151
|
Abstract
Molecular abnormalities that shape human neoplasms dissociate their phenotypic landscape from that of the healthy counterpart. Through the lens of a microscope, tumour pathology optically captures such aberrations projected onto a tissue slide and has categorized human epithelial neoplasms into distinct histological subtypes based on the diverse morphogenetic and molecular programmes that they manifest. Tumour histology often reflects tumour aggressiveness, patient prognosis and therapeutic vulnerability, and thus has been used as a de facto diagnostic tool and for making clinical decisions. However, it remains elusive how the diverse histological subtypes arise and translate into pleiotropic biological phenotypes. Molecular analysis of clinical tumour tissues and their culture, including patient-derived organoids, and add-back genetic reconstruction of tumorigenic pathways using gene engineering in culture models and rodents further elucidated molecular mechanisms that underlie morphological variations. Such mechanisms include genetic mutations and epigenetic alterations in cellular identity codes that erode hard-wired morphological programmes and histologically digress tumours from the native tissues. Interestingly, tumours acquire the ability to grow independently of the niche-driven stem cell ecosystem along with these morphological alterations, providing a biological rationale for histological diversification during tumorigenesis. This Review comprehensively summarizes our current understanding of such plasticity in the histological and lineage commitment fostered cooperatively by molecular alterations and the tumour environment, and describes basic and clinical implications for future cancer therapy.
Collapse
Affiliation(s)
- Masayuki Fujii
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| | - Shigeki Sekine
- Division of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan
| | - Toshiro Sato
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
152
|
Solanki S, Shah YM. Hypoxia-Induced Signaling in Gut and Liver Pathobiology. ANNUAL REVIEW OF PATHOLOGY 2024; 19:291-317. [PMID: 37832943 DOI: 10.1146/annurev-pathmechdis-051122-094743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
Oxygen (O2) is essential for cellular metabolism and biochemical reactions. When the demand for O2 exceeds the supply, hypoxia occurs. Hypoxia-inducible factors (HIFs) are essential to activate adaptive and survival responses following hypoxic stress. In the gut (intestines) and liver, the presence of oxygen gradients or physiologic hypoxia is necessary to maintain normal homeostasis. While physiologic hypoxia is beneficial and aids in normal functions, pathological hypoxia is harmful as it exacerbates inflammatory responses and tissue dysfunction and is a hallmark of many cancers. In this review, we discuss the role of gut and liver hypoxia-induced signaling, primarily focusing on HIFs, in the physiology and pathobiology of gut and liver diseases. Additionally, we examine the function of HIFs in various cell types during gut and liver diseases, beyond intestinal epithelial and hepatocyte HIFs. This review highlights the importance of understanding hypoxia-induced signaling in the pathogenesis of gut and liver diseases and emphasizes the potential of HIFs as therapeutic targets.
Collapse
Affiliation(s)
- Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA;
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA;
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
153
|
Liu Y, Han K, Liu H, Jia G, Comer L, Wang G, Pan Z, Zhao Y, Jiang S, Jiao N, Huang L, Yang W, Li Y. Macleaya cordata isoquinoline alkaloids attenuate Escherichia coli lipopolysaccharide-induced intestinal epithelium injury in broiler chickens by co-regulating the TLR4/MyD88/NF-κB and Nrf2 signaling pathways. Front Immunol 2024; 14:1335359. [PMID: 38299145 PMCID: PMC10828024 DOI: 10.3389/fimmu.2023.1335359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
This study sought to explore the effects and potential mechanisms of dietary supplementation with isoquinoline alkaloids (IA) from Macleaya cordata to alleviate lipopolysaccharide (LPS)-induced intestinal epithelium injury in broilers. A total of 486 1-day-old broilers were assigned at random to a control (CON) group, LPS group, and LPS+IA group in a 21-d study. The CON and LPS groups received a basal diet, while the LPS+IA group received a basal diet supplemented with 0.6 mg/kg IA. At 17, 19, and 21 days of age, the LPS and LPS+BP groups were injected intraperitoneally with LPS, and the CON group was intraperitoneally injected equivalent amount of saline solution. The results manifested that LPS injection caused intestinal inflammation and lipid peroxidation, disrupted intestinal barrier and function, and increased the abundance of harmful microorganisms. However, dietary IA supplementation alleviated LPS-induced adverse changes in intestinal morphology, apoptosis, mucosal barrier integrity, cecum microorganisms, and homeostasis disorder by decreasing inflammatory cytokines and enhancing antioxidant-related genes expressions; inhibited LPS-induced increases in TLR4 and NF-κB expressions and decreases in Nrf2 and GPX1 genes expressions. Our findings indicated that Macleaya cordata IA addition attenuated LPS-induced intestinal epithelium injury and disorder of intestinal homeostasis by enhancing the anti-inflammatory and antioxidant capacity of broiler chickens possibly via co-regulating TLR4/MyD88/NF-κB and Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Yang Liu
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kai Han
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Hua Liu
- College of Animal Science and Technology, Hunan Agriculture University, Changsha, China
| | - Gang Jia
- Institute of Animal Nutrition, Key Laboratory for Animal Disease-Resistance Nutrition of China, Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Luke Comer
- The Nutrition and Animal Microbiota Ecosystems Laboratory, Division of Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Heverlee, Belgium
| | - Guanlin Wang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Zizhu Pan
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Yiqian Zhao
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Shuzhen Jiang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Ning Jiao
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Libo Huang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Weiren Yang
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
| | - Yang Li
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Department of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai’an, China
- The Nutrition and Animal Microbiota Ecosystems Laboratory, Division of Animal and Human Health Engineering, Department of Biosystems, KU Leuven, Heverlee, Belgium
| |
Collapse
|
154
|
Sakahara M, Okamoto T, Srivastava U, Natsume Y, Yamanaka H, Suzuki Y, Obama K, Nagayama S, Yao R. Paneth-like cells produced from OLFM4 + stem cells support OLFM4 + stem cell growth in advanced colorectal cancer. Commun Biol 2024; 7:27. [PMID: 38182890 PMCID: PMC10770338 DOI: 10.1038/s42003-023-05504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 10/25/2023] [Indexed: 01/07/2024] Open
Abstract
Tumor tissues consist of heterogeneous cells that originate from stem cells; however, their cell fate determination program remains incompletely understood. Using patient-derived organoids established from patients with advanced colorectal cancer (CRC), we evaluated the potential of olfactomedin 4 (OLFM4)+ stem cells to produce a bifurcated lineage of progenies with absorptive and secretory properties. In the early phases of organoid reconstruction, OLFM4+ cells preferentially gave rise to secretory cells. Additionally, we found that Paneth-like cells, which do not exist in the normal colon, were induced in response to Notch signaling inhibition. Video recordings of single OLFM4+ cells revealed that organoids containing Paneth-like cells were effectively propagated and that their selective ablation led to organoid collapse. In tumor tissues, Paneth-like cells were identified only in the region where tumor cells lost cell adhesion. These findings indicate that Paneth-like cells are directly produced by OLFM4+ stem cells and that their interaction contributes to tumor formation by providing niche factors. This study reveals the importance of the cell fate specification program for building a complete tumor cellular ecosystem, which might be targeted with novel therapeutics.
Collapse
Affiliation(s)
- Mizuho Sakahara
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takuya Okamoto
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Upasna Srivastava
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Yasuko Natsume
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hitomi Yamanaka
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Kazutaka Obama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Satoshi Nagayama
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Surgery, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Ryoji Yao
- Department of Cell Biology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
155
|
Fu SC, Qu JY, Li LX, Yang XX, Li YQ, Zuo XL. Excessive Mitochondrial Fission Suppresses Mucosal Repair by Impairing Butyrate Metabolism in Colonocytes. Inflamm Bowel Dis 2024; 30:114-124. [PMID: 37454276 DOI: 10.1093/ibd/izad132] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Indexed: 07/18/2023]
Abstract
BACKGROUND Mucosal healing is one of the principal therapeutic targets for ulcerative colitis (UC). Mitochondria are dynamic organelles that undergo constant fusion and fission; however, the process that is most conducive to mucosal healing remains unclear. This study investigated the role of mitochondrial fission in mucosal healing in UC patients. METHODS Quantitative polymerase chain reaction, Western blotting, and immunostaining were used to detect mitochondrial fission in UC patients and a dextran sulfate sodium-induced colitis model. Colonic organoids were used to investigate the role of mitochondrial fission in butyrate metabolism. Enzyme activity assays were performed to identify the key proteins involved in this mechanism. RESULTS It was found that inhibition of mitochondrial fission promoted mucosal healing in mice and that there was an increase in mitochondrial fission in colonic epithelial cells of UC patients. Excessive fission inhibits stem cell proliferation by impairing butyrate metabolism in colonic organoids. The mitochondrial fission antagonist P110 failed to promote mucosal healing in antibiotic-treated mice, and the addition of exogenous butyrate reversed this effect. Increased butyrate exposure in the colonic stem cell niche has also been observed in UC patients. Mechanistically, enzyme activity assays on colonic organoids revealed that excessive fission inhibits mitochondrial acetoacetyl-CoA thiolase activity via reactive oxygen species. CONCLUSIONS Collectively, these data indicate that excessive mitochondrial fission suppresses mucosal repair by inhibiting butyrate metabolism and provides a potential target for mucosal healing in patients with ulcerative colitis.
Collapse
Affiliation(s)
- Shi-Chen Fu
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital, Shandong University, Jinan, China
| | - Jun-Yan Qu
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital, Shandong University, Jinan, China
| | - Li-Xiang Li
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital, Shandong University, Jinan, China
| | - Xiao-Xiao Yang
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital, Shandong University, Jinan, China
| | - Yan-Qing Li
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital, Shandong University, Jinan, China
| | - Xiu-Li Zuo
- Department of Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Laboratory of Translational Gastroenterology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Robot Engineering Laboratory for Precise Diagnosis and Therapy of GI Tumor, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
156
|
Beumer J, Clevers H. Hallmarks of stemness in mammalian tissues. Cell Stem Cell 2024; 31:7-24. [PMID: 38181752 PMCID: PMC10769195 DOI: 10.1016/j.stem.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/03/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024]
Abstract
All adult tissues experience wear and tear. Most tissues can compensate for cell loss through the activity of resident stem cells. Although the cellular maintenance strategies vary greatly between different adult (read: postnatal) tissues, the function of stem cells is best defined by their capacity to replace lost tissue through division. We discuss a set of six complementary hallmarks that are key enabling features of this basic function. These include longevity and self-renewal, multipotency, transplantability, plasticity, dependence on niche signals, and maintenance of genome integrity. We discuss these hallmarks in the context of some of the best-understood adult stem cell niches.
Collapse
Affiliation(s)
- Joep Beumer
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| | - Hans Clevers
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Basel, Switzerland.
| |
Collapse
|
157
|
Khan MUA, Stojanović GM, Abdullah MFB, Dolatshahi-Pirouz A, Marei HE, Ashammakhi N, Hasan A. Fundamental properties of smart hydrogels for tissue engineering applications: A review. Int J Biol Macromol 2024; 254:127882. [PMID: 37951446 DOI: 10.1016/j.ijbiomac.2023.127882] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
Tissue engineering is an advanced and potential biomedical approach to treat patients suffering from lost or failed an organ or tissue to repair and regenerate damaged tissues that increase life expectancy. The biopolymers have been used to fabricate smart hydrogels to repair damaged tissue as they imitate the extracellular matrix (ECM) with intricate structural and functional characteristics. These hydrogels offer desired and controllable qualities, such as tunable mechanical stiffness and strength, inherent adaptability and biocompatibility, swellability, and biodegradability, all crucial for tissue engineering. Smart hydrogels provide a superior cellular environment for tissue engineering, enabling the generation of cutting-edge synthetic tissues due to their special qualities, such as stimuli sensitivity and reactivity. Numerous review articles have presented the exceptional potential of hydrogels for various biomedical applications, including drug delivery, regenerative medicine, and tissue engineering. Still, it is essential to write a comprehensive review article on smart hydrogels that successfully addresses the essential challenging issues in tissue engineering. Hence, the recent development on smart hydrogel for state-of-the-art tissue engineering conferred progress, highlighting significant challenges and future perspectives. This review discusses recent advances in smart hydrogels fabricated from biological macromolecules and their use for advanced tissue engineering. It also provides critical insight, emphasizing future research directions and progress in tissue engineering.
Collapse
Affiliation(s)
- Muhammad Umar Aslam Khan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar.
| | - Goran M Stojanović
- Department of Electronics, Faculty of Technical Sciences, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Mohd Faizal Bin Abdullah
- Oral and Maxillofacial Surgery Unit, School of Dental Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kota Bharu, Kelantan, Malaysia; Oral and Maxillofacial Surgery Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kota Bharu, Kelantan, Malaysia.
| | | | - Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, College of Engineering and Human Medicine, Michigan State University, East Lansing, MI 48824, USA.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| |
Collapse
|
158
|
Xu J, Zheng B, Xie C, Zhao Y, Wu H, Wang Y, Guan X, Lei X, Liu D, Lou X, Chen X, Huang Y. Inhibition of FABP5 attenuates inflammatory bowel disease by modulating macrophage alternative activation. Biochem Pharmacol 2024; 219:115974. [PMID: 38081366 DOI: 10.1016/j.bcp.2023.115974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023]
Abstract
Fatty acid binding protein 5 (FABP5) is an intracellular chaperone of fatty acid molecules that regulates lipid metabolism and cell growth. However, its role in intestinal inflammation remains enigmatic. Through examination of human tissue samples and single-cell data, we observed a significant upregulation of FABP5 within the mucosa of patients afflicted with ulcerative colitis (UC) and Crohn's disease (CD), predominantly localized in intestinal macrophages. Herein, we investigate the regulation of FABP5-IN-1, a FABP5 inhibitor, on various cells of the gut in an inflammatory environment. Our investigations confirmed that FABP5 ameliorates DSS-induced colitis in mice by impeding the differentiation of macrophages into M1 macrophages in vitro and in vivo. Furthermore, following FABP5-IN-1 intervention, we observed a notable restoration of intestinal goblet cells and tuft cells, even under inflammatory conditions. Additionally, FABP5-IN-1 exhibits a protective effect against DSS-induced colitis by promoting the polarization of macrophages towards the M2 phenotype in vivo. In summary, FABP5-IN-1 confers protection against DSS-induced acute colitis through a multifaceted approach, encompassing the reduction of inflammatory macrophage infiltration, macrophage polarization, regulating Th17/Treg cells to play an anti-inflammatory role in IBD. The implications for IBD are underscored by the comprehensive in vivo and in vitro experiments presented in this article, thereby positioning FABP5 as a promising and novel therapeutic target for the treatment of IBD.
Collapse
Affiliation(s)
- Jingping Xu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Bolin Zheng
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, PR China
| | - Chunlan Xie
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen 361102, PR China
| | - Yao Zhao
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Hailun Wu
- First Affiliated Hospital, Nanchang University, Nanchang 330000, Jiangxi Province, PR China
| | - Yiting Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Xiaoli Guan
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Xintao Lei
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Dexin Liu
- Department of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou 362000, PR China
| | - Xiaoying Lou
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China
| | - Xiaohui Chen
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen 361003, PR China.
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510655, PR China; Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University,Guangzhou, Guangdong 510655, PR China.
| |
Collapse
|
159
|
Takase Y, Takahashi T. Method for Two-Dimensional Epithelial Monolayer Formation Derived from Mouse Three-Dimensional Small Intestinal Organoids. Methods Mol Biol 2024; 2749:73-84. [PMID: 38133775 DOI: 10.1007/978-1-0716-3609-1_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
The intestinal epithelium is composed of two distinct structures, namely, the villi and crypts. The base of the crypts contains intestinal stem cells (ISCs), which support the high regenerative capacity of the intestinal epithelium. With the establishment of the three-dimensional (3D) organoid culture method, the cellular and molecular mechanisms of differentiation, proliferation, and maintenance of ISCs have been widely analyzed. However, the sphere-like morphology of the 3D organoids prevents access to the apical side of the epithelium. To overcome this limitation, two-dimensional (2D) monolayer cultures derived from 3D organoids have been attempted; however, 2D culture methods for the mouse small intestine have not been well established. In this study, we developed a simple method that uses only commercially available materials, for the formation of 2D epithelial monolayers from mouse 3D small intestinal organoids. Using this method, confluent 2D epithelial monolayers were established within 4 days. This monolayer showed stable tight junction and included ISCs and differentiated intestinal cells. It also showed physiologically relevant transepithelial electrical resistance values. On the basis of these findings, this method opens a novel platform for analyzing the physiology of the intestinal epithelium, its interaction with microbes, and mechanisms of villus formation.
Collapse
Affiliation(s)
- Yuta Takase
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan.
| | - Toshio Takahashi
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| |
Collapse
|
160
|
Reyes-Rivera J, Grillo-Alvarado V, Soriano-López AE, García-Arrarás JE. Evidence of interactions among apoptosis, cell proliferation, and dedifferentiation in the rudiment during whole-organ intestinal regeneration in the sea cucumber. Dev Biol 2024; 505:99-109. [PMID: 37925124 PMCID: PMC11163280 DOI: 10.1016/j.ydbio.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 09/05/2023] [Accepted: 11/01/2023] [Indexed: 11/06/2023]
Abstract
Sea cucumbers have an extraordinary regenerative capability. Under stressful conditions, Holothuria glaberrima can eviscerate their internal organs, including the digestive tract. From the mesentery, a rudiment grows and gives rise to a new intestine within a few weeks. In the last decades, the cellular events that occur during intestinal regeneration have been characterized, including apoptosis, cell proliferation, and muscle cell dedifferentiation. Nevertheless, their contribution to the formation and early growth of the rudiment is still unknown. Furthermore, these cellular events' relationship and potential interdependence remain a mystery. Using modulators to inhibit apoptosis and cell proliferation, we tested whether rudiment growth or other regenerative cellular events like muscle cell dedifferentiation were affected. We found that inhibition of apoptosis by zVAD and cell proliferation by aphidicolin and mitomycin did not affect the overall size of the rudiment seven days post-evisceration (7-dpe). Interestingly, animals treated with aphidicolin showed higher levels of muscle cell dedifferentiation in the distal mesentery, which could act as a compensatory mechanism. On the other hand, inhibition of apoptosis led to a decrease in cell proliferation in the rudiment and a delay in the spatiotemporal progression of muscle cell dedifferentiation throughout the rudiment-mesentery structure. Our findings suggest that neither apoptosis nor cell proliferation significantly contributes to early rudiment growth during intestinal regeneration in the sea cucumber. Nevertheless, apoptosis may play an essential role in modulating cell proliferation in the rudiment (a process known as apoptosis-induced proliferation) and the timing for the progression of muscle cell dedifferentiation. These findings provide new insights into the role and relationship of cellular events during intestinal regeneration in an emerging regeneration model.
Collapse
Affiliation(s)
- Josean Reyes-Rivera
- Department of Biology, University of Puerto Rico, Río Piedras, PR, USA; Molecular and Cell Biology Department, University of California, Berkeley, CA, USA
| | | | | | | |
Collapse
|
161
|
Coutry N, Gasmi I, Herbert F, Jay P. Mechanisms of intestinal dysbiosis: new insights into tuft cell functions. Gut Microbes 2024; 16:2379624. [PMID: 39042424 PMCID: PMC11268228 DOI: 10.1080/19490976.2024.2379624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
Symbiosis between the host and intestinal microbial communities is essential for human health. Disruption in this symbiosis is linked to gastrointestinal diseases, including inflammatory bowel diseases, as well as extra-gastrointestinal diseases. Unbalanced gut microbiome or gut dysbiosis contributes in multiple ways to disease frequency, severity and progression. Microbiome taxonomic profiling and metabolomics approaches greatly improved our understanding of gut dysbiosis features; however, the precise mechanisms involved in gut dysbiosis establishment still need to be clarified. The aim of this review is to present new actors and mechanisms underlying gut dysbiosis formation following parasitic infection or in a context of altered Paneth cells, revealing the existence of a critical crosstalk between Paneth and tuft cells to control microbiome composition.
Collapse
Affiliation(s)
- Nathalie Coutry
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Imène Gasmi
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Fabien Herbert
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Philippe Jay
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Inserm, Montpellier, France
| |
Collapse
|
162
|
Rodriguez-Colman MJ, Dansen TB, Burgering BMT. FOXO transcription factors as mediators of stress adaptation. Nat Rev Mol Cell Biol 2024; 25:46-64. [PMID: 37710009 DOI: 10.1038/s41580-023-00649-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2023] [Indexed: 09/16/2023]
Abstract
The forkhead box protein O (FOXO, consisting of FOXO1, FOXO3, FOXO4 and FOXO6) transcription factors are the mammalian orthologues of Caenorhabditis elegans DAF-16, which gained notoriety for its capability to double lifespan in the absence of daf-2 (the gene encoding the worm insulin receptor homologue). Since then, research has provided many mechanistic details on FOXO regulation and FOXO activity. Furthermore, conditional knockout experiments have provided a wealth of data as to how FOXOs control development and homeostasis at the organ and organism levels. The lifespan-extending capabilities of DAF-16/FOXO are highly correlated with their ability to induce stress response pathways. Exogenous and endogenous stress, such as cellular redox stress, are considered the main drivers of the functional decline that characterizes ageing. Functional decline often manifests as disease, and decrease in FOXO activity indeed negatively impacts on major age-related diseases such as cancer and diabetes. In this context, the main function of FOXOs is considered to preserve cellular and organismal homeostasis, through regulation of stress response pathways. Paradoxically, the same FOXO-mediated responses can also aid the survival of dysfunctional cells once these eventually emerge. This general property to control stress responses may underlie the complex and less-evident roles of FOXOs in human lifespan as opposed to model organisms such as C. elegans.
Collapse
Affiliation(s)
| | - Tobias B Dansen
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Boudewijn M T Burgering
- Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
- Oncode Institute, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
163
|
Li C, Sun Y, He T, Lu Y, Szeto IMY, Duan S, Zhang Y, Liu B, Zhang Y, Zhang W, He J, Li Y. Synergistic effect of lactoferrin and osteopontin on intestinal barrier injury. Int J Biol Macromol 2023; 253:127416. [PMID: 37838132 DOI: 10.1016/j.ijbiomac.2023.127416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
Several studies indicate that the disruption of the intestinal epithelial barrier can lead to inflammatory bowel disease (IBD). Recent evidence has increasingly demonstrated that lactoferrin (LF) and osteopontin (OPN) can alleviate intestinal barrier injury. However, the potential synergistic effects of these two proteins and the mechanisms underlying their effects remain unclear. To address this question, we developed a lipopolysaccharide-induced intestinal barrier injury model in C57BL/6 N mice. Our findings demonstrated that the combination of LF and OPN at a 1:5 ratio exerts the strongest protective effect on the intestinal barrier, and it is more effective than LF or OPN alone. This protection is evidenced by the decrease in serum diamine oxidase (DAO) activity (1.66-fold decrease) and D-lactic content (1.51-fold decrease) and the reduced rate of FITC-labeled glucan transport across the jejunum (3.18-fold decrease). Moreover, the protein combination significantly promoted villi length (1.66-fold increase) and crypt depth (1.57-fold increase), improved tight junction protein structure and expression, and boosted the number of absorptive cells (4.34-fold increase) in the intestinal epithelium. Furthermore, the combination promoted crypt cell proliferation and differentiation via Notch signaling. In summary, our findings provide scientific evidence supporting the use of dietary intervention strategies for preventing IBD.
Collapse
Affiliation(s)
- Chuangang Li
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yanan Sun
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Tingchao He
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot 010110, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yao Lu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China; National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Sufang Duan
- Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot 010110, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yifan Zhang
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Biao Liu
- College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China; Inner Mongolia Yili Industrial Group, Co. Ltd., Yili Maternal and Infant Nutrition Institute (YMINI), Beijing 100070, China
| | - Yiran Zhang
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Wen Zhang
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Yixuan Li
- Key Laboratory of Functional Daily, Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
164
|
Stolovich-Rain M, Fridlich O, Azulai S, Klochendler A, Anzi S, Magenheim J, Stein I, Mushasha F, Glaser B, Pikarsky E, Ben-Zvi D, Dor Y. Extensive elimination of acinar cells during normal postnatal pancreas growth. Cell Rep 2023; 42:113457. [PMID: 37995187 DOI: 10.1016/j.celrep.2023.113457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 09/01/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023] Open
Abstract
While programmed cell death plays important roles during morphogenetic stages of development, post-differentiation organ growth is considered an efficient process whereby cell proliferation increases cell number. Here we demonstrate that early postnatal growth of the pancreas unexpectedly involves massive acinar cell elimination. Measurements of cell proliferation and death in the human pancreas in comparison to the actual increase in cell number predict daily elimination of 0.7% of cells, offsetting 88% of cell formation over the first year of life. Using mouse models, we show that death is associated with mitosis, through a failure of dividing cells to generate two viable daughters. In p53-deficient mice, acinar cell death and proliferation are reduced, while organ size is normal, suggesting that p53-dependent developmental apoptosis triggers compensatory proliferation. We propose that excess cell turnover during growth of the pancreas, and presumably other organs, facilitates robustness to perturbations and supports maintenance of tissue architecture.
Collapse
Affiliation(s)
- Miri Stolovich-Rain
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ori Fridlich
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Shira Azulai
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Agnes Klochendler
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Shira Anzi
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Judith Magenheim
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ilan Stein
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel; Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Fatima Mushasha
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Benjamin Glaser
- Endocrinology and Metabolism Service, Department of Internal Medicine, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Eli Pikarsky
- The Concern Foundation Laboratories at The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, The Hebrew University, Jerusalem, Israel; Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Danny Ben-Zvi
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Yuval Dor
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada (IMRIC), The Hebrew University-Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
165
|
Funk MC, Gleixner JG, Heigwer F, Vonficht D, Valentini E, Aydin Z, Tonin E, Del Prete S, Mahara S, Throm Y, Hetzer J, Heide D, Stegle O, Odom DT, Feldmann A, Haas S, Heikenwalder M, Boutros M. Aged intestinal stem cells propagate cell-intrinsic sources of inflammaging in mice. Dev Cell 2023; 58:2914-2929.e7. [PMID: 38113852 DOI: 10.1016/j.devcel.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 05/03/2023] [Accepted: 11/13/2023] [Indexed: 12/21/2023]
Abstract
Low-grade chronic inflammation is a hallmark of ageing, associated with impaired tissue function and disease development. However, how cell-intrinsic and -extrinsic factors collectively establish this phenotype, termed inflammaging, remains poorly understood. We addressed this question in the mouse intestinal epithelium, using mouse organoid cultures to dissect stem cell-intrinsic and -extrinsic sources of inflammaging. At the single-cell level, we found that inflammaging is established differently along the crypt-villus axis, with aged intestinal stem cells (ISCs) strongly upregulating major histocompatibility complex class II (MHC-II) genes. Importantly, the inflammaging phenotype was stably propagated by aged ISCs in organoid cultures and associated with increased chromatin accessibility at inflammation-associated loci in vivo and ex vivo, indicating cell-intrinsic inflammatory memory. Mechanistically, we show that the expression of inflammatory genes is dependent on STAT1 signaling. Together, our data identify that intestinal inflammaging in mice is promoted by a cell-intrinsic mechanism, stably propagated by ISCs, and associated with a disbalance in immune homeostasis.
Collapse
Affiliation(s)
- Maja C Funk
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Jan G Gleixner
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), Division of Computational Genomics and Systems Genetics, 69120 Heidelberg, Germany; Genome Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69117 Heidelberg, Germany
| | - Florian Heigwer
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany; Department of Life Sciences and Engineering, University of Applied Sciences Bingen, 55411 Bingen am Rhein, Germany
| | - Dominik Vonficht
- Faculty of Biosciences, Heidelberg University, 69117 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine, (HI-STEM gGmbH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), Division of Stem Cells and Cancer, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Erica Valentini
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Zeynep Aydin
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Elena Tonin
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Stefania Del Prete
- German Cancer Research Center (DKFZ), Division Regulatory Genomics and Cancer Evolution, 69120 Heidelberg, Germany
| | - Sylvia Mahara
- German Cancer Research Center (DKFZ), Junior Research Group Mechanisms of Genome Control, 69120 Heidelberg, Germany
| | - Yannick Throm
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany
| | - Jenny Hetzer
- German Cancer Research Center (DKFZ), Division Chronic Inflammation and Cancer, 69120 Heidelberg, Germany
| | - Danijela Heide
- German Cancer Research Center (DKFZ), Division Chronic Inflammation and Cancer, 69120 Heidelberg, Germany
| | - Oliver Stegle
- German Cancer Research Center (DKFZ), Division of Computational Genomics and Systems Genetics, 69120 Heidelberg, Germany; Genome Biology Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Duncan T Odom
- German Cancer Research Center (DKFZ), Division Regulatory Genomics and Cancer Evolution, 69120 Heidelberg, Germany
| | - Angelika Feldmann
- German Cancer Research Center (DKFZ), Junior Research Group Mechanisms of Genome Control, 69120 Heidelberg, Germany
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine, (HI-STEM gGmbH), 69120 Heidelberg, Germany; German Cancer Research Center (DKFZ), Division of Stem Cells and Cancer, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, 10178 Berlin, Germany; Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, 10115 Berlin, Germany
| | - Mathias Heikenwalder
- German Cancer Research Center (DKFZ), Division Chronic Inflammation and Cancer, 69120 Heidelberg, Germany; M3 Research Center, Medical Faculty Tübingen, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
| | - Michael Boutros
- German Cancer Research Center (DKFZ), Division Signaling and Functional Genomics, Heidelberg University, BioQuant & Department of Cell and Molecular Biology, Medical Faculty Mannheim, Heidelberg University, Institute for Human Genetics, Medical Faculty Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
166
|
Farhadipour M, Arnauts K, Clarysse M, Thijs T, Liszt K, Van der Schueren B, Ceulemans LJ, Deleus E, Lannoo M, Ferrante M, Depoortere I. SCFAs switch stem cell fate through HDAC inhibition to improve barrier integrity in 3D intestinal organoids from patients with obesity. iScience 2023; 26:108517. [PMID: 38125020 PMCID: PMC10730380 DOI: 10.1016/j.isci.2023.108517] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/25/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Stem cells are a keystone of intestinal homeostasis, but their function could be shifted during energy imbalance or by crosstalk with microbial metabolites in the stem cell niche. This study reports the effect of obesity and microbiota-derived short-chain fatty acids (SCFAs) on intestinal stem cell (ISC) fate in human crypt-derived intestinal organoids (enteroids). ISC fate decision was impaired in obesity, resulting in smaller enteroids with less outward protruding crypts. Our key finding is that SCFAs switch ISC commitment to the absorptive enterocytes, resulting in reduced intestinal permeability in obese enteroids. Mechanistically, SCFAs act as HDAC inhibitors in stem cells to enhance Notch signaling, resulting in transcriptional activation of the Notch target gene HES1 to promote enterocyte differentiation. In summary, targeted reprogramming of ISC fate, using HDAC inhibitors, may represent a potential, robust therapeutic strategy to improve gut integrity in obesity.
Collapse
Affiliation(s)
- Mona Farhadipour
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
| | - Kaline Arnauts
- Inflammatory Bowel Disease, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
| | - Mathias Clarysse
- Leuven Intestinal Failure and Transplantation (LIFT) Center, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Theo Thijs
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
| | - Kathrin Liszt
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
| | | | - Laurens J. Ceulemans
- Leuven Intestinal Failure and Transplantation (LIFT) Center, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ellen Deleus
- Department of Abdominal Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Matthias Lannoo
- Department of Abdominal Surgery, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Marc Ferrante
- Inflammatory Bowel Disease, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
167
|
Li Y, Li W, Zeng Z, Han Y, Chen Q, Dong X, Wang Z, Feng S, Cao W. Lasso peptide MccY alleviates non-typhoidal salmonellae-induced mouse gut inflammation via regulation of intestinal barrier function and gut microbiota. Microbiol Spectr 2023; 11:e0178423. [PMID: 37819128 PMCID: PMC10714986 DOI: 10.1128/spectrum.01784-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 10/13/2023] Open
Abstract
IMPORTANCE Diseases caused by Enterobacteriaceae multidrug-resistant strains have become increasingly difficult to manage. It is necessary to verify the new antibacterial drug MccY effect on non-typhoid Salmonella infection in mice since it is regarded as a promising microcin. The results demonstrated that MccY has a potential therapeutic application value in the protection against Salmonella-induced intestinal damage and alleviating related intestinal dysbiosis and metabolic disorders. MccY could be a promising candidate as an antimicrobial or anti-inflammatory agent for treating infectious diseases.
Collapse
Affiliation(s)
- Yu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Wenjing Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhiwei Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yu Han
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qinxi Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinyi Dong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zepeng Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Saixiang Feng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangdong, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
| | - Weisheng Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, Guangdong, China
- Key Laboratory of Zoonosis of Ministry of Agriculture and Rural Affairs, Guangzhou, China
- Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture and Rural Affairs, Guangzhou, China
- National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, Guangzhou, China
| |
Collapse
|
168
|
Qin X, Cardoso Rodriguez F, Sufi J, Vlckova P, Claus J, Tape CJ. An oncogenic phenoscape of colonic stem cell polarization. Cell 2023; 186:5554-5568.e18. [PMID: 38065080 DOI: 10.1016/j.cell.2023.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/14/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023]
Abstract
Cancer cells are regulated by oncogenic mutations and microenvironmental signals, yet these processes are often studied separately. To functionally map how cell-intrinsic and cell-extrinsic cues co-regulate cell fate, we performed a systematic single-cell analysis of 1,107 colonic organoid cultures regulated by (1) colorectal cancer (CRC) oncogenic mutations, (2) microenvironmental fibroblasts and macrophages, (3) stromal ligands, and (4) signaling inhibitors. Multiplexed single-cell analysis revealed a stepwise epithelial differentiation phenoscape dictated by combinations of oncogenes and stromal ligands, spanning from fibroblast-induced Clusterin (CLU)+ revival colonic stem cells (revCSCs) to oncogene-driven LRIG1+ hyper-proliferative CSCs (proCSCs). The transition from revCSCs to proCSCs is regulated by decreasing WNT3A and TGF-β-driven YAP signaling and increasing KRASG12D or stromal EGF/Epiregulin-activated MAPK/PI3K flux. We find that APC loss and KRASG12D collaboratively limit access to revCSCs and disrupt stromal-epithelial communication-trapping epithelia in the proCSC fate. These results reveal that oncogenic mutations dominate homeostatic differentiation by obstructing cell-extrinsic regulation of cell-fate plasticity.
Collapse
Affiliation(s)
- Xiao Qin
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Ferran Cardoso Rodriguez
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Jahangir Sufi
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Petra Vlckova
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Jeroen Claus
- Phospho Biomedical Animation, The Greenhouse Studio 6, London N17 9QU, UK
| | - Christopher J Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
169
|
Zhang F, Chen M, Liu X, Ji X, Li S, Jin E. New insights into the unfolded protein response (UPR)-anterior gradient 2 (AGR2) pathway in the regulation of intestinal barrier function in weaned piglets. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:225-232. [PMID: 38033605 PMCID: PMC10685161 DOI: 10.1016/j.aninu.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/05/2023] [Accepted: 08/11/2023] [Indexed: 12/02/2023]
Abstract
Sustained dysfunction of the intestinal barrier caused by early weaning is a major factor that induces postweaning diarrhea in weaned piglets. In both healthy and diseased states, the intestinal barrier is regulated by goblet cells. Alterations in the characteristics of goblet cells are linked to intestinal barrier dysfunction and inflammatory conditions during pathogenic infections. In this review, we summarize the current understanding of the mechanisms of the unfolded protein response (UPR) and anterior gradient 2 (AGR2) in maintaining intestinal barrier function and how modifications to these systems affect mucus barrier characteristics and goblet cell dysregulation. We highlight a novel mechanism underlying the UPR-AGR2 pathway, which affects goblet cell differentiation and maturation and the synthesis and secretion of mucin by regulating epidermal growth factor receptor and mucin 2. This study provides a theoretical basis and new insights into the regulation of intestinal health in weaned piglets.
Collapse
Affiliation(s)
- Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Mengxian Chen
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xiaodan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Shenghe Li
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| |
Collapse
|
170
|
Endo R, Sugimoto S, Shirosaki K, Kato H, Wada M, Kanai T, Sato T. Clinical challenges of short bowel syndrome and the path forward for organoid-based regenerative medicine. Regen Ther 2023; 24:64-73. [PMID: 37868721 PMCID: PMC10584670 DOI: 10.1016/j.reth.2023.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/25/2023] [Accepted: 06/01/2023] [Indexed: 10/24/2023] Open
Abstract
Short bowel syndrome (SBS) is a rare condition, the main symptom of which is malabsorption following extensive resection of the small intestine. Treatment for SBS is mainly supportive, consisting of supplementation, prevention and treatment of complications, and promotion of intestinal adaptation. While development of parenteral nutrition and drugs promoting intestinal adaptation has improved clinical outcomes, the prognosis of patients with SBS remains poor. Intestinal transplantation is the only curative therapy but its outcome is unsatisfactory. In the absence of definitive therapy, novel treatment is urgently needed. With the advent of intestinal organoids, research on the intestine has developed remarkably in recent years. Concepts such as the "tissue-engineered small intestine" and "small intestinalized colon," which create a functional small intestine by combining organoids with other technologies, are potentially novel regenerative therapeutic approaches for SBS. Although they are still under development and there are substantial issues to be resolved, the problems that have prevented establishment of the complex function and structure of the small intestine are gradually being overcome. This review discusses the current treatments for SBS, the fundamentals of the intestine and organoids, the current status of these new technologies, and future perspectives.
Collapse
Affiliation(s)
- Ryoma Endo
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Shinya Sugimoto
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Koji Shirosaki
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Pediatric Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Hirochika Kato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Motoshi Wada
- Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
171
|
Wang D, Jiang Q, Dong Z, Meng T, Hu F, Wang J, Yuan H. Nanocarriers transport across the gastrointestinal barriers: The contribution to oral bioavailability via blood circulation and lymphatic pathway. Adv Drug Deliv Rev 2023; 203:115130. [PMID: 37913890 DOI: 10.1016/j.addr.2023.115130] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/03/2023]
Abstract
Oral administration is the preferred route of drug delivery in clinical practice due to its noninvasiveness, safety, convenience, and high patient compliance. The gastrointestinal tract (GIT) plays a crucial role in facilitating the targeted delivery of oral drugs. However, the GIT presents multiple barriers that impede drug absorption, including the gastric barrier in the stomach and the mucus and epithelial barriers in the intestine. In recent decades, nanotechnology has emerged as a promising approach for overcoming these challenges by utilizing nanocarrier-based drug delivery systems such as liposomes, micelles, polymeric nanoparticles, solid lipid nanoparticles, and inorganic nanoparticles. Encapsulating drugs within nanocarriers not only protects them from degradation but also enhances their transport and absorption across the GIT, ultimately improving oral bioavailability. The aim of this review is to elucidate the mechanisms underlying nanocarrier-mediated transportation across the GIT into systemic circulation via both the blood circulation and lymphatic pathway.
Collapse
Affiliation(s)
- Ding Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Qi Jiang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Zhefan Dong
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China
| | - Jianwei Wang
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, 310009, PR China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, PR China; China Jinhua Institute of Zhejiang University, Jinhua 321299, PR China.
| |
Collapse
|
172
|
Wei X, Yu S, Zhang T, Liu L, Wang X, Wang X, Chan YS, Wang Y, Meng S, Chen YG. MicroRNA-200 Loaded Lipid Nanoparticles Promote Intestinal Epithelium Regeneration in Canonical MicroRNA-Deficient Mice. ACS NANO 2023; 17:22901-22915. [PMID: 37939210 PMCID: PMC10690841 DOI: 10.1021/acsnano.3c08030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/10/2023]
Abstract
Intestinal epithelium undergoes regeneration after injuries, and the disruption of this process can lead to inflammatory bowel disease and tumorigenesis. Intestinal stem cells (ISCs) residing in the crypts are crucial for maintaining the intestinal epithelium's homeostasis and promoting regeneration upon injury. However, the precise role of DGCR8, a critical component in microRNA (miRNA) biogenesis, in intestinal regeneration remains poorly understood. In this study, we provide compelling evidence demonstrating the indispensable role of epithelial miRNAs in the regeneration of the intestine in mice subjected to 5-FU or irradiation-induced injury. Through a comprehensive pooled screen of miRNA function in Dgcr8-deficient organoids, we observe that the loss of the miR-200 family leads to the hyperactivation of the p53 pathway, thereby reducing ISCs and impairing epithelial regeneration. Notably, downregulation of the miR-200 family and hyperactivation of the p53 pathway are verified in colonic tissues from patients with active ulcerative colitis (UC). Most importantly, the transient supply of miR-200 through the oral delivery of lipid nanoparticles (LNPs) carrying miR-200 restores ISCs and promotes intestinal regeneration in mice following acute injury. Our study implies the miR-200/p53 pathway as a promising therapeutic target for active UC patients with diminished levels of the miR-200 family. Furthermore, our findings suggest that the clinical application of LNP-miRNAs could enhance the efficacy, safety, and acceptability of existing therapeutic modalities for intestinal diseases.
Collapse
Affiliation(s)
- Xiyang Wei
- Guangzhou
Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou
National Laboratory, Guangzhou 510005, China
| | - Shicheng Yu
- Guangzhou
Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou
National Laboratory, Guangzhou 510005, China
| | | | - Liansheng Liu
- Guangzhou
Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou
National Laboratory, Guangzhou 510005, China
| | - Xu Wang
- Guangzhou
National Laboratory, Guangzhou 510005, China
| | - Xiaodan Wang
- The
State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for
Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yun-Shen Chan
- Guangzhou
National Laboratory, Guangzhou 510005, China
| | - Yangming Wang
- Institute
of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Shu Meng
- Guangzhou
National Laboratory, Guangzhou 510005, China
| | - Ye-Guang Chen
- Guangzhou
National Laboratory, Guangzhou 510005, China
- The
State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for
Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
- School
of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
173
|
Colozza G, Lee H, Merenda A, Wu SHS, Català-Bordes A, Radaszkiewicz TW, Jordens I, Lee JH, Bamford AD, Farnhammer F, Low TY, Maurice MM, Bryja V, Kim J, Koo BK. Intestinal Paneth cell differentiation relies on asymmetric regulation of Wnt signaling by Daam1/2. SCIENCE ADVANCES 2023; 9:eadh9673. [PMID: 38000028 PMCID: PMC10672176 DOI: 10.1126/sciadv.adh9673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023]
Abstract
The mammalian intestine is one of the most rapidly self-renewing tissues, driven by stem cells residing at the crypt bottom. Paneth cells form a major element of the niche microenvironment providing various growth factors to orchestrate intestinal stem cell homeostasis, such as Wnt3. Different Wnt ligands can selectively activate β-catenin-dependent (canonical) or -independent (noncanonical) signaling. Here, we report that the Dishevelled-associated activator of morphogenesis 1 (Daam1) and its paralogue Daam2 asymmetrically regulate canonical and noncanonical Wnt (Wnt/PCP) signaling. Daam1/2 interacts with the Wnt inhibitor RNF43, and Daam1/2 double knockout stimulates canonical Wnt signaling by preventing RNF43-dependent degradation of the Wnt receptor, Frizzled (Fzd). Single-cell RNA sequencing analysis revealed that Paneth cell differentiation is impaired by Daam1/2 depletion because of defective Wnt/PCP signaling. Together, we identified Daam1/2 as an unexpected hub molecule coordinating both canonical and noncanonical Wnt, which is fundamental for specifying an adequate number of Paneth cells.
Collapse
Affiliation(s)
- Gabriele Colozza
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Heetak Lee
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
- Center for Genome Engineering, Institute for Basic Science, 55, Expo-ro, Yuseong-gu, Daejeon 34126, Republic of Korea
| | | | - Szu-Hsien Sam Wu
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Andrea Català-Bordes
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
| | - Tomasz W. Radaszkiewicz
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Ingrid Jordens
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Ji-Hyun Lee
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
- Center for Genome Engineering, Institute for Basic Science, 55, Expo-ro, Yuseong-gu, Daejeon 34126, Republic of Korea
| | - Aileen-Diane Bamford
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Fiona Farnhammer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
- Division of Metabolism and Division of Oncology, University Children’s Hospital Zurich and Children’s Research Center, University of Zurich, 8032 Zurich, Switzerland
| | - Teck Yew Low
- UKM Medical Molecular Biology Institute (UMBI), University Kebangsaan Malaysia (UKM), Jalan Yaacob Latiff, Bandar Tun Razak, 56000 Kuala Lumpur, Malaysia
| | - Madelon M. Maurice
- Oncode Institute and Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Vítězslav Bryja
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
- Department of Cytokinetics, Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czech Republic
| | - Jihoon Kim
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), 1030 Vienna, Austria
- Center for Genome Engineering, Institute for Basic Science, 55, Expo-ro, Yuseong-gu, Daejeon 34126, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| |
Collapse
|
174
|
Jolly A, Fernández B, Mundo SL, Elguezabal N. Modeling Paratuberculosis in Laboratory Animals, Cells, or Tissues: A Focus on Their Applications for Pathogenesis, Diagnosis, Vaccines, and Therapy Studies. Animals (Basel) 2023; 13:3553. [PMID: 38003170 PMCID: PMC10668694 DOI: 10.3390/ani13223553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Paratuberculosis is a chronic granulomatous enteritis caused by Mycobacterium avium subsp. Paratuberculosis that affects a wide variety of domestic and wild animals. It is considered one of the diseases with the highest economic impact on the ruminant industry. Despite many efforts and intensive research, paratuberculosis control still remains controversial, and the existing diagnostic and immunoprophylactic tools have great limitations. Thus, models play a crucial role in understanding the pathogenesis of infection and disease, and in testing novel vaccine candidates. Ruminant animal models can be restricted by several reasons, related to space requirements, the cost of the animals, and the maintenance of the facilities. Therefore, we review the potential and limitations of the different experimental approaches currently used in paratuberculosis research, focusing on laboratory animals and cell-based models. The aim of this review is to offer a vision of the models that have been used, and what has been achieved or discovered with each one, so that the reader can choose the best model to answer their scientific questions and prove their hypotheses. Also, we bring forward new approaches that we consider worth exploring in the near future.
Collapse
Affiliation(s)
- Ana Jolly
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
| | - Bárbara Fernández
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
- Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
- Instituto de Investigación y Tecnología en Reproducción Animal (INITRA), Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
| | - Silvia Leonor Mundo
- Cátedra de Inmunología, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina; (B.F.); (S.L.M.)
- Instituto de Investigaciones en Producción Animal (INPA), CONICET-Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
- Instituto de Investigación y Tecnología en Reproducción Animal (INITRA), Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Av. Chorroarín 280, Buenos Aires C1427CWO, Argentina
| | - Natalia Elguezabal
- Departamento de Sanidad Animal, NEIKER-Instituto Vasco de Investigación y Desarrollo Agrario-Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
| |
Collapse
|
175
|
Vemuri K, Kumar S, Chen L, Verzi MP. Dynamic RNA Polymerase II Recruitment Drives Differentiation of the Intestine under the direction of HNF4. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566322. [PMID: 37986803 PMCID: PMC10659318 DOI: 10.1101/2023.11.08.566322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Terminal differentiation requires a massive restructuring of the transcriptome. During intestinal differentiation, the expression patterns of nearly 4000 genes are altered as cells transition from progenitor cells in crypts to differentiated cells in villi. We identified dynamic recruitment of RNA Polymerase II (Pol II) to gene promoters as the primary driver of transcriptomic shifts during intestinal differentiation in vivo. Changes in enhancer-promoter looping interactions accompany dynamic Pol II recruitment and are dependent upon HNF4, a pro-differentiation transcription factor. Using genetic loss-of- function, ChIP-seq and IP mass spectrometry, we demonstrate that HNF4 collaborates with chromatin remodelers and loop-stabilizing proteins and facilitates Pol II recruitment at hundreds of genes pivotal to differentiation. We also explore alternate mechanisms which drive differentiation gene expression and find pause-release of Pol II and post- transcriptional mRNA stability regulate smaller subsets of differentially expressed genes. These studies provide insights into the mechanisms of differentiation in a renewing adult tissue.
Collapse
Affiliation(s)
- Kiranmayi Vemuri
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
| | - Sneha Kumar
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
| | - Lei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| | - Michael P. Verzi
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08903, USA
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition & Health, Rutgers University, New Brunswick, NJ 08901, USA
- NIEHS Center for Environmental Exposures and Disease (CEED), Rutgers EOHSI Piscataway, NJ 08854, USA
- Lead Contact
| |
Collapse
|
176
|
Fritsch SD, Sukhbaatar N, Gonzales K, Sahu A, Tran L, Vogel A, Mazic M, Wilson JL, Forisch S, Mayr H, Oberle R, Weiszmann J, Brenner M, Vanhoutte R, Hofmann M, Pirnes-Karhu S, Magnes C, Kühnast T, Weckwerth W, Bock C, Klavins K, Hengstschläger M, Moissl-Eichinger C, Schabbauer G, Egger G, Pirinen E, Verhelst SHL, Weichhart T. Metabolic support by macrophages sustains colonic epithelial homeostasis. Cell Metab 2023; 35:1931-1943.e8. [PMID: 37804836 DOI: 10.1016/j.cmet.2023.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 06/23/2023] [Accepted: 09/14/2023] [Indexed: 10/09/2023]
Abstract
The intestinal epithelium has a high turnover rate and constantly renews itself through proliferation of intestinal crypt cells, which depends on insufficiently characterized signals from the microenvironment. Here, we showed that colonic macrophages were located directly adjacent to epithelial crypt cells in mice, where they metabolically supported epithelial cell proliferation in an mTORC1-dependent manner. Specifically, deletion of tuberous sclerosis complex 2 (Tsc2) in macrophages activated mTORC1 signaling that protected against colitis-induced intestinal damage and induced the synthesis of the polyamines spermidine and spermine. Epithelial cells ingested these polyamines and rewired their cellular metabolism to optimize proliferation and defense. Notably, spermine directly stimulated proliferation of colon epithelial cells and colon organoids. Genetic interference with polyamine production in macrophages altered global polyamine levels in the colon and modified epithelial cell proliferation. Our results suggest that macrophages act as "commensals" that provide metabolic support to promote efficient self-renewal of the colon epithelium.
Collapse
Affiliation(s)
| | - Nyamdelger Sukhbaatar
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Karine Gonzales
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Alishan Sahu
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Department of Pathology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| | - Andrea Vogel
- Institute for Vascular Biology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Mario Mazic
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Jayne Louise Wilson
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Stephan Forisch
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Hannah Mayr
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Raimund Oberle
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Jakob Weiszmann
- Department of Ecogenomics and Systems Biology, University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Martin Brenner
- Department of Ecogenomics and Systems Biology, University of Vienna, Vienna, Austria; Department of Pharmaceutical Sciences/ Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Roeland Vanhoutte
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Melanie Hofmann
- Institute for Vascular Biology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Sini Pirnes-Karhu
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Christoph Magnes
- HEALTH-Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft mbH, Graz, Austria
| | - Torben Kühnast
- Diagnostic and Research Department of Microbiology, Hygiene and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Wolfram Weckwerth
- Department of Ecogenomics and Systems Biology, University of Vienna, Vienna, Austria; Vienna Metabolomics Center (VIME), University of Vienna, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Institute of Artificial Intelligence, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Kristaps Klavins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Markus Hengstschläger
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria
| | - Christine Moissl-Eichinger
- Diagnostic and Research Department of Microbiology, Hygiene and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Gernot Schabbauer
- Institute for Vascular Biology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute Applied Diagnostics (LBI AD), Vienna, Austria
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Research Unit of Biomedicine and Internal Medicine, Faculty of Medicine, University of Oulu, Oulu, Finland; Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Steven H L Verhelst
- Laboratory of Chemical Biology, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Thomas Weichhart
- Center for Pathobiochemsitry & Genetics, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
177
|
Abo H, Sultana MF, Kawashima H. Dual function of angiogenin-4 inducing intestinal stem cells and apoptosis. Front Cell Dev Biol 2023; 11:1181145. [PMID: 38020881 PMCID: PMC10651741 DOI: 10.3389/fcell.2023.1181145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
The intestinal epithelium is the first line of host defense, and its homeostasis is dependent on soluble factors that comprise the crypt niche. Antimicrobial proteins are one of the mediators to maintain gut homeostasis. Angiogenin-4 (Ang4) is a member of the ribonuclease A superfamily and plays a pivotal role in antimicrobial activity against gut microbiota. However, the functions of Ang4 within the intestinal crypt niche, particularly its involvement in the development of intestinal epithelial cells (IECs), remain unknown. Here, we demonstrate that Ang4 plays a significant role in maintaining Lgr5+ intestinal stem cells (ISCs) and induces apoptosis of IECs in a concentration-dependent manner. We revealed that Ang4 is highly expressed by Paneth cells in the small intestine, as well as regenerating islet-derived family member-4 (Reg4) expressing goblet cells in the colon, and both cell subsets highly contribute to ISC maintenance. Functional analysis using intestinal organoids revealed that Ang4 induces Wnt and Notch signaling, increases Lgr5+ stem cell expansion, and promotes organoid growth. Furthermore, high concentrations of Ang4 induced apoptosis in the IEC cell line and organoids. Collectively, we propose that Ang4 is a dual functional protein and is a novel member of the crypt niche factor that promotes the expansion of ISCs and induces apoptosis.
Collapse
Affiliation(s)
- Hirohito Abo
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| | - Mst. Farzana Sultana
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
- Department of Pharmacy, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Hiroto Kawashima
- Laboratory of Microbiology and Immunology, Graduate School of Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
178
|
Tian F, Chen T, Xu W, Fan Y, Feng X, Huang Q, Chen J. Curcumin Compensates GLP-1 Deficiency via the Microbiota-Bile Acids Axis and Modulation in Functional Crosstalk between TGR5 and FXR in ob/ob Mice. Mol Nutr Food Res 2023; 67:e2300195. [PMID: 37712101 DOI: 10.1002/mnfr.202300195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/22/2023] [Indexed: 09/16/2023]
Abstract
SCOPE Glucagon-like peptide-1 (GLP-1) deficiency occurs in obesity-related pathologies due to defects in the intestinal lumen. And expanding the L-cell population has emerged as a promising avenue to elevate GLP-1 secretion to tackle metabolic disorders. Curcumin (Cur), the principal active component of spice turmeric, possesses well-established anti-obesity properties. To clarify, the study investigates whether Cur promotes GLP-1 secretion built upon the L-cell expansion. METHODS AND RESULTS Cur (60 mg kg-1 ) is administered orally to male ob/ob mice for 8 weeks. Cur ameliorates obesity and impaires glucose tolerance through increasing energy expenditure in ob/ob mice, accompanied by the maintenance of crypt architecture and gut permeability. It refines the microbial structure and bile acid (BA) profiles, resulting in deoxycholic acid (DCA) accumulation by weakening the enrichment of Lactobacillus. Further analyses show radically different properties of Cur on the intestine function of TGR5 and FXR (i.e., activation and repression). Cur amplifies L-cell number to promote GLP-1 secretion in ob/ob mice. CONCLUSIONS The findings suggest that Cur may act as a natural TGR5 agonist and FXR antagonist to improve obesity by enhancing GLP-1 release from L-cell expansion via the gut microbiota-BAs-TGR5/FXR axis, and it may serve as a promising therapeutic agent to compensate obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Fengyuan Tian
- General Practice, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
| | - Tianxi Chen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
| | - Wangda Xu
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
| | - Yichang Fan
- First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
| | - Xiaohong Feng
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
| | - Qi Huang
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
| | - Jie Chen
- Department of Endocrinology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310000, P. R. China
| |
Collapse
|
179
|
Boers R, Boers J, Tan B, van Leeuwen ME, Wassenaar E, Sanchez EG, Sleddens E, Tenhagen Y, Mulugeta E, Laven J, Creyghton M, Baarends W, van IJcken WFJ, Gribnau J. Retrospective analysis of enhancer activity and transcriptome history. Nat Biotechnol 2023; 41:1582-1592. [PMID: 36823354 PMCID: PMC10635829 DOI: 10.1038/s41587-023-01683-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 01/20/2023] [Indexed: 02/25/2023]
Abstract
Cell state changes in development and disease are controlled by gene regulatory networks, the dynamics of which are difficult to track in real time. In this study, we used an inducible DCM-RNA polymerase subunit b fusion protein which labels active genes and enhancers with a bacterial methylation mark that does not affect gene transcription and is propagated in S-phase. This DCM-RNA polymerase fusion protein enables transcribed genes and active enhancers to be tagged and then examined at later stages of development or differentiation. We apply this DCM-time machine (DCM-TM) technology to study intestinal homeostasis, revealing rapid and coordinated activation of enhancers and nearby genes during enterocyte differentiation. We provide new insights in absorptive-secretory lineage decision-making in intestinal stem cell (ISC) differentiation and show that ISCs retain a unique chromatin landscape required to maintain ISC identity and delineate future expression of differentiation-associated genes. DCM-TM has wide applicability in tracking cell states, providing new insights in the regulatory networks underlying cell state changes.
Collapse
Affiliation(s)
- Ruben Boers
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Joachim Boers
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Beatrice Tan
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marieke E van Leeuwen
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Evelyne Wassenaar
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Erlantz Gonzalez Sanchez
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Esther Sleddens
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Yasha Tenhagen
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Eskeatnaf Mulugeta
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Joop Laven
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Menno Creyghton
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Willy Baarends
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Wilfred F J van IJcken
- Erasmus Center for Biomics, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Joost Gribnau
- Department of Developmental Biology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands.
| |
Collapse
|
180
|
Kalashyan M, Raghunathan K, Oller H, Bayer MT, Jimenez L, Roland JT, Kolobova E, Hagen SJ, Goldsmith JD, Shub MD, Goldenring JR, Kaji I, Thiagarajah JR. Patient-derived enteroids provide a platform for the development of therapeutic approaches in microvillus inclusion disease. J Clin Invest 2023; 133:e169234. [PMID: 37643022 PMCID: PMC10575727 DOI: 10.1172/jci169234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
Microvillus inclusion disease (MVID), caused by loss-of-function mutations in the motor protein myosin Vb (MYO5B), is a severe infantile disease characterized by diarrhea, malabsorption, and acid/base instability, requiring intensive parenteral support for nutritional and fluid management. Human patient-derived enteroids represent a model for investigation of monogenic epithelial disorders but are a rare resource from MVID patients. We developed human enteroids with different loss-of function MYO5B variants and showed that they recapitulated the structural changes found in native MVID enterocytes. Multiplex immunofluorescence imaging of patient duodenal tissues revealed patient-specific changes in localization of brush border transporters. Functional analysis of electrolyte transport revealed profound loss of Na+/H+ exchange (NHE) activity in MVID patient enteroids with near-normal chloride secretion. The chloride channel-blocking antidiarrheal drug crofelemer dose-dependently inhibited agonist-mediated fluid secretion. MVID enteroids exhibited altered differentiation and maturation versus healthy enteroids. γ-Secretase inhibition with DAPT recovered apical brush border structure and functional Na+/H+ exchange activity in MVID enteroids. Transcriptomic analysis revealed potential pathways involved in the rescue of MVID cells including serum/glucocorticoid-regulated kinase 2 (SGK2) and NHE regulatory factor 3 (NHERF3). These results demonstrate the utility of patient-derived enteroids for developing therapeutic approaches to MVID.
Collapse
Affiliation(s)
- Meri Kalashyan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Krishnan Raghunathan
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Haley Oller
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Marie-Theres Bayer
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lissette Jimenez
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Congenital Enteropathy Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
| | - Joseph T. Roland
- Section of Surgical Sciences and
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Elena Kolobova
- Section of Surgical Sciences and
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Susan J. Hagen
- Department of Surgery, Division of Surgical Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Jeffrey D. Goldsmith
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
- Department of Pathology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mitchell D. Shub
- Department of Child Health, University of Arizona College of Medicine–Phoenix, and Division of Gastroenterology, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - James R. Goldenring
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
- Section of Surgical Sciences and
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville VA Medical Center, Nashville, Tennessee, USA
| | - Izumi Kaji
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
- Section of Surgical Sciences and
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jay R. Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Congenital Enteropathy Program, Boston Children’s Hospital, Boston, Massachusetts, USA
- PediCODE Consortium, as detailed in Supplemental Acknowledgments
- Harvard Digestive Disease Center, Boston, Massachusetts, USA
| |
Collapse
|
181
|
Zhang R, Chen S, Yang Z, Zhang N, Guo K, Lv K, Zhou Z, Gao M, Hu X, Su Y, He J, Wang F. Actin polymerization inhibition by targeting ARPC2 affects intestinal stem cell homeostasis. BURNS & TRAUMA 2023; 11:tkad038. [PMID: 37849945 PMCID: PMC10578047 DOI: 10.1093/burnst/tkad038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/28/2023] [Accepted: 05/25/2023] [Indexed: 10/19/2023]
Abstract
Background The rapid turnover of the intestinal epithelium is driven by the proliferation and differentiation of intestinal stem cells (ISCs). The dynamics of the F-actin cytoskeleton are critical for maintaining intercellular force and the signal transduction network. However, it remains unclear how direct interference with actin polymerization impacts ISC homeostasis. This study aims to reveal the regulatory effects of the F-actin cytoskeleton on the homeostasis of intestinal epithelium, as well as the potential risks of benproperine (BPP) as an anti-tumor drug. Methods Phalloidin fluorescence staining was utilized to test F-actin polymerization. Flow cytometry and IHC staining were employed to discriminate different types of intestinal epithelial cells. Cell proliferation was assessed through bromo-deoxyuridine (BrdU) and 5-ethynyl-2'-deoxyuridine (EdU) incorporation assays. The proliferation and differentiation of intestinal stem cells were replicated in vitro through organoid culture. Epithelial migration was evaluated through BrdU pulse labeling and chasing in mice. Results The F-actin content was observed to significantly increase as crypt cells migrated into the villus region. Additionally, actin polymerization in secretory cells, especially in Paneth cells (PCs), was much higher than that in neighboring ISCs. Treatment with the newly identified actin-related protein 2/3 complex subunit 2 (ARPC2) inhibitor BPP led to a dose-dependent increase or inhibition of intestinal organoid growth in vitro and crypt cell proliferation in vivo. Compared with the vehicle group, BPP treatment decreased the expression of Lgr5 ISC feature genes in vivo and in organoid culture. Meanwhile, PC differentiation derived from ISCs and progenitors was decreased by inhibition of F-actin polymerization. Mechanistically, BPP-induced actin polymerization inhibition may activate the Yes1-associated transcriptional regulator pathway, which affects ISC proliferation and differentiation. Accordingly, BPP treatment affected intestinal epithelial cell migration in a dose-dependent manner. Conclusion Our findings indicate that the regulation of cytoskeleton reorganization can affect ISC homeostasis. In addition, inhibiting ARPC2 with the Food and Drug Administration-approved drug BPP represents a novel approach to influencing the turnover of intestinal epithelial cells.
Collapse
Affiliation(s)
- Ruzhen Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
- College of Life Sciences, Chongqing Normal University,Chongqing, 401331China
| | - Sheng Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Zhifan Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Ning Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Kenan Guo
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing 400038, China
| | - Keyi Lv
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Zimo Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Meijiao Gao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Xiancheng Hu
- College of Life Sciences, Chongqing Normal University,Chongqing, 401331China
| | - Yongping Su
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Jianming He
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research (Hebei), Shijiazhuang, 050011
| | - Fengchao Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
182
|
Peng J, Huang W, Liang Y, Zhang W, Zhang Y, Yang M, Zheng S, Lv Y, Gou Z, Cheng C, Gao H, Wang W, Peng J, Huang Y. Optimal dietary energy and protein levels for breeding pigeons in the winter "2 + 3" lactation pattern. Poult Sci 2023; 102:102964. [PMID: 37573846 PMCID: PMC10428047 DOI: 10.1016/j.psj.2023.102964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/21/2023] [Accepted: 07/21/2023] [Indexed: 08/15/2023] Open
Abstract
The nutritional requirements of breeding pigeons depend on their physiological period, breeding pattern, and environmental conditions. Despite works on reduced litter size in winter production to combat high mortality and the poor welfare of squabs, there are few studies on the related nutritional requirements of these pigeons. A total of 432 pairs of European Mimas pigeons were randomly divided into 9 groups in which 3 crude protein (CP) levels (15, 16.5, and 18%) and 3 metabolizable energy (ME) levels (12.2 MJ/kg, 12.4 MJ/kg, and 12.6 MJ/kg) were tested to determine the optimal energy and protein requirements of breeding pigeons in the winter "2 + 3" breeding pattern. The results showed that ME and CP levels had little effect on the body weight, feed intake, and egg quality of breeding pigeons during the lactation period. An 18% CP diet significantly increased the laying rate and hatchability (P < 0.05), but there was no difference in the laying rate with 18% CP and 16.5% CP during the whole reproductive cycle (P > 0.05). There was a significant interaction between ME and CP levels, and the laying interval of breeding pigeons in group 9 (18% CP; 12.6 MJ/kg) was significantly shortened (P < 0.05). For squabs, the ME level had no effect on growth performance, slaughter performance, or meat quality. The body weight of 21-day-old squabs in the 18% CP group increased by 3.16% compared with that of the 15% CP group, but there was no difference between the 18% CP and 16.5% CP groups. Compared with other experimental groups, group 7 (18% CP; 12.2 MJ/kg) had the fastest growth rate in squabs (P < 0.05), and the corresponding slaughter weight was also the heaviest (P < 0.05). We further found that the height of the squab intestinal epithelium was significantly increased in both the 16.5% CP and 18% CP groups of squabs (P < 0.01), but male breeding pigeons showed a certain degree of oxidative stress with an increase in CP level. In conclusion, the effects of 15 to 18% CP levels and 12.2 to 12.6 MJ/kg ME levels on the reproductive metabolism of breeding pigeons and the growth and development of squabs in the "2 + 3" breeding pattern during winter are small. For economic efficiency, we suggest that the CP level can be reduced to 16.5% while the ME level should not be less than 12.2 MJ/kg in practical production.
Collapse
Affiliation(s)
- Jie Peng
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Weiying Huang
- College of Animal Science and Technology and Animal Medicine, Huazhong Agricultural University, Wuchang, Wuhan 430000, China
| | - Yayan Liang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Wei Zhang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Yanlin Zhang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Menglin Yang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Shiqi Zheng
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Yantao Lv
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Zhongyong Gou
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture and Rural Affairs, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Guangzhou, Guangdong 510640, China
| | - Chuanshang Cheng
- Key Laboratory of Animal Nutrition and Feed Science, Ministry of Agriculture and Rural Affairs, WENS Research Institute (Technology center), Yunfu 527300, China
| | - Hongyan Gao
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Wei Wang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China
| | - Jian Peng
- College of Animal Science and Technology and Animal Medicine, Huazhong Agricultural University, Wuchang, Wuhan 430000, China
| | - Yanhua Huang
- Innovative Institute of Animal Healthy Breeding, College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, Guangdong 510225, China; Guangdong Laboratory for Lingnan Modern Agricultural, Guangzhou 510642, China.
| |
Collapse
|
183
|
Costa AVD, Rattes IC, Goes CP, Lobo LHG, Barreto LBE, Gama P. Breastfeeding lifespan control of growth, maintenance, and metabolism of small intestinal epithelium. J Cell Physiol 2023; 238:2304-2315. [PMID: 37555566 DOI: 10.1002/jcp.31089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/08/2023] [Accepted: 07/12/2023] [Indexed: 08/10/2023]
Abstract
Gastrointestinal epithelial cells respond to milk-born molecules throughout breastfeeding, influencing growth, and development. The rapid renewal of the small intestine depends on the proliferation in the crypt that drives cell fates. We used early weaning model to investigate immediate and late effects of breastfeeding on proliferation, differentiation of jejunal epithelial cells. Wistar rats were either allowed to suckle (S) until 21 postnatal days or submitted to early weaning (EW) at 15 days. By comparing ages (18, 60, and 120 days), we found that EW decreased Ki67 indices and villi height at 18 and 60 days (p < 0.05), and at 120 days they were similar between diets. Proliferative reduction and augmented expression of Cdkn1b (p27 gene) were parallel. In the stem cell niche, EW increased the number and activity (Defa24) of Paneth cells at 18 and 60 days (p < 0.05), and Lgr5 and Ascl2 genes showed inverted responses between ages. Among target cells, EW decreased goblet cell number at 18 and 60 days (p < 0.05) and increased it at 120 days (p < 0.05), whereas enteroendocrine marker genes were differentially altered. EW reduced enterocytes density at 18 days (p < 0.05), and at 120 days this population was decreased (vs. 60 days). Among cell fate crypt-controlling genes, Notch and Atoh1 were the main targets of EW. Metabolically, intraperitoneal glucose tolerance was immediately reduced (18 days), being reverted until 120 days (p < 0.05). Currently, we showed that breastfeeding has a lifespan influence on intestinal mucosa and on its stem cell compartment. We suggest that, although jejunum absorptive function is granted after early weaning, the long lasting changes in gene expression might prime the mucosa with a different sensitivity to gut disorders that still have to be further explored.
Collapse
Affiliation(s)
- Aline Vasques da Costa
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Isadora Campos Rattes
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Carolina Purcell Goes
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Larissa Honda Greco Lobo
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Laylla Barreto E Barreto
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patricia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
184
|
Cui C, Wang X, Zheng Y, Li L, Wang F, Wei H, Peng J. Paneth cells protect intestinal stem cell niche to alleviate deoxynivalenol-induced intestinal injury. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115457. [PMID: 37688865 DOI: 10.1016/j.ecoenv.2023.115457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/10/2023] [Accepted: 09/05/2023] [Indexed: 09/11/2023]
Abstract
Deoxynivalenol (DON) is a common toxin in grains and feeds, and DON exposure triggers severe small intestinal injury and inflammation, which harms the health of humans and livestock. DON treatment leads to a decrease in Paneth cells, whereas the role of Paneth cells in DON-induced intestinal injury is poorly understood. We utilized dithizone (40 mg/kg) to keep murine Paneth cell number at a low level. The results showed that dithizone-mediated long-term disruption of Paneth cells aggravated intestinal injury, intestinal stem cell (ISC) loss, and microbiota disorder in DON (2 mg/kg)-treated mice. Unexpectedly, the number of goblet cells and proliferative cells was boosted in mice treated with dithizone and DON. After dithizone and DON treatments, the Firmicutes/Bacteroidetes (F/B) ratio was reduced, and the increased abundance of Dubosiella and the decreased abundance of Lactobacillus were observed in mice. The functional recovery of Paneth cells by lysozyme (200 U/day) supplementation improved intestinal injury and ISC loss in mice after DON challenge. In addition, lysozyme also promoted the growth and ISC activity of intestinal organoids. Taken together, these results demonstrate the protective role of Paneth cells in DON-induced intestinal injury. Our study raises a novel target, Paneth cell, for the treatment of DON exposure.
Collapse
Affiliation(s)
- Chenbin Cui
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xinru Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yao Zheng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Lindeng Li
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Fangke Wang
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongkui Wei
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Peng
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; The Cooperative Innovation Center for Sustainable Pig Production, Wuhan 400700, China.
| |
Collapse
|
185
|
Singh AK, Rai A, Weber A, Gericke M, Janssen KP, Moser M, Posern G. MRTF-A gain-of-function in mice impairs homeostatic renewal of the intestinal epithelium. Cell Death Dis 2023; 14:639. [PMID: 37770456 PMCID: PMC10539384 DOI: 10.1038/s41419-023-06158-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023]
Abstract
The actin-regulated transcription factor MRTF-A represents a central relay in mechanotransduction and controls a subset of SRF-dependent target genes. However, gain-of-function studies in vivo are lacking. Here we characterize a conditional MRTF-A transgenic mouse model. While MRTF-A gain-of-function impaired embryonic development, induced expression of constitutively active MRTF-A provoked rapid hepatocyte ballooning and liver failure in adult mice. Specific expression in the intestinal epithelium caused an erosive architectural distortion, villus blunting, cryptal hyperplasia and colonic inflammation, resulting in transient weight loss. Organoids from transgenic mice repeatedly induced in vitro showed impaired self-renewal and defective cryptal compartments. Mechanistically, MRTF-A gain-of-function decreased proliferation and increased apoptosis, but did not induce fibrosis. MRTF-A targets including Acta2 and Pai-1 were induced, whereas markers of stem cells and differentiated cells were reduced. Our results suggest that activated MRTF-A in the intestinal epithelium shifts the balance between proliferation, differentiation and apoptosis.
Collapse
Affiliation(s)
- Anurag Kumar Singh
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany.
| | - Amrita Rai
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Anja Weber
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany
| | - Martin Gericke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120, Halle (Saale), Germany
- Institute of Anatomy, Medical Faculty, Leipzig University, 04103, Leipzig, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, Technical University Munich, 81675, Munich, Germany
| | - Markus Moser
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
- Institute of Experimental Hematology, School of Medicine, Technical University Munich, 81675, Munich, Germany
| | - Guido Posern
- Institute for Physiological Chemistry, Medical Faculty, Martin Luther University Halle-Wittenberg, 06114, Halle (Saale), Germany.
| |
Collapse
|
186
|
Fang F, Liu Y, Xiong Y, Li X, Li G, Jiang Y, Hou X, Song J. Slowed Intestinal Transit Induced by Less Mucus in Intestinal Goblet Cell Piezo1-Deficient Mice through Impaired Epithelial Homeostasis. Int J Mol Sci 2023; 24:14377. [PMID: 37762681 PMCID: PMC10531822 DOI: 10.3390/ijms241814377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 09/29/2023] Open
Abstract
Mucus secreted by goblet cells (GCs) may play an important role in intestinal transit function. Our previous study found that Piezo1 protein is essential for GC function; however, the effect of GC Piezo1 on intestinal transit function is unclear. Our study aimed to investigate the effect of Piezo1 in GCs on intestinal transit and the potential mechanism. We compared intestinal mucus, fecal form, intestinal transit time, intestinal epithelial cell composition, and stem cell function in WT and GC-specific Piezo1-deficient (Piezo1ΔGC) mice. Our results revealed a correlation between mucus and intestinal transit: the less mucus there was, the slower the intestinal transit. Piezo1 deficiency in GCs led to decreased mucus synthesis and also disrupted the ecological niche of colon stem cells (CSCs). Through organoid culture, we found that the capacity of proliferation and differentiation in Piezo1ΔGC mouse CSCs was significantly decreased, which also led to a reduced source of GCs. Further studies found that the reduced Wnt and Notch signals in colon crypts might be the potential mechanism. These results indicated the importance of GC Piezo1 in intestinal transit function, which acts by maintaining the homeostasis of intestinal epithelial cells and mucus.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jun Song
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (F.F.); (Y.L.); (Y.X.); (X.L.); (G.L.); (Y.J.); (X.H.)
| |
Collapse
|
187
|
Li Y, Liu S, Zhou K, Wang Y, Chen Y, Hu W, Li S, Li H, Wang Y, Wang Q, He D, Xu H. Neuromedin U programs eosinophils to promote mucosal immunity of the small intestine. Science 2023; 381:1189-1196. [PMID: 37708282 DOI: 10.1126/science.ade4177] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 08/18/2023] [Indexed: 09/16/2023]
Abstract
Eosinophils are granulocytes that play an essential role in type 2 immunity and regulate multiple homeostatic processes in the small intestine (SI). However, the signals that regulate eosinophil activity in the SI at steady state remain poorly understood. Through transcriptome profiling of eosinophils from various mouse tissues, we found that a subset of SI eosinophils expressed neuromedin U (NMU) receptor 1 (NMUR1). Fate-mapping analyses showed that NMUR1 expression in SI eosinophils was programmed by the local microenvironment and further enhanced by inflammation. Genetic perturbation and eosinophil-organoid coculture experiments revealed that NMU-mediated eosinophil activation promotes goblet cell differentiation. Thus, NMU regulates epithelial cell differentiation and barrier immunity by stimulating NMUR1-expressing eosinophils in the SI, which highlights the importance of neuroimmune-epithelial cross-talk in maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Yu Li
- School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Shaorui Liu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Kewen Zhou
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yinsheng Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yan Chen
- Center for Inflammatory Bowel Diseases, Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Wen Hu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang, China
| | - Shuyan Li
- Department of Nursing, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Hui Li
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yan Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Qiuying Wang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Danyang He
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
| | - Heping Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| |
Collapse
|
188
|
Kourula S, Derksen M, Jardi F, Jonkers S, van Heerden M, Verboven P, Theuns V, Van Asten S, Huybrechts T, Kunze A, Frazer-Mendelewska E, Lai KW, Overmeer R, Roos JL, Vries RGJ, Boj SF, Monshouwer M, Pourfarzad F, Snoeys J. Intestinal organoids as an in vitro platform to characterize disposition, metabolism, and safety profile of small molecules. Eur J Pharm Sci 2023; 188:106481. [PMID: 37244450 DOI: 10.1016/j.ejps.2023.106481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 05/29/2023]
Abstract
Intestinal organoids derived from LGR5+ adult stem cells allow for long-term culturing, more closely resemble human physiology than traditional intestinal models, like Caco-2, and have been established for several species. Here we evaluated intestinal organoids for drug disposition, metabolism, and safety applications. Enterocyte-enriched human duodenal organoids were cultured as monolayers to enable bidirectional transport studies. 3D enterocyte-enriched human duodenal and colonic organoids were incubated with probe substrates of major intestinal drug metabolizing enzymes (DMEs). To distinguish human intestinal toxic (high incidence of diarrhea in clinical trials and/or black box warning related to intestinal side effects) from non-intestinal toxic compounds, ATP-based cell viability was used as a readout, and compounds were ranked based on their IC50 values in relation to their 30-times maximal total plasma concentration (Cmax). To assess if rat and dog organoids reproduced the respective in vivo intestinal safety profiles, ATP-based viability was assessed in rat and dog organoids and compared to in vivo intestinal findings when available. Human duodenal monolayers discriminated high and low permeable compounds and demonstrated functional activity for the main efflux transporters Multi drug resistant protein 1 (MDR1, P-glycoprotein P-gp) and Breast cancer resistant protein (BCRP). Human 3D duodenal and colonic organoids also showed metabolic activity for the main intestinal phase I and II DMEs. Organoids derived from specific intestinal segments showed activity differences in line with reported DMEs expression. Undifferentiated human organoids accurately distinguished all but one compound from the test set of non-toxic and toxic drugs. Cytotoxicity in rat and dog organoids correlated with preclinical toxicity findings and observed species sensitivity differences between human, rat, and dog organoids. In conclusion, the data suggest intestinal organoids are suitable in vitro tools for drug disposition, metabolism, and intestinal toxicity endpoints. The possibility to use organoids from different species, and intestinal segment holds great potential for cross-species and regional comparisons.
Collapse
Affiliation(s)
- Stephanie Kourula
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium.
| | - Merel Derksen
- HUB Organoids, Yalelaan 62, 3584 CM Utrecht, The Netherlands
| | - Ferran Jardi
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Sophie Jonkers
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Marjolein van Heerden
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Peter Verboven
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Veronique Theuns
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Stijn Van Asten
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Tinne Huybrechts
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Annett Kunze
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | | | - Ka Wai Lai
- HUB Organoids, Yalelaan 62, 3584 CM Utrecht, The Netherlands
| | - René Overmeer
- HUB Organoids, Yalelaan 62, 3584 CM Utrecht, The Netherlands
| | - Jamie Lee Roos
- HUB Organoids, Yalelaan 62, 3584 CM Utrecht, The Netherlands
| | | | - Sylvia F Boj
- HUB Organoids, Yalelaan 62, 3584 CM Utrecht, The Netherlands
| | - Mario Monshouwer
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| | | | - Jan Snoeys
- Preclinical Sciences & Translational Safety, Janssen R&D, Turnhoutseweg 30, 2340, Beerse, Belgium
| |
Collapse
|
189
|
Bao W, You Y, Ni J, Hou H, Lyu J, Feng G, Wang Y, You K, Zhang S, Zhang L, Cao X, Wang X, Li H, Li H, Xu J, Liu C, Luo X, Du P, Chen D, Shen X. Inhibiting sorting nexin 10 promotes mucosal healing through SREBP2-mediated stemness restoration of intestinal stem cells. SCIENCE ADVANCES 2023; 9:eadh5016. [PMID: 37647408 PMCID: PMC10468130 DOI: 10.1126/sciadv.adh5016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023]
Abstract
Intestinal stem cell (ISC) is a promising therapeutic target for inflammatory bowel disease. Cholesterol availability is critical for ISC stemness. Low plasma cholesterol is a typical feature of Crohn's disease (CD); however, its impact on mucosal healing remains unclear. Here, we identified an essential role of sorting nexin 10 (SNX10) in maintaining the stemness of ISCs. SNX10 expression in intestinal tissues positively correlates with the severity of human CD and mouse colitis. Conditional SNX10 knockout in intestinal epithelial cells or ISCs promotes intestinal mucosal repair by maintaining the ISC population associated with increased intracellular cholesterol synthesis. Disassociation of ERLIN2 with SCAP by SNX10 deletion enhances the activation of SREBP2, resulting in increased cholesterol biosynthesis. DC-SX029, a small-molecule inhibitor of SNX10, was used to verify the druggable potential of SNX10 for the treatment of patients with CD. Our study provides a strategy for mucosal healing through SREBP2-mediated stemness restoration of ISCs.
Collapse
Affiliation(s)
- Weilian Bao
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, China
| | - Yan You
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiahui Ni
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Hui Hou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiaren Lyu
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Guize Feng
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Yirui Wang
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Keyuan You
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Sulin Zhang
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lijie Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xinyue Cao
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Xu Wang
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Haidong Li
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Hong Li
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Chenying Liu
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Colorectal Cancer Research Center, Shanghai, China
| | - Xiaomin Luo
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Peng Du
- Department of Colorectal and Anal Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Colorectal Cancer Research Center, Shanghai, China
| | - Daofeng Chen
- Department of Natural Medicine, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaoyan Shen
- Department of Pharmacology and the Key Laboratory of Smart Drug Delivery Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
190
|
Zheng X, Betjes MA, Ender P, Goos YJ, Huelsz-Prince G, Clevers H, van Zon JS, Tans SJ. Organoid cell fate dynamics in space and time. SCIENCE ADVANCES 2023; 9:eadd6480. [PMID: 37595032 PMCID: PMC10438469 DOI: 10.1126/sciadv.add6480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/20/2023] [Indexed: 08/20/2023]
Abstract
Organoids are a major new tool to study tissue renewal. However, characterizing the underlying differentiation dynamics remains challenging. Here, we developed TypeTracker, which identifies cell fates by AI-enabled cell tracking and propagating end point fates back along the branched lineage trees. Cells that ultimately migrate to the villus commit to their new type early, when still deep inside the crypt, with important consequences: (i) Secretory cells commit before terminal division, with secretory fates emerging symmetrically in sister cells. (ii) Different secretory types descend from distinct stem cell lineages rather than an omnipotent secretory progenitor. (iii) The ratio between secretory and absorptive cells is strongly affected by proliferation after commitment. (iv) Spatial patterning occurs after commitment through type-dependent cell rearrangements. This "commit-then-sort" model contrasts with the conventional conveyor belt picture, where cells differentiate by moving up the crypt-villus axis and hence raises new questions about the underlying commitment and sorting mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center, Uppsalalaan 8, Utrecht 3584 CT, Netherlands
| | | | - Sander J Tans
- Bionanoscience Department, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, Netherlands
- AMOLF, Amsterdam, Netherlands.
| |
Collapse
|
191
|
Wei S, Li M, Song W, Liu J, Yu S, Wang Y, Zhang M, Du H, Liu Y, Liu H, Fu W, Li B, Chen YG. The cyclooxygenase-expressing mesenchyme resists intestinal epithelial injury by paracrine signaling. CELL REGENERATION (LONDON, ENGLAND) 2023; 12:30. [PMID: 37574502 PMCID: PMC10423710 DOI: 10.1186/s13619-023-00174-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023]
Abstract
Paracrine signals play pivotal roles in organ homeostasis. Mesenchymal stromal cells (MSCs) play a key role in regulating epithelium homeostasis in the intestine while their paracrine effects are poorly characterized. Here, we identified prostaglandin E2 (PGE2) secreted by cyclooxygenase (COX)-expressing MSCs as a vital factor to maintain the intestinal mucosal barrier. We found that MSCs-induced organoid swelling through paracrine effect in vitro, a process due to enhanced water adsorption and is mediated by the COX-PGE2-EP4 axis. To further explore the regulatory effect of this axis on the intestinal epithelial barrier in vivo, we established the conditional knockout mouse model to specifically delete COX in MSCs and found that PGE2 reduction downregulated the gene Muc2 and induced a gastric metaplasia-like phenotype. Moreover, PGE2 defects increased the susceptibility of intestinal epithelium to colitis. Our study uncovers the paracrine signaling of COX-expressing MSCs in intestinal mucosal barrier maintenance, providing a basis for understanding the role of mesenchymal cells in the pathophysiological function of the intestine.
Collapse
Affiliation(s)
- Siting Wei
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Meng Li
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Wanlu Song
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jiaye Liu
- Department of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shicheng Yu
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Yalong Wang
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Mengxian Zhang
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Huijun Du
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuan Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Huidong Liu
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wei Fu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuro- Psychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
192
|
Sheng W, Ji G, Zhang L. Immunomodulatory effects of inulin and its intestinal metabolites. Front Immunol 2023; 14:1224092. [PMID: 37638034 PMCID: PMC10449545 DOI: 10.3389/fimmu.2023.1224092] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
"Dietary fiber" (DF) refers to a type of carbohydrate that cannot be digested fully. DF is not an essential nutrient, but it plays an important part in enhancing digestive capacity and maintaining intestinal health. Therefore, DF supplementation in the daily diet is highly recommended. Inulin is a soluble DF, and commonly added to foods. Recently, several studies have found that dietary supplementation of inulin can improve metabolic function and regulate intestinal immunity. Inulin is fermented in the colon by the gut microbiota and a series of metabolites is generated. Among these metabolites, short-chain fatty acids provide energy to intestinal epithelial cells and participate in regulating the differentiation of immune cells. Inulin and its intestinal metabolites contribute to host immunity. This review summarizes the effect of inulin and its metabolites on intestinal immunity, and the underlying mechanisms of inulin in preventing diseases such as type 2 diabetes mellitus, inflammatory bowel disease, chronic kidney disease, and certain cancer types.
Collapse
Affiliation(s)
| | | | - Li Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
193
|
Liu L, Wang Y, Yu S, Liu H, Li Y, Hua S, Chen Y. Transforming Growth Factor Beta Promotes Inflammation and Tumorigenesis in Smad4-Deficient Intestinal Epithelium in a YAP-Dependent Manner. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300708. [PMID: 37261975 PMCID: PMC10427365 DOI: 10.1002/advs.202300708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/18/2023] [Indexed: 06/03/2023]
Abstract
Transforming growth factor beta (TGF-β), a multifunctional cytokine, plays critical roles in immune responses. However, the precise role of TGF-β in colitis and colitis-associated cancer remains poorly defined. Here, it is demonstrated that TGF-β promotes the colonic inflammation and related tumorigenesis in the absence of Smad family member 4 (Smad4). Smad4 loss in intestinal epithelium aggravates colitis and colitis-associated neoplasia induced by dextran sulfate sodium (DSS) and azoxymethane/dextran sulfate sodium (AOM/DSS), leading to over-activated immune responses and increased TGF-β1 levels. In Smad4-deficient organoids, TGF-β1 stimulates spheroid formation and impairs intestinal stem cell proliferation and lineage specification. YAP, whose expression is directly upregulated by TGF-β1 after Smad4 deletion, mediates the effect of TGF-β1 by interacting with Smad2/3. Attenuation of YAP/TAZ prevents TGF-β1-induced spheroid formation in Smad4-/- organoids and alleviates colitis and colitis-associated cancer in Smad4-deficient mice. Collectively, these results highlight an integral role of the TGF-β/Smad4 axis in restraining intestinal inflammation and tumorigenesis and suggest TGF-β or YAP signaling as therapeutic targets for these gastrointestinal diseases intervention.
Collapse
Affiliation(s)
- Liansheng Liu
- Guangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhou510530China
- Guangzhou LaboratoryGuangzhou510700China
| | - Yalong Wang
- Guangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhou510530China
- Guangzhou LaboratoryGuangzhou510700China
| | - Shicheng Yu
- Guangzhou Institutes of Biomedicine and HealthUniversity of Chinese Academy of SciencesChinese Academy of SciencesGuangzhou510530China
- Guangzhou LaboratoryGuangzhou510700China
| | - Huidong Liu
- The State Key Laboratory of Membrane BiologyTsinghua‐Peking Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijing100084China
| | - Yehua Li
- The State Key Laboratory of Membrane BiologyTsinghua‐Peking Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijing100084China
| | - Shan Hua
- Guangzhou LaboratoryGuangzhou510700China
- Center for Life SciencesSchool of Life SciencesYunnan UniversityKunming650500China
| | - Ye‐Guang Chen
- Guangzhou LaboratoryGuangzhou510700China
- The State Key Laboratory of Membrane BiologyTsinghua‐Peking Center for Life SciencesSchool of Life SciencesTsinghua UniversityBeijing100084China
- Jiangxi Medical CollegeNanchang UniversityNanchang330031China
| |
Collapse
|
194
|
Montes-Olivas S, Legge D, Lund A, Fletcher AG, Williams AC, Marucci L, Homer M. In-silico and in-vitro morphometric analysis of intestinal organoids. PLoS Comput Biol 2023; 19:e1011386. [PMID: 37578984 PMCID: PMC10473498 DOI: 10.1371/journal.pcbi.1011386] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 09/01/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023] Open
Abstract
Organoids offer a powerful model to study cellular self-organisation, the growth of specific tissue morphologies in-vitro, and to assess potential medical therapies. However, the intrinsic mechanisms of these systems are not entirely understood yet, which can result in variability of organoids due to differences in culture conditions and basement membrane extracts used. Improving the standardisation of organoid cultures is essential for their implementation in clinical protocols. Developing tools to assess and predict the behaviour of these systems may produce a more robust and standardised biological model to perform accurate clinical studies. Here, we developed an algorithm to automate crypt-like structure counting on intestinal organoids in both in-vitro and in-silico images. In addition, we modified an existing two-dimensional agent-based mathematical model of intestinal organoids to better describe the system physiology, and evaluated its ability to replicate budding structures compared to new experimental data we generated. The crypt-counting algorithm proved useful in approximating the average number of budding structures found in our in-vitro intestinal organoid culture images on days 3 and 7 after seeding. Our changes to the in-silico model maintain the potential to produce simulations that replicate the number of budding structures found on days 5 and 7 of in-vitro data. The present study aims to aid in quantifying key morphological structures and provide a method to compare both in-vitro and in-silico experiments. Our results could be extended later to 3D in-silico models.
Collapse
Affiliation(s)
- Sandra Montes-Olivas
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
| | - Danny Legge
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Abbie Lund
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
| | - Alexander G. Fletcher
- School of Mathematics and Statistics, University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Ann C. Williams
- Colorectal Tumour Biology Group, School of Cellular and Molecular Medicine, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Lucia Marucci
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- BrisSynBio, Bristol, United Kingdom
| | - Martin Homer
- Department of Engineering Mathematics, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
195
|
Szabó L, Seubert AC, Kretzschmar K. Modelling adult stem cells and their niche in health and disease with epithelial organoids. Semin Cell Dev Biol 2023; 144:20-30. [PMID: 36127261 DOI: 10.1016/j.semcdb.2022.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 10/14/2022]
Abstract
Adult stem cells are responsible for homoeostasis and regeneration of epithelial tissues. Stem cell function is regulated by both cell autonomous mechanisms as well as the niche. Deregulated stem cell function contributes to diseases such as cancer. Epithelial organoid cultures generated from tissue-resident adult stem cells have allowed unprecedented insights into the biology of epithelial tissues. The subsequent adaptation of organoid technology enabled the modelling of the communication of stem cells with their cellular and non-cellular niche as well as diseases. Starting from its first model described in 2009, the murine small intestinal organoid, we discuss here how epithelial organoid cultures have been become a prime in vitro research tool for cell and developmental biology, bioengineering, and biomedicine in the last decade.
Collapse
Affiliation(s)
- Lili Szabó
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Würzburg, IZKF/MSNZ, Würzburg, Germany
| | - Anna C Seubert
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Würzburg, IZKF/MSNZ, Würzburg, Germany
| | - Kai Kretzschmar
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Würzburg, IZKF/MSNZ, Würzburg, Germany.
| |
Collapse
|
196
|
Kolev HM, Kaestner KH. Mammalian Intestinal Development and Differentiation-The State of the Art. Cell Mol Gastroenterol Hepatol 2023; 16:809-821. [PMID: 37507088 PMCID: PMC10520362 DOI: 10.1016/j.jcmgh.2023.07.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
The development of the mammalian intestine, from its earliest origins as a morphologically uniform sheet of endoderm cells during gastrulation into the complex organ system that is essential for the life of the organism, is a truly fascinating process. During midgestation development, reciprocal interactions between endoderm-derived epithelium and mesoderm-derived mesenchyme enable villification, or the conversion of a radially symmetric pseudostratified epithelium into the functional subdivision of crypts and villi. Once a mature crypt-villus axis is established, proliferation and differentiation of new epithelial cells continue throughout life. Spatially localized signals including the wingless and Int-1, fibroblast growth factor, and Hippo systems, among others, ensure that new cells are being born continuously in the crypt. As cells exit the crypt compartment, a gradient of bone morphogenetic protein signaling limits proliferation to allow for the specification of multiple mature cell types. The first major differentiation decision is dependent on Notch signaling, which specifies epithelial cells into absorptive and secretory lineages. The secretory lineage is subdivided further into Paneth, goblet, tuft, and enteroendocrine cells via a complex network of transcription factors. Although some of the signaling molecules are produced by epithelial cells, critical components are derived from specialized crypt-adjacent mesenchymal cells termed telocytes, which are marked by Forkhead box l1, GLI Family Zinc Finger 1, and platelet-derived growth factor receptor α. The crucial nature of these processes is evidenced by the multitude of intestinal disorders such as colorectal cancer, short-bowel syndrome, and inflammatory bowel disease, which all reflect perturbations of the development and/or differentiation of the intestine.
Collapse
Affiliation(s)
- Hannah M Kolev
- Department of Genetics and Center for Molecular Studies in Digestive and Liver Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Klaus H Kaestner
- Department of Genetics and Center for Molecular Studies in Digestive and Liver Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
197
|
Ahmad Sophien AN, Jusop AS, Tye GJ, Tan YF, Wan Kamarul Zaman WS, Nordin F. Intestinal stem cells and gut microbiota therapeutics: hype or hope? Front Med (Lausanne) 2023; 10:1195374. [PMID: 37547615 PMCID: PMC10400779 DOI: 10.3389/fmed.2023.1195374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023] Open
Abstract
The vital role of the intestines as the main site for the digestion and absorption of nutrients for the body continues subconsciously throughout one's lifetime, but underneath all the complex processes lie the intestinal stem cells and the gut microbiota that work together to maintain the intestinal epithelium. Intestinal stem cells (ISC) are multipotent stem cells from which all intestinal epithelial cells originate, and the gut microbiota refers to the abundant collection of various microorganisms that reside in the gastrointestinal tract. Both reside in the intestines and have many mechanisms and pathways in place with the ultimate goal of co-managing human gastrointestinal tract homeostasis. Based on the abundance of research that is focused on either of these two topics, this suggests that there are many methods by which both players affect one another. Therefore, this review aims to address the relationship between ISC and the gut microbiota in the context of regenerative medicine. Understanding the principles behind both aspects is therefore essential in further studies in the field of regenerative medicine by making use of the underlying designed mechanisms.
Collapse
Affiliation(s)
- Ahmad Naqiuddin Ahmad Sophien
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Amirah Syamimi Jusop
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Gee Jun Tye
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Gelugor, Malaysia
| | - Yuen-Fen Tan
- PPUKM-MAKNA Cancer Center, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
- M. Kandiah Faculty of Medicine and Health Sciences (MK FMHS), Universiti Tunku Abdul Rahman, Kajang, Malaysia
| | - Wan Safwani Wan Kamarul Zaman
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
- Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Fazlina Nordin
- Centre for Tissue Engineering and Regenerative Medicine (CTERM), Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
198
|
Andres SF, Zhang Y, Kuhn M, Scottoline B. Building better barriers: how nutrition and undernutrition impact pediatric intestinal health. Front Immunol 2023; 14:1192936. [PMID: 37545496 PMCID: PMC10401430 DOI: 10.3389/fimmu.2023.1192936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/26/2023] [Indexed: 08/08/2023] Open
Abstract
Chronic undernutrition is a major cause of death for children under five, leaving survivors at risk for adverse long-term consequences. This review focuses on the role of nutrients in normal intestinal development and function, from the intestinal epithelium, to the closely-associated mucosal immune system and intestinal microbiota. We examine what is known about the impacts of undernutrition on intestinal physiology, with focus again on the same systems. We provide a discussion of existing animal models of undernutrition, and review the evidence demonstrating that correcting undernutrition alone does not fully ameliorate effects on intestinal function, the microbiome, or growth. We review efforts to treat undernutrition that incorporate data indicating that improved recovery is possible with interventions focused not only on delivery of sufficient energy, macronutrients, and micronutrients, but also on efforts to correct the abnormal intestinal microbiome that is a consequence of undernutrition. Understanding of the role of the intestinal microbiome in the undernourished state and correction of the phenotype is both complex and a subject that holds great potential to improve recovery. We conclude with critical unanswered questions in the field, including the need for greater mechanistic research, improved models for the impacts of undernourishment, and new interventions that incorporate recent research gains. This review highlights the importance of understanding the mechanistic effects of undernutrition on the intestinal ecosystem to better treat and improve long-term outcomes for survivors.
Collapse
Affiliation(s)
- Sarah F. Andres
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Yang Zhang
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Madeline Kuhn
- Division of Pediatric Gastroenterology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| | - Brian Scottoline
- Division of Neonatology, Department of Pediatrics, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
199
|
Tong DQ, Lu ZJ, Zeng N, Wang XQ, Yan HC, Gao CQ. Dietary supplementation with probiotics increases growth performance, improves the intestinal mucosal barrier and activates the Wnt/β-catenin pathway activity in chicks. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:4649-4659. [PMID: 36930725 DOI: 10.1002/jsfa.12562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/21/2022] [Accepted: 03/17/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Probiotics comprise effective feed additives that can replace antibiotics in animal livestock production. However, mono-strain probiotics appear less effective because of their instability. Therefore, the present study aimed to investigate dietary supplementation with compound probiotics (CPP) on growth performance, diarrhea rate and intestinal mucosal barrier, as well as the possible molecular mechanism, in chicks. In total, 360 1-day-old chicks of the Hy-Line Brown Chicks were randomly divided into the control group (CON, basal diet), chlortetracycline group (500 mg kg-1 CTC) and compound probiotics group (1000 mg kg-1 CPP, consisting of Bacillus subtilis, Bacillus licheniformis, Enterococcus faecium and yeast). The experiment period was 56 days. RESULTS The results showed that, in comparison with the CON group, CPP significantly increased the average daily feed intake and average daily gain of chicks and reduced diarrhea (P < 0.05). The probiotic group exhibited increased immune organ (i.e. spleen and thymus) mass and increased levels of serum immunoglobulin (Ig)A, IgM and IgG (P < 0.05) compared to the CTC group. In addition, the jejunal mass and morphology were improved in the probiotic group (P < 0.05). Moreover, CPP reinforced jejunal barrier function, as indicated by increased transepithelial electrical resistance, protein expression of occludin and claudin-1, and diamine oxidase levels in the jejunum (P < 0.05). Likewise, enhanced fluorescence signals of proliferating cell nuclear antigen-labeled mitotic cells and villin-labeled absorptive cells in the jejunum (P < 0.05) suggested that CPP promoted intestinal stem cells activity. Mechanistically, the Wnt/β-catenin signaling pathway, including β-catenin, TCF4, c-Myc, cyclin D1 and Lgr5, was amplified in the jejunum by CPP addition (P < 0.05). CONCLUSION The present study demonstrated that dietary supplementation with CPP reinforced the jejunal epithelial integrity by activating Wnt/β-catenin signaling and enhanced immune function in chicks. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Di-Qing Tong
- College of Animal Science, South China Agricultural University/Guangdong Provincial Key Laboratory of Animal Nutrition Control/Guangdong Laboratory for Lingnan Modern Agriculture/State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China
| | - Zhu-Jin Lu
- College of Animal Science, South China Agricultural University/Guangdong Provincial Key Laboratory of Animal Nutrition Control/Guangdong Laboratory for Lingnan Modern Agriculture/State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China
| | - Nan Zeng
- College of Animal Science, South China Agricultural University/Guangdong Provincial Key Laboratory of Animal Nutrition Control/Guangdong Laboratory for Lingnan Modern Agriculture/State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China
| | - Xiu-Qi Wang
- College of Animal Science, South China Agricultural University/Guangdong Provincial Key Laboratory of Animal Nutrition Control/Guangdong Laboratory for Lingnan Modern Agriculture/State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China
| | - Hui-Chao Yan
- College of Animal Science, South China Agricultural University/Guangdong Provincial Key Laboratory of Animal Nutrition Control/Guangdong Laboratory for Lingnan Modern Agriculture/State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China
| | - Chun-Qi Gao
- College of Animal Science, South China Agricultural University/Guangdong Provincial Key Laboratory of Animal Nutrition Control/Guangdong Laboratory for Lingnan Modern Agriculture/State Key Laboratory of Livestock and Poultry Breeding, Guangzhou, China
| |
Collapse
|
200
|
Creff J, Nowosad A, Prel A, Pizzoccaro A, Aguirrebengoa M, Duquesnes N, Callot C, Jungas T, Dozier C, Besson A. p57 Kip2 acts as a transcriptional corepressor to regulate intestinal stem cell fate and proliferation. Cell Rep 2023; 42:112659. [PMID: 37327110 DOI: 10.1016/j.celrep.2023.112659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/01/2022] [Accepted: 06/01/2023] [Indexed: 06/18/2023] Open
Abstract
p57Kip2 is a cyclin/CDK inhibitor and a negative regulator of cell proliferation. Here, we report that p57 regulates intestinal stem cell (ISC) fate and proliferation in a CDK-independent manner during intestinal development. In the absence of p57, intestinal crypts exhibit an increased proliferation and an amplification of transit-amplifying cells and of Hopx+ ISCs, which are no longer quiescent, while Lgr5+ ISCs are unaffected. RNA sequencing (RNA-seq) analyses of Hopx+ ISCs show major gene expression changes in the absence of p57. We found that p57 binds to and inhibits the activity of Ascl2, a transcription factor critical for ISC specification and maintenance, by participating in the recruitment of a corepressor complex to Ascl2 target gene promoters. Thus, our data suggest that, during intestinal development, p57 plays a key role in maintaining Hopx+ ISC quiescence and repressing the ISC phenotype outside of the crypt bottom by inhibiting the transcription factor Ascl2 in a CDK-independent manner.
Collapse
Affiliation(s)
- Justine Creff
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Ada Nowosad
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Anne Prel
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Anne Pizzoccaro
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Marion Aguirrebengoa
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Nicolas Duquesnes
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Caroline Callot
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Thomas Jungas
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Christine Dozier
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Arnaud Besson
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France.
| |
Collapse
|