoum, Khartoum, Sudan
| |
Collapse
|
6
|
Hagenbuch B, Stieger B, Locher KP. Organic anion transporting polypeptides: Pharmacology, toxicology, structure, and transport mechanisms. Pharmacol Rev 2025; 77:100023. [PMID: 40148036 DOI: 10.1016/j.pharmr.2024.100023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Organic anion transporting polypeptides (OATPs) are membrane proteins that mediate the uptake of a wide range of substrates across the plasma membrane of various cells and tissues. They are classified into 6 subfamilies, OATP1 through OATP6. Humans contain 12 OATPs encoded by 11 solute carrier of organic anion transporting polypeptide (SLCO) genes: OATP1A2, OATP1B1, OATP1B3, the splice variant OATP1B3-1B7, OATP1C1, OATP2A1, OATP2B1, OATP3A1, OATP4A1, OATP4C1, OATP5A1, and OATP6A1. Most of these proteins are expressed in epithelial cells, where they mediate the uptake of structurally unrelated organic anions, cations, and even neutral compounds into the cytoplasm. The best-characterized members are OATP1B1 and OATP1B3, which have an important role in drug metabolism by mediating drug uptake into the liver and are involved in drug-drug interactions. In this review, we aimed to (1) provide a historical perspective on the identification of OATPs and their nomenclature and discuss their phylogenic relationships and molecular characteristics; (2) review the current knowledge of the broad substrate specificity and their role in drug disposition and drug-drug interactions, with a special emphasis on human hepatic OATPs; (3) summarize the different experimental systems that are used to study the function of OATPs and discuss their advantages and disadvantages; (4) review the available experimental 3-dimensional structures and examine how they can help elucidate the transport mechanisms of OATPs; and (5) finally, summarize the current knowledge of the regulation of OATP expression, discuss clinically important single-nucleotide polymorphisms, and outline challenges of physiologically based pharmacokinetic modeling and in vitro to in vivo extrapolation. SIGNIFICANCE STATEMENT: Organic anion transporting polypeptides (OATPs) are a family of 12 uptake transporters in the solute carrier superfamily. Several members, particularly the liver-expressed OATP1B1 and OATP1B3, are important drug transporters. They mediate the uptake of several endobiotics and xenobiotics, including statins and numerous other drugs, into hepatocytes, and their inhibition by other drugs or reduced expression due to single-nucleotide polymorphisms can lead to adverse drug effects. Their recently solved 3-dimensional structures should help to elucidate their transport mechanisms and broad substrate specificities.
Collapse
Affiliation(s)
- Bruno Hagenbuch
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas.
| | - Bruno Stieger
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| | - Kaspar P Locher
- Institute of Molecular Biology and Biophysics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
7
|
Josvai M, Polyak E, Kalluri M, Robertson S, Crone WC, Suzuki M. An engineered in vitro model of the human myotendinous junction. Acta Biomater 2024; 180:279-294. [PMID: 38604466 PMCID: PMC11088524 DOI: 10.1016/j.actbio.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 03/12/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
The myotendinous junction (MTJ) is a vulnerable region at the interface of skeletal muscle and tendon that forms an integrated mechanical unit. This study presents a technique for the spatially restrictive co-culture of human embryonic stem cell (hESC)-derived skeletal myocytes and primary tenocytes for two-dimensional modeling of the MTJ. Micropatterned lanes of extracellular matrix and a 2-well culture chamber define the initial regions of occupation. On day 1, both lines occupy less than 20 % of the initially vacant interstitial zone, referred to henceforth as the junction. Myocyte-tenocyte interdigitations are observed by day 7. Immunocytochemistry reveals enhanced organization and alignment of patterned myocyte and tenocyte features, as well as differential expression of multiple MTJ markers. On day 24, electrically stimulated junction myocytes demonstrate negative contractile strains, while positive tensile strains are exhibited by mechanically passive tenocytes at the junction. Unpatterned tenocytes distal to the junction experience significantly decreased strains in comparison to cells at the interface. Unpatterned myocytes have impaired organization and uncoordinated contractile behavior. These findings suggest that this platform is capable of inducing myocyte-tenocyte junction formation and mechanical coupling similar to the native MTJ, showing transduction of force across the cell-cell interface. STATEMENT OF SIGNIFICANCE: The myotendinous junction (MTJ) is an integrated structure that transduces force across the muscle-tendon boundary, making the region vulnerable to strain injury. Despite the clinical relevance, previous in vitro models of the MTJ lack the structure and mechanical accuracy of the native tissue and have difficulty transmitting force across the cell-cell interface. This study demonstrates an in vitro model of the MTJ, using spatially restrictive cues to inform human myocyte-tenocyte interactions and architecture. The model expressed MTJ markers and developed anisotropic myocyte-tenocyte integrations that resemble the native tissue and allow for force transduction from contracting myocytes to passive tenocyte regions. As such, this study presents a system capable of investigating development, injury, and pathology in the human MTJ.
Collapse
Affiliation(s)
- Mitchell Josvai
- Department of Biomedical Engineering, University of Wisconsin-Madison, Engineering Centers Building, 2126, 1550 Engineering Dr, Madison WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, 330 N Orchard St, Madison, WI 53715, USA
| | - Erzsebet Polyak
- Department of Comparative Biosciences, University of Wisconsin-Madison, Veterinary Medicine Bldg, 2015 Linden Dr, Madison, WI 53706, USA
| | - Meghana Kalluri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Engineering Centers Building, 2126, 1550 Engineering Dr, Madison WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, 330 N Orchard St, Madison, WI 53715, USA
| | - Samantha Robertson
- Department of Comparative Biosciences, University of Wisconsin-Madison, Veterinary Medicine Bldg, 2015 Linden Dr, Madison, WI 53706, USA
| | - Wendy C Crone
- Department of Biomedical Engineering, University of Wisconsin-Madison, Engineering Centers Building, 2126, 1550 Engineering Dr, Madison WI 53706, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, 330 N Orchard St, Madison, WI 53715, USA; The Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA; Department of Nuclear Engineering and Engineering Physics, University of Wisconsin-Madison, 1500 Engineering Drive, Madison, WI 53706, USA; Department of Mechanical Engineering, University of Wisconsin-Madison, 1513 University Avenue, Madison, WI 53706, USA.
| | - Masatoshi Suzuki
- Department of Biomedical Engineering, University of Wisconsin-Madison, Engineering Centers Building, 2126, 1550 Engineering Dr, Madison WI 53706, USA; Department of Comparative Biosciences, University of Wisconsin-Madison, Veterinary Medicine Bldg, 2015 Linden Dr, Madison, WI 53706, USA; The Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA.
| |
Collapse
|
8
|
Carpentier A. Cell Culture Models for Hepatitis B and D Viruses Infection: Old Challenges, New Developments and Future Strategies. Viruses 2024; 16:716. [PMID: 38793598 PMCID: PMC11125795 DOI: 10.3390/v16050716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Chronic Hepatitis B and D Virus (HBV and HDV) co-infection is responsible for the most severe form of viral Hepatitis, the Hepatitis Delta. Despite an efficient vaccine against HBV, the HBV/HDV infection remains a global health burden. Notably, no efficient curative treatment exists against any of these viruses. While physiologically distinct, HBV and HDV life cycles are closely linked. HDV is a deficient virus that relies on HBV to fulfil is viral cycle. As a result, the cellular response to HDV also influences HBV replication. In vitro studying of HBV and HDV infection and co-infection rely on various cell culture models that differ greatly in terms of biological relevance and amenability to classical virology experiments. Here, we review the various cell culture models available to scientists to decipher HBV and HDV virology and host-pathogen interactions. We discuss their relevance and how they may help address the remaining questions, with one objective in mind: the development of new therapeutic approaches allowing viral clearance in patients.
Collapse
Affiliation(s)
- Arnaud Carpentier
- Institute for Experimental Virology, TWINCORE Centre for Experimental and Clinical Infection Research, a Joint Venture between Hannover Medical School (MHH) and Helmholtz Centre for Infection Research (HZI), Feodor-Lynen-Strasse 7, 30625 Hannover, Germany;
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| |
Collapse
|
9
|
March S, Nerurkar N, Jain A, Andrus L, Kim D, Whittaker CA, Tan EK, Thiberge S, Fleming HE, Mancio-Silva L, Rice CM, Bhatia SN. Autonomous circadian rhythms in the human hepatocyte regulate hepatic drug metabolism and inflammatory responses. SCIENCE ADVANCES 2024; 10:eadm9281. [PMID: 38657074 PMCID: PMC11042741 DOI: 10.1126/sciadv.adm9281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024]
Abstract
Critical aspects of physiology and cell function exhibit self-sustained ~24-hour variations termed circadian rhythms. In the liver, circadian rhythms play fundamental roles in maintaining organ homeostasis. Here, we established and characterized an in vitro liver experimental system in which primary human hepatocytes display self-sustained oscillations. By generating gene expression profiles of these hepatocytes over time, we demonstrated that their transcriptional state is dynamic across 24 hours and identified a set of cycling genes with functions related to inflammation, drug metabolism, and energy homeostasis. We designed and tested a treatment protocol to minimize atorvastatin- and acetaminophen-induced hepatotoxicity. Last, we documented circadian-dependent induction of pro-inflammatory cytokines when triggered by LPS, IFN-β, or Plasmodium infection in human hepatocytes. Collectively, our findings emphasize that the phase of the circadian cycle has a robust impact on the efficacy and toxicity of drugs, and we provide a test bed to study the timing and magnitude of inflammatory responses over the course of infection in human liver.
Collapse
Affiliation(s)
- Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Niketa Nerurkar
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Anisha Jain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Linda Andrus
- Laboratory of Virology and Infectious Disease, The Rockefeller University, NY, New York, USA
| | - Daniel Kim
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Charles A. Whittaker
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Edward K.W. Tan
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Sabine Thiberge
- Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, Centre de Production et d’Infection des Anophèles, 75015 Paris, France
| | - Heather E. Fleming
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Liliana Mancio-Silva
- Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, NY, New York, USA
| | - Sangeeta N. Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Wyss Institute at Harvard University, 201 Brookline Ave, Boston, MA 02215, USA
| |
Collapse
|
10
|
Huang Q, Yang T, Song Y, Sun W, Xu J, Cheng Y, Yin R, Zhu L, Zhang M, Ma L, Li H, Zhang H. A three-dimensional (3D) liver-kidney on a chip with a biomimicking circulating system for drug safety evaluation. LAB ON A CHIP 2024; 24:1715-1726. [PMID: 38328873 DOI: 10.1039/d3lc00980g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The liver and kidney are the major detoxifying organs in the human body and play an important role in pharmacokinetics. Drug-induced hepatotoxicity and nephrotoxicity can cause irreversible damage to the liver and kidney and are a major cause of drug failure in later stages. Both animal models and conventional cell culture have a number of limitations, such as animal ethics and gene mismatching and there is an urgent need to develop a new drug toxicity evaluation approach. In this paper, a 3D liver-kidney on a chip with a biomimicking circulating system (LKOCBCS) was constructed to obtain kidney and liver models in vitro for drug safety evaluation. LKOCBCS, which has a parallel circulating system mimicking biological circulation, consists of 3D biomimetic tissue of liver lobules similar to that of the human liver constructed by 3D bioprinting and renal proximal tubule barriers fabricated by ultrafast laser assisted etching. The proposed LKOCBCS facilitates the communication between the liver and the kidney, including the exchange of nutrients, compounds, and metabolites. The results revealed that the glucose concentration and cell metabolism stabilized after 7 days. A dynamically repeated low-dose administration of cyclosporine A (CsA) was fed to the system, and hepatotoxicity and nephrotoxicity were observed on day 3 according to the changes in toxicity markers. The high levels of drug induced biomarkers expressed in LKOCBCS indicate that this system is more sensitive than the monoculture liver chip and it is highly potential in replacing animal models for effective drug toxicity screening.
Collapse
Affiliation(s)
- Qihong Huang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Tianhao Yang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Yunpeng Song
- School of Physics and Electronic Science, East China Normal University, Shanghai 200062, China.
| | - Wenxuan Sun
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Jian Xu
- School of Physics and Electronic Science, East China Normal University, Shanghai 200062, China.
| | - Ya Cheng
- School of Physics and Electronic Science, East China Normal University, Shanghai 200062, China.
| | - Ruixue Yin
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Lili Zhu
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Mengting Zhang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Lei Ma
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Honglin Li
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Hongbo Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
11
|
Wang W, Chen Y, Chen Y, Liu E, Li J, An N, Xu J, Gu S, Dang X, Yi J, An Q, Hu X, Yin W. Supernatant of platelet- Klebsiella pneumoniae coculture induces apoptosis-like death in Klebsiella pneumoniae. Microbiol Spectr 2024; 12:e0127923. [PMID: 38289116 PMCID: PMC10913751 DOI: 10.1128/spectrum.01279-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 12/13/2023] [Indexed: 03/06/2024] Open
Abstract
Multidrug-resistant Klebsiella pneumoniae strains, especially carbapenem-resistant K. pneumoniae, have become a rapidly emerging crisis worldwide, greatly limiting current therapeutic options and posing new challenges to infection management. Therefore, it is imperative to develop novel and effective biological agents for the treatment of multidrug-resistant K. pneumoniae infections. Platelets play an important role in the development of inflammation and immune responses. The main component responsible for platelet antibacterial activity lies in the supernatant stimulated by gram-positive bacteria. However, little research has been conducted on the interaction of gram-negative bacteria with platelets. Therefore, we aimed to explore the bacteriostatic effect of the supernatant derived from platelet-K. pneumoniae coculture and the mechanism underlying this effect to further assess the potential of platelet-bacterial coculture supernatant. We conducted this study on the gram-negative bacteria K. pneumoniae and CRKP and detected turbidity changes in K. pneumoniae and CRKP cultures when grown with platelet-K. pneumoniae coculture supernatant added to the culture medium. We found that platelet-K. pneumoniae coculture supernatant significantly inhibited the growth of K. pneumoniae and CRKP in vitro. Furthermore, transfusion of platelet-K. pneumoniae coculture supernatant alleviated the symptoms of K. pneumoniae and CRKP infection in a murine model. Additionally, we observed apoptosis-like changes, such as phosphatidylserine exposure, chromosome condensation, DNA fragmentation, and overproduction of reactive oxygen species in K. pneumoniae following treatment with the supernatant. Our study demonstrates that the platelet-K. pneumoniae coculture supernatant can inhibit K. pneumoniae growth by inducing an apoptosis-like death, which is important for the antibacterial strategies development in the future.IMPORTANCEWith the widespread use of antibiotics, bacterial resistance is increasing, and a variety of multi-drug resistant Gram-negative bacteria have emerged, which brings great challenges to the treatment of infections caused by Gram-negative bacteria. Therefore, finding new strategies to inhibit Gram-negative bacteria and even multi-drug- resistant Gram-negative bacteria is crucial for treating infections caused by Gram-negative bacteria, improving the abuse of antibiotics, and maintaining the balance between bacteria and antibiotics. K. pneumoniae is a common clinical pathogen, and drug-resistant CRKP is increasingly difficult to cure, which brings great clinical challenges. In this study, we found that the platelet-K. pneumoniae coculture supernatant can inhibit K. pneumoniae growth by inducing an apoptosis-like death. This finding has inspired the development of future antimicrobial strategies, which are expected to improve the clinical treatment of Gram-negative bacteria and control the development of multidrug-resistant strains.
Collapse
Affiliation(s)
- Wenting Wang
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
- Faculty of Life Science College, Southwest Forestry University, Kunming, Yunnan, China
| | - Yaozhen Chen
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yutong Chen
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Erxiong Liu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jing Li
- Faculty of Life Science College, Southwest Forestry University, Kunming, Yunnan, China
| | - Ning An
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jinmei Xu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Shunli Gu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xuan Dang
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Jing Yi
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Qunxing An
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Xingbin Hu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Wen Yin
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
12
|
Wang E, Andrade MJ, Smith Q. Vascularized liver-on-a-chip model to investigate nicotine-induced dysfunction. BIOMICROFLUIDICS 2023; 17:064108. [PMID: 38155919 PMCID: PMC10754629 DOI: 10.1063/5.0172677] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023]
Abstract
The development of physiologically relevant in vitro systems for simulating disease onset and progression and predicting drug metabolism holds tremendous value in reducing drug discovery time and cost. However, many of these platforms lack accuracy in replicating the tissue architecture and multicellular interactions. By leveraging three-dimensional cell culture, biomimetic soft hydrogels, and engineered stimuli, in vitro models have continued to progress. Nonetheless, the incorporation of the microvasculature has been met with many challenges, specifically with the addition of parenchymal cell types. Here, a systematic approach to investigating the initial seeding density of endothelial cells and its effects on interconnected networks was taken and combined with hepatic spheroids to form a liver-on-a-chip model. Leveraging this system, nicotine's effects on microvasculature and hepatic function were investigated. The findings indicated that nicotine led to interrupted adherens junctions, decreased guanosine triphosphate cyclohydrolase 1 expression, impaired angiogenesis, and lowered barrier function, all key factors in endothelial dysfunction. With the combination of the optimized microvascular networks, a vascularized liver-on-a-chip was formed, providing functional xenobiotic metabolism and synthesis of both albumin and urea. This system provides insight into potential hepatotoxicity caused by various drugs and allows for assessing vascular dysfunction in a high throughput manner.
Collapse
Affiliation(s)
- Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, USA
| | - Melisa J. Andrade
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, USA
| | | |
Collapse
|
13
|
Nicholas J, Kolli SK, Subramani PA, De SL, Ogbondah MM, Barnes SJ, Ntumngia FB, Adams JH. Comparative analyses of functional antibody-mediated inhibition with anti-circumsporozoite monoclonal antibodies against transgenic Plasmodium berghei. Malar J 2023; 22:335. [PMID: 37936181 PMCID: PMC10629016 DOI: 10.1186/s12936-023-04765-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Acquired functional inhibitory antibodies are one of several humoral immune mechanisms used to neutralize foreign pathogens. In vitro bioassays are useful tools for quantifying antibody-mediated inhibition and evaluating anti-parasite immune antibodies. However, a gap remains in understanding of how antibody-mediated inhibition in vitro translates to inhibition in vivo. In this study, two well-characterized transgenic Plasmodium berghei parasite lines, PbmCh-luc and Pb-PfCSP(r), and murine monoclonal antibodies (mAbs) specific to P. berghei and Plasmodium falciparum circumsporozoite protein (CSP), 3D11 and 2A10, respectively, were used to evaluate antibody-mediated inhibition of parasite development in both in vitro and in vivo functional assays. METHODS IC50 values of mAbs were determined using an established inhibition of liver-stage development assay (ILSDA). For the in vivo inhibition assay, mice were passively immunized by transfer of the mAbs and subsequently challenged with 5.0 × 103 sporozoites via tail vein injection. The infection burden in both assays was quantified by luminescence and qRT-PCR of P. berghei 18S rRNA normalized to host GAPDH. RESULTS The IC50 values quantified by relative luminescence of mAbs 3D11 and 2A10 were 0.396 µg/ml and 0.093 µg/ml, respectively, against transgenic lines in vitro. Using the highest (> 90%) inhibitory antibody concentrations in a passive transfer, an IC50 of 233.8 µg/ml and 181.5 µg/ml for mAbs 3D11 and 2A10, respectively, was observed in vivo. At 25 µg (250 µg/ml), the 2A10 antibody significantly inhibited liver burden in mice compared to control. Additionally, qRT-PCR of P. berghei 18S rRNA served as a secondary validation of liver burden quantification. CONCLUSIONS Results from both experimental models, ILSDA and in vivo challenge, demonstrated that increased concentrations of the homologous anti-CSP repeat mAbs increased parasite inhibition. However, differences in antibody IC50 values between parasite lines did not allow a direct correlation between the inhibition of sporozoite invasion in vitro by ILSDA and the inhibition of mouse liver stage burden. Further studies are needed to establish the conditions for confident predictions for the in vitro ILSDA to be a predictor of in vivo outcomes using this model system.
Collapse
Affiliation(s)
- Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Surendra Kumar Kolli
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Pradeep Annamalai Subramani
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Sai Lata De
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Infectious Disease & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Madison M Ogbondah
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Samantha J Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - John H Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
14
|
Joshi A, Singh N. Generation of Patterned Cocultures in 2D and 3D: State of the Art. ACS OMEGA 2023; 8:34249-34261. [PMID: 37780002 PMCID: PMC10536108 DOI: 10.1021/acsomega.3c02713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023]
Abstract
Cells inside the body are embedded into a highly structured microenvironment that consists of cells that lie in direct or close contact with other cell types that regulate the overall tissue function. Therefore, coculture models are versatile tools that can generate tissue engineering constructs with improved mimicking of in vivo conditions. While there are many reviews that have focused on pattering a single cell type, very few reviews have been focused on techniques for coculturing multiple cell types on a single substrate with precise control. In this regard, this Review covers various technologies that have been utilized for the development of these patterned coculture models while mentioning the limitations associated with each of them. Further, the application of these models to various tissue engineering applications has been discussed.
Collapse
Affiliation(s)
- Akshay Joshi
- Centre
for Biomedical Engineering, Indian Institute
of Technology Delhi, Hauz Khas, New Delhi, Delhi 110016, India
| | - Neetu Singh
- Centre
for Biomedical Engineering, Indian Institute
of Technology Delhi, Hauz Khas, New Delhi, Delhi 110016, India
- Biomedical
Engineering Unit, All India Institute of
Medical Sciences, Ansari
Nagar, New Delhi, Delhi 110029, India
| |
Collapse
|
15
|
Wang J, Sun S, Deng H. Chemical reprogramming for cell fate manipulation: Methods, applications, and perspectives. Cell Stem Cell 2023; 30:1130-1147. [PMID: 37625410 DOI: 10.1016/j.stem.2023.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023]
Abstract
Chemical reprogramming offers an unprecedented opportunity to control somatic cell fate and generate desired cell types including pluripotent stem cells for applications in biomedicine in a precise, flexible, and controllable manner. Recent success in the chemical reprogramming of human somatic cells by activating a regeneration-like program provides an alternative way of producing stem cells for clinical translation. Likewise, chemical manipulation enables the capture of multiple (stem) cell states, ranging from totipotency to the stabilization of somatic fates in vitro. Here, we review progress in using chemical approaches for cell fate manipulation in addition to future opportunities in this promising field.
Collapse
Affiliation(s)
- Jinlin Wang
- MOE Engineering Research Center of Regenerative Medicine, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China; Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing, China
| | - Shicheng Sun
- Changping Laboratory, 28 Life Science Park Road, Beijing, China; Murdoch Children's Research Institute, Royal Children's Hospital, Flemington Road, Parkville, VIC, Australia.
| | - Hongkui Deng
- MOE Engineering Research Center of Regenerative Medicine, School of Basic Medical Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center and the MOE Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China; Changping Laboratory, 28 Life Science Park Road, Beijing, China.
| |
Collapse
|
16
|
Cliffe FE, Madden C, Costello P, Devitt S, Mukkunda SR, Keshava BB, Fearnhead HO, Vitkauskaite A, Dehkordi MH, Chingwaru W, Przyjalgowski M, Rebrova N, Lyons M. Mera: A scalable high throughput automated micro-physiological system. SLAS Technol 2023; 28:230-242. [PMID: 36708805 DOI: 10.1016/j.slast.2023.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023]
Abstract
There is an urgent need for scalable Microphysiological Systems (MPS's)1 that can better predict drug efficacy and toxicity at the preclinical screening stage. Here we present Mera, an automated, modular and scalable system for culturing and assaying microtissues with interconnected fluidics, inbuilt environmental control and automated image capture. The system presented has multiple possible fluidics modes. Of these the primary mode is designed so that cells may be matured into a desired microtissue type and in the secondary mode the fluid flow can be re-orientated to create a recirculating circuit composed of inter-connected channels to allow drugging or staining. We present data demonstrating the prototype system Mera using an Acetaminophen/HepG2 liver microtissue toxicity assay with Calcein AM and Ethidium Homodimer (EtHD1) viability assays. We demonstrate the functionality of the automated image capture system. The prototype microtissue culture plate wells are laid out in a 3 × 3 or 4 × 10 grid format with viability and toxicity assays demonstrated in both formats. In this paper we set the groundwork for the Mera system as a viable option for scalable microtissue culture and assay development.
Collapse
Affiliation(s)
- Finola E Cliffe
- Hooke Bio Ltd, L4A Smithstown Industrial Estate, Shannon, Co. Clare V14 XH92, Ireland
| | - Conor Madden
- Hooke Bio Ltd, L4A Smithstown Industrial Estate, Shannon, Co. Clare V14 XH92, Ireland
| | - Patrick Costello
- Hooke Bio Ltd, L4A Smithstown Industrial Estate, Shannon, Co. Clare V14 XH92, Ireland
| | - Shane Devitt
- Hooke Bio Ltd, L4A Smithstown Industrial Estate, Shannon, Co. Clare V14 XH92, Ireland
| | - Sumir Ramesh Mukkunda
- Hooke Bio Ltd, L4A Smithstown Industrial Estate, Shannon, Co. Clare V14 XH92, Ireland
| | | | - Howard O Fearnhead
- Pharmacology and Therapeutics, Biomedical Sciences, Dangan, NUI Galway, Galway, Ireland
| | - Aiste Vitkauskaite
- Pharmacology and Therapeutics, Biomedical Sciences, Dangan, NUI Galway, Galway, Ireland
| | - Mahshid H Dehkordi
- Pharmacology and Therapeutics, Biomedical Sciences, Dangan, NUI Galway, Galway, Ireland
| | - Walter Chingwaru
- Pharmacology and Therapeutics, Biomedical Sciences, Dangan, NUI Galway, Galway, Ireland
| | - Milosz Przyjalgowski
- Centre for Advanced Photonics and Process Analysis, Munster Technological University, Cork T12 P928, Ireland
| | - Natalia Rebrova
- Centre for Advanced Photonics and Process Analysis, Munster Technological University, Cork T12 P928, Ireland
| | - Mark Lyons
- Hooke Bio Ltd, L4A Smithstown Industrial Estate, Shannon, Co. Clare V14 XH92, Ireland.
| |
Collapse
|
17
|
Carpentier N, Urbani L, Dubruel P, Van Vlierberghe S. The native liver as inspiration to create superior in vitro hepatic models. Biomater Sci 2023; 11:1091-1115. [PMID: 36594602 DOI: 10.1039/d2bm01646j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Drug induced liver injury (DILI) is one of the major reasons of drug withdrawal during the different phases of drug development. The later in the drug development a drug is discovered to be toxic, the higher the economical as well as the ethical impact will be. In vitro models for early detection of drug liver toxicity are under constant development, however to date a superior model of the liver is still lacking. Ideally, a highly reliable model should be established to maintain the different hepatic cell functionalities to the greatest extent possible, during a period of time long enough to allow for tracking of the toxicity of compounds. In the case of DILI, toxicity can appear even after months of exposure. To reach this goal, an in vitro model should be developed that mimics the in vivo liver environment, function and response to external stimuli. The different approaches for the development of liver models currently used in the field of tissue engineering will be described in this review. Combining different technologies, leading to optimal materials, cells and 3D-constructs will ultimately lead to an ideal superior model that fully recapitulates the liver.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Luca Urbani
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE5 9NT, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium.
| |
Collapse
|
18
|
Liu Z, Nan H, Chiou YS, Zhan Z, Lobie PE, Hu C. Selective Formation of Osteogenic and Vasculogenic Tissues for Cartilage Regeneration. Adv Healthc Mater 2023; 12:e2202008. [PMID: 36353894 DOI: 10.1002/adhm.202202008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/02/2022] [Indexed: 11/11/2022]
Abstract
Tissue-engineered periosteum substitutes (TEPSs) incorporating hierarchical architecture with osteoprogenitor and vascular niches are drawing much attention as a promising tool to support functional cells in defined zones and nourish the cortical bone. Current TEPSs usually lack technologies to closely observe cell performance, especially at the cell contact interface between distinct compartments containing defined biological configurations and functions. Here, an electrodeposition strategy is reported, which enables the selective formation of TEPSs with osteoprogenitor and vascular niches in a multiphasic scaffold in combination with different human cell types for cartilage regeneration in an in vivo osteochondral defect model. Human umbilical vein endothelial cells (HUVECs), dermal fibroblasts (HDFs), and bone marrow mesenchymal stem cells (hMSCs) are used to mirror both the vascular and osteogenic niches, respectively. It is observed that the intrinsic viscoelastic nature of the porous solid matrix is essential to successfully induce angiogenesis. Coculture of hMSCs with functional cells (HUVECs/HDFs) in TEPSs also effectively promoted periosteal regeneration, including osteogenic and angiogenic processes. The osteoarthritis cartilage histopathology assessment and histologic/histochemical grading system data indicate that the TEPSs containing hMSCs/HUVECs/HDFs exhibit superior potential for cartilage regeneration.
Collapse
Affiliation(s)
- Zeyang Liu
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.,Guangdong Provincial Key Laboratory of Human-Augmentation and Rehabilitation Robotics in Universities, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Haochen Nan
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.,Guangdong Provincial Key Laboratory of Human-Augmentation and Rehabilitation Robotics in Universities, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yi Shiou Chiou
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.,Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Zhen Zhan
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.,Guangdong Provincial Key Laboratory of Human-Augmentation and Rehabilitation Robotics in Universities, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Peter E Lobie
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518055, China
| | - Chengzhi Hu
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China.,Guangdong Provincial Key Laboratory of Human-Augmentation and Rehabilitation Robotics in Universities, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
19
|
Nardella F, Dobrescu I, Hassan H, Rodrigues F, Thiberge S, Mancio-Silva L, Tafit A, Jallet C, Cadet-Daniel V, Goussin S, Lorthiois A, Menon Y, Molinier N, Pechalrieu D, Long C, Sautel F, Matondo M, Duchateau M, Médard G, Witkowski B, Scherf A, Halby L, Arimondo PB. Hemisynthetic alkaloids derived from trilobine are antimalarials with sustained activity in multidrug-resistant Plasmodium falciparum. iScience 2023; 26:105940. [PMID: 36718363 PMCID: PMC9883252 DOI: 10.1016/j.isci.2023.105940] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/23/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Malaria eradication requires the development of new drugs to combat drug-resistant parasites. We identified bisbenzylisoquinoline alkaloids isolated from Cocculus hirsutus that are active against Plasmodium falciparum blood stages. Synthesis of a library of 94 hemi-synthetic derivatives allowed to identify compound 84 that kills multi-drug resistant clinical isolates in the nanomolar range (median IC50 ranging from 35 to 88 nM). Chemical optimization led to compound 125 with significantly improved preclinical properties. 125 delays the onset of parasitemia in Plasmodium berghei infected mice and inhibits P. falciparum transmission stages in vitro (culture assays), and in vivo using membrane feeding assay in the Anopheles stephensi vector. Compound 125 also impairs P. falciparum development in sporozoite-infected hepatocytes, in the low micromolar range. Finally, by chemical pull-down strategy, we characterized the parasite interactome with trilobine derivatives, identifying protein partners belonging to metabolic pathways that are not targeted by the actual antimalarial drugs or implicated in drug-resistance mechanisms.
Collapse
Affiliation(s)
- Flore Nardella
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Irina Dobrescu
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Haitham Hassan
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Fabien Rodrigues
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Sabine Thiberge
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France,Center for Production and Infection of Anopheles (CEPIA), Center for Animal Resources and Research, Institut Pasteur, 75015 Paris, France
| | - Liliana Mancio-Silva
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Ambre Tafit
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Corinne Jallet
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Véronique Cadet-Daniel
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Stéphane Goussin
- Center for Production and Infection of Anopheles (CEPIA), Center for Animal Resources and Research, Institut Pasteur, 75015 Paris, France
| | - Audrey Lorthiois
- Center for Production and Infection of Anopheles (CEPIA), Center for Animal Resources and Research, Institut Pasteur, 75015 Paris, France
| | - Yoann Menon
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - Nicolas Molinier
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - Dany Pechalrieu
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - Christophe Long
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - François Sautel
- USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France
| | - Mariette Matondo
- Proteomics Platform, Mass Spectrometry for Biology Unit, Institut Pasteur, Université de Paris-Cité, CNRS USR 2000, 28 rue du Dr Roux, 75015 Paris, France
| | - Magalie Duchateau
- Proteomics Platform, Mass Spectrometry for Biology Unit, Institut Pasteur, Université de Paris-Cité, CNRS USR 2000, 28 rue du Dr Roux, 75015 Paris, France
| | - Guillaume Médard
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, 85354 Freising, Germany
| | - Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Artur Scherf
- Biology of Host-Parasite Interaction, Department of Parasites and Insect Vectors, Institut Pasteur, Université de Paris-Cité, CNRS EMR 9195, INSERM Unit U1201, 25-28 Rue du Dr Roux, 75015 Paris, France,Corresponding author
| | - Ludovic Halby
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France
| | - Paola B. Arimondo
- Epigenetic Chemical Biology, Department of Structural Biology and Chemistry, Institut Pasteur, Université de Paris-Cité, UMR n°3523, CNRS, 28 Rue du Dr Roux, 75015 Paris, France,USR CNRS-Pierre Fabre No. 3388 ETaC, Centre de Recherche et Développement Pierre Fabre, 3 Avenue Hubert Curien, 31035 Toulouse Cedex 01, France,Corresponding author
| |
Collapse
|
20
|
Widerspick L, Steffen JF, Tappe D, Muñoz-Fontela C. Animal Model Alternatives in Filovirus and Bornavirus Research. Viruses 2023; 15:158. [PMID: 36680198 PMCID: PMC9863967 DOI: 10.3390/v15010158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
The order Mononegavirales contains a variety of highly pathogenic viruses that may infect humans, including the families Filoviridae, Bornaviridae, Paramyxoviridae, and Rhabodoviridae. Animal models have historically been important to study virus pathogenicity and to develop medical countermeasures. As these have inherent shortcomings, the rise of microphysiological systems and organoids able to recapitulate hallmarks of the diseases caused by these viruses may have enormous potential to add to or partially replace animal modeling in the future. Indeed, microphysiological systems and organoids are already used in the pharmaceutical R&D pipeline because they are prefigured to overcome the translational gap between model systems and clinical studies. Moreover, they may serve to alleviate ethical concerns related to animal research. In this review, we discuss the value of animal model alternatives in human pathogenic filovirus and bornavirus research. The current animal models and their limitations are presented followed by an overview of existing alternatives, such as organoids and microphysiological systems, which might help answering open research questions.
Collapse
Affiliation(s)
- Lina Widerspick
- Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Riems, 38124 Braunschweig, Germany
| | | | - Dennis Tappe
- Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany
- National Reference Center for Tropical Pathogens, Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany
| | - César Muñoz-Fontela
- Bernhard-Nocht-Institute for Tropical Medicine, 20359 Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Luebeck-Borstel-Riems, 38124 Braunschweig, Germany
| |
Collapse
|
21
|
Xiang K, Zhuang H. Liver Organoid Potential Application for Hepatitis E Virus Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1417:133-139. [PMID: 37223863 DOI: 10.1007/978-981-99-1304-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Despite the advances in hepatitis E virus (HEV) cell infection models' development, HEV infection efficacy in these cell models is still low, which hampers the further study of molecular mechanism of HEV infection and replication and even the interaction between HEV and host. Along with the advances in the technology for liver organoids generation, major efforts will be made to develop liver organoids for HEV infection. Here, we summarize the entire new and impressive cell culture system of liver organoids and discuss their potential application in HEV infection and pathogenesis. Liver organoids can be generated from tissue-resident cells isolated from biopsies of adult tissues or from iPSCs/ESCs differentiation, which can expand the large-scale experiments such as antiviral drug screening. Different types of liver cells working together can recapitulate the liver organ maintaining the physiological and biochemical microenvironments to support cell morphogenesis, migration, and response to viral infections. Efforts to optimize the protocols for liver organoids generation will speed up the research for HEV infection and pathogenesis and even the antiviral drug identification and evaluation.
Collapse
Affiliation(s)
- Kuanhui Xiang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.
| | - Hui Zhuang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
22
|
Valdiviezo A, Brown GE, Michell AR, Trinconi CM, Bodke VV, Khetani SR, Luo YS, Chiu WA, Rusyn I. Reanalysis of Trichloroethylene and Tetrachloroethylene Metabolism to Glutathione Conjugates Using Human, Rat, and Mouse Liver in Vitro Models to Improve Precision in Risk Characterization. ENVIRONMENTAL HEALTH PERSPECTIVES 2022; 130:117009. [PMID: 36445294 PMCID: PMC9707501 DOI: 10.1289/ehp12006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/16/2022] [Accepted: 11/15/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Both trichloroethylene (TCE) and tetrachloroethylene (PCE) are high-priority chemicals subject to numerous human health risk evaluations by a range of agencies. Metabolism of TCE and PCE determines their ultimate toxicity; important uncertainties exist in quantitative characterization of metabolism to genotoxic moieties through glutathione (GSH) conjugation and species differences therein. OBJECTIVES This study aimed to address these uncertainties using novel in vitro liver models, interspecies comparison, and a sensitive assay for quantification of GSH conjugates of TCE and PCE, S-(1,2-dichlorovinyl)glutathione (DCVG) and S-(1,2,2-trichlorovinyl) glutathione (TCVG), respectively. METHODS Liver in vitro models used herein were suspension, 2-D culture, and micropatterned coculture (MPCC) with primary human, rat, and mouse hepatocytes, as well as human induced pluripotent stem cell (iPSC)-derived hepatocytes (iHep). RESULTS We found that, although efficiency of metabolism varied among models, consistent with known differences in their metabolic capacity, formation rates of DCVG and TCVG generally followed the patterns human ≥ rat ≥ mouse , and primary hepatocytes > iHep . Data derived from MPCC were most consistent with estimates from physiologically based pharmacokinetic models calibrated to in vivo data. DISCUSSION For TCE, the new data provided additional empirical support for inclusion of GSH conjugation-mediated kidney effects as critical for the derivation of noncancer toxicity values. For PCE, the data reduced previous uncertainties regarding the extent of TCVG formation in humans; this information was used to update several candidate kidney-specific noncancer toxicity values. Overall, MPCC-derived data provided physiologically relevant estimates of GSH-mediated metabolism of TCE and PCE to reduce uncertainties in interspecies extrapolation that constrained previous risk evaluations, thereby increasing the precision of risk characterizations of these high-priority toxicants. https://doi.org/10.1289/EHP12006.
Collapse
Affiliation(s)
- Alan Valdiviezo
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Grace E. Brown
- Department of Biomedical Engineering, University of Illinois Chicago, Illinois, USA
| | - Ashlin R. Michell
- Department of Biomedical Engineering, University of Illinois Chicago, Illinois, USA
| | | | - Vedant V. Bodke
- Department of Biomedical Engineering, University of Illinois Chicago, Illinois, USA
| | - Salman R. Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, Illinois, USA
| | - Yu-Syuan Luo
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Weihsueh A. Chiu
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
23
|
Valdiviezo A, Kato Y, Baker ES, Chiu WA, Rusyn I. Evaluation of Metabolism of a Defined Pesticide Mixture through Multiple In Vitro Liver Models. TOXICS 2022; 10:566. [PMID: 36287846 PMCID: PMC9609317 DOI: 10.3390/toxics10100566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
The evaluation of exposure to multiple contaminants in a mixture presents a number of challenges. For example, the characterization of chemical metabolism in a mixture setting remains a research area with critical knowledge gaps. Studies of chemical metabolism typically utilize suspension cultures of primary human hepatocytes; however, this model is not suitable for studies of more extended exposures and donor-to-donor variability in a metabolic capacity is unavoidable. To address this issue, we utilized several in vitro models based on human-induced pluripotent stem cell (iPSC)-derived hepatocytes (iHep) to characterize the metabolism of an equimolar (1 or 5 µM) mixture of 20 pesticides. We used iHep suspensions and 2D sandwich cultures, and a microphysiological system OrganoPlate® 2-lane 96 (MimetasTM) that also included endothelial cells and THP-1 cell-derived macrophages. When cell culture media were evaluated using gas and liquid chromatography coupled to tandem mass spectrometry methods, we found that the parent molecule concentrations diminished, consistent with metabolic activity. This effect was most pronounced in iHep suspensions with a 1 µM mixture, and was lowest in OrganoPlate® 2-lane 96 for both mixtures. Additionally, we used ion mobility spectrometry-mass spectrometry (IMS-MS) to screen for metabolite formation in these cultures. These analyses revealed the presence of five primary metabolites that allowed for a more comprehensive evaluation of chemical metabolism in vitro. These findings suggest that iHep-based suspension assays maintain higher metabolic activity compared to 2D sandwich and OrganoPlate® 2-lane 96 model. Moreover, this study illustrates that IMS-MS can characterize in vitro metabolite formation following exposure to mixtures of environmental contaminants.
Collapse
Affiliation(s)
- Alan Valdiviezo
- Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Yuki Kato
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Laboratory for Drug Discovery and Development, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Erin S. Baker
- Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - Weihsueh A. Chiu
- Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
24
|
Scoon WA, Mancio-Silva L, Suder EL, Villacorta-Martin C, Lindstrom-Vautrin J, Bernbaum JG, Mazur S, Johnson RF, Olejnik J, Flores EY, Mithal A, Wang F, Hume AJ, Kaserman JE, March-Riera S, Wilson AA, Bhatia SN, Mühlberger E, Mostoslavsky G. Ebola virus infection induces a delayed type I IFN response in bystander cells and the shutdown of key liver genes in human iPSC-derived hepatocytes. Stem Cell Reports 2022; 17:2286-2302. [PMID: 36084636 PMCID: PMC9561183 DOI: 10.1016/j.stemcr.2022.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 01/26/2023] Open
Abstract
Liver damage and an exacerbated inflammatory response are hallmarks of Ebola virus (EBOV) infection. Little is known about the intrinsic response to infection in human hepatocytes and their contribution to inflammation. Here, we present an induced pluripotent stem cell (iPSC)-derived hepatocyte-like cell (HLC) platform to define the hepato-intrinsic response to EBOV infection. We used this platform to show robust EBOV infection, with characteristic ultrastructural changes and evidence for viral replication. Transcriptomics analysis revealed a delayed response with minimal early transcriptomic changes, followed by a general downregulation of hepatic function and upregulation of interferon signaling, providing a potential mechanism by which hepatocytes participate in disease severity and liver damage. Using RNA-fluorescence in situ hybridization (FISH), we showed that IFNB1 and CXCL10 were mainly expressed in non-infected bystander cells. We did not observe an inflammatory signature during infection. In conclusion, iPSC-HLCs are an immune competent platform to study responses to EBOV infection.
Collapse
Affiliation(s)
- Whitney A. Scoon
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, 620 Albany Street, Boston, MA 02118, USA,Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118, USA
| | - Liliana Mancio-Silva
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, MA 02139, USA
| | - Ellen L. Suder
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, 620 Albany Street, Boston, MA 02118, USA,Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA
| | - Jonathan Lindstrom-Vautrin
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA
| | - John G. Bernbaum
- Integrated Research Facility, Division of Clinical Research, National Institute for Allergy and Infectious Disease, National Institutes of Health, Frederick, MD 21702, USA
| | - Steve Mazur
- Integrated Research Facility, Division of Clinical Research, National Institute for Allergy and Infectious Disease, National Institutes of Health, Frederick, MD 21702, USA
| | - Reed F. Johnson
- Integrated Research Facility, Division of Clinical Research, National Institute for Allergy and Infectious Disease, National Institutes of Health, Frederick, MD 21702, USA,Emerging Viral Pathogens Section, Laboratory of Immunoregulation, Division of Intramural Research, National Institute for Allergy and Infectious Disease, National Institutes of Health, Frederick, MD 21702, USA
| | - Judith Olejnik
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, 620 Albany Street, Boston, MA 02118, USA,Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118, USA
| | - Elizabeth Y. Flores
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA,National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, 620 Albany Street, Boston, MA 02118, USA,Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118, USA
| | - Aditya Mithal
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA,Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118, USA
| | - Feiya Wang
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA
| | - Adam J. Hume
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, 620 Albany Street, Boston, MA 02118, USA,Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118, USA
| | - Joseph E. Kaserman
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Sandra March-Riera
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, MA 02139, USA
| | - Andrew A. Wilson
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA,The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Sangeeta N. Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, MA 02139, USA,Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA,Broad Institute, Cambridge, MA 02139, USA,Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Elke Mühlberger
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, 620 Albany Street, Boston, MA 02118, USA; Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118, USA.
| | - Gustavo Mostoslavsky
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, 670 Albany Street, Suite 209, Boston, MA 02118, USA; National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, 620 Albany Street, Boston, MA 02118, USA; Department of Microbiology, Boston University School of Medicine, 620 Albany Street, Boston, MA 02118, USA; Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, 670 Albany Street, Suite 209, Boston, MA 02118, USA.
| |
Collapse
|
25
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
26
|
Xu X, Jiang S, Gu L, Li B, Xu F, Li C, Chen P. High-throughput bioengineering of homogenous and functional human-induced pluripotent stem cells-derived liver organoids via micropatterning technique. Front Bioeng Biotechnol 2022; 10:937595. [PMID: 36032707 PMCID: PMC9399390 DOI: 10.3389/fbioe.2022.937595] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/05/2022] [Indexed: 11/15/2022] Open
Abstract
Human pluripotent stem cell-derived liver organoids are emerging as more human-relevant in vitro models for studying liver diseases and hepatotoxicity than traditional hepatocyte cultures and animal models. The generation of liver organoids is based on the Matrigel dome method. However, the organoids constructed by this method display significant heterogeneity in their morphology, size, and maturity. Additionally, the formed organoid is randomly encapsulated in the Matrigel dome, which is not convenient for in situ staining and imaging. Here, we demonstrate an approach to generate a novel type of liver organoids via micropatterning technique. This approach enables the reproducible and high-throughput formation of bioengineered fetal liver organoids with uniform morphology and deterministic size and location in a multiwell plate. The liver organoids constructed by this technique closely recapitulate some critical features of human liver development at the fetal stage, including fetal liver-specific gene and protein expression, glycogen storage, lipid accumulation, and protein secretion. Additionally, the organoids allow whole-mount in-situ staining and imaging. Overall, this new type of liver organoids is compatible with the pharmaceutical industry’s widely-used preclinical drug discovery tools and will facilitate liver drug screening and hepatotoxic assessment.
Collapse
Affiliation(s)
- Xiaodong Xu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Shanqing Jiang
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Longjun Gu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Bin Li
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Fang Xu
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Changyong Li
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Pu Chen
- Tissue Engineering and Organ Manufacturing (TEOM) Lab, Department of Biomedical Engineering, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
- Institute of Hepatobiliary Diseases of Wuhan University, Hubei Engineering Center of Natural Polymers-based Medical Materials, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Pu Chen,
| |
Collapse
|
27
|
Abstract
Liver regeneration is a well-orchestrated process that is typically studied in animal models. Although previous animal studies have offered many insights into liver regeneration, human biology is less well understood. To this end, we developed a three-dimensional (3D) platform called structurally vascularized hepatic ensembles for analyzing regeneration (SHEAR) to model multiple aspects of human liver regeneration. SHEAR enables control over hemodynamic alterations to mimic those that occur during liver injury and regeneration and supports the administration of biochemical inputs such as cytokines and paracrine interactions with endothelial cells. We found that exposing the endothelium-lined channel to fluid flow led to increased secretion of regeneration-associated factors. Stimulation with relevant cytokines not only amplified the secretory response, but also induced cell-cycle entry of primary human hepatocytes (PHHs) embedded within the device. Further, we identified endothelial-derived mediators that are sufficient to initiate proliferation of PHHs in this context. Collectively, the data presented here underscore the importance of multicellular models that can recapitulate high-level tissue functions and demonstrate that the SHEAR device can be used to discover and validate conditions that promote human liver regeneration.
Collapse
|
28
|
Aparici Herraiz I, Caires HR, Castillo-Fernández Ó, Sima N, Méndez-Mora L, Risueño RM, Sattabongkot J, Roobsoong W, Hernández-Machado A, Fernandez-Becerra C, Barrias CC, del Portillo HA. Advancing Key Gaps in the Knowledge of Plasmodium vivax Cryptic Infections Using Humanized Mouse Models and Organs-on-Chips. Front Cell Infect Microbiol 2022; 12:920204. [PMID: 35873153 PMCID: PMC9302440 DOI: 10.3389/fcimb.2022.920204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium vivax is the most widely distributed human malaria parasite representing 36.3% of disease burden in the South-East Asia region and the most predominant species in the region of the Americas. Recent estimates indicate that 3.3 billion of people are under risk of infection with circa 7 million clinical cases reported each year. This burden is certainly underestimated as the vast majority of chronic infections are asymptomatic. For centuries, it has been widely accepted that the only source of cryptic parasites is the liver dormant stages known as hypnozoites. However, recent evidence indicates that niches outside the liver, in particular in the spleen and the bone marrow, can represent a major source of cryptic chronic erythrocytic infections. The origin of such chronic infections is highly controversial as many key knowledge gaps remain unanswered. Yet, as parasites in these niches seem to be sheltered from immune response and antimalarial drugs, research on this area should be reinforced if elimination of malaria is to be achieved. Due to ethical and technical considerations, working with the liver, bone marrow and spleen from natural infections is very difficult. Recent advances in the development of humanized mouse models and organs-on-a-chip models, offer novel technological frontiers to study human diseases, vaccine validation and drug discovery. Here, we review current data of these frontier technologies in malaria, highlighting major challenges ahead to study P. vivax cryptic niches, which perpetuate transmission and burden.
Collapse
Affiliation(s)
- Iris Aparici Herraiz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Hugo R. Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Óscar Castillo-Fernández
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Núria Sima
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Lourdes Méndez-Mora
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | - Ruth M. Risueño
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Aurora Hernández-Machado
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Centre de Recerca Matemàtica (CRM), Barcelona, Spain
| | - Carmen Fernandez-Becerra
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Cristina C. Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Hernando A. del Portillo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- *Correspondence: Hernando A. del Portillo,
| |
Collapse
|
29
|
Mancio-Silva L, Gural N, Real E, Wadsworth MH, Butty VL, March S, Nerurkar N, Hughes TK, Roobsoong W, Fleming HE, Whittaker CA, Levine SS, Sattabongkot J, Shalek AK, Bhatia SN. A single-cell liver atlas of Plasmodium vivax infection. Cell Host Microbe 2022; 30:1048-1060.e5. [PMID: 35443155 DOI: 10.1016/j.chom.2022.03.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/31/2022] [Accepted: 03/25/2022] [Indexed: 12/15/2022]
Abstract
Malaria-causing Plasmodium vivax parasites can linger in the human liver for weeks to years and reactivate to cause recurrent blood-stage infection. Although they are an important target for malaria eradication, little is known about the molecular features of replicative and non-replicative intracellular liver-stage parasites and their host cell dependence. Here, we leverage a bioengineered human microliver platform to culture patient-derived P. vivax parasites for transcriptional profiling. Coupling enrichment strategies with bulk and single-cell analyses, we capture both parasite and host transcripts in individual hepatocytes throughout the course of infection. We define host- and state-dependent transcriptional signatures and identify unappreciated populations of replicative and non-replicative parasites that share features with sexual transmissive forms. We find that infection suppresses the transcription of key hepatocyte function genes and elicits an anti-parasite innate immune response. Our work provides a foundation for understanding host-parasite interactions and reveals insights into the biology of P. vivax dormancy and transmission.
Collapse
Affiliation(s)
- Liliana Mancio-Silva
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France.
| | - Nil Gural
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Eliana Real
- Institut Pasteur, Université Paris Cité, Inserm U1201, CNRS EMR9195, Unité de Biologie des Interactions Hôte-Parasite, 75015 Paris, France
| | - Marc H Wadsworth
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA
| | - Vincent L Butty
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Niketa Nerurkar
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Travis K Hughes
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok 10400, Thailand
| | - Heather E Fleming
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Charlie A Whittaker
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Stuart S Levine
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; BioMicro Center, MIT, Cambridge, MA 02139, USA
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine Mahidol University, Bangkok 10400, Thailand
| | - Alex K Shalek
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Department of Chemistry, MIT, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, MIT, and Harvard, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA.
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; The Wyss Institute for Biologically Inspired Engineering Harvard University Boston, MA 02215, USA.
| |
Collapse
|
30
|
Tasnim F, Huang X, Lee CZW, Ginhoux F, Yu H. Recent Advances in Models of Immune-Mediated Drug-Induced Liver Injury. FRONTIERS IN TOXICOLOGY 2022; 3:605392. [PMID: 35295156 PMCID: PMC8915912 DOI: 10.3389/ftox.2021.605392] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatic inflammation is a key feature of a variety of liver diseases including drug-induced liver injury (DILI), orchestrated by the innate immune response (Kupffer cells, monocytes, neutrophils, dendritic cells) and the adaptive immune system (T cells and natural killer T cells). In contrast to acute DILI, prediction of immune-mediated DILI (im-DILI) has been more challenging due to complex disease pathogenesis, lack of reliable models and limited knowledge of underlying mechanisms. This review summarizes in vivo and in vitro systems that have been used to model im-DILI. In particular, the review focuses on state-of-the-art in vitro human-based multicellular models which have been developed to supplement the use of in vivo models due to interspecies variation and increasing ethical concerns regarding animal use. Advantages of the co-cultures in maintaining hepatocyte functions and importantly, introducing heterotypic cell-cell interactions to mimic inflammatory hepatic microenvironment are discussed. Challenges regarding cell source and incorporation of different cells with physical cell-cell contact are outlined and potential solutions are proposed. It is likely that better understanding of the interplay of immune cells in liver models will allow for the development of more accurate systems to better predict hepatotoxicity and stratification of drugs that can cause immune-mediated effects.
Collapse
Affiliation(s)
- Farah Tasnim
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Institute of Bioengineering and Nanotechnology, The Nanos, Singapore, Singapore
| | - Xiaozhong Huang
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Institute of Bioengineering and Nanotechnology, The Nanos, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christopher Zhe Wei Lee
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Singapore Immunology Network, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Florent Ginhoux
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Singapore Immunology Network, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China.,Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Hanry Yu
- Innovations in Food & Chemical Safety Programme, ASTAR, Singapore, Singapore.,Institute of Bioengineering and Nanotechnology, The Nanos, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National University of Singapore (NUS) Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, Singapore, Singapore.,T-Labs, Mechanobiology Institute, Singapore, Singapore.,Critical Analytics for Manufacturing Personalised-Medicine Interdisciplinary Research Groups (CAMP-IRG), Singapore-Massachusetts Institute of Technology Alliance for Research and Technology, Singapore, Singapore
| |
Collapse
|
31
|
Pantasis S, Friemel J, Brütsch SM, Hu Z, Krautbauer S, Liebisch G, Dengjel J, Weber A, Werner S, Bordoli MR. Vertebrate lonesome kinase modulates the hepatocyte secretome to prevent perivascular liver fibrosis and inflammation. J Cell Sci 2022; 135:275016. [PMID: 35293576 PMCID: PMC9016620 DOI: 10.1242/jcs.259243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/08/2022] [Indexed: 11/20/2022] Open
Abstract
Vertebrate lonesome kinase (VLK) is the only known extracellular tyrosine kinase, but its physiological functions are largely unknown. We show that VLK is highly expressed in hepatocytes of neonatal mice, but downregulated during adulthood. To determine the role of VLK in liver homeostasis and regeneration, we generated mice with a hepatocyte-specific knockout of the VLK gene (Pkdcc). Cultured progenitor cells established from primary hepatocytes of Pkdcc knockout mice produced a secretome, which promoted their own proliferation in 3D spheroids and proliferation of cultured fibroblasts. In vivo, Pkdcc knockout mice developed liver steatosis with signs of inflammation and perivascular fibrosis upon aging, combined with expansion of liver progenitor cells. In response to chronic CCl4-induced liver injury, the pattern of deposited collagen was significantly altered in these mice. The liver injury marker alpha-fetoprotein (AFP) was increased in the secretome of VLK-deficient cultured progenitor cells and in liver tissues of aged or CCl4-treated knockout mice. These results support a key role for VLK and extracellular protein phosphorylation in liver homeostasis and repair through paracrine control of liver cell function and regulation of appropriate collagen deposition. This article has an associated First Person interview with the first author of the paper. Summary: The secreted protein kinase VLK is released from hepatocytes and protects the liver from perivascular fibrosis and inflammation.
Collapse
Affiliation(s)
- Sophia Pantasis
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology ETH, Otto-Stern Weg 7, CH-8093, Zurich, Switzerland
| | - Juliane Friemel
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Salome Mirjam Brütsch
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology ETH, Otto-Stern Weg 7, CH-8093, Zurich, Switzerland
| | - Zehan Hu
- Department of Biology, Université de Fribourg, Chemin du Musée 10, CH-1700, Fribourg, Switzerland
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Joern Dengjel
- Department of Biology, Université de Fribourg, Chemin du Musée 10, CH-1700, Fribourg, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology ETH, Otto-Stern Weg 7, CH-8093, Zurich, Switzerland
| | - Mattia Renato Bordoli
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology ETH, Otto-Stern Weg 7, CH-8093, Zurich, Switzerland
| |
Collapse
|
32
|
Wang X, Guo C, Guo L, Wang M, Liu M, Song Y, Jiao H, Wei X, Zhao Z, Kaplan DL. Radially Aligned Porous Silk Fibroin Scaffolds as Functional Templates for Engineering Human Biomimetic Hepatic Lobules. ACS APPLIED MATERIALS & INTERFACES 2022; 14:201-213. [PMID: 34929079 DOI: 10.1021/acsami.1c18215] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Bioengineering functional hepatic tissue constructs that physiologically replicate the human native liver tissue in vitro is sought for clinical research and drug discovery. However, the intricate architecture and specific biofunctionality possessed by the native liver tissue remain challenging to mimic in vitro. In the present study, a versatile strategy to fabricate lobular-like silk protein scaffolds with radially aligned lamellar sheets, interconnected channels, and a converging central cavity was designed and implemented. A proof-of-concept study to bioengineer biomimetic hepatic lobules was conducted through coculturing human hepatocytes and primary endothelial cells on these lobular-like scaffolds. Relatively long-term viability of hepatocyte/endothelial cells was found along with cell alignment and organization in vitro. The hepatocytes showed special epithelial polarity and bile duct formation, similar to the liver plate, while the aligned endothelial cells generated endothelial networks, similar to natural hepatic sinuses. This endowed the three-dimensional (3D) tissue constructs with the capability to recapitulate hepatic-like parenchymal-mesenchymal growth patterns in vitro. More importantly, the cocultured hepatocytes outperformed monocultures or monolayer cultures, displaying significantly enhanced hepatocyte functions, including functional gene expression, albumin (ALB) secretion, urea synthesis, and metabolic activity. Thus, this functional unit with a biomimetic phenotype provides a novel technology for bioengineering biomimetic hepatic lobules in vitro, with potential utility as a building block for bioartificial liver (BAL) engineering or as a robust tool for drug metabolism investigation.
Collapse
Affiliation(s)
- Xiuli Wang
- Department of Histology & Embryology, College of Basic Medical Sciences, Dalian Medical University, Liaoning 116044, China
- Biomedical Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| | - Chengchen Guo
- Biomedical Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
- School of Engineering, Westlake University, Hangzhou, Zhejiang 310023, China
| | - Lina Guo
- Department of Histology & Embryology, College of Basic Medical Sciences, Dalian Medical University, Liaoning 116044, China
| | - Mingqi Wang
- Department of Histology & Embryology, College of Basic Medical Sciences, Dalian Medical University, Liaoning 116044, China
| | - Ming Liu
- Department of Histology & Embryology, College of Basic Medical Sciences, Dalian Medical University, Liaoning 116044, China
| | - Yizhe Song
- Department of Histology & Embryology, College of Basic Medical Sciences, Dalian Medical University, Liaoning 116044, China
| | - Hui Jiao
- Department of Histology & Embryology, College of Basic Medical Sciences, Dalian Medical University, Liaoning 116044, China
| | - Xiaoqing Wei
- Department of Histology & Embryology, College of Basic Medical Sciences, Dalian Medical University, Liaoning 116044, China
| | - Zinan Zhao
- Department of Histology & Embryology, College of Basic Medical Sciences, Dalian Medical University, Liaoning 116044, China
| | - David L Kaplan
- Biomedical Engineering Department, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
33
|
Segovia-Zafra A, Di Zeo-Sánchez DE, López-Gómez C, Pérez-Valdés Z, García-Fuentes E, Andrade RJ, Lucena MI, Villanueva-Paz M. Preclinical models of idiosyncratic drug-induced liver injury (iDILI): Moving towards prediction. Acta Pharm Sin B 2021; 11:3685-3726. [PMID: 35024301 PMCID: PMC8727925 DOI: 10.1016/j.apsb.2021.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Idiosyncratic drug-induced liver injury (iDILI) encompasses the unexpected harms that prescription and non-prescription drugs, herbal and dietary supplements can cause to the liver. iDILI remains a major public health problem and a major cause of drug attrition. Given the lack of biomarkers for iDILI prediction, diagnosis and prognosis, searching new models to predict and study mechanisms of iDILI is necessary. One of the major limitations of iDILI preclinical assessment has been the lack of correlation between the markers of hepatotoxicity in animal toxicological studies and clinically significant iDILI. Thus, major advances in the understanding of iDILI susceptibility and pathogenesis have come from the study of well-phenotyped iDILI patients. However, there are many gaps for explaining all the complexity of iDILI susceptibility and mechanisms. Therefore, there is a need to optimize preclinical human in vitro models to reduce the risk of iDILI during drug development. Here, the current experimental models and the future directions in iDILI modelling are thoroughly discussed, focusing on the human cellular models available to study the pathophysiological mechanisms of the disease and the most used in vivo animal iDILI models. We also comment about in silico approaches and the increasing relevance of patient-derived cellular models.
Collapse
Affiliation(s)
- Antonio Segovia-Zafra
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - Daniel E. Di Zeo-Sánchez
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Carlos López-Gómez
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Zeus Pérez-Valdés
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Raúl J. Andrade
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - M. Isabel Lucena
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
- Platform ISCIII de Ensayos Clínicos, UICEC-IBIMA, Málaga 29071, Spain
| | - Marina Villanueva-Paz
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| |
Collapse
|
34
|
Maher SP, Vantaux A, Chaumeau V, Chua ACY, Cooper CA, Andolina C, Péneau J, Rouillier M, Rizopoulos Z, Phal S, Piv E, Vong C, Phen S, Chhin C, Tat B, Ouk S, Doeurk B, Kim S, Suriyakan S, Kittiphanakun P, Awuku NA, Conway AJ, Jiang RHY, Russell B, Bifani P, Campo B, Nosten F, Witkowski B, Kyle DE. Probing the distinct chemosensitivity of Plasmodium vivax liver stage parasites and demonstration of 8-aminoquinoline radical cure activity in vitro. Sci Rep 2021; 11:19905. [PMID: 34620901 PMCID: PMC8497498 DOI: 10.1038/s41598-021-99152-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022] Open
Abstract
Improved control of Plasmodium vivax malaria can be achieved with the discovery of new antimalarials with radical cure efficacy, including prevention of relapse caused by hypnozoites residing in the liver of patients. We screened several compound libraries against P. vivax liver stages, including 1565 compounds against mature hypnozoites, resulting in one drug-like and several probe-like hits useful for investigating hypnozoite biology. Primaquine and tafenoquine, administered in combination with chloroquine, are currently the only FDA-approved antimalarials for radical cure, yet their activity against mature P. vivax hypnozoites has not yet been demonstrated in vitro. By developing an extended assay, we show both drugs are individually hypnozonticidal and made more potent when partnered with chloroquine, similar to clinically relevant combinations. Post-hoc analyses of screening data revealed excellent performance of ionophore controls and the high quality of single point assays, demonstrating a platform able to support screening of greater compound numbers. A comparison of P. vivax liver stage activity data with that of the P. cynomolgi blood, P. falciparum blood, and P. berghei liver stages reveals overlap in schizonticidal but not hypnozonticidal activity, indicating that the delivery of new radical curative agents killing P. vivax hypnozoites requires an independent and focused drug development test cascade.
Collapse
Affiliation(s)
- Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr. Suite 370, Athens, GA, 30602, USA.
| | - Amélie Vantaux
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Victor Chaumeau
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 68/30 Bantung Rd., Mae Sot, Tak, 63110, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Adeline C Y Chua
- Infectious Diseases Laboratories (ID Labs), Agency for Science, Technology and Research (A*STAR), Immunos, Biopolis, Singapore, 138648, Singapore
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Caitlin A Cooper
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr. Suite 370, Athens, GA, 30602, USA
| | - Chiara Andolina
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 68/30 Bantung Rd., Mae Sot, Tak, 63110, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Julie Péneau
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Mélanie Rouillier
- Medicines for Malaria Venture (MMV), Route de Pré-Bois 20, 1215, Geneva, Switzerland
| | - Zaira Rizopoulos
- Medicines for Malaria Venture (MMV), Route de Pré-Bois 20, 1215, Geneva, Switzerland
| | - Sivchheng Phal
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Eakpor Piv
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Chantrea Vong
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Sreyvouch Phen
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Chansophea Chhin
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Baura Tat
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Sivkeng Ouk
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Bros Doeurk
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Saorin Kim
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia
| | - Sangrawee Suriyakan
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 68/30 Bantung Rd., Mae Sot, Tak, 63110, Thailand
| | - Praphan Kittiphanakun
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 68/30 Bantung Rd., Mae Sot, Tak, 63110, Thailand
| | - Nana Akua Awuku
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr. Suite 370, Athens, GA, 30602, USA
| | - Amy J Conway
- Department of Global Health, College of Public Health, Center for Global Health and Infectious Disease Research, University of South Florida, 3720 Spectrum Blvd Suite 402, Tampa, FL, 33612, USA
| | - Rays H Y Jiang
- Department of Global Health, College of Public Health, Center for Global Health and Infectious Disease Research, University of South Florida, 3720 Spectrum Blvd Suite 402, Tampa, FL, 33612, USA
| | - Bruce Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Pablo Bifani
- Infectious Diseases Laboratories (ID Labs), Agency for Science, Technology and Research (A*STAR), Immunos, Biopolis, Singapore, 138648, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, WC1E 7HT, UK
| | - Brice Campo
- Medicines for Malaria Venture (MMV), Route de Pré-Bois 20, 1215, Geneva, Switzerland
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 68/30 Bantung Rd., Mae Sot, Tak, 63110, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research Building, University of Oxford, Old Road Campus, Oxford, UK
| | - Benoît Witkowski
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, 5 Boulevard Monivong, PO Box 983, Phnom Penh, 12201, Cambodia.
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 DW Brooks Dr. Suite 370, Athens, GA, 30602, USA.
| |
Collapse
|
35
|
Schepers AG, Shan J, Cox AG, Huang A, Evans H, Walesky C, Fleming HE, Goessling W, Bhatia SN. Identification of NQO2 As a Protein Target in Small Molecule Modulation of Hepatocellular Function. ACS Chem Biol 2021; 16:1770-1778. [PMID: 34427427 DOI: 10.1021/acschembio.1c00503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The utility of in vitro human disease models is mainly dependent on the availability and functional maturity of tissue-specific cell types. We have previously screened for and identified small molecules that can enhance hepatocyte function in vitro. Here, we characterize the functional effects of one of the hits, FH1, on primary human hepatocytes in vitro, and also in vivo on primary hepatocytes in a zebrafish model. Furthermore, we conducted an analogue screen to establish the structure-activity relationship of FH1. We performed affinity-purification proteomics that identified NQO2 to be a potential binding target for this small molecule, revealing a possible link between inflammatory signaling and hepatocellular function in zebrafish and human hepatocyte model systems.
Collapse
Affiliation(s)
- Arnout G. Schepers
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Netherlands Cancer Institute, 1066 CX Amsterdam, Netherlands
| | - Jing Shan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Andrew G. Cox
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ada Huang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Helen Evans
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Chad Walesky
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Heather E. Fleming
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Wolfram Goessling
- Genetics Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Harvard Stem Cell Institute, Cambridge, Massachusetts 02138, United States
- Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
- Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts 02114, United States
- Harvard−MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Sangeeta N. Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Harvard−MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02139, United States
- Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
36
|
De Crignis E, Hossain T, Romal S, Carofiglio F, Moulos P, Khalid MM, Rao S, Bazrafshan A, Verstegen MM, Pourfarzad F, Koutsothanassis C, Gehart H, Kan TW, Palstra RJ, Boucher C, IJzermans JN, Huch M, Boj SF, Vries R, Clevers H, van der Laan LJ, Hatzis P, Mahmoudi T. Application of human liver organoids as a patient-derived primary model for HBV infection and related hepatocellular carcinoma. eLife 2021; 10:e60747. [PMID: 34328417 PMCID: PMC8384419 DOI: 10.7554/elife.60747] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
The molecular events that drive hepatitis B virus (HBV)-mediated transformation and tumorigenesis have remained largely unclear, due to the absence of a relevant primary model system. Here we propose the use of human liver organoids as a platform for modeling HBV infection and related tumorigenesis. We first describe a primary ex vivo HBV-infection model derived from healthy donor liver organoids after challenge with recombinant virus or HBV-infected patient serum. HBV-infected organoids produced covalently closed circular DNA (cccDNA) and HBV early antigen (HBeAg), expressed intracellular HBV RNA and proteins, and produced infectious HBV. This ex vivo HBV-infected primary differentiated hepatocyte organoid platform was amenable to drug screening for both anti-HBV activity and drug-induced toxicity. We also studied HBV replication in transgenically modified organoids; liver organoids exogenously overexpressing the HBV receptor sodium taurocholate co-transporting polypeptide (NTCP) after lentiviral transduction were not more susceptible to HBV, suggesting the necessity for additional host factors for efficient infection. We also generated transgenic organoids harboring integrated HBV, representing a long-term culture system also suitable for viral production and the study of HBV transcription. Finally, we generated HBV-infected patient-derived liver organoids from non-tumor cirrhotic tissue of explants from liver transplant patients. Interestingly, transcriptomic analysis of patient-derived liver organoids indicated the presence of an aberrant early cancer gene signature, which clustered with the hepatocellular carcinoma (HCC) cohort on The Cancer Genome Atlas Liver Hepatocellular Carcinoma dataset and away from healthy liver tissue, and may provide invaluable novel biomarkers for the development of HCC and surveillance in HBV-infected patients.
Collapse
Affiliation(s)
- Elisa De Crignis
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Tanvir Hossain
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Shahla Romal
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Fabrizia Carofiglio
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Panagiotis Moulos
- Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
| | - Mir Mubashir Khalid
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Shringar Rao
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ameneh Bazrafshan
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Monique Ma Verstegen
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | | | | | - Helmuth Gehart
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Tsung Wai Kan
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Robert-Jan Palstra
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Charles Boucher
- Department of Viroscience, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Jan Nm IJzermans
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Meritxell Huch
- Max Plank Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sylvia F Boj
- Foundation Hubrecht Organoid Technology (HUB), Utrecht, Netherlands
| | - Robert Vries
- Foundation Hubrecht Organoid Technology (HUB), Utrecht, Netherlands
| | - Hans Clevers
- Hubrecht Institute-KNAW, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Luc Jw van der Laan
- Department of Surgery, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Pantelis Hatzis
- Biomedical Sciences Research Center 'Alexander Fleming', Vari, Greece
| | - Tokameh Mahmoudi
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Urology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Pathology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
37
|
Ya S, Ding W, Li S, Du K, Zhang Y, Li C, Liu J, Li F, Li P, Luo T, He L, Xu A, Gao D, Qiu B. On-Chip Construction of Liver Lobules with Self-Assembled Perfusable Hepatic Sinusoid Networks. ACS APPLIED MATERIALS & INTERFACES 2021; 13:32640-32652. [PMID: 34225454 DOI: 10.1021/acsami.1c00794] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Although various liver chips have been developed using emerging organ-on-a-chip techniques, it remains an enormous challenge to replicate the liver lobules with self-assembled perfusable hepatic sinusoid networks. Herein we develop a lifelike bionic liver lobule chip (LLC), on which the perfusable hepatic sinusoid networks are achieved using a microflow-guided angiogenesis methodology; additionally, during and after self-assembly, oxygen concentration is regulated to mimic physiologically dissolved levels supplied by actual hepatic arterioles and venules. This liver lobule design thereby produces more bionic liver microstructures, higher metabolic abilities, and longer lasting hepatocyte function than other liver-on-a-chip techniques that are able to deliver. We found that the flow through the unique micropillar design in the cell coculture zone guides the radiating assembly of the hepatic sinusoid, the oxygen concentration affects the morphology of the sinusoid by proliferation, and the oxygen gradient plays a key role in prolonging hepatocyte function. The expected breadth of applications our LLC is suited to is demonstrated by means of preliminarily testing chronic and acute hepatotoxicity of drugs and replicating growth of tumors in situ. This work provides new insights into designing more extensive bionic vascularized liver chips, while achieving longer lasting ex-vivo hepatocyte function.
Collapse
Affiliation(s)
- Shengnan Ya
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Weiping Ding
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Lab for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shibo Li
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Kun Du
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yuanyuan Zhang
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Chengpan Li
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Jing Liu
- School of Biology, Food and Environment Engineering, Hefei University, Hefei, Anhui 230601, China
| | - Fenfen Li
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Lab for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ping Li
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Tianzhi Luo
- School of Engineering Science, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Liqun He
- School of Engineering Science, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ao Xu
- Division of Life Sciences and Medicine, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Bensheng Qiu
- The Centers for Biomedical Engineering, University of Science and Technology of China, Hefei, Anhui 230027, China
- Hefei National Lab for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
38
|
Khoshdel-Rad N, Zahmatkesh E, Bikmulina P, Peshkova M, Kosheleva N, Bezrukov EA, Sukhanov RB, Solovieva A, Shpichka A, Timashev P, Vosough M. Modeling Hepatotropic Viral Infections: Cells vs. Animals. Cells 2021; 10:1726. [PMID: 34359899 PMCID: PMC8305759 DOI: 10.3390/cells10071726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
The lack of an appropriate platform for a better understanding of the molecular basis of hepatitis viruses and the absence of reliable models to identify novel therapeutic agents for a targeted treatment are the two major obstacles for launching efficient clinical protocols in different types of viral hepatitis. Viruses are obligate intracellular parasites, and the development of model systems for efficient viral replication is necessary for basic and applied studies. Viral hepatitis is a major health issue and a leading cause of morbidity and mortality. Despite the extensive efforts that have been made on fundamental and translational research, traditional models are not effective in representing this viral infection in a laboratory. In this review, we discuss in vitro cell-based models and in vivo animal models, with their strengths and weaknesses. In addition, the most important findings that have been retrieved from each model are described.
Collapse
Affiliation(s)
- Niloofar Khoshdel-Rad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (N.K.-R.); (E.Z.)
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Ensieh Zahmatkesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (N.K.-R.); (E.Z.)
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| | - Polina Bikmulina
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (P.B.); (M.P.); (A.S.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Maria Peshkova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (P.B.); (M.P.); (A.S.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Nastasia Kosheleva
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- FSBSI ‘Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Evgeny A. Bezrukov
- Department of Urology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.A.B.); (R.B.S.)
| | - Roman B. Sukhanov
- Department of Urology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.A.B.); (R.B.S.)
| | - Anna Solovieva
- Department of Polymers and Composites, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (P.B.); (M.P.); (A.S.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (P.B.); (M.P.); (A.S.)
- World-Class Research Center “Digital biodesign and personalized healthcare”, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
- Department of Polymers and Composites, N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, 119991 Moscow, Russia;
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (N.K.-R.); (E.Z.)
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| |
Collapse
|
39
|
Mwakingwe-Omari A, Healy SA, Lane J, Cook DM, Kalhori S, Wyatt C, Kolluri A, Marte-Salcedo O, Imeru A, Nason M, Ding LK, Decederfelt H, Duan J, Neal J, Raiten J, Lee G, Hume JCC, Jeon JE, Ikpeama I, Kc N, Chakravarty S, Murshedkar T, Church LWP, Manoj A, Gunasekera A, Anderson C, Murphy SC, March S, Bhatia SN, James ER, Billingsley PF, Sim BKL, Richie TL, Zaidi I, Hoffman SL, Duffy PE. Two chemoattenuated PfSPZ malaria vaccines induce sterile hepatic immunity. Nature 2021; 595:289-294. [PMID: 34194041 PMCID: PMC11127244 DOI: 10.1038/s41586-021-03684-z] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 06/01/2021] [Indexed: 02/06/2023]
Abstract
The global decline in malaria has stalled1, emphasizing the need for vaccines that induce durable sterilizing immunity. Here we optimized regimens for chemoprophylaxis vaccination (CVac), for which aseptic, purified, cryopreserved, infectious Plasmodium falciparum sporozoites (PfSPZ) were inoculated under prophylactic cover with pyrimethamine (PYR) (Sanaria PfSPZ-CVac(PYR)) or chloroquine (CQ) (PfSPZ-CVac(CQ))-which kill liver-stage and blood-stage parasites, respectively-and we assessed vaccine efficacy against homologous (that is, the same strain as the vaccine) and heterologous (a different strain) controlled human malaria infection (CHMI) three months after immunization ( https://clinicaltrials.gov/ , NCT02511054 and NCT03083847). We report that a fourfold increase in the dose of PfSPZ-CVac(PYR) from 5.12 × 104 to 2 × 105 PfSPZs transformed a minimal vaccine efficacy (low dose, two out of nine (22.2%) participants protected against homologous CHMI), to a high-level vaccine efficacy with seven out of eight (87.5%) individuals protected against homologous and seven out of nine (77.8%) protected against heterologous CHMI. Increased protection was associated with Vδ2 γδ T cell and antibody responses. At the higher dose, PfSPZ-CVac(CQ) protected six out of six (100%) participants against heterologous CHMI three months after immunization. All homologous (four out of four) and heterologous (eight out of eight) infectivity control participants showed parasitaemia. PfSPZ-CVac(CQ) and PfSPZ-CVac(PYR) induced a durable, sterile vaccine efficacy against a heterologous South American strain of P. falciparum, which has a genome and predicted CD8 T cell immunome that differs more strongly from the African vaccine strain than other analysed African P. falciparum strains.
Collapse
Affiliation(s)
- Agnes Mwakingwe-Omari
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Center for Vaccine Research, GlaxoSmithKline, Rockville, MD, USA
| | - Sara A Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacquelyn Lane
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David M Cook
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sahand Kalhori
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Charles Wyatt
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aarti Kolluri
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Omely Marte-Salcedo
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alemush Imeru
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Martha Nason
- Biostatistical Research Branch, National Institute of Allergy and Infectious Diseases, Rockville, MD, USA
| | - Lei K Ding
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hope Decederfelt
- Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Junhui Duan
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jillian Neal
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacob Raiten
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Grace Lee
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jen C C Hume
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jihyun E Jeon
- Clinical Center Pharmacy Department, National Institutes of Health, Bethesda, MD, USA
| | - Ijeoma Ikpeama
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Natasha Kc
- Sanaria, Rockville, MD, USA
- Protein Potential, Rockville, MD, USA
| | | | | | | | | | | | - Charles Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sean C Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Seattle Malaria Clinical Trials Center, Fred Hutch Cancer Research Center, Seattle, WA, USA
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Broad Institute, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - B Kim Lee Sim
- Sanaria, Rockville, MD, USA
- Protein Potential, Rockville, MD, USA
| | | | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
40
|
Suscovich TJ, Fallon JK, Das J, Demas AR, Crain J, Linde CH, Michell A, Natarajan H, Arevalo C, Broge T, Linnekin T, Kulkarni V, Lu R, Slein MD, Luedemann C, Marquette M, March S, Weiner J, Gregory S, Coccia M, Flores-Garcia Y, Zavala F, Ackerman ME, Bergmann-Leitner E, Hendriks J, Sadoff J, Dutta S, Bhatia SN, Lauffenburger DA, Jongert E, Wille-Reece U, Alter G. Mapping functional humoral correlates of protection against malaria challenge following RTS,S/AS01 vaccination. Sci Transl Med 2021; 12:12/553/eabb4757. [PMID: 32718991 DOI: 10.1126/scitranslmed.abb4757] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/02/2020] [Indexed: 12/13/2022]
Abstract
Vaccine development has the potential to be accelerated by coupling tools such as systems immunology analyses and controlled human infection models to define the protective efficacy of prospective immunogens without expensive and slow phase 2b/3 vaccine studies. Among human challenge models, controlled human malaria infection trials have long been used to evaluate candidate vaccines, and RTS,S/AS01 is the most advanced malaria vaccine candidate, reproducibly demonstrating 40 to 80% protection in human challenge studies in malaria-naïve individuals. Although antibodies are critical for protection after RTS,S/AS01 vaccination, antibody concentrations are inconsistently associated with protection across studies, and the precise mechanism(s) by which vaccine-induced antibodies provide protection remains enigmatic. Using a comprehensive systems serological profiling platform, the humoral correlates of protection against malaria were identified and validated across multiple challenge studies. Rather than antibody concentration, qualitative functional humoral features robustly predicted protection from infection across vaccine regimens. Despite the functional diversity of vaccine-induced immune responses across additional RTS,S/AS01 vaccine studies, the same antibody features, antibody-mediated phagocytosis and engagement of Fc gamma receptor 3A (FCGR3A), were able to predict protection across two additional human challenge studies. Functional validation using monoclonal antibodies confirmed the protective role of Fc-mediated antibody functions in restricting parasite infection both in vitro and in vivo, suggesting that these correlates may mechanistically contribute to parasite restriction and can be used to guide the rational design of an improved vaccine against malaria.
Collapse
Affiliation(s)
- Todd J Suscovich
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | | | - Jishnu Das
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Allison R Demas
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA.,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jonathan Crain
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Caitlyn H Linde
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Ashlin Michell
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Harini Natarajan
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Claudia Arevalo
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Thomas Broge
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Thomas Linnekin
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Viraj Kulkarni
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Richard Lu
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | - Matthew D Slein
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA
| | | | - Meghan Marquette
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sandra March
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Joshua Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Scott Gregory
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | - Yevel Flores-Garcia
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Fidel Zavala
- Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Elke Bergmann-Leitner
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Jenny Hendriks
- Janssen Vaccines & Prevention B.V., 2333CN Leiden, Netherlands
| | - Jerald Sadoff
- Janssen Vaccines & Prevention B.V., 2333CN Leiden, Netherlands
| | - Sheetij Dutta
- Malaria Vaccine Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA.,Broad Institute, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Douglas A Lauffenburger
- Koch Institute for Integrative Cancer Research, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | - Galit Alter
- Ragon Institute of MGH, Harvard, and MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
41
|
Ren G, Zheng X, Sharma V, Letson J, Nestor-Kalinoski AL, Furuta S. Loss of Nitric Oxide Induces Fibrogenic Response in Organotypic 3D Co-Culture of Mammary Epithelia and Fibroblasts-An Indicator for Breast Carcinogenesis. Cancers (Basel) 2021; 13:cancers13112815. [PMID: 34198735 PMCID: PMC8201212 DOI: 10.3390/cancers13112815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Fibrosis, which is often caused by chronic diseases and environmental substances, is closely associated with cancer. Thus, the development of a robust method allowing for deep studies of the linkage between fibrosis and cancer is essential. Here, we tested whether our novel three-dimensional (3D) co-culture of breast epithelia and fibroblasts would be a suitable model for that purpose. We compared the phenotypic effects of L-NAME, an inhibitor of nitric oxide (NO) production, on 3D mono- and co-cultures. We previously reported that prolonged NO depletion with L-NAME caused fibrosis and tumorigenesis in mouse mammary glands. Such in vivo effects of L-NAME were well recapitulated in 3D co-cultures, but not in 3D mono-cultures of epithelia and fibroblasts. These results support not only the essential roles of the presence of the stroma in cancer development, but also the utility of this co-culture in studying the causal relationship between fibrosis and cancer. Abstract Excessive myofibroblast activation, which leads to dysregulated collagen deposition and the stiffening of the extracellular matrix (ECM), plays pivotal roles in cancer initiation and progression. Cumulative evidence attests to the cancer-causing effects of a number of fibrogenic factors found in the environment, diseases and drugs. While identifying such factors largely depends on epidemiological studies, it would be of great importance to develop a robust in vitro method to demonstrate the causal relationship between fibrosis and cancer. Here, we tested whether our recently developed organotypic three-dimensional (3D) co-culture would be suitable for that purpose. This co-culture system utilizes the discontinuous ECM to separately culture mammary epithelia and fibroblasts in the discrete matrices to model the complexity of the mammary gland. We observed that pharmaceutical deprivation of nitric oxide (NO) in 3D co-cultures induced myofibroblast differentiation of the stroma as well as the occurrence of epithelial–mesenchymal transition (EMT) of the parenchyma. Such in vitro response to NO deprivation was unique to co-cultures and closely mimicked the phenotype of NO-depleted mammary glands exhibiting stromal desmoplasia and precancerous lesions undergoing EMT. These results suggest that this novel 3D co-culture system could be utilized in the deep mechanistic studies of the linkage between fibrosis and cancer.
Collapse
Affiliation(s)
- Gang Ren
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Xunzhen Zheng
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Vandana Sharma
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Joshua Letson
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
| | - Andrea L. Nestor-Kalinoski
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA;
| | - Saori Furuta
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA; (G.R.); (X.Z.); (V.S.); (J.L.)
- Correspondence:
| |
Collapse
|
42
|
Arez F, Rodrigues AF, Brito C, Alves PM. Bioengineered Liver Cell Models of Hepatotropic Infections. Viruses 2021; 13:773. [PMID: 33925701 PMCID: PMC8146083 DOI: 10.3390/v13050773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatitis viruses and liver-stage malaria are within the liver infections causing higher morbidity and mortality rates worldwide. The highly restricted tropism of the major human hepatotropic pathogens-namely, the human hepatitis B and C viruses and the Plasmodium falciparum and Plasmodium vivax parasites-has hampered the development of disease models. These models are crucial for uncovering the molecular mechanisms underlying the biology of infection and governing host-pathogen interaction, as well as for fostering drug development. Bioengineered cell models better recapitulate the human liver microenvironment and extend hepatocyte viability and phenotype in vitro, when compared with conventional two-dimensional cell models. In this article, we review the bioengineering tools employed in the development of hepatic cell models for studying infection, with an emphasis on 3D cell culture strategies, and discuss how those tools contributed to the level of recapitulation attained in the different model layouts. Examples of host-pathogen interactions uncovered by engineered liver models and their usefulness in drug development are also presented. Finally, we address the current bottlenecks, trends, and prospect toward cell models' reliability, robustness, and reproducibility.
Collapse
MESH Headings
- Animals
- Bioengineering/methods
- Cell Culture Techniques
- Disease Models, Animal
- Disease Susceptibility
- Drug Discovery
- Hepatitis/drug therapy
- Hepatitis/etiology
- Hepatitis/metabolism
- Hepatitis/pathology
- Hepatitis, Viral, Human/etiology
- Hepatitis, Viral, Human/metabolism
- Hepatitis, Viral, Human/pathology
- Hepatocytes/metabolism
- Hepatocytes/parasitology
- Hepatocytes/virology
- Host-Pathogen Interactions
- Humans
- Liver/metabolism
- Liver/parasitology
- Liver/virology
- Liver Diseases, Parasitic/etiology
- Liver Diseases, Parasitic/metabolism
- Liver Diseases, Parasitic/pathology
Collapse
Affiliation(s)
- Francisca Arez
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Ana F. Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (F.A.); (A.F.R.); (C.B.)
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
43
|
Cromwell EF, Leung M, Hammer M, Thai A, Rajendra R, Sirenko O. Disease Modeling with 3D Cell-Based Assays Using a Novel Flowchip System and High-Content Imaging. SLAS Technol 2021; 26:237-248. [PMID: 33783259 DOI: 10.1177/24726303211000688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
There is an increasing interest in using three-dimensional (3D) cell structures for modeling tumors, organs, and tissue to accelerate translational research. We describe here a novel automated organoid assay system (the Pu·MA System) combined with microfluidic-based flowchips that can facilitate 3D cell-based assays. The flowchip is composed of sample wells, which contain organoids, connected to additional multiple wells that can hold various assay reagents. Organoids are positioned in a protected chamber in sample wells, and fluids are exchanged from side reservoirs using pressure-driven flow. Media exchange, sample staining, wash steps, and other processes can be performed without disruption to or loss of 3D sample. The bottom of the sample chamber is thin, optically clear plastic compatible with high-content imaging (HCI). The whole system can be kept in an incubator, allowing long-term cellular assays to be performed. We present two examples of use of the system for biological research. In the first example, cytotoxicity effects of anticancer drugs were evaluated on HeLa and HepG2 spheroids using HCI and vascular endothelial growth factor expression. In the second application, the flowchip system was used for the functional evaluation of Ca2+ oscillations in neurospheroids. Neurospheres were incubated with neuroactive compounds, and neuronal activity was assessed using Ca2+-sensitive dyes and fast kinetic fluorescence imaging. This novel assay system using microfluidics enables automation of 3D cell-based cultures that mimic in vivo conditions, performs multidosing protocols and multiple media exchanges, provides gentle handling of spheroids and organoids, and allows a wide range of assay detection modalities.
Collapse
|
44
|
Livingstone MC, Bitzer AA, Giri A, Luo K, Sankhala RS, Choe M, Zou X, Dennison SM, Li Y, Washington W, Ngauy V, Tomaras GD, Joyce MG, Batchelor AH, Dutta S. In vitro and in vivo inhibition of malaria parasite infection by monoclonal antibodies against Plasmodium falciparum circumsporozoite protein (CSP). Sci Rep 2021; 11:5318. [PMID: 33674699 PMCID: PMC7970865 DOI: 10.1038/s41598-021-84622-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/11/2021] [Indexed: 01/23/2023] Open
Abstract
Plasmodium falciparum malaria contributes to a significant global disease burden. Circumsporozoite protein (CSP), the most abundant sporozoite stage antigen, is a prime vaccine candidate. Inhibitory monoclonal antibodies (mAbs) against CSP map to either a short junctional sequence or the central (NPNA)n repeat region. We compared in vitro and in vivo activities of six CSP-specific mAbs derived from human recipients of a recombinant CSP vaccine RTS,S/AS01 (mAbs 317 and 311); an irradiated whole sporozoite vaccine PfSPZ (mAbs CIS43 and MGG4); or individuals exposed to malaria (mAbs 580 and 663). RTS,S mAb 317 that specifically binds the (NPNA)n epitope, had the highest affinity and it elicited the best sterile protection in mice. The most potent inhibitor of sporozoite invasion in vitro was mAb CIS43 which shows dual-specific binding to the junctional sequence and (NPNA)n. In vivo mouse protection was associated with the mAb reactivity to the NANPx6 peptide, the in vitro inhibition of sporozoite invasion activity, and kinetic parameters measured using intact mAbs or their Fab fragments. Buried surface area between mAb and its target epitope was also associated with in vivo protection. Association and disconnects between in vitro and in vivo readouts has important implications for the design and down-selection of the next generation of CSP based interventions.
Collapse
Affiliation(s)
- Merricka C Livingstone
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Alexis A Bitzer
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Alish Giri
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Kun Luo
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Rajeshwer S Sankhala
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Misook Choe
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Xiaoyan Zou
- Malaria Department, Naval Medical Research Center, Silver Spring, MD, USA
| | - S Moses Dennison
- Center for Human Systems Immunology, Duke University Medical Center, Durham, NC, USA
- Departments of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Yuanzhang Li
- Statistics and Epidemiology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - William Washington
- Statistics and Epidemiology Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Viseth Ngauy
- Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Georgia D Tomaras
- Center for Human Systems Immunology, Duke University Medical Center, Durham, NC, USA
- Departments of Surgery, Duke University Medical Center, Durham, NC, USA
- Departments of Immunology, Duke University Medical Center, Durham, NC, USA
- Departments of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - M Gordon Joyce
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Adrian H Batchelor
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Sheetij Dutta
- Structural Vaccinology Lab, Malaria Biologics Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| |
Collapse
|
45
|
刘 婷, 葛 玉, 袁 敏, 熊 巧, 赵 建. [A review on cell-based models of human liver disease in vitro]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2021; 38:178-184. [PMID: 33899443 PMCID: PMC10307582 DOI: 10.7507/1001-5515.202004027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/31/2020] [Indexed: 11/03/2022]
Abstract
Unhealthy diet, habits and drug abuse cause a variety of liver diseases, including steatohepatitis, liver fibrosis, liver cirrhosis and liver cancer, which seriously affect human health. The fabrication of highly simulated cell models in vitro is important in the treatment of liver diseases and drug development. This article summarized the common strategies for the construction of liver pathology models in vitro. It introduced four typical cell models in vitro related to liver disease and provided a reference for the study of liver disease models.
Collapse
Affiliation(s)
- 婷 刘
- 上海理工大学 医疗器械与食品学院(上海 200093)School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R.China
- 中国科学院 上海微系统与信息技术研究所 传感技术联合国家重点实验室(上海 200050)State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P.R.China
| | - 玉卿 葛
- 中国科学院 上海微系统与信息技术研究所 传感技术联合国家重点实验室(上海 200050)State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P.R.China
| | - 敏 袁
- 上海理工大学 医疗器械与食品学院(上海 200093)School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R.China
| | - 巧 熊
- 海军军医大学附属长海医院 泌尿外科(上海 200433)Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R.China
| | - 建龙 赵
- 中国科学院 上海微系统与信息技术研究所 传感技术联合国家重点实验室(上海 200050)State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P.R.China
| |
Collapse
|
46
|
Tissue Chips and Microphysiological Systems for Disease Modeling and Drug Testing. MICROMACHINES 2021; 12:mi12020139. [PMID: 33525451 PMCID: PMC7911320 DOI: 10.3390/mi12020139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
Tissue chips (TCs) and microphysiological systems (MPSs) that incorporate human cells are novel platforms to model disease and screen drugs and provide an alternative to traditional animal studies. This review highlights the basic definitions of TCs and MPSs, examines four major organs/tissues, identifies critical parameters for organization and function (tissue organization, blood flow, and physical stresses), reviews current microfluidic approaches to recreate tissues, and discusses current shortcomings and future directions for the development and application of these technologies. The organs emphasized are those involved in the metabolism or excretion of drugs (hepatic and renal systems) and organs sensitive to drug toxicity (cardiovascular system). This article examines the microfluidic/microfabrication approaches for each organ individually and identifies specific examples of TCs. This review will provide an excellent starting point for understanding, designing, and constructing novel TCs for possible integration within MPS.
Collapse
|
47
|
Rose S, Ezan F, Cuvellier M, Bruyère A, Legagneux V, Langouët S, Baffet G. Generation of proliferating human adult hepatocytes using optimized 3D culture conditions. Sci Rep 2021; 11:515. [PMID: 33436872 PMCID: PMC7804446 DOI: 10.1038/s41598-020-80019-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/10/2020] [Indexed: 02/08/2023] Open
Abstract
Generating the proliferation of differentiated normal adult human hepatocytes is a major challenge and an expected central step in understanding the microenvironmental conditions that regulate the phenotype of human hepatocytes in vitro. In this work, we described optimized 3D culture conditions of primary human hepatocytes (PHH) to trigger two waves of proliferation and we identified matrix stiffness and cell-cell interactions as the main actors necessary for this proliferation. We demonstrated that DNA replication and overexpression of cell cycle markers are modulate by the matrix stiffness while PHH cultured in 3D without prior cellular interactions did not proliferate. Besides, we showed that PHH carry out an additional cell cycle after transient inhibition of MAPK MER1/2-ERK1/2 signaling pathway. Collagen cultured hepatocytes are organized as characteristic hollow spheroids able to maintain survival, cell polarity and hepatic differentiation for long-term culture periods of at least 28 days. Remarkably, we demonstrated by transcriptomic analysis and functional experiments that proliferating cells are mature hepatocytes with high detoxication capacities. In conclusion, the advanced 3D model described here, named Hepoid, is particularly relevant for obtaining normal human proliferating hepatocytes. By allowing concomitant proliferation and differentiation, it constitutes a promising tool for many pharmacological and biotechnological applications.
Collapse
Affiliation(s)
- Sophie Rose
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Frédéric Ezan
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Marie Cuvellier
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Arnaud Bruyère
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Vincent Legagneux
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France
| | - Sophie Langouët
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France.
| | - Georges Baffet
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, environnement et travail)-UMR_S 1085, 35043, Rennes Cedex, France.
| |
Collapse
|
48
|
Chen AX, Chhabra A, Song HHG, Fleming HE, Chen CS, Bhatia SN. Controlled Apoptosis of Stromal Cells to Engineer Human Microlivers. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910442. [PMID: 33776613 PMCID: PMC7996305 DOI: 10.1002/adfm.201910442] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/29/2020] [Indexed: 05/02/2023]
Abstract
Engineered tissue models comprise a variety of multiplexed ensembles in which combinations of epithelial, stromal, and immune cells give rise to physiologic function. Engineering spatiotemporal control of cell-cell and cell-matrix interactions within these 3D multicellular tissues would represent a significant advance for tissue engineering. In this work, a new method, entitled CAMEO (Controlled Apoptosis in Multicellular tissues for Engineered Organogenesis) enables the non-invasive triggering of controlled apoptosis to eliminate genetically-engineered cells from a pre-established culture. Using this approach, the contribution of stromal cells to the phenotypic stability of primary human hepatocytes is examined. 3D hepatic microtissues, in which fibroblasts can enhance phenotypic stability and accelerate aggregation into spheroids, were found to rely only transiently on fibroblast interaction to support multiple axes of liver function, such as protein secretion and drug detoxification. Due to its modularity, CAMEO has the promise to be readily extendable to other applications that are tied to the complexity of 3D tissue biology, from understanding in vitro organoid models to building artificial tissue grafts.
Collapse
Affiliation(s)
- Amanda X Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arnav Chhabra
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - H-H Greco Song
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Heather E Fleming
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
49
|
Sun P, Zhang G, Su X, Jin C, Yu B, Yu X, Lv Z, Ma H, Zhang M, Wei W, Li W. Maintenance of Primary Hepatocyte Functions In Vitro by Inhibiting Mechanical Tension-Induced YAP Activation. Cell Rep 2020; 29:3212-3222.e4. [PMID: 31801084 DOI: 10.1016/j.celrep.2019.10.128] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/30/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocytes are the primary functional cells of the liver, performing its metabolic, detoxification, and endocrine functions. Functional hepatocytes are extremely valuable in drug discovery and evaluation, as well as in cell therapy for liver diseases. However, it has been a long-standing challenge to maintain the functions of hepatocytes in vitro. Even freshly isolated hepatocytes lose essential functions after short-term culture for reasons that are still not well understood. In the present study, we find that mechanical tension-induced yes-associated protein activation triggers hepatocyte dedifferentiation. Alleviation of mechanical tension by confining cell spreading is sufficient to inhibit hepatocyte dedifferentiation. Based on this finding, we identify a small molecular cocktail through reiterative chemical screening that can maintain hepatocyte functions over the long term and in vivo repopulation capacity by targeting actin polymerization and actomyosin contraction. Our work reveals the mechanisms underlying hepatocyte dedifferentiation and establishes feasible approaches to maintain hepatocyte functions.
Collapse
Affiliation(s)
- Pingxin Sun
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Guanyu Zhang
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Xiaohui Su
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China; Stem Cell and Regenerative Medicine Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Caixia Jin
- Department of Regenerative Medicine, College of Medicine, Tongji University, Shanghai 200433, China
| | - Bing Yu
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Xinlu Yu
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Zhuman Lv
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Haoxin Ma
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Mingliang Zhang
- Department of Histoembryology, Genetics, and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wanguo Wei
- Stem Cell and Regenerative Medicine Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Wenlin Li
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China; Department of Regenerative Medicine, College of Medicine, Tongji University, Shanghai 200433, China; Shanghai Key Laboratory of Cell Engineering, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
50
|
Henslee EA, Dunlop CM, de Mel CM, Carter EA, Abdallat RG, Camelliti P, Labeed FH. DEP-Dots for 3D cell culture: low-cost, high-repeatability, effective 3D cell culture in multiple gel systems. Sci Rep 2020; 10:14603. [PMID: 32884022 PMCID: PMC7471335 DOI: 10.1038/s41598-020-71265-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 07/13/2020] [Indexed: 11/16/2022] Open
Abstract
It is known that cells grown in 3D are more tolerant to drug treatment than those grown in dispersion, but the mechanism for this is still not clear; cells grown in 3D have opportunities to develop inter-cell communication, but are also closely packed which may impede diffusion. In this study we examine methods for dielectrophoresis-based cell aggregation of both suspension and adherent cell lines, and compare the effect of various drugs on cells grown in 3D and 2D. Comparing viability of pharmacological interventions on 3D cell clusters against both suspension cells and adherent cells grown in monolayer, as well as against a unicellular organism with no propensity for intracellular communication, we suggest that 3D aggregates of adherent cells, compared to suspension cells, show a substantially different drug response to cells grown in monolayer, which increases as the IC50 is approached. Further, a mathematical model of the system for each agent demonstrates that changes to drug response are due to inherent changes in the system of adherent cells from the 2D to 3D state. Finally, differences in the electrophysiological membrane properties of the adherent cell type suggest this parameter plays an important role in the differences found in the 3D drug response.
Collapse
Affiliation(s)
- Erin A Henslee
- Centre for Biomedical Engineering, Department of Mechanical Engineering Sciences, University of Surrey, Guildford, GU2 7XH, Surrey, UK.,Department of Engineering, Wake Forest University, Wake Downtown, Winston-Salem, NC, 27109, USA
| | - Carina M Dunlop
- Department of Mathematics, University of Surrey, Guildford, GU2 7XH, Surrey, UK
| | - Christine M de Mel
- Centre for Biomedical Engineering, Department of Mechanical Engineering Sciences, University of Surrey, Guildford, GU2 7XH, Surrey, UK
| | - Emily A Carter
- Centre for Biomedical Engineering, Department of Mechanical Engineering Sciences, University of Surrey, Guildford, GU2 7XH, Surrey, UK
| | - Rula G Abdallat
- Centre for Biomedical Engineering, Department of Mechanical Engineering Sciences, University of Surrey, Guildford, GU2 7XH, Surrey, UK.,Department of Biomedical Engineering, Faculty of Engineering, The Hashemite University, PO Box 330127, Zarqa, 13133, Jordan
| | - Patrizia Camelliti
- School of Biosciences and Medicine, University of Surrey, Guildford, GU2 7XH, Surrey, UK
| | - Fatima H Labeed
- Centre for Biomedical Engineering, Department of Mechanical Engineering Sciences, University of Surrey, Guildford, GU2 7XH, Surrey, UK.
| |
Collapse
|