151
|
Jejunum-derived NF-κB reporter organoids as 3D models for the study of TNF-alpha-induced inflammation. Sci Rep 2022; 12:14425. [PMID: 36002565 PMCID: PMC9829906 DOI: 10.1038/s41598-022-18556-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 08/16/2022] [Indexed: 01/10/2023] Open
Abstract
Inflammation is an important process for epithelial barrier protection but when uncontrolled, it can also lead to tissue damage. The nuclear factor-kappa light chain enhancer of activated B cells (NF-κB) signaling pathway is particularly relevant in the intestine, as it seems to play a dual role. Whereas NF-κB protects intestinal epithelium against various noxious stimuli, the same pathway mediates intestinal inflammatory diseases by inducing pro-inflammatory gene expression. The availability of appropriate in vitro models of the intestinal epithelium is crucial for further understanding the contribution of NF-κB in physiological and pathological processes and advancing in the development of drugs and therapies against gut diseases. Here we established, characterized, and validated three-dimensional cultures of intestinal organoids obtained from biopsies of NF-κB-RE-Luc mice. The NF-κB-RE-Luc intestinal organoids derived from different intestine regions recreated the cellular composition of the tissue and showed a reporter responsiveness similar to the in vivo murine model. When stimulated with TNF-α, jejunum-derived NF-κB-RE-Luc-reporter organoids, provided a useful model to evaluate the anti-inflammatory effects of natural and synthetic compounds. These reporter organoids are valuable tools to explore the epithelial TNF-α-induced NF-κB contribution in the small intestine, being a reliable alternative method while helping to reduce the use of laboratory animals for experimentation.
Collapse
|
152
|
Luo H, Li M, Wang F, Yang Y, Wang Q, Zhao Y, Du F, Chen Y, Shen J, Zhao Q, Zeng J, Wang S, Chen M, Li X, Li W, Sun Y, Gu L, Wen Q, Xiao Z, Wu X. The role of intestinal stem cell within gut homeostasis: Focusing on its interplay with gut microbiota and the regulating pathways. Int J Biol Sci 2022; 18:5185-5206. [PMID: 35982910 PMCID: PMC9379405 DOI: 10.7150/ijbs.72600] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/29/2022] [Indexed: 12/05/2022] Open
Abstract
Intestinal stem cells (ISCs) play an important role in maintaining intestinal homeostasis via promoting a healthy gut barrier. Within the stem cell niche, gut microbiota linking the crosstalk of dietary influence and host response has been identified as a key regulator of ISCs. Emerging insights from recent research reveal that ISC and gut microbiota interplay regulates epithelial self-renewal. This article reviews the recent knowledge on the key role of ISC in their local environment (stem cell niche) associating with gut microbiota and their metabolites as well as the signaling pathways. The current progress of intestinal organoid culture is further summarized. Subsequently, the key challenges and future directions are discussed.
Collapse
Affiliation(s)
- Haoming Luo
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Fang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Yifei Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Qin Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou 646000, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou 646000, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou 646000, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China.,South Sichuan Institute of Translational Medicine, Luzhou 646000, Sichuan, China
| | - Qianyun Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Jiuping Zeng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, Sichuan, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Meijuan Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Yuhong Sun
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Li Gu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Qinglian Wen
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Zhangang Xiao
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China.,Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
153
|
Guo Y, Raev S, Kick MK, Raque M, Saif LJ, Vlasova AN. Rotavirus C Replication in Porcine Intestinal Enteroids Reveals Roles for Cellular Cholesterol and Sialic Acids. Viruses 2022; 14:v14081825. [PMID: 36016447 PMCID: PMC9416568 DOI: 10.3390/v14081825] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/14/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022] Open
Abstract
Rotaviruses (RVs) are a significant cause of severe diarrheal illness in infants and young animals, including pigs. Group C rotavirus (RVC) is an emerging pathogen increasingly reported in pigs and humans worldwide, and is currently recognized as the major cause of gastroenteritis in neonatal piglets that results in substantial economic losses to the pork industry. However, little is known about RVC pathogenesis due to the lack of a robust cell culture system, with the exception of the RVC Cowden strain. Here, we evaluated the permissiveness of porcine crypt-derived 3D and 2D intestinal enteroid (PIE) culture systems for RVC infection. Differentiated 3D and 2D PIEs were infected with porcine RVC (PRVC) Cowden G1P[1], PRVC104 G3P[18], and PRVC143 G6P[5] virulent strains, and the virus replication was measured by qRT-PCR. Our results demonstrated that all RVC strains replicated in 2D-PIEs poorly, while 3D-PIEs supported a higher level of replication, suggesting that RVC selectively infects terminally differentiated enterocytes, which were less abundant in the 2D vs. 3D PIE cultures. While cellular receptors for RVC are unknown, target cell surface carbohydrates, including histo-blood-group antigens (HBGAs) and sialic acids (SAs), are believed to play a role in cell attachment/entry. The evaluation of the selective binding of RVCs to different HBGAs revealed that PRVC Cowden G1P[1] replicated to the highest titers in the HBGA-A PIEs, while PRVC104 or PRVC143 achieved the highest titers in the HBGA-H PIEs. Further, contrasting outcomes were observed following sialidase treatment (resulting in terminal SA removal), which significantly enhanced Cowden and RVC143 replication, but inhibited the growth of PRVC104. These observations suggest that different RVC strains may recognize terminal (PRVC104) as well as internal (Cowden and RVC143) SAs on gangliosides. Finally, several cell culture additives, such as diethylaminoethyl (DEAE)-dextran, cholesterol, and bile extract, were tested to establish if they could enhance RVC replication. We observed that only DEAE-dextran significantly enhanced RVC attachment, but it had no effect on RVC replication. Additionally, the depletion of cellular cholesterol by MβCD inhibited Cowden replication, while the restoration of the cellular cholesterol partially reversed the MβCD effects. These results suggest that cellular cholesterol plays an important role in the replication of the PRVC strain tested. Overall, our study has established a novel robust and physiologically relevant system to investigate RVC pathogenesis. We also generated novel, experimentally derived evidence regarding the role of host glycans, DEAE, and cholesterol in RVC replication, which is critical for the development of control strategies.
Collapse
Affiliation(s)
- Yusheng Guo
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Sergei Raev
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Maryssa K. Kick
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Molly Raque
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Linda J. Saif
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Ohio Agricultural Research and Development Center, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Wooster, OH 44691, USA
- Correspondence:
| |
Collapse
|
154
|
Villablanca EJ, Selin K, Hedin CRH. Mechanisms of mucosal healing: treating inflammatory bowel disease without immunosuppression? NATURE REVIEWS. GASTROENTEROLOGY & HEPATOLOGY 2022. [PMID: 35440774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
Almost all currently available treatments for inflammatory bowel disease (IBD) act by inhibiting inflammation, often blocking specific inflammatory molecules. However, given the infectious and neoplastic disease burden associated with chronic immunosuppressive therapy, the goal of attaining mucosal healing without immunosuppression is attractive. The absence of treatments that directly promote mucosal healing and regeneration in IBD could be linked to the lack of understanding of the underlying pathways. The range of potential strategies to achieve mucosal healing is diverse. However, the targeting of regenerative mechanisms has not yet been achieved for IBD. Stem cells provide hope as a regenerative treatment and are used in limited clinical situations. Growth factors are available for the treatment of short bowel syndrome but have not yet been applied in IBD. The therapeutic application of organoid culture and stem cell therapy to generate new intestinal tissue could provide a novel mechanism to restore barrier function in IBD. Furthermore, blocking key effectors of barrier dysfunction (such as MLCK or damage-associated molecular pattern molecules) has shown promise in experimental IBD. Here, we review the diversity of molecular targets available to directly promote mucosal healing, experimental models to identify new potential pathways and some of the anticipated potential therapies for IBD.
Collapse
Affiliation(s)
- Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| | - Katja Selin
- Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte R H Hedin
- Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden. .,Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
155
|
Villablanca EJ, Selin K, Hedin CRH. Mechanisms of mucosal healing: treating inflammatory bowel disease without immunosuppression? Nat Rev Gastroenterol Hepatol 2022; 19:493-507. [PMID: 35440774 DOI: 10.1038/s41575-022-00604-y] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
Almost all currently available treatments for inflammatory bowel disease (IBD) act by inhibiting inflammation, often blocking specific inflammatory molecules. However, given the infectious and neoplastic disease burden associated with chronic immunosuppressive therapy, the goal of attaining mucosal healing without immunosuppression is attractive. The absence of treatments that directly promote mucosal healing and regeneration in IBD could be linked to the lack of understanding of the underlying pathways. The range of potential strategies to achieve mucosal healing is diverse. However, the targeting of regenerative mechanisms has not yet been achieved for IBD. Stem cells provide hope as a regenerative treatment and are used in limited clinical situations. Growth factors are available for the treatment of short bowel syndrome but have not yet been applied in IBD. The therapeutic application of organoid culture and stem cell therapy to generate new intestinal tissue could provide a novel mechanism to restore barrier function in IBD. Furthermore, blocking key effectors of barrier dysfunction (such as MLCK or damage-associated molecular pattern molecules) has shown promise in experimental IBD. Here, we review the diversity of molecular targets available to directly promote mucosal healing, experimental models to identify new potential pathways and some of the anticipated potential therapies for IBD.
Collapse
Affiliation(s)
- Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| | - Katja Selin
- Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte R H Hedin
- Gastroenterology unit, Department of Gastroenterology, Dermatovenereology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden. .,Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
156
|
Smith EM, Grassel CL, Papadimas A, Foulke-Abel J, Barry EM. The role of CFA/I in adherence and toxin delivery by ETEC expressing multiple colonization factors in the human enteroid model. PLoS Negl Trop Dis 2022; 16:e0010638. [PMID: 35881640 PMCID: PMC9355178 DOI: 10.1371/journal.pntd.0010638] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/05/2022] [Accepted: 07/07/2022] [Indexed: 01/10/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a primary causative agent of diarrhea in travelers and young children in low-to-middle-income countries (LMICs). ETEC adhere to intestinal epithelia via colonization factors (CFs) and secrete heat-stable toxin (ST) and/or heat-labile toxin (LT), causing dysregulated cellular ion transport and water secretion. ETEC isolates often harbor genes encoding more than one CF that are targets as vaccine antigens. CFA/I is a major CF that is associated with ETEC that causes moderate-to-severe diarrhea and plays an important role in pathogenesis. The Global Enteric Multicenter Study finding that 78% of CFA/I-expressing ETEC also encode the minor CF CS21 prompted investigation of the combined role of these two CFs. Western blots and electron microscopy demonstrated growth media-dependent and strain-dependent differences in CFA/I and CS21 expression. The critical role of CFA/I in adherence by ETEC strains expressing CFA/I and CS21 was demonstrated using the human enteroid model and a series of CFA/I- and CS21-specific mutants. Furthermore, only anti-CFA/I antibodies inhibited adherence by global ETEC isolates expressing CFA/I and CS21. Delivery of ST and resulting cGMP secretion was measured in supernatants from infected enteroid monolayers, and strain-specific ST delivery and time-dependent cGMP production was observed. Interestingly, cGMP levels were similar across wildtype and CF-deficient strains, reflecting a limitation of this static aerobic infection model. Despite adherence by ETEC and delivery of ST, the enteroid monolayer integrity was not disrupted, as shown by the lack of decrease in transepithelial electrical resistance and the lack of IL-8 cytokines produced during infection. Taken together, these data demonstrate that targeting CFA/I in global clinical CFA/I-CS21 strains is sufficient for adherence inhibition, supporting a vaccine strategy that focuses on blocking major CFs. In addition, the human enteroid model has significant utility for the study of ETEC pathogenesis and evaluation of vaccine-induced functional antibody responses.
Collapse
Affiliation(s)
- Emily M. Smith
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Christen L. Grassel
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Antonia Papadimas
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Jennifer Foulke-Abel
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Eileen M. Barry
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
157
|
Lyu Y, Chen Y. Digested Human Colostrum Reduces Interleukin-8 Production in Induced Human Intestinal Epithelial Cells. Nutrients 2022; 14:nu14142787. [PMID: 35889744 PMCID: PMC9324903 DOI: 10.3390/nu14142787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/23/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Little is known about the impact of human colostrum on infant intestinal health following digestion. The aim of this study was to compare the effect of digested versus undigested human colostrum on inflammation and cytotoxicity in human intestinal epithelial cells (Caco2BBe) stimulated with lipopolysaccharides (LPS) or tumor necrosis factor (TNF). Colostrum samples (days 2–8 postpartum) from ten mothers of preterm infant were applied. Caco2BBe cells were pretreated by digested or undigested colostrum before stimulation with LPS or TNF. The inflammatory response was determined by measuring the production of interleukin-8 (IL-8) from cells using enzyme linked immunosorbent assay (ELISA). Cytotoxicity was examined by measuring the release of lactate dehydrogenase (LDH) from the cells. Digested colostrum significantly reduced IL-8 production under LPS and TNF stimulation compared with undigested colostrum. Individual colostrum samples exhibited wide variance in the ability to suppress IL-8 production and cytotoxicity in Caco2BBe cells. In vitro-digested human colostrum suppressed an inflammatory response more than undigested human colostrum in an induced intestinal cell culture model.
Collapse
|
158
|
Oost MJ, Ijaz A, van Haarlem DA, van Summeren K, Velkers FC, Kraneveld AD, Venema K, Jansen CA, Pieters RHH, Ten Klooster JP. Chicken-derived RSPO1 and WNT3 contribute to maintaining longevity of chicken intestinal organoid cultures. Sci Rep 2022; 12:10563. [PMID: 35732901 PMCID: PMC9217957 DOI: 10.1038/s41598-022-14875-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/14/2022] [Indexed: 11/09/2022] Open
Abstract
Intestinal organoids are advanced cellular models, which are widely used in mammalian studies to mimic and study in vivo intestinal function and host–pathogen interactions. Growth factors WNT3 and RSPO1 are crucial for the growth of intestinal organoids. Chicken intestinal organoids are currently cultured with mammalian Wnt3a and Rspo1, however, maintaining their longevity has shown to be challenging. Based on the limited homology between mammalian and avian RSPO1, we expect that chicken-derived factors are required for the organoid cultures. Isolated crypts from embryonic tissue of laying hens were growing in the presence of chicken WNT3 and RSPO1, whereas growth in the presence of mammalian Wnt3a and Rspo1 was limited. Moreover, the growth was increased by using Prostaglandin E2 (PGE2) and a Forkhead box O1-inhibitor (FOXO1-inhibitor), allowing to culture these organoids for 15 passages. Furthermore, stem cells maintained their ability to differentiate into goblets, enterocytes and enteroendocrine cells in 2D structures. Overall, we show that chicken intestinal organoids can be cultured for multiple passages using chicken-derived WNT3 and RSPO1, PGE2, and FOXO1-inhibitor.
Collapse
Affiliation(s)
- Miriam J Oost
- Centre for Healthy Eating and Food Innovation, Faculty of Science and Engineering, Maastricht University, Campus Venlo, Venlo, The Netherlands
| | - Adil Ijaz
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Daphne A van Haarlem
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Kitty van Summeren
- Innovative Testing in Life Sciences and Chemistry, Research Centre Healthy and Sustainable Living, University of Applied Sciences Utrecht, Utrecht, The Netherlands
| | - Francisca C Velkers
- Division Farm Animal Health, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Koen Venema
- Centre for Healthy Eating and Food Innovation, Faculty of Science and Engineering, Maastricht University, Campus Venlo, Venlo, The Netherlands
| | - Christine A Jansen
- Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.,Department of Animal Sciences, Cell Biology and Immunology Group, Wageningen University & Research, Wageningen, The Netherlands
| | - Raymond H H Pieters
- Innovative Testing in Life Sciences and Chemistry, Research Centre Healthy and Sustainable Living, University of Applied Sciences Utrecht, Utrecht, The Netherlands.,Department of Population Health Sciences, Institute for Risk Assessment Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jean Paul Ten Klooster
- Innovative Testing in Life Sciences and Chemistry, Research Centre Healthy and Sustainable Living, University of Applied Sciences Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
159
|
Canet-Jourdan C, Pagès DL, Nguyen-Vigouroux C, Cartry J, Zajac O, Desterke C, Lopez JB, Gutierrez-Mateyron E, Signolle N, Adam J, Raingeaud J, Polrot M, Gonin P, Mathieu JRR, Souquere S, Pierron G, Gelli M, Dartigues P, Ducreux M, Barresi V, Jaulin F. Patient-derived organoids identify an apico-basolateral polarity switch associated with survival in colorectal cancer. J Cell Sci 2022; 135:276070. [PMID: 35703098 DOI: 10.1242/jcs.259256] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
The metastatic progression of cancer remains a major issue in patient treatment. Yet, the molecular and cellular mechanisms underlying this process remains unclear. Here, we use primary explants and organoids from patients harboring mucinous colorectal carcinoma (MUC CRC), a poor prognosis histological form of digestive cancers, to study the architecture, invasive behavior and chemoresistance of tumor cell intermediates. We report that these tumors maintain a robust apico-basolateral polarity as they spread in the peritumoral stroma or organotypic collagen-I gels. We identified two distinct topologies: MUC CRCs either display a conventional "apical-in" polarity or, more frequently, harbor an inverted "apical-out" topology. Transcriptomic analyses combined with interference experiments on organoids showed that TGFb and focal adhesion signaling pathways are the main drivers of polarity orientation. Finally, this apical-out topology is associated with increased resistance to chemotherapeutic treatments in organoids and decreased patient survival in the clinic. Thus, patient-derived organoids have the potential to bridge histological, cellular and molecular analyses to decrypt onco-morphogenic programs and stratify cancer patients.
Collapse
Affiliation(s)
| | | | | | - Jérôme Cartry
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| | - Olivier Zajac
- Institut Curie, PSL Research University, CNRS UMR 144, F-75005 Paris, France
| | | | | | | | - Nicolas Signolle
- INSERM Unit U981, Experimental Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Julien Adam
- INSERM Unit U981, Experimental Pathology, Gustave Roussy, 94805 Villejuif, France
| | - Joel Raingeaud
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| | - Mélanie Polrot
- Plateforme d'Evaluation Préclinique, AMMICA UMS 3655/ US 23, Gustave Roussy, Villejuif, F-94805, France
| | - Patrick Gonin
- Plateforme d'Evaluation Préclinique, AMMICA UMS 3655/ US 23, Gustave Roussy, Villejuif, F-94805, France
| | | | | | | | - Maximiliano Gelli
- Department of Medical Oncology, Gustave Roussy, Villejuif, F-94805, France
| | - Peggy Dartigues
- Pathology Department, Gustave Roussy, Villejuif, F-94805, France
| | - Michel Ducreux
- Department of Medical Oncology, Gustave Roussy, Villejuif, F-94805, France.,Paris-Saclay University, Saint-Aubin, F-91190, France
| | - Valeria Barresi
- Department of Diagnostics and Public Health, University of Verona, Verona 37129, Italia
| | - Fanny Jaulin
- INSERM U-1279, Gustave Roussy, Villejuif, F-94805, France
| |
Collapse
|
160
|
Hao M, Cao Z, Wang Z, Xin J, Kong B, Xu J, Zhang L, Chen P. Patient-Derived Organoid Model in the Prediction of Chemotherapeutic Drug Response in Colorectal Cancer. ACS Biomater Sci Eng 2022; 8:3515-3525. [PMID: 35696669 DOI: 10.1021/acsbiomaterials.2c00354] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
As an emerging technology in precision medicine, the patient-derived organoid (PDO) technology has been indicated to provide novel modalities to judge the sensitivity of individual tumors to cancer drugs. In this work, an in vitro model of colorectal cancer (CRC) was established using the PDO culture, and it is demonstrated that the PDO samples preserved, to a great extent, the histologic features and marker expression of the original tumor tissues. Subsequently, cancer drugs 5-FU, oxaliplatin, and irinotecan were selected and screened on five CRC PDO samples, while the patient-derived organoid xenograft (PDOX) model was applied for comparison. The receiver operating characteristic (ROC) curve was drawn according to the IC50 data from the PDO model and the relative tumor proliferation rate (T/C%) from PDOX. Interestingly, the area under the ROC curve was 0.84 (95% CI, 0.64-1.04, P value = 0.028), which suggested that the IC50 of cancer drugs from the PDO model was strongly correlated with PDOX responses. In addition, the optimal sensitivity cutoff value for drug screening in CRC PDOs was identified at 10.35 μM, which could act as a reference value for efficacy evaluation of 5-FU, oxaliplatin, and irinotecan in the colorectal cancer drug screening. Since there are no unified criteria to judge the sensitivity of drugs in vitro, our work provides a method for establishing in vitro evaluation criteria via PDO and PDOX model using the patient tissues received from local hospitals, exhibiting potential in clinical cancer therapy and precision medicine.
Collapse
Affiliation(s)
- Minglu Hao
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China
| | - Zhipeng Cao
- NanoPeptide (Qingdao) Biotechnology Ltd., Qingdao 266100, China
| | - Zhiwei Wang
- The Affiliated Qingdao Central Hospital, Qingdao University, Qingdao 266000, China.,Qingdao Central Hospital, Qingdao 266042, China
| | - Jianjun Xin
- The Affiliated Qingdao Central Hospital, Qingdao University, Qingdao 266000, China.,Qingdao Central Hospital, Qingdao 266042, China
| | - Biao Kong
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai 200438, China
| | - Jing Xu
- The Affiliated Qingdao Central Hospital, Qingdao University, Qingdao 266000, China.,Qingdao Central Hospital, Qingdao 266042, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Pu Chen
- Advanced Materials Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250014, China.,Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
161
|
Analysis of mouse intestinal organoid culture with conditioned media isolated from mucosal enteric glial cells. STAR Protoc 2022; 3:101351. [PMID: 35516847 PMCID: PMC9062428 DOI: 10.1016/j.xpro.2022.101351] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
This protocol describes the isolation and culture of 3D intestinal crypt organoids with stromal niche cells. We show a murine organoid culture system that utilizes conditioned media isolated from primary, mucosal enteric glial cell culture. We describe three assays of analyzing this organoid culture: flow cytometry, gene expression, and organoid morphology analyses. This protocol can also be used to study the mechanisms of stem cell interaction with other stromal niche cell types such as mesenchymal cells and innate immune cells. For complete details on the use and execution of this protocol, please refer to Baghdadi et al. (2021). Organoid protocol to study intestinal stem cell interactions with mucosal glial cells Optimized protocol to isolate mucosal glial cells and collect their conditioned media Efficient analysis of organoid morphology, gene expression and cellular composition
Collapse
|
162
|
Pahapale GJ, Tao J, Nikolic M, Gao S, Scarcelli G, Sun SX, Romer LH, Gracias DH. Directing Multicellular Organization by Varying the Aspect Ratio of Soft Hydrogel Microwells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104649. [PMID: 35434926 PMCID: PMC9189654 DOI: 10.1002/advs.202104649] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/08/2022] [Indexed: 06/03/2023]
Abstract
Multicellular organization with precise spatial definition is essential to various biological processes, including morphogenesis, development, and healing in vascular and other tissues. Gradients and patterns of chemoattractants are well-described guides of multicellular organization, but the influences of 3D geometry of soft hydrogels are less well defined. Here, the discovery of a new mode of endothelial cell self-organization guided by combinatorial effects of stiffness and geometry, independent of protein or chemical patterning, is described. Endothelial cells in 2 kPa microwells are found to be ≈30 times more likely to migrate to the edge to organize in ring-like patterns than in stiff 35 kPa microwells. This organization is independent of curvature and significantly more pronounced in 2 kPa microwells with aspect ratio (perimeter/depth) < 25. Physical factors of cells and substrates that drive this behavior are systematically investigated and a mathematical model that explains the organization by balancing the dynamic interaction between tangential cytoskeletal tension, cell-cell, and cell-substrate adhesion is presented. These findings demonstrate the importance of combinatorial effects of geometry and stiffness in complex cellular organization that can be leveraged to facilitate the engineering of bionics and integrated model organoid systems with customized nutrient vascular networks.
Collapse
Affiliation(s)
- Gayatri J. Pahapale
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jiaxiang Tao
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Milos Nikolic
- Maryland Biophysics ProgramInstitute for Physical Science and TechnologyUniversity of MarylandCollege ParkMD20742USA
| | - Sammy Gao
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Giuliano Scarcelli
- Maryland Biophysics ProgramInstitute for Physical Science and Technology and Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Sean X. Sun
- Department of Mechanical EngineeringCell Biologyand Institute of NanoBioTechnology (INBT)Johns Hopkins UniversityBaltimoreMD21218USA
| | - Lewis H. Romer
- Department of Cell BiologyAnesthesiology and Critical Care MedicineBiomedical EngineeringPediatricsand Center for Cell DynamicsJohns Hopkins School of MedicineBaltimoreMD21205USA
| | - David H. Gracias
- Department of Chemical and Biomolecular EngineeringMaterials Science and EngineeringChemistry and Laboratory for Computational Sensing and Robotics (LCSR)Johns Hopkins UniversityBaltimoreMD21218USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins School of MedicineBaltimoreMD21205USA
| |
Collapse
|
163
|
Ma P, Fang P, Ren T, Fang L, Xiao S. Porcine Intestinal Organoids: Overview of the State of the Art. Viruses 2022; 14:1110. [PMID: 35632851 PMCID: PMC9147602 DOI: 10.3390/v14051110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 02/01/2023] Open
Abstract
The intestinal tract is a crucial part of the body for growth and development, and its dysregulation can cause several diseases. The lack of appropriate in vitro models hampers the development of effective preventions and treatments against these intestinal tract diseases. Intestinal organoids are three-dimensional (3D) polarized structures composed of different types of cells capable of self-organization and self-renewal, resembling their organ of origin in architecture and function. Porcine intestinal organoids (PIOs) have been cultured and are used widely in agricultural, veterinary, and biomedical research. Based on the similarity of the genomic sequence, anatomic morphology, and drug metabolism with humans and the difficulty in obtaining healthy human tissue, PIOs are also considered ideal models relative to rodents. In this review, we summarize the current knowledge on PIOs, emphasizing their culturing, establishment and development, and applications in the study of host-microbe interactions, nutritional development, drug discovery, and gene editing potential.
Collapse
Affiliation(s)
- Panpan Ma
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (P.M.); (T.R.); (L.F.); (S.X.)
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Puxian Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (P.M.); (T.R.); (L.F.); (S.X.)
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Tianze Ren
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (P.M.); (T.R.); (L.F.); (S.X.)
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Liurong Fang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (P.M.); (T.R.); (L.F.); (S.X.)
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| | - Shaobo Xiao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (P.M.); (T.R.); (L.F.); (S.X.)
- The Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Cooperative Innovation Center for Sustainable Pig Production, Wuhan 430070, China
| |
Collapse
|
164
|
Jacquemin G, Wurmser A, Huyghe M, Sun W, Homayed Z, Merle C, Perkins M, Qasrawi F, Richon S, Dingli F, Arras G, Loew D, Vignjevic D, Pannequin J, Fre S. Paracrine signalling between intestinal epithelial and tumour cells induces a regenerative programme. eLife 2022; 11:e76541. [PMID: 35543624 PMCID: PMC9094746 DOI: 10.7554/elife.76541] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
Tumours are complex ecosystems composed of different types of cells that communicate and influence each other. While the critical role of stromal cells in affecting tumour growth is well established, the impact of mutant cancer cells on healthy surrounding tissues remains poorly defined. Here, using mouse intestinal organoids, we uncover a paracrine mechanism by which intestinal cancer cells reactivate foetal and regenerative YAP-associated transcriptional programmes in neighbouring wildtype epithelial cells, rendering them adapted to thrive in the tumour context. We identify the glycoprotein thrombospondin-1 (THBS1) as the essential factor that mediates non-cell-autonomous morphological and transcriptional responses. Importantly, Thbs1 is associated with bad prognosis in several human cancers. This study reveals the THBS1-YAP axis as the mechanistic link mediating paracrine interactions between epithelial cells in intestinal tumours.
Collapse
Affiliation(s)
- Guillaume Jacquemin
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215ParisFrance
- Sorbonne University, UPMC University of Paris VIParisFrance
| | - Annabelle Wurmser
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215ParisFrance
| | - Mathilde Huyghe
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215ParisFrance
| | - Wenjie Sun
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215ParisFrance
| | - Zeinab Homayed
- IGF, University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Candice Merle
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215ParisFrance
| | - Meghan Perkins
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215ParisFrance
| | - Fairouz Qasrawi
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215ParisFrance
| | - Sophie Richon
- Institut Curie, PSL Research University, CNRS UMR 144ParisFrance
| | - Florent Dingli
- Institut Curie, PSL Research University, Laboratory of Mass Spectrometry and ProteomicsParisFrance
| | - Guillaume Arras
- Institut Curie, PSL Research University, Laboratory of Mass Spectrometry and ProteomicsParisFrance
| | - Damarys Loew
- Institut Curie, PSL Research University, Laboratory of Mass Spectrometry and ProteomicsParisFrance
| | | | - Julie Pannequin
- IGF, University of Montpellier, CNRS, INSERMMontpellierFrance
| | - Silvia Fre
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215ParisFrance
| |
Collapse
|
165
|
Kakni P, López-Iglesias C, Truckenmüller R, Habibović P, Giselbrecht S. Reversing Epithelial Polarity in Pluripotent Stem Cell-Derived Intestinal Organoids. Front Bioeng Biotechnol 2022; 10:879024. [PMID: 35547177 PMCID: PMC9081652 DOI: 10.3389/fbioe.2022.879024] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/08/2022] [Indexed: 11/29/2022] Open
Abstract
The inner surface of the intestine is a dynamic system, composed of a single layer of polarized epithelial cells. The development of intestinal organoids was a major breakthrough since they robustly recapitulate intestinal architecture, regional specification and cell composition in vitro. However, the cyst-like organization hinders direct access to the apical side of the epithelium, thus limiting their use in functional assays. For the first time, we show an intestinal organoid model from pluripotent stem cells with reversed polarity where the apical side faces the surrounding culture media and the basal side faces the lumen. These inside-out organoids preserve a distinct apico-basolateral orientation for a long period and differentiate into the major intestinal cell types. This novel model lays the foundation for developing new in vitro functional assays particularly targeting the apical surface of the epithelium and thus offers a new research tool to study nutrient/drug uptake, metabolism and host-microbiome/pathogen interactions.
Collapse
Affiliation(s)
- Panagiota Kakni
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Carmen López-Iglesias
- Microscopy CORE Lab, Maastricht Multimodal Molecular Imaging Institute (M4I), Maastricht University, Maastricht, Netherlands
| | - Roman Truckenmüller
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Pamela Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Stefan Giselbrecht
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
- *Correspondence: Stefan Giselbrecht,
| |
Collapse
|
166
|
Wang J, Chen C, Wang L, Xie M, Ge X, Wu S, He Y, Mou X, Ye C, Sun Y. Patient-Derived Tumor Organoids: New Progress and Opportunities to Facilitate Precision Cancer Immunotherapy. Front Oncol 2022; 12:872531. [PMID: 35449581 PMCID: PMC9016336 DOI: 10.3389/fonc.2022.872531] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/14/2022] [Indexed: 12/18/2022] Open
Abstract
Cancer immunotherapy has revolutionized the field of cancer treatment in recent years. However, not all patients receiving cancer immunotherapy exhibit durable responses, and reliable, high-throughput testing platforms are urgently needed to guide personalized cancer immunotherapy. The ability of patient-derived tumor organoids to recapitulate pivotal features of original cancer tissues makes them useful as a preclinical model for cancer research and precision medicine. Nevertheless, many challenges exist in the translation of tumor organoid research to clinical decision making. Herein we discuss the applications of patient-derived tumor organoid models and the advances and potential of using complex immune-organoid systems as testing platforms to facilitate precision cancer immunotherapy. In addition, we highlight intriguing applications of tumor organoids with novel multi-omics in preclinical cancer research, highlighting genetic editing, proteomics, and liquid biopsy.
Collapse
Affiliation(s)
- Ji Wang
- Center for Plastic & Reconstructive Surgery, Department of Plastic & Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Chao Chen
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| | - Lu Wang
- Center for Plastic & Reconstructive Surgery, Department of Plastic & Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Mingjun Xie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, China
| | - Xinyang Ge
- College of Letters and Science, University of California, Los Angeles, Los Angeles, CA, United States
| | - Sufan Wu
- Center for Plastic & Reconstructive Surgery, Department of Plastic & Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Yong He
- Cancer Center, Zhejiang University, Hangzhou, China.,State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, China.,Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, College of Mechanical Engineering, Zhejiang University, Hangzhou, China
| | - Xiaozhou Mou
- Center for Plastic & Reconstructive Surgery, Department of Plastic & Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Chenyang Ye
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China.,Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Sun
- Center for Plastic & Reconstructive Surgery, Department of Plastic & Reconstructive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
167
|
Wu DT, Jeffreys N, Diba M, Mooney DJ. Viscoelastic Biomaterials for Tissue Regeneration. Tissue Eng Part C Methods 2022; 28:289-300. [PMID: 35442107 PMCID: PMC9347380 DOI: 10.1089/ten.tec.2022.0040] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The extracellular matrix (ECM) mechanical properties regulate key cellular processes in tissue development and regeneration. The majority of scientific investigation has focused on ECM elasticity as the primary mechanical regulator of cell and tissue behavior. However, all living tissues are viscoelastic, exhibiting both solid- and liquid-like mechanical behavior. Despite increasing evidence regarding the role of ECM viscoelasticity in directing cellular behavior, this aspect is still largely overlooked in the design of biomaterials for tissue regeneration. Recently, with the emergence of various bottom-up material design strategies, new approaches can deliver unprecedented control over biomaterial properties at multiple length scales, thus enabling the design of viscoelastic biomaterials that mimic various aspect of the native tissue ECM microenvironment. This review describes key considerations for the design of viscoelastic biomaterials for tissue regeneration. We provide an overview of the role of matrix viscoelasticity in directing cell behavior towards regenerative outcomes, highlight recent strategies utilizing viscoelastic hydrogels for regenerative therapies, and outline remaining challenges, potential solutions, and emerging applications for viscoelastic biomaterials in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- David Tiansui Wu
- Harvard University John A Paulson School of Engineering and Applied Sciences, 124077, Cambridge, Massachusetts, United States.,Harvard University Wyss Institute for Biologically Inspired Engineering, 465574, Boston, Massachusetts, United States.,Harvard School of Dental Medicine, 124048, Oral Medicine, Infection, and Immunity, Boston, Massachusetts, United States;
| | - Nicholas Jeffreys
- Harvard University John A Paulson School of Engineering and Applied Sciences, 124077, Cambridge, Massachusetts, United States.,Harvard University Wyss Institute for Biologically Inspired Engineering, 465574, Boston, Massachusetts, United States;
| | - Mani Diba
- Harvard University John A Paulson School of Engineering and Applied Sciences, 124077, Cambridge, Massachusetts, United States;
| | - David J Mooney
- Harvard University John A Paulson School of Engineering and Applied Sciences, 124077, Cambridge, Massachusetts, United States.,Harvard University Wyss Institute for Biologically Inspired Engineering, 465574, Boston, Massachusetts, United States;
| |
Collapse
|
168
|
Extending the viability of human precision-cut intestinal slice model for drug metabolism studies. Arch Toxicol 2022; 96:1815-1827. [PMID: 35428896 PMCID: PMC9095520 DOI: 10.1007/s00204-022-03295-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/24/2022] [Indexed: 11/09/2022]
Abstract
Human Precision-cut intestinal slices (hPCIS) are used to study intestinal physiology, pathophysiology, drug efficacy, toxicology, kinetics, and metabolism. However, the use of this ex vivo model is restricted to approximately a 24 h timeframe because of declining viability of the hPCIS during traditional culture. We hypothesized that we could extend the hPCIS viability by using organoid medium. Therefore, we cultured hPCIS for up to 72 h in organoid media [expansion medium (Emed) and differentiation medium (Dmed)]. After incubation, we assessed culture-induced changes on viability markers, specific cell type markers and we assessed the metabolic activity of enterocytes by measuring midazolam metabolite formation. We show that the adenosine triphosphate (ATP)/protein ratio of Emed-cultured hPCIS and morphology of both Emed- and Dmed-cultured hPCIS was improved compared to WME-cultured hPCIS. Emed-cultured hPCIS showed an increased expression of proliferation and stem cell markers, whereas Dmed-cultured hPCIS showed an increased expression of proliferation and enterocyte markers, along with increased midazolam metabolism. Using the Emed, the viability of hPCIS could be extended for up to 72 h, and proliferating stem cells remained preserved. Using Dmed, hPCS also remained viable for up to 72 h, and specifically rescued the metabolizing enterocytes during culture. In conclusion, by using two different organoid culture media, we could extend the hPCIS viability for up to 72 h of incubation and specifically steer stem cells or enterocytes towards their original function, metabolism, and proliferation, potentially allowing pharmacokinetic and toxicology studies beyond the 24 h timeframe.
Collapse
|
169
|
Wang Z, Zhao S, Lin X, Chen G, Kang J, Ma Z, Wang Y, Li Z, Xiao X, He A, Xiang D. Application of Organoids in Carcinogenesis Modeling and Tumor Vaccination. Front Oncol 2022; 12:855996. [PMID: 35371988 PMCID: PMC8968694 DOI: 10.3389/fonc.2022.855996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Organoids well recapitulate organ-specific functions from their tissue of origin and remain fundamental aspects of organogenesis. Organoids are widely applied in biomedical research, drug discovery, and regenerative medicine. There are various cultivated organoid systems induced by adult stem cells and pluripotent stem cells, or directly derived from primary tissues. Researchers have drawn inspiration by combination of organoid technology and tissue engineering to produce organoids with more physiological relevance and suitable for translational medicine. This review describes the value of applying organoids for tumorigenesis modeling and tumor vaccination. We summarize the application of organoids in tumor precision medicine. Extant challenges that need to be conquered to make this technology be more feasible and precise are discussed.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shasha Zhao
- State Key Laboratory of Oncogenes and Related Genes, the Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaolin Lin
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guanglong Chen
- Department of General Surgery, Zhengzhou University, Affiliated Cancer Hospital (Henan Cancer Hospital), Zhengzhou, China
| | - Jiawei Kang
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | | | - Yiming Wang
- Shanghai OneTar Biomedicine, Shanghai, China
| | - Zhi Li
- Department of General Surgery, Zhengzhou University, Affiliated Cancer Hospital (Henan Cancer Hospital), Zhengzhou, China
| | - Xiuying Xiao
- Department of Oncology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Aina He
- Department of Oncology, The Sixth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dongxi Xiang
- State Key Laboratory of Oncogenes and Related Genes, Department of Biliary-Pancreatic Surgery, The Renji Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
170
|
Chen L, Dai Z, Ge C, Huang D, Zhou X, Pan K, Xu W, Fu J, lin Du J. Specific Metabolic Response of Patient-derived Organoids to Curcumin of Colorectal Cancer. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1203:123260. [DOI: 10.1016/j.jchromb.2022.123260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/17/2022] [Accepted: 04/17/2022] [Indexed: 12/12/2022]
|
171
|
Donà F, Eli S, Mapelli M. Insights Into Mechanisms of Oriented Division From Studies in 3D Cellular Models. Front Cell Dev Biol 2022; 10:847801. [PMID: 35356279 PMCID: PMC8959941 DOI: 10.3389/fcell.2022.847801] [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: 01/03/2022] [Accepted: 02/17/2022] [Indexed: 12/13/2022] Open
Abstract
In multicellular organisms, epithelial cells are key elements of tissue organization. In developing tissues, cellular proliferation and differentiation are under the tight regulation of morphogenetic programs, that ensure the correct organ formation and functioning. In these processes, mitotic rates and division orientation are crucial in regulating the velocity and the timing of the forming tissue. Division orientation, specified by mitotic spindle placement with respect to epithelial apico-basal polarity, controls not only the partitioning of cellular components but also the positioning of the daughter cells within the tissue, and hence the contacts that daughter cells retain with the surrounding microenvironment. Daughter cells positioning is important to determine signal sensing and fate, and therefore the final function of the developing organ. In this review, we will discuss recent discoveries regarding the mechanistics of planar divisions in mammalian epithelial cells, summarizing technologies and model systems used to study oriented cell divisions in vitro such as three-dimensional cysts of immortalized cells and intestinal organoids. We also highlight how misorientation is corrected in vivo and in vitro, and how it might contribute to the onset of pathological conditions.
Collapse
Affiliation(s)
- Federico Donà
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Susanna Eli
- IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | |
Collapse
|
172
|
Chu JN, Traverso G. Foundations of gastrointestinal-based drug delivery and future developments. Nat Rev Gastroenterol Hepatol 2022; 19:219-238. [PMID: 34785786 PMCID: PMC12053541 DOI: 10.1038/s41575-021-00539-w] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/07/2021] [Indexed: 12/12/2022]
Abstract
Gastrointestinal-based drug delivery is considered the preferred mode of drug administration owing to its convenience for patients, which improves adherence. However, unique characteristics of the gastrointestinal tract (such as the digestive environment and constraints on transport across the gastrointestinal mucosa) limit the absorption of drugs. As a result, many medications, in particular biologics, still exist only or predominantly in injectable form. In this Review, we examine the fundamentals of gastrointestinal drug delivery to inform clinicians and pharmaceutical scientists. We discuss general principles, including the challenges that need to be overcome for successful drug formulation, and describe the unique features to consider for each gastrointestinal compartment when designing drug formulations for topical and systemic applications. We then discuss emerging technologies that seek to address remaining obstacles to successful gastrointestinal-based drug delivery.
Collapse
Affiliation(s)
- Jacqueline N Chu
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giovanni Traverso
- Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
173
|
Costa B, Estrada MF, Barroso MT, Fior R. Zebrafish Patient-Derived Avatars from Digestive Cancers for Anti-cancer Therapy Screening. Curr Protoc 2022; 2:e415. [PMID: 35436037 DOI: 10.1002/cpz1.415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Patient-derived xenografts (PDXs), also called "avatars," are generated by the implantation of human primary tumor cells or tissues into a host animal. Given the complexity and unique characteristics of each tumor, PDXs are models of choice in cancer research and precision medicine. In this context, the zebrafish PDX model (zPDX or zAvatar) has been recognized as a promising in vivo model to directly challenge patient cells with anti-cancer therapies in a personalized manner. The assay relies on the injection of tumor cells from patients into zebrafish embryos to then test and identify the best available drug combination for a particular patient. Compared to mouse PDXs, zAvatar assays take less time and do not require in vitro or in vivo cell expansion. The present article describes how to generate zAvatars from resected digestive cancer from surgeries and how to then use them for anti-cancer therapy screening. We describe the steps for tumor sample collection and cryopreservation, sample preparation and fluorescent labeling for microinjection into zebrafish embryos, drug administration, and analysis of tumor behavior by single-cell confocal imaging. We provide detailed protocols and helpful tips for performing this assay, and we address the technical challenges associated with the workflow. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Patient tumor sample collection and cryopreservation Basic Protocol 2: Generation of zAvatars and anti-cancer treatment Basic Protocol 3: Whole-mount immunofluorescence Basic Protocol 4: Confocal imaging and analysis.
Collapse
Affiliation(s)
- Bruna Costa
- Champalimaud Foundation, Champalimaud Research, Lisbon, Portugal
| | - Marta F Estrada
- Champalimaud Foundation, Champalimaud Research, Lisbon, Portugal
| | | | - Rita Fior
- Champalimaud Foundation, Champalimaud Research, Lisbon, Portugal
| |
Collapse
|
174
|
ABSTRACTS (BY NUMBER). Tissue Eng Part A 2022. [DOI: 10.1089/ten.tea.2022.29025.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
175
|
Chrisnandy A, Blondel D, Rezakhani S, Broguiere N, Lutolf MP. Synthetic dynamic hydrogels promote degradation-independent in vitro organogenesis. NATURE MATERIALS 2022; 21:479-487. [PMID: 34782747 DOI: 10.1038/s41563-021-01136-7] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 09/21/2021] [Indexed: 06/13/2023]
Abstract
Epithelial organoids are most efficiently grown from mouse-tumour-derived, reconstituted extracellular matrix hydrogels, whose poorly defined composition, batch-to-batch variability and immunogenicity limit clinical applications. Efforts to replace such ill-defined matrices for organoid culture have largely focused on non-adaptable hydrogels composed of covalently crosslinked hydrophilic macromolecules. However, the excessive forces caused by tissue expansion in such elastic gels severely restrict organoid growth and morphogenesis. Chemical or enzymatic degradation schemes can partially alleviate this problem, but due to their irreversibility, long-term applicability is limited. Here we report a family of synthetic hydrogels that promote extensive organoid morphogenesis through dynamic rearrangements mediated by reversible hydrogen bonding. These tunable matrices are stress relaxing and thus promote efficient crypt budding in intestinal stem-cell epithelia through increased symmetry breaking and Paneth cell formation dependent on yes-associated protein 1. As such, these well-defined gels provide promising versatile matrices for fostering elaborate in vitro morphogenesis.
Collapse
Affiliation(s)
- Antonius Chrisnandy
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Delphine Blondel
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Saba Rezakhani
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nicolas Broguiere
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), EPFL, Lausanne, Switzerland.
- Roche Institute for Translational Bioengineering (ITB), Pharma Research and Early Development (pRED), F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
|
176
|
Donath S, Angerstein L, Gentemann L, Müller D, Seidler AE, Jesinghaus C, Bleich A, Heisterkamp A, Buettner M, Kalies S. Investigation of Colonic Regeneration via Precise Damage Application Using Femtosecond Laser-Based Nanosurgery. Cells 2022; 11:1143. [PMID: 35406708 PMCID: PMC8998079 DOI: 10.3390/cells11071143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 11/23/2022] Open
Abstract
Organoids represent the cellular composition of natural tissue. So called colonoids, organoids derived from colon tissue, are a good model for understanding regeneration. However, next to the cellular composition, the surrounding matrix, the cell-cell interactions, and environmental factors have to be considered. This requires new approaches for the manipulation of a colonoid. Of key interest is the precise application of localized damage and the following cellular reaction. We have established multiphoton imaging in combination with femtosecond laser-based cellular nanosurgery in colonoids to ablate single cells in the colonoids' crypts, the proliferative zones, and the differentiated zones. We observed that half of the colonoids recovered within six hours after manipulation. An invagination of the damaged cell and closing of the structure was observed. In about a third of the cases of targeted crypt damage, it caused a stop in crypt proliferation. In the majority of colonoids ablated in the crypt, the damage led to an increase in Wnt signalling, indicated via a fluorescent lentiviral biosensor. qRT-PCR analysis showed increased expression of various proliferation and Wnt-associated genes in response to damage. Our new model of probing colonoid regeneration paves the way to better understand organoid dynamics on a single cell level.
Collapse
Affiliation(s)
- Sören Donath
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (L.A.); (L.G.); (D.M.); (A.E.S.); (C.J.); (A.H.); (S.K.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
| | - Leon Angerstein
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (L.A.); (L.G.); (D.M.); (A.E.S.); (C.J.); (A.H.); (S.K.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
| | - Lara Gentemann
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (L.A.); (L.G.); (D.M.); (A.E.S.); (C.J.); (A.H.); (S.K.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
| | - Dominik Müller
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (L.A.); (L.G.); (D.M.); (A.E.S.); (C.J.); (A.H.); (S.K.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
- REBIRTH Research Center for Translational Regenerative Medicine, 30625 Hannover, Germany
| | - Anna E. Seidler
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (L.A.); (L.G.); (D.M.); (A.E.S.); (C.J.); (A.H.); (S.K.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
| | - Christian Jesinghaus
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (L.A.); (L.G.); (D.M.); (A.E.S.); (C.J.); (A.H.); (S.K.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
| | - André Bleich
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
- REBIRTH Research Center for Translational Regenerative Medicine, 30625 Hannover, Germany
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Alexander Heisterkamp
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (L.A.); (L.G.); (D.M.); (A.E.S.); (C.J.); (A.H.); (S.K.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
| | - Manuela Buettner
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
- REBIRTH Research Center for Translational Regenerative Medicine, 30625 Hannover, Germany
- Institute for Laboratory Animal Science, Hannover Medical School, 30625 Hannover, Germany
| | - Stefan Kalies
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany; (L.A.); (L.G.); (D.M.); (A.E.S.); (C.J.); (A.H.); (S.K.)
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), 30625 Hannover, Germany; (A.B.); (M.B.)
- REBIRTH Research Center for Translational Regenerative Medicine, 30625 Hannover, Germany
| |
Collapse
|
177
|
Martinelli I, Tayebati SK, Tomassoni D, Nittari G, Roy P, Amenta F. Brain and Retinal Organoids for Disease Modeling: The Importance of In Vitro Blood–Brain and Retinal Barriers Studies. Cells 2022; 11:cells11071120. [PMID: 35406683 PMCID: PMC8997725 DOI: 10.3390/cells11071120] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 11/16/2022] Open
Abstract
Brain and retinal organoids are functional and dynamic in vitro three-dimensional (3D) structures derived from pluripotent stem cells that spontaneously organize themselves to their in vivo counterparts. Here, we review the main literature data of how these organoids have been developed through different protocols and how they have been technically analyzed. Moreover, this paper reviews recent advances in using organoids to model neurological and retinal diseases, considering their potential for translational applications but also pointing out their limitations. Since the blood–brain barrier (BBB) and blood–retinal barrier (BRB) are understood to play a fundamental role respectively in brain and eye functions, both in health and in disease, we provide an overview of the progress in the development techniques of in vitro models as reliable and predictive screening tools for BBB and BRB-penetrating compounds. Furthermore, we propose potential future directions for brain and retinal organoids, in which dedicated biobanks will represent a novel tool for neuroscience and ophthalmology research.
Collapse
Affiliation(s)
- Ilenia Martinelli
- School of Medicinal and Health Products Sciences, University of Camerino, 62032 Camerino, Italy; (S.K.T.); (G.N.); (F.A.)
- Correspondence:
| | - Seyed Khosrow Tayebati
- School of Medicinal and Health Products Sciences, University of Camerino, 62032 Camerino, Italy; (S.K.T.); (G.N.); (F.A.)
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (P.R.)
| | - Giulio Nittari
- School of Medicinal and Health Products Sciences, University of Camerino, 62032 Camerino, Italy; (S.K.T.); (G.N.); (F.A.)
| | - Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy; (D.T.); (P.R.)
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, University of Camerino, 62032 Camerino, Italy; (S.K.T.); (G.N.); (F.A.)
| |
Collapse
|
178
|
Yavitt FM, Kirkpatrick BE, Blatchley MR, Anseth KS. 4D Materials with Photoadaptable Properties Instruct and Enhance Intestinal Organoid Development. ACS Biomater Sci Eng 2022; 8:4634-4638. [PMID: 35298149 DOI: 10.1021/acsbiomaterials.1c01450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intestinal organoids are self-organized tissue constructs, grown in vitro, that resemble the structure and function of the intestine and are often considered promising as a prospective platform for drug testing and disease modeling. Organoid development in vitro is typically instructed by exogenous cues delivered from the media, but cellular responses also depend on properties of the surrounding microenvironmental niche, such as mechanical stiffness and extracellular matrix (ECM) ligands. In recent years, synthetic hydrogel platforms have been engineered to resemble the in vivo niche, with the goal of generating physiologically relevant environments that can promote mature and reproducible organoid development. However, a few of these approaches consider the importance of intestinal organoid morphology or how morphology changes during development, as cues that may dictate organoid functionality. For example, intestinal organoids grown in vitro often lack the physical boundary conditions found in vivo that are responsible for shaping a collection of cells into developmentally relevant morphologies, resulting in organoids that often differ in structure and cellular organization from the parent organ. This disconnect relates, in part, to a lack of appropriate adaptable and programmable materials for cell culture, especially those that enable control over colony growth and differentiation in space and time (i.e., 4D materials). We posit that the future of organoid culture platforms may benefit from advances in photoadaptable chemistries and integration into biomaterials scaffolds, thereby allowing greater user-directed control over both the macro- and microscale material properties. In this way, synthetic materials can begin to better replicate changes in the ECM during development or regeneration in vivo. Recapitulation of cellular and tissue morphological changes, along with an appreciation for the appropriate developmental time scales, should help instruct the next generation of organoid models to facilitate predictable outcomes.
Collapse
Affiliation(s)
| | - Bruce E Kirkpatrick
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | | | | |
Collapse
|
179
|
Luo L, Ma Y, Zheng Y, Su J, Huang G. Application Progress of Organoids in Colorectal Cancer. Front Cell Dev Biol 2022; 10:815067. [PMID: 35273961 PMCID: PMC8902504 DOI: 10.3389/fcell.2022.815067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/31/2022] [Indexed: 12/24/2022] Open
Abstract
Currently, colorectal cancer is still the third leading cause of cancer-related mortality, and the incidence is rising. It is a long time since the researchers used cancer cell lines and animals as the study subject. However, these models possess various limitations to reflect the cancer progression in the human body. Organoids have more clinical significance than cell lines, and they also bridge the gap between animal models and humans. Patient-derived organoids are three-dimensional cultures that simulate the tumor characteristics in vivo and recapitulate tumor cell heterogeneity. Therefore, the emergence of colorectal cancer organoids provides an unprecedented opportunity for colorectal cancer research. It retains the molecular and cellular composition of the original tumor and has a high degree of homology and complexity with patient tissues. Patient-derived colorectal cancer organoids, as personalized tumor organoids, can more accurately simulate colorectal cancer patients’ occurrence, development, metastasis, and predict drug response in colorectal cancer patients. Colorectal cancer organoids show great potential for application, especially preclinical drug screening and prediction of patient response to selected treatment options. Here, we reviewed the application of colorectal cancer organoids in disease model construction, basic biological research, organoid biobank construction, drug screening and personalized medicine, drug development, drug toxicity and safety, and regenerative medicine. In addition, we also displayed the current limitations and challenges of organoids and discussed the future development direction of organoids in combination with other technologies. Finally, we summarized and analyzed the current clinical trial research of organoids, especially the clinical trials of colorectal cancer organoids. We hoped to lay a solid foundation for organoids used in colorectal cancer research.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.,Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Yucui Ma
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Yilin Zheng
- Clinical Research Center, Shantou Central Hospital, Shantou, China
| | - Jiating Su
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Guoxin Huang
- Clinical Research Center, Shantou Central Hospital, Shantou, China
| |
Collapse
|
180
|
Ma L, Yu J, Zhang H, Zhao B, Zhang J, Yang D, Luo F, Wang B, Jin B, Liu J. Effects of Immune Cells on Intestinal Stem Cells: Prospects for Therapeutic Targets. Stem Cell Rev Rep 2022; 18:2296-2314. [DOI: 10.1007/s12015-022-10347-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
|
181
|
Taelman J, Diaz M, Guiu J. Human Intestinal Organoids: Promise and Challenge. Front Cell Dev Biol 2022; 10:854740. [PMID: 35359445 PMCID: PMC8962662 DOI: 10.3389/fcell.2022.854740] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
The study of human intestinal biology in healthy and diseased conditions has always been challenging. Primary obstacles have included limited tissue accessibility, inadequate in vitro maintenance and ethical constrains. The development of three-dimensional organoid cultures has transformed this entirely. Intestinal organoids are self-organized three-dimensional structures that partially recapitulate the identity, cell heterogeneity and cell behaviour of the original tissue in vitro. This includes the capacity of stem cells to self-renew, as well as to differentiate towards major intestinal lineages. Therefore, over the past decade, the use of human organoid cultures has been instrumental to model human intestinal development, homeostasis, disease, and regeneration. Intestinal organoids can be derived from pluripotent stem cells (PSC) or from adult somatic intestinal stem cells (ISC). Both types of organoid sources harbour their respective strengths and weaknesses. In this mini review, we describe the applications of human intestinal organoids, discussing the differences, advantages, and disadvantages of PSC-derived and ISC-derived organoids.
Collapse
Affiliation(s)
- Jasin Taelman
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
| | - Mònica Diaz
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
| | - Jordi Guiu
- Cell Plasticity and Regeneration Group, Regenerative Medicine Program, Institut d’Investigació Biomèdica de Bellvitge–IDIBELL, L’Hospitalet de Llobregat, Spain
- Program for advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L’Hospitalet de Llobregat, Spain
- *Correspondence: Jordi Guiu,
| |
Collapse
|
182
|
Brice DP, Murray GI, Wilson HM, Porter RJ, Berry S, Durum SK, McLean MH. Interleukin-27 Regulates the Function of the Gastrointestinal Epithelial Barrier in a Human Tissue-Derived Organoid Model. BIOLOGY 2022; 11:biology11030427. [PMID: 35336801 PMCID: PMC8945023 DOI: 10.3390/biology11030427] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 11/25/2022]
Abstract
A treatment with direct healing effects on the gastrointestinal epithelial barrier is desirable for inflammatory bowel disease (IBD). Interleukin-27 (IL-27) is an immunoregulatory cytokine, and oral delivery is an effective treatment in murine models of IBD. We aimed to define IL-27 effects on the human gastrointestinal epithelial barrier. We characterised gene and protein expression of permeability mediators in a human colon-derived organoid model. Functional permeability was determined in an organoid-derived 2D monolayer by transepithelial electrical resistance. IL-27 effects on epithelial innate immune responses were assessed through expression of cytokines, anti-microbial peptides and MUC genes. IL-27 effects on wound healing and proliferation were determined in human colon epithelial cell lines. IL-27 led to restoration of permeability regulation following inflammatory cytokine insult (p = 0.001), associated with differential expression of tight junction mediators with decrease in claudin 2 (p = 0.024) and increase in claudin 4 (p < 0.001), E-cadherin (p < 0.001) and zona occludens (p = 0.0014). IL-27 evoked differential gene expression of epithelial-derived innate immune responses (reduced IL1B and IL18, and increased IL33, HBD1, MUC1 and MUC2; p < 0.012). IL-27 induced epithelial barrier wound healing through restitution (p < 0.001), and increased proliferation (p < 0.001) following injury. Overall, IL-27 provokes mucosal healing of the human gastrointestinal epithelial barrier.
Collapse
Affiliation(s)
- Daniel P. Brice
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (D.P.B.); (G.I.M.); (H.M.W.); (S.B.)
| | - Graeme I. Murray
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (D.P.B.); (G.I.M.); (H.M.W.); (S.B.)
| | - Heather M. Wilson
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (D.P.B.); (G.I.M.); (H.M.W.); (S.B.)
| | - Ross J. Porter
- Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Susan Berry
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (D.P.B.); (G.I.M.); (H.M.W.); (S.B.)
| | - Scott K. Durum
- Cytokines and Immunity Section, Laboratory of Cancer Immunometabolism, National Cancer Institute (NCI), National Institute of Health (NIH), Frederick, MD 21702, USA;
| | - Mairi H. McLean
- Division of Molecular & Clinical Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
- Correspondence:
| |
Collapse
|
183
|
Foo MA, You M, Chan SL, Sethi G, Bonney GK, Yong WP, Chow EKH, Fong ELS, Wang L, Goh BC. Clinical translation of patient-derived tumour organoids- bottlenecks and strategies. Biomark Res 2022; 10:10. [PMID: 35272694 PMCID: PMC8908618 DOI: 10.1186/s40364-022-00356-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Multiple three-dimensional (3D) tumour organoid models assisted by multi-omics and Artificial Intelligence (AI) have contributed greatly to preclinical drug development and precision medicine. The intrinsic ability to maintain genetic and phenotypic heterogeneity of tumours allows for the reconciliation of shortcomings in traditional cancer models. While their utility in preclinical studies have been well established, little progress has been made in translational research and clinical trials. In this review, we identify the major bottlenecks preventing patient-derived tumour organoids (PDTOs) from being used in clinical setting. Unsuitable methods of tissue acquisition, disparities in establishment rates and a lengthy timeline are the limiting factors for use of PDTOs in clinical application. Potential strategies to overcome this include liquid biopsies via circulating tumour cells (CTCs), an automated organoid platform and optical metabolic imaging (OMI). These proposed solutions accelerate and optimize the workflow of a clinical organoid drug screening. As such, PDTOs have the potential for potential applications in clinical oncology to improve patient outcomes. If remarkable progress is made, cancer patients can finally benefit from this revolutionary technology.
Collapse
Affiliation(s)
- Malia Alexandra Foo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Mingliang You
- Hangzhou Cancer Institute, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, 31002, China.,Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 31002, China
| | - Shing Leng Chan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Surgery, National University Hospital, Singapore, Singapore
| | - Gautam Sethi
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Glenn K Bonney
- Department of Surgery, National University Hospital, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei-Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore, Singapore
| | - Edward Kai-Hua Chow
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Eliza Li Shan Fong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Boon-Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore, Singapore.
| |
Collapse
|
184
|
Klingler S, Hsu KS, Hua G, Martin ML, Adileh M, Baslan T, Zhang Z, Paty PB, Fuks Z, Brown AM, Kolesnick R. Disruption of the crypt niche promotes outgrowth of mutated colorectal tumor stem cells. JCI Insight 2022; 7:153793. [PMID: 35260534 PMCID: PMC8983138 DOI: 10.1172/jci.insight.153793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 01/26/2022] [Indexed: 12/14/2022] Open
Abstract
Recent data establish a logarithmic expansion of leucine rich repeat containing G protein coupled receptor 5–positive (Lgr5+) colonic epithelial stem cells (CESCs) in human colorectal cancer (CRC). Complementary studies using the murine 2-stage azoxymethane–dextran sulfate sodium (AOM-DSS) colitis-associated tumor model indicate early acquisition of Wnt pathway mutations drives CESC expansion during adenoma progression. Here, subdivision of the AOM-DSS model into in vivo and in vitro stages revealed DSS induced physical separation of CESCs from stem cell niche cells and basal lamina, a source of Wnt signals, within hours, disabling the stem cell program. While AOM delivery in vivo under non-adenoma-forming conditions yielded phenotypically normal mucosa and organoids derived thereof, niche injury ex vivo by progressive DSS dose escalation facilitated outgrowth of Wnt-independent dysplastic organoids. These organoids contained 10-fold increased Lgr5+ CESCs with gain-of-function Wnt mutations orthologous to human CRC driver mutations. We posit CRC originates by niche injury–induced outgrowth of normally suppressed mutated stem cells, consistent with models of adaptive oncogenesis.
Collapse
Affiliation(s)
- Stefan Klingler
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kuo-Shun Hsu
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Guoqiang Hua
- Institute of Radiation Medicine, Fudan University, Shanghai, China
| | - Maria Laura Martin
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Mohammad Adileh
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | | | - Zvi Fuks
- Department of Radiation Oncology, and
| | - Anthony Mc Brown
- Department of Cell & Developmental Biology, Weill Cornell Medicine, New York, New York, USA
| | - Richard Kolesnick
- Laboratory of Signal Transduction, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
185
|
Boye TL, Steenholdt C, Jensen KB, Nielsen OH. Molecular manipulations and intestinal stem cell-derived organoids in inflammatory bowel disease. Stem Cells 2022; 40:447-457. [DOI: 10.1093/stmcls/sxac014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022]
Abstract
Abstract
The pathogenesis of inflammatory bowel diseases (IBD) involves genetic predisposition, environmental factors, and a broadly dysregulated intestinal immune response to the commensal intestinal microflora. The interface between genetic predisposition and environmental factors is reflected in the epigenetic regulation at the transcriptional level. Treatment targets now involve mucosal and histological healing, but the future might additionally include normalization of intestinal cellular functions also at the molecular level, for example comprising complete restoration of phenotypic, genotypic, and epigenetic states. Recent developments in patient-derived epithelial intestinal stem cell (ISC) organoid technologies have opened exciting new therapeutic opportunities to potentially attain molecular healing by combining stem cell therapy with molecular manipulations using (epi)drugs and/or CRISPR/Cas9 genome editing. Here, we are the first to discuss the possibility for phenotypic, genotypic, and epigenetic restoration via molecular manipulations and stem cell therapy in IBD from a clinical perspective.
Collapse
Affiliation(s)
- Theresa Louise Boye
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, DK-2730 Herlev, Denmark
| | - Casper Steenholdt
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, DK-2730 Herlev, Denmark
| | - Kim Bak Jensen
- Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, DK-2730 Herlev, Denmark
| |
Collapse
|
186
|
Tian F, Huang S, Xu W, Chen L, Su J, Ni H, Feng X, Chen J, Wang X, Huang Q. Compound K attenuates hyperglycemia by enhancing glucagon-like peptide-1 secretion through activating TGR5 via the remodeling of gut microbiota and bile acid metabolism. J Ginseng Res 2022; 46:780-789. [PMID: 36312739 PMCID: PMC9597441 DOI: 10.1016/j.jgr.2022.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/25/2022] [Accepted: 03/29/2022] [Indexed: 01/06/2023] Open
Abstract
Background Incretin impairment, characterized by insufficient secretion of L-cell-derived glucagon-like peptide-1 (GLP-1), is a defining step of type 2 diabetes mellitus (T2DM). Ginsenoside compound K (CK) can stimulate GLP-1 secretion; however, the potential mechanism underlying this effect has not been established. Methods CK (40 mg/kg) was administered orally to male db/db mice for 4 weeks. The body weight, oral glucose tolerance, GLP-1 secretion, gut microbiota sequencing, bile acid (BA) profiles, and BA synthesis markers of each subject were then analyzed. Moreover, TGR5 expression was evaluated by immunoblotting and immunofluorescence, and L-cell lineage markers involved in L-cell abundance were analyzed. Results CK ameliorated obesity and impaired glucose tolerance in db/db mice by altering the gut microbiota, especially Ruminococcaceae family, and this changed microbe was positively correlated with secondary BA synthesis. Additionally, CK treatment resulted in the up-regulation of CYP7B1 and CYP27A1 and the down-regulation of CYP8B1, thereby shifting BA biosynthesis from the classical pathway to the alternative pathway. CK altered the BA pool by mainly increasing LCA and DCA. Furthermore, CK induced L-cell number expansion leading to enhanced GLP-1 release through TGR5 activation. These increases were supported by the upregulation of genes governing GLP-1 secretion and L-cell differentiation. Conclusions The results indicate that CK improves glucose homeostasis by increasing L-cell numbers, which enhances GLP-1 release through a mechanism partially mediated by the gut microbiota-BA-TGR5 pathway. Therefore, that therapeutic attempts with CK might be useful for patients with T2DM.
Collapse
Affiliation(s)
- Fengyuan Tian
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Shuo Huang
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wangda Xu
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Lan Chen
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jianming Su
- Department of Emergency, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Haixiang Ni
- Department of Endocrinology, First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Xiaohong Feng
- Department of Endocrinology, First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Jie Chen
- Department of Endocrinology, First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Xi Wang
- Central Laboratory, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- Corresponding author. Central Laboratory, First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, 310006, China.
| | - Qi Huang
- Department of Endocrinology, First Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, China
- Corresponding author. Department of Endocrinology, First Affiliated Hospital of Zhejiang, Chinese Medicine University. Hangzhou, 310006, China.
| |
Collapse
|
187
|
3D in vitro morphogenesis of human intestinal epithelium in a gut-on-a-chip or a hybrid chip with a cell culture insert. Nat Protoc 2022; 17:910-939. [PMID: 35110737 PMCID: PMC9675318 DOI: 10.1038/s41596-021-00674-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022]
Abstract
Human intestinal morphogenesis establishes 3D epithelial microarchitecture and spatially organized crypt-villus characteristics. This unique structure is necessary to maintain intestinal homeostasis by protecting the stem cell niche in the basal crypt from exogenous microbial antigens and their metabolites. Also, intestinal villi and secretory mucus present functionally differentiated epithelial cells with a protective barrier at the intestinal mucosal surface. Thus, re-creating the 3D epithelial structure is critical to building in vitro intestine models. Notably, an organomimetic gut-on-a-chip can induce spontaneous 3D morphogenesis of an intestinal epithelium with enhanced physiological function and biomechanics. Here we provide a reproducible protocol to robustly induce intestinal morphogenesis in a microfluidic gut-on-a-chip as well as in a Transwell-embedded hybrid chip. We describe detailed methods for device fabrication, culture of Caco-2 or intestinal organoid epithelial cells in conventional setups as well as on microfluidic platforms, induction of 3D morphogenesis and characterization of established 3D epithelium using multiple imaging modalities. This protocol enables the regeneration of functional intestinal microarchitecture by controlling basolateral fluid flow within 5 d. Our in vitro morphogenesis method employs physiologically relevant shear stress and mechanical motions, and does not require complex cellular engineering or manipulation, which may be advantageous over other existing techniques. We envision that our proposed protocol may have a broad impact on biomedical research communities, providing a method to regenerate in vitro 3D intestinal epithelial layers for biomedical, clinical and pharmaceutical applications.
Collapse
|
188
|
Ren J, Li N, Pei S, Lian Y, Li L, Peng Y, Liu Q, Guo J, Wang X, Han Y, Zhang G, Wang H, Li Y, Jiang J, Li Q, Tan M, Peng J, Hu G, Xiao Y, Li X, Lin M, Qin J. Histone methyltransferase WHSC1 loss dampens MHC-I antigen presentation pathway to impair IFN-γ-stimulated anti-tumor immunity. J Clin Invest 2022; 132:153167. [PMID: 35230972 PMCID: PMC9012282 DOI: 10.1172/jci153167] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 02/23/2022] [Indexed: 11/17/2022] Open
Abstract
IFN-γ–stimulated MHC class I (MHC-I) antigen presentation underlies the core of antitumor immunity. However, sustained IFN-γ signaling also enhances the programmed death ligand 1 (PD-L1) checkpoint pathway to dampen antitumor immunity. It remains unclear how these opposing effects of IFN-γ are regulated. Here, we report that loss of the histone dimethyltransferase WHSC1 impaired the antitumor effect of IFN-γ signaling by transcriptional downregulation of the MHC-I machinery without affecting PD-L1 expression in colorectal cancer (CRC) cells. Whsc1 loss promoted tumorigenesis via a non-cell-autonomous mechanism in an Apcmin/+ mouse model, CRC organoids, and xenografts. Mechanistically, we found that the IFN-γ/STAT1 signaling axis stimulated WHSC1 expression and, in turn, that WHSC1 directly interacted with NLRC5 to promote MHC-I gene expression, but not that of PD-L1. Concordantly, silencing Whsc1 diminished MHC-I levels, impaired antitumor immunity, and blunted the effect of immune checkpoint blockade. Patient cohort analysis revealed that WHSC1 expression positively correlated with enhanced MHC-I expression, tumor-infiltrating T cells, and favorable disease outcomes. Together, our findings establish a tumor-suppressive function of WHSC1 that relays IFN-γ signaling to promote antigen presentation on CRC cells and provide a rationale for boosting WHSC1 activity in immunotherapy.
Collapse
Affiliation(s)
- Jiale Ren
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Ni Li
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Siyu Pei
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Yannan Lian
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Li Li
- Department of General Surgery, Department of Gastroenterology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuchong Peng
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qiuli Liu
- Department of Urology, Daping Hospital, Army Medical University, Shanghai, China
| | - Jiacheng Guo
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Xuege Wang
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Ying Han
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Guoying Zhang
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Hanling Wang
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Yaqi Li
- Department of Oncology, Fudan University, Shanghai, China
| | - Jun Jiang
- Department of Urology, Daping Hospital, Army Medical University, Shanghai, China
| | - Qintong Li
- Department of Obstetrics, Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Junjie Peng
- Department of Oncology, Fudan University, Shanghai, China
| | - Guohong Hu
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Yichuan Xiao
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| | - Xiong Li
- Center for Clinical Precision Pharmacy, The First Affiliated Hospital, School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Moubin Lin
- Department of General Surgery, Department of Gastroenterology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Qin
- Shanghai Jiao Tong University School of Medicine (SJTUSM) & Chinese Academy, Shanghai Institute of Nutrition and Health, Shanghai, China
| |
Collapse
|
189
|
Xi W, Saleh J, Yamada A, Tomba C, Mercier B, Janel S, Dang T, Soleilhac M, Djemat A, Wu H, Romagnolo B, Lafont F, Mège RM, Chen Y, Delacour D. Modulation of designer biomimetic matrices for optimized differentiated intestinal epithelial cultures. Biomaterials 2022; 282:121380. [DOI: 10.1016/j.biomaterials.2022.121380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 01/07/2022] [Accepted: 01/16/2022] [Indexed: 12/22/2022]
|
190
|
D'Alessio S, Ungaro F, Noviello D, Lovisa S, Peyrin-Biroulet L, Danese S. Revisiting fibrosis in inflammatory bowel disease: the gut thickens. Nat Rev Gastroenterol Hepatol 2022; 19:169-184. [PMID: 34876680 DOI: 10.1038/s41575-021-00543-0] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2021] [Indexed: 12/11/2022]
Abstract
Intestinal fibrosis, which is usually the consequence of chronic inflammation, is a common complication of inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis. In the past few years, substantial advances have been made in the areas of pathogenesis, diagnosis and management of intestinal fibrosis. Of particular interest have been inflammation-independent mechanisms behind the gut fibrotic process, genetic and environmental risk factors (such as the role of the microbiota), and the generation of new in vitro and in vivo systems to study fibrogenesis in the gut. A huge amount of work has also been done in the area of biomarkers to predict or detect intestinal fibrosis, including novel cross-sectional imaging techniques. In parallel, researchers are embarking on developing and validating clinical trial end points and protocols to test novel antifibrotic agents, although no antifibrotic therapies are currently available. This Review presents the state of the art on the most recently identified pathogenic mechanisms of this serious IBD-related complication, focusing on possible targets of antifibrotic therapies, management strategies, and factors that might predict fibrosis progression or response to treatment.
Collapse
Affiliation(s)
| | - Federica Ungaro
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Daniele Noviello
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Sara Lovisa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,IBD Centre, Laboratory of Gastrointestinal Immunopathology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Laurent Peyrin-Biroulet
- INSERM NGERE, University of Lorraine, Vandoeuvre-les-Nancy, Nancy, France.,Nancy University Hospital, Vandoeuvre-les-Nancy, Nancy, France
| | - Silvio Danese
- Department of Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy. .,University Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
191
|
Hasebe T, Fujimoto K, Ishizuya-Oka A. Essential roles of YAP-TEAD complex in adult stem cell development during thyroid hormone-induced intestinal remodeling of Xenopus laevis. Cell Tissue Res 2022; 388:313-329. [PMID: 35211820 DOI: 10.1007/s00441-022-03600-5] [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: 08/17/2021] [Accepted: 02/16/2022] [Indexed: 11/24/2022]
Abstract
During amphibian metamorphosis which is triggered by thyroid hormone (TH), the small intestine is extensively remodeled from the larval to adult form. In the Xenopus laevis intestine, some of the larval epithelial cells dedifferentiate into adult stem cells, which newly form the adult epithelium similar to the mammalian one. We have previously shown that TH-activated Shh, Wnt and Notch signaling pathways play important roles in adult epithelial development. Here we focus on the Hippo signaling pathway, which is known to interact with these pathways in the mammalian intestine. Our quantitative RT-PCR analysis indicates that the expression of genes involved in this pathway including YAP1, TAZ, TEAD1 and core kinases is differently regulated by TH in the metamorphosing intestine. Additionally, we show by in situ hybridization and immunohistochemistry that the transcriptional co-activator YAP1, a major effector of the Hippo signaling, is expressed in the adult stem cells and connective tissue cells surrounding them and that YAP1 protein is localized in either nucleus or cytoplasm of the stem cells. We further show that YAP1 binds its binding partner TEAD1 (transcription factor) in vivo and that their interaction is inhibited by verteporfin (VP). More importantly, by using VP in organ culture of the tadpole intestine, we experimentally demonstrate that the inhibition of YAP1-TEAD1 interaction decreases both TH-induced stem cells expressing LGR5 and nearby connective tissue cells in number and proliferation, leading to the failure of adult epithelial development. Our results indicate that YAP-TEAD complex is required for stem cell development during intestinal remodeling.
Collapse
Affiliation(s)
- Takashi Hasebe
- Department of Biology, Nippon Medical School, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-0023, Japan.
| | - Kenta Fujimoto
- Department of Biology, Nippon Medical School, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-0023, Japan
| | - Atsuko Ishizuya-Oka
- Department of Biology, Nippon Medical School, 1-7-1 Kyonan-cho, Musashino, Tokyo, 180-0023, Japan
| |
Collapse
|
192
|
What Do In Vitro and In Vivo Models Tell Us about Anisakiasis? New Tools Still to Be Explored. Pathogens 2022; 11:pathogens11030285. [PMID: 35335609 PMCID: PMC8953344 DOI: 10.3390/pathogens11030285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 02/04/2023] Open
Abstract
Anisakiasis is a zoonosis caused by the ingestion of raw or undercooked seafood infected with third-stage larvae (L3) of the marine nematode Anisakis. Based on L3 localization in human accidental hosts, gastric, intestinal or ectopic (extra-gastrointestinal) anisakiasis can occur, in association with mild to severe symptoms of an allergic nature. Given the increasing consumption of fish worldwide, the European Food Safety Authority declared Anisakis as an emerging pathogen. Despite its importance for public health and economy, the scientific literature is largely characterized by taxonomic, systematic and ecological studies, while investigations on clinical aspects, such as the inflammatory and immune response during anisakiasis, using a proper model that simulates the niche of infection are still very scarce. The aims of this review are to describe the clinical features of anisakiasis, to report the main evidence from the in vivo and in vitro studies carried out to date, highlighting limitations, and to propose future perspectives in the study field of anisakiasis.
Collapse
|
193
|
Synthetic developmental biology: Engineering approaches to guide multicellular organization. Stem Cell Reports 2022; 17:715-733. [PMID: 35276092 PMCID: PMC9023767 DOI: 10.1016/j.stemcr.2022.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 11/30/2022] Open
Abstract
Multicellular organisms of various complexities self-organize in nature. Organoids are in vitro 3D structures that display important aspects of the anatomy and physiology of their in vivo counterparts and that develop from pluripotent or tissue-specific stem cells through a self-organization process. In this review, we describe the multidisciplinary concept of “synthetic developmental biology” where engineering approaches are employed to guide multicellular organization in an experimental setting. We introduce a novel classification of engineering approaches based on the extent of microenvironmental manipulation applied to organoids. In the final section, we discuss how engineering tools might help overcome current limitations in organoid construction.
Collapse
|
194
|
Chiang HY, Lu HH, Sudhakar JN, Chen YW, Shih NS, Weng YT, Shui JW. IL-22 initiates an IL-18-dependent epithelial response circuit to enforce intestinal host defence. Nat Commun 2022; 13:874. [PMID: 35169117 PMCID: PMC8847568 DOI: 10.1038/s41467-022-28478-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/26/2022] [Indexed: 12/19/2022] Open
Abstract
IL-18 is emerging as an IL-22-induced and epithelium-derived cytokine which contributes to host defence against intestinal infection and inflammation. In contrast to its known role in Goblet cells, regulation of barrier function at the molecular level by IL-18 is much less explored. Here we show that IL-18 is a bona fide IL-22-regulated gate keeper for intestinal epithelial barrier. IL-22 promotes crypt immunity both via induction of phospho-Stat3 binding to the Il-18 gene promoter and via Il-18 independent mechanisms. In organoid culture, while IL-22 primarily increases organoid size and inhibits expression of stem cell genes, IL-18 preferentially promotes organoid budding and induces signature genes of Lgr5+ stem cells via Akt-Tcf4 signalling. During adherent-invasive E. coli (AIEC) infection, systemic administration of IL-18 corrects compromised T-cell IFNγ production and restores Lysozyme+ Paneth cells in Il-22-/- mice, but IL-22 administration fails to restore these parameters in Il-18-/- mice, thereby placing IL-22-Stat3 signalling upstream of the IL-18-mediated barrier defence function. IL-18 in return regulates Stat3-mediated anti-microbial response in Paneth cells, Akt-Tcf4-triggered expansion of Lgr5+ stem cells to facilitate tissue repair, and AIEC clearance by promoting IFNγ+ T cells.
Collapse
Affiliation(s)
- Hung-Yu Chiang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hsueh-Han Lu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | | - Yu-Wen Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Nien-Shin Shih
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Ting Weng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jr-Wen Shui
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
195
|
Arostegui D, Wallach T. The Cutting Edge of Gastroenteritis: Advances in Understanding of Enteric Infection. J Pediatr Gastroenterol Nutr 2022; 74:180-185. [PMID: 34560728 DOI: 10.1097/mpg.0000000000003304] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
ABSTRACT In recent years, multiple advances have been made in the care, diagnosis, and mechanistic understanding of acute gastroenteritis (AGE). In this review, we discuss the current state of the art of diagnosis and management, as well as how changes in practice can improve care and decrease costs. We will discuss present study demonstrating the effect of AGE on the microbiome and how that may be linked to secondary effects or long-term changes. We will explore the use of novel technologies to further our capacity to understand how gastrointestinal infections occur and promulgate. Finally, will discuss advances in our understanding of how gastrointestinal infections capacitate other changes such as post-viral motility or other post viral intestinal dysfunction.
Collapse
Affiliation(s)
- Dalia Arostegui
- SUNY Downstate Department of Pediatrics, Division of Pediatric Gastroenterology, Brooklyn, NY
| | | |
Collapse
|
196
|
Research Priorities for Kidney-Related Research-An Agenda to Advance Kidney Care: A Position Statement From the National Kidney Foundation. Am J Kidney Dis 2022; 79:141-152. [PMID: 34627932 DOI: 10.1053/j.ajkd.2021.08.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 08/26/2021] [Indexed: 02/01/2023]
Abstract
Despite the high prevalence and economic burden of chronic kidney disease (CKD) in the United States, federal funding for kidney-related research, prevention, and education activities under the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) remains substantially lower compared to other chronic diseases. More federal support is needed to promote critical research that will expand knowledge of kidney health and disease, develop new and effective therapies, and reduce health disparities. In 2021, the National Kidney Foundation (NKF) convened 2 Research Roundtables (preclinical and clinical research), comprising nephrology leaders from prominent US academic institutions and the pharmaceutical industry, key bodies with expertise in research, and including individuals with CKD and their caregivers and kidney donors. The goal of these roundtables was to identify priorities for preclinical and clinical kidney-related research. The research priorities identified by the Research Roundtables and presented in this position statement outline attainable opportunities for groundbreaking and critically needed innovations that will benefit patients with kidney disease in the next 5-10 years. Research priorities fall within 4 preclinical science themes (expand data science capability, define kidney disease mechanisms and utilize genetic tools to identify new therapeutic targets, develop better models of human disease, and test cell-specific drug delivery systems and utilize gene editing) and 3 clinical science themes (expand number and inclusivity of clinical trials, develop and test interventions to reduce health disparities, and support implementation science). These priorities in kidney-related research, if supported by additional funding by federal agencies, will increase our understanding of the development and progression of kidney disease among diverse populations, attract additional industry investment, and lead to new and more personalized treatments.
Collapse
|
197
|
Arian CM, Imaoka T, Yang J, Kelly EJ, Thummel KE. Gutsy science: In vitro systems of the human intestine to model oral drug disposition. Pharmacol Ther 2022; 230:107962. [PMID: 34478775 PMCID: PMC8821120 DOI: 10.1016/j.pharmthera.2021.107962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 02/03/2023]
Abstract
The intestine has important gate-keeping functions that can profoundly affect the systemic blood exposure of orally administered drugs. Thus, characterizing a new molecular entity's (NME) disposition within the intestine is of utmost importance in drug development. While currently used in vitro systems, such as Ussing chamber, precision-cut intestinal slices, immortalized cell lines, and primary enterocytes provide substantial knowledge about drug absorption and the intestinal first-pass effect, they remain sub-optimal for quantitatively predicting this process and the oral bioavailability of many drugs. Use of novel in vitro systems such as intestinal organoids and intestinal microphysiological systems have provided substantial advances over the past decade, expanding our understanding of intestinal physiology, pathology, and development. However, application of these emerging in vitro systems in the pharmaceutical science is in its infancy. Preliminary work has demonstrated that these systems more accurately recapitulate the physiology and biochemistry of the intact intestine, as it relates to oral drug disposition, and thus they hold considerable promise as preclinical testing platforms of the future. Here we review currently used and emerging in vitro models of the human intestine employed in pharmaceutical science research. We also highlight aspects of these emerging tools that require further study.
Collapse
Affiliation(s)
- Christopher M Arian
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Tomoki Imaoka
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Jade Yang
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Edward J Kelly
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA
| | - Kenneth E Thummel
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
198
|
Anjum M, Laitila A, Ouwehand AC, Forssten SD. Current Perspectives on Gastrointestinal Models to Assess Probiotic-Pathogen Interactions. Front Microbiol 2022; 13:831455. [PMID: 35173703 PMCID: PMC8841803 DOI: 10.3389/fmicb.2022.831455] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
There are different models available that mimic the human intestinal epithelium and are thus available for studying probiotic and pathogen interactions in the gastrointestinal tract. Although, in vivo models make it possible to study the overall effects of a probiotic on a living subject, they cannot always be conducted and there is a general commitment to reduce the use of animal models. Hence, in vitro methods provide a more rapid tool for studying the interaction between probiotics and pathogens; as well as being ethically superior, faster, and less expensive. The in vitro models are represented by less complex traditional models, standard 2D models compromised of culture plates as well as Transwell inserts, and newer 3D models like organoids, enteroids, as well as organ-on-a-chip. The optimal model selected depends on the research question. Properly designed in vitro and/or in vivo studies are needed to examine the mechanism(s) of action of probiotics on pathogens to obtain physiologically relevant results.
Collapse
Affiliation(s)
| | | | | | - Sofia D. Forssten
- International Flavors and Fragrances, Health and Biosciences, Danisco Sweeteners Oy, Kantvik, Finland
| |
Collapse
|
199
|
Cholico GN, Nault R, Zacharewski TR. Genome-Wide ChIPseq Analysis of AhR, COUP-TF, and HNF4 Enrichment in TCDD-Treated Mouse Liver. Int J Mol Sci 2022; 23:1558. [PMID: 35163483 PMCID: PMC8836158 DOI: 10.3390/ijms23031558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 02/01/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor known for mediating the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds. Although the canonical mechanism of AhR activation involves heterodimerization with the aryl hydrocarbon receptor nuclear translocator, other transcriptional regulators that interact with AhR have been identified. Enrichment analysis of motifs in AhR-bound genomic regions implicated co-operation with COUP transcription factor (COUP-TF) and hepatocyte nuclear factor 4 (HNF4). The present study investigated AhR, HNF4α and COUP-TFII genomic binding and effects on gene expression associated with liver-specific function and cell differentiation in response to TCDD. Hepatic ChIPseq data from male C57BL/6 mice at 2 h after oral gavage with 30 µg/kg TCDD were integrated with bulk RNA-sequencing (RNAseq) time-course (2-72 h) and dose-response (0.01-30 µg/kg) datasets to assess putative AhR, HNF4α and COUP-TFII interactions associated with differential gene expression. Functional enrichment analysis of differentially expressed genes (DEGs) identified differential binding enrichment for AhR, COUP-TFII, and HNF4α to regions within liver-specific genes, suggesting intersections associated with the loss of liver-specific functions and hepatocyte differentiation. Analysis found that the repression of liver-specific, HNF4α target and hepatocyte differentiation genes, involved increased AhR and HNF4α binding with decreased COUP-TFII binding. Collectively, these results suggested TCDD-elicited loss of liver-specific functions and markers of hepatocyte differentiation involved interactions between AhR, COUP-TFII and HNF4α.
Collapse
Affiliation(s)
| | | | - Tim R. Zacharewski
- Biochemistry & Molecular Biology, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; (G.N.C.); (R.N.)
| |
Collapse
|
200
|
Lu HJ, Li J, Yang G, Yi CJ, Zhang D, Yu F, Ma Z. Circular RNAs in stem cells: from basic research to clinical implications. Biosci Rep 2022; 42:BSR20212510. [PMID: 34908111 PMCID: PMC8738868 DOI: 10.1042/bsr20212510] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Circular RNAs (circRNAs) are a special class of endogenous RNAs with a wide variety of pathophysiological functions via diverse mechanisms, including transcription, microRNA (miRNA) sponge, protein sponge/decoy, and translation. Stem cells are pluripotent cells with unique properties of self-renewal and differentiation. Dysregulated circRNAs identified in various stem cell types can affect stem cell self-renewal and differentiation potential by manipulating stemness. However, the emerging roles of circRNAs in stem cells remain largely unknown. This review summarizes the major functions and mechanisms of action of circRNAs in stem cell biology and disease progression. We also highlight circRNA-mediated common pathways in diverse stem cell types and discuss their diagnostic significance with respect to stem cell-based therapy.
Collapse
Affiliation(s)
- Hui-Juan Lu
- The First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, China
| | - Juan Li
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guodong Yang
- Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang, Hubei 438000, China
| | - Cun-Jian Yi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, China
| | - Daping Zhang
- The First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
| | - Fenggang Yu
- Institute of Life Science, Yinfeng Biological Group, Jinan 250000, China
| | - Zhaowu Ma
- The First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, China
- School of Basic Medicine, Health Science Center, Yangtze University, 1 Nanhuan Road, Jingzhou, Hubei 434023, China
| |
Collapse
|