62,493 results on '"Paneth A"'
Search Results
2. Absence of Paneth Cell Metaplasia to Predict Clinical Relapse in Ulcerative Colitis with Endoscopically Quiescent Mucosa
- Author
-
Horiuchi, Kazuki, Higashiyama, Masaaki, Tahara, Hiroyuki, Yoshidome, Yuta, Ayaki, Kana, Nishimura, Hiroyuki, Tomioka, Akira, Narimatsu, Kazuyuki, Komoto, Shunsuke, Tomita, Kengo, and Hokari, Ryota
- Published
- 2024
- Full Text
- View/download PDF
3. Paneth cell ontogeny in term and preterm ovine models
- Author
-
Bautista, Geoanna M, Cera, Anjali J, Schoenauer, Rebecca J, Persiani, Michele, Lakshminrusimha, Satyan, Chandrasekharan, Praveen, Gugino, Sylvia F, Underwood, Mark A, and McElroy, Steven J
- Subjects
Paneth cell ,immature intestine ,perinatal bradycardic stress ,preterm ,ovine ,Veterinary Sciences ,Veterinary sciences - Abstract
IntroductionPaneth cells are critically important to intestinal health, including protecting intestinal stem cells, shaping the intestinal microbiome, and regulating host immunity. Understanding Paneth cell biology in the immature intestine is often modeled in rodents with little information in larger mammals such as sheep. Previous studies have only established the distribution pattern of Paneth cells in healthy adult sheep. Our study aimed to examine the ontogeny, quantification, and localization of Paneth cells in fetal and newborn lambs at different gestational ages and with perinatal transient asphyxia. We hypothesized that ovine Paneth cell distribution at birth resembles the pattern seen in humans (highest concentrations in the ileum) and that ovine Paneth cell density is gestation-dependent.MethodsIntestinal samples were obtained from 126-127 (preterm, with and without perinatal transient asphyxia) and 140-141 (term) days gestation sheep. Samples were quantified per crypt in at least 100 crypts per animal and confirmed as Paneth cells through in immunohistochemistry.ResultsPaneth cells had significantly higher density in the ileum compared to the jejunum and were absent in the colon.DiscussionExposure to perinatal transient asphyxia acutely decreased Paneth cell numbers. These novel data support the possibility of utilizing ovine models for understanding Paneth cell biology in the fetus and neonate.
- Published
- 2024
4. Development of a deep learning algorithm for Paneth cell density quantification for inflammatory bowel disease.
- Author
-
Kang, Liang-I, Sarullo, Kathryn, Marsh, Jon, Lu, Liang, Khonde, Pooja, Ma, Changqing, Haritunians, Talin, Mujukian, Angela, Mengesha, Emebet, McGovern, Dermot, Stappenbeck, Thaddeus, Swamidass, S, and Liu, Ta-Chiang
- Subjects
Crohns disease ,Pathology ,Prognosis ,Whole slide images - Abstract
BACKGROUND: Alterations in ileal Paneth cell (PC) density have been described in gut inflammatory diseases such as Crohns disease (CD) and could be used as a biomarker for disease prognosis. However, quantifying PCs is time-intensive, a barrier for clinical workflow. Deep learning (DL) has transformed the development of robust and accurate tools for complex image evaluation. Our aim was to use DL to quantify PCs for use as a quantitative biomarker. METHODS: A retrospective cohort of whole slide images (WSI) of ileal tissue samples from patients with/without inflammatory bowel disease (IBD) was used for the study. A pathologist-annotated training set of WSI were used to train a U-net two-stage DL model to quantify PC number, crypt number, and PC density. For validation, a cohort of 48 WSIs were manually quantified by study pathologists and compared to the DL algorithm, using root mean square error (RMSE) and the coefficient of determination (r2) as metrics. To test the value of PC quantification as a biomarker, resection specimens from patients with CD (n = 142) and without IBD (n = 48) patients were analysed with the DL model. Finally, we compared time to disease recurrence in patients with CD with low versus high DL-quantified PC density using Log-rank test. FINDINGS: Initial one-stage DL model showed moderate accuracy in predicting PC density in cross-validation tests (RMSE = 1.880, r2 = 0.641), but adding a second stage significantly improved accuracy (RMSE = 0.802, r2 = 0.748). In the validation of the two-stage model compared to expert pathologists, the algorithm showed good performance up to RMSE = 1.148, r2 = 0.708. The retrospective cross-sectional cohort had mean ages of 62.1 years in the patients without IBD and 38.6 years for the patients with CD. In the non-IBD cohort, 43.75% of the patients were male, compared to 49.3% of the patients with CD. Analysis by the DL model showed significantly higher PC density in non-IBD controls compared to the patients with CD (4.04 versus 2.99 PC/crypt). Finally, the algorithm quantification of PCs density in patients with CD showed patients with the lowest 25% PC density (Quartile 1) have significantly shorter recurrence-free interval (p = 0.0399). INTERPRETATION: The current model performance demonstrates the feasibility of developing a DL-based tool to measure PC density as a predictive biomarker for future clinical practice. FUNDING: This study was funded by the National Institutes of Health (NIH).
- Published
- 2024
5. Infection and inflammation stimulate expansion of a CD74+ Paneth cell subset to regulate disease progression.
- Author
-
Balasubramanian, Iyshwarya, Bandyopadhyay, Sheila, Flores, Juan, Bianchi-Smak, Jared, Lin, Xiang, Liu, Haoran, Sun, Shengxiang, Golovchenko, Natasha, Liu, Yue, Wang, Dahui, Patel, Radha, Joseph, Ivor, Suntornsaratoon, Panan, Vargas, Justin, Green, Peter, Bhagat, Govind, Lagana, Stephen, Ying, Wang, Zhang, Yi, Wang, Zhihan, Singh, Sukhwinder, Zhou, Zhongren, Kollias, George, Farr, Laura, Moonah, Shannon, Yu, Shiyan, Wei, Zhi, Bonder, Edward, Zhang, Lanjing, Kiela, Pawel, Edelblum, Karen, Ferraris, Ronaldo, Liu, Ta-Chiang, Gao, Nan, and Li, Wei Vivian
- Subjects
CD74 ,Lyz1 ,Paneth cell ,heterogeneity ,lysozyme ,Humans ,Animals ,Mice ,Paneth Cells ,Intestine ,Small ,Inflammation ,Microbiota ,Cytokines - Abstract
Paneth cells (PCs), a specialized secretory cell type in the small intestine, are increasingly recognized as having an essential role in host responses to microbiome and environmental stresses. Whether and how commensal and pathogenic microbes modify PC composition to modulate inflammation remain unclear. Using newly developed PC-reporter mice under conventional and gnotobiotic conditions, we determined PC transcriptomic heterogeneity in response to commensal and invasive microbes at single cell level. Infection expands the pool of CD74+ PCs, whose number correlates with auto or allogeneic inflammatory disease progressions in mice. Similar correlation was found in human inflammatory disease tissues. Infection-stimulated cytokines increase production of reactive oxygen species (ROS) and expression of a PC-specific mucosal pentraxin (Mptx2) in activated PCs. A PC-specific ablation of MyD88 reduced CD74+ PC population, thus ameliorating pathogen-induced systemic disease. A similar phenotype was also observed in mice lacking Mptx2. Thus, infection stimulates expansion of a PC subset that influences disease progression.
- Published
- 2023
6. Advances in Exploring the Interaction between Paneth Cells and Intestinal Stem Cells and Possible Targets for Traditional Chinese Medicine Based on Mitochondrial Function and Energy Metabolism
- Author
-
LUO Guoliang, ZHANG Xindan, TANG Taichun, ZHU Yan, WANG Shuting, CHEN Min
- Subjects
intestinal stem cells ,paneth cells ,mitochondria ,energy metabolism ,traditional chinese medicine ,Medicine - Abstract
Intestinal stem cells and Paneth cells interact with each other to maintain intestinal homeostasis and function, the interaction between intestinal stem cells and Paneth cells is regulated by mitochondria-related functions and energy conversion. The unique advantages of traditional Chinese medicine (TCM) in the treatment of intestinal diseases are also reflected in its effects on mitochondria and energy metabolism. This paper explores the influence of mitochondrial function and energy metabolism on the interaction between intestinal stem cells and Paneth cells, as well as the possible targets related to mitochondria in the treatment of intestinal diseases by TCM, summarizes the regulatory effects of TCM on mitochondria and energy metabolism, thereby restoring intestinal homeostasis and alleviating disease, with an aim to provide reference and theoretical basis for the research related to the TCM treatment of intestinal stem cells and Paneth cells based on mitochondria.
- Published
- 2024
- Full Text
- View/download PDF
7. LRRK2 is not required for lysozyme expression in Paneth cells
- Author
-
Tasegian, Anna, Dikovskaya, Dina, Scott, Molly M., Chawla, Amanpreet Singh, Pemberton, Rebecca, Helps, Thomas, Meus, Tosca, McLean, Mairi H., and Swamy, Mahima
- Published
- 2024
- Full Text
- View/download PDF
8. Reply to: LRRK2 is not required for lysozyme expression in Paneth cells
- Author
-
Zhang, Chengye, Han, Lizhuang, Dong, Kemeng, Zhang, Qin, and Liu, Zhihua
- Published
- 2024
- Full Text
- View/download PDF
9. Paneth-like cells produced from OLFM4+ stem cells support OLFM4+ stem cell growth in advanced colorectal cancer
- Author
-
Sakahara, Mizuho, Okamoto, Takuya, Srivastava, Upasna, Natsume, Yasuko, Yamanaka, Hitomi, Suzuki, Yutaka, Obama, Kazutaka, Nagayama, Satoshi, and Yao, Ryoji
- Published
- 2024
- Full Text
- View/download PDF
10. Metastasis of colon cancer requires Dickkopf-2 to generate cancer cells with Paneth cell properties
- Author
-
Jae Hun Shin, Jooyoung Park, Jaechul Lim, Jaekwang Jeong, Ravi K Dinesh, Stephen E Maher, Jeonghyun Kim, Soyeon Park, Jun Young Hong, John Wysolmerski, Jungmin Choi, and Alfred LM Bothwell
- Subjects
metastasis ,colon cancer ,DKK2 ,Paneth cell properties ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Metastasis is the leading cause of cancer-related mortality. Paneth cells provide stem cell niche factors in homeostatic conditions, but the underlying mechanisms of cancer stem cell niche development are unclear. Here, we report that Dickkopf-2 (DKK2) is essential for the generation of cancer cells with Paneth cell properties during colon cancer metastasis. Splenic injection of Dkk2 knockout (KO) cancer organoids into C57BL/6 mice resulted in a significant reduction of liver metastases. Transcriptome analysis showed reduction of Paneth cell markers such as lysozymes in KO organoids. Single-cell RNA sequencing analyses of murine metastasized colon cancer cells and patient samples identified the presence of lysozyme positive cells with Paneth cell properties including enhanced glycolysis. Further analyses of transcriptome and chromatin accessibility suggested hepatocyte nuclear factor 4 alpha (HNF4A) as a downstream target of DKK2. Chromatin immunoprecipitation followed by sequencing analysis revealed that HNF4A binds to the promoter region of Sox9, a well-known transcription factor for Paneth cell differentiation. In the liver metastatic foci, DKK2 knockout rescued HNF4A protein levels followed by reduction of lysozyme positive cancer cells. Taken together, DKK2-mediated reduction of HNF4A protein promotes the generation of lysozyme positive cancer cells with Paneth cell properties in the metastasized colon cancers.
- Published
- 2024
- Full Text
- View/download PDF
11. Peptide YY: A Paneth cell antimicrobial peptide that maintains Candida gut commensalism.
- Author
-
Pierre, Joseph, Peters, Brian, La Torre, Diana, Sidebottom, Ashley, Tao, Yun, Zhu, Xiaorong, Cham, Candace, Wang, Ling, Kambal, Amal, Harris, Katharine, Silva, Julian, Zaborina, Olga, Alverdy, John, Herzog, Herbert, Witchley, Jessica, Leone, Vanessa, Chang, Eugene, and Noble, Suzanne
- Subjects
Animals ,Antifungal Agents ,Antimicrobial Peptides ,Candida ,Paneth Cells ,Peptide Fragments ,Peptide YY ,Symbiosis ,Humans ,Mice - Abstract
The mammalian gut secretes a family of multifunctional peptides that affect appetite, intestinal secretions, and motility whereas others regulate the microbiota. We have found that peptide YY (PYY1-36), but not endocrine PYY3-36, acts as an antimicrobial peptide (AMP) expressed by gut epithelial paneth cells (PC). PC-PYY is packaged into secretory granules and is secreted into and retained by surface mucus, which optimizes PC-PYY activity. Although PC-PYY shows some antibacterial activity, it displays selective antifungal activity against virulent Candida albicans hyphae-but not the yeast form. PC-PYY is a cationic molecule that interacts with the anionic surfaces of fungal hyphae to cause membrane disruption and transcriptional reprogramming that selects for the yeast phenotype. Hence, PC-PYY is an antifungal AMP that contributes to the maintenance of gut fungal commensalism.
- Published
- 2023
12. Azathioprine promotes intestinal epithelial cell differentiation into Paneth cells and alleviates ileal Crohn’s disease severity
- Author
-
Ragab, Mohab, Schlichting, Heidi, Hicken, Maren, Mester, Patricia, Hirose, Misa, Almeida, Larissa N., Christiansen, Lea, Ibrahim, Saleh, Tews, Hauke Christian, Divanovic, Senad, Sina, Christian, and Derer, Stefanie
- Published
- 2024
- Full Text
- View/download PDF
13. A Deep Learning-Based Approach to Estimate Paneth Cell Granule Area in Celiac Disease
- Author
-
Alharbi, Ebtihal, Rajaram, Ajay, Tech, M., Cote, Kevin, Farag, Mina, Maleki, Farhad, Gao, Zu-Hua, Maedler-Kron, Chelsea, Marcus, Victoria, and Fiset, Pierre Olivier
- Subjects
Celiac disease -- Models -- Development and progression ,Intestine, Small -- Physiological aspects -- Health aspects ,Machine learning -- Usage -- Health aspects ,Epithelial cells -- Physiological aspects -- Health aspects ,Health - Abstract
* Context.--Changes in Paneth cell numbers can be associated with chronic inflammatory diseases of the gastrointestinal tract. So far, no consensus has been achieved on the number of Paneth cells and their relevance to celiac disease (CD). Objectives.--To compare crypt and Paneth cell granule areas between patients with CD and those without CD (non-CD) using an artificial intelligence-based solution. Design.--Hematoxylin-eosin-stained sections of duodenal biopsies from 349 patients at the McGill University Health Centre were analyzed. Of these, 185 had a history of CD and 164 were controls. Slides were digitized, and NoCodeSeg, a code-free workflow using open-source software (QuPath, DeepMIB), was implemented to train deep learning models to segment crypts and Paneth cell granules. The total area of the entire analyzed tissue, epithelium, crypts, and Paneth cell granules was documented for all slides, and comparisons were performed. Results.--A mean intersection-over-union score of 88.76% and 91.30% was achieved for crypt areas and Paneth cell granule segmentations, respectively. On normalization to total tissue area, the crypt to total tissue area in CD was increased and the Paneth cell granule area to total tissue area decreased when compared to non-CD controls. Conclusions.--Crypt hyperplasia was confirmed in CD compared to non-CD controls. The area of Paneth cell granules, an indirect measure of Paneth cell function, decreased with increasing severity of CD. More importantly, our study analyzed complete hematoxylin-eosin slide sections using an efficient and easy to use coding-free artificial intelligence workflow. doi: 10.5858/arpa.2023-0074-OA), Paneth cells were first described by Paneth in 1888. (1) They are normally found in the small intestine, right colon, and appendix. They are usually located at the base of [...]
- Published
- 2024
- Full Text
- View/download PDF
14. PTPN2 Is a Critical Regulator of Ileal Paneth Cell Viability and Function in Mice
- Author
-
Canale, Vinicius, Spalinger, Marianne R, Alvarez, Rocio, Sayoc-Becerra, Anica, Sanati, Golshid, Manz, Salomon, Chatterjee, Pritha, Santos, Alina N, Lei, Hillmin, Jahng, Sharon, Chu, Timothy, Shawki, Ali, Hanson, Elaine, Eckmann, Lars, Ouellette, André J, and McCole, Declan F
- Subjects
Medical Biotechnology ,Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Digestive Diseases ,Inflammatory Bowel Disease ,Aetiology ,2.1 Biological and endogenous factors ,Oral and gastrointestinal ,Mice ,Animals ,Paneth Cells ,Protein Tyrosine Phosphatase ,Non-Receptor Type 2 ,Cell Survival ,Inflammatory Bowel Diseases ,Ileum ,Mice ,Knockout ,Antimicrobial Peptides ,TCPTP ,Lysozyme ,Intestinal Epithelial Cells ,Microbiome ,Biochemistry and cell biology ,Clinical sciences - Abstract
Background & aimsLoss-of-function variants in the PTPN2 gene are associated with increased risk of inflammatory bowel disease. We recently showed that Ptpn2 is critical for intestinal epithelial cell (IEC) barrier maintenance, IEC-macrophage communication, and modulation of the gut microbiome in mice, restricting expansion of a small intestinal pathobiont associated with inflammatory bowel disease. Here, we aimed to identify how Ptpn2 loss affects ileal IEC subtypes and their function in vivo.MethodsConstitutive Ptpn2 wild-type, heterozygous, and knockout (KO) mice, as well as mice with inducible deletion of Ptpn2 in IECs, were used in the study. Investigation was performed using imaging techniques, flow cytometry, enteroid culture, and analysis of gene and protein levels of IEC markers.ResultsPartial transcriptome analysis showed that expression of Paneth cell-associated antimicrobial peptides Lyz1, Pla2g2a, and Defa6 was down-regulated markedly in Ptpn2-KO mice compared with wild-type and heterozygous. In parallel, Paneth cell numbers were reduced, their endoplasmic reticulum architecture was disrupted, and the endoplasmic reticulum stress protein, C/EBP-homologous protein (CHOP), was increased in Ptpn2-KO mice. Despite reduced Paneth cell number, flow cytometry showed increased expression of the Paneth cell-stimulatory cytokines interleukin 22 and interferon γ+ in CD4+ T cells isolated from Ptpn2-KO ileum. Key findings in constitutive Ptpn2-KO mice were confirmed in epithelium-specific Ptpn2ΔIEC mice, which also showed impaired lysozyme protein levels in Paneth cells compared with Ptpn2fl/fl control mice.ConclusionsConstitutive Ptpn2 deficiency affects Paneth cell viability and compromises Paneth cell-specific antimicrobial peptide production. The observed effects may contribute to the increased susceptibility to intestinal infection and dysbiosis in these mice.
- Published
- 2023
15. Paneth cell TNF signaling induces gut bacterial translocation and sepsis.
- Author
-
Wallaeys, Charlotte, Garcia-Gonzalez, Natalia, Timmermans, Steven, Vandewalle, Jolien, Vanderhaeghen, Tineke, De Beul, Somara, Dufoor, Hester, Eggermont, Melanie, Moens, Elise, Bosteels, Victor, De Rycke, Riet, Thery, Fabien, Impens, Francis, Verbanck, Serge, Lienenklaus, Stefan, Janssens, Sophie, Blumberg, Richard S., Iwawaki, Takao, and Libert, Claude
- Abstract
The cytokine tumor necrosis factor (TNF) plays important roles in limiting infection but is also linked to sepsis. The mechanisms underlying these paradoxical roles are unclear. Here, we show that TNF limits the antimicrobial activity of Paneth cells (PCs), causing bacterial translocation from the gut to various organs. This TNF-induced lethality does not occur in mice with a PC-specific deletion in the TNF receptor, P55. In PCs, TNF stimulates the IFN pathway and ablates the steady-state unfolded protein response (UPR), effects not observed in mice lacking P55 or IFNAR1. TNF triggers the transcriptional downregulation of IRE1 key genes Ern1 and Ern2 , which are key mediators of the UPR. This UPR deficiency causes a significant reduction in antimicrobial peptide production and PC antimicrobial activity, causing bacterial translocation to organs and subsequent polymicrobial sepsis, organ failure, and death. This study highlights the roles of PCs in bacterial control and therapeutic targets for sepsis. [Display omitted] • PC-specific TNFR1 (P55) mutant mice are protected against lethal TNF effects • PC-P55 signaling triggers an interferon signature and UPR failure in these cells • This UPR failure in PCs is IFNAR1 and P55 dependent but microbiome independent • PC-P55 UPR failure reduces AMP activity, causing bacterial escape and sepsis Wallaeys et al. studied Paneth cells (PCs) during TNF-induced inflammation. TNF signaling through its P55 receptor on PCs triggers an interferon signature and disrupts the unfolded protein response, causing reduced production of antimicrobial peptides and decreased antimicrobial activity. This PC dysfunction causes gut bacterial translocation and sepsis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Flagella-driven motility is a target of human Paneth cell defensin activity
- Author
-
Akahoshi, Douglas T, Natwick, Dean E, Yuan, Weirong, Lu, Wuyuan, Collins, Sean R, and Bevins, Charles L
- Subjects
Microbiology ,Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Prevention ,Emerging Infectious Diseases ,Biotechnology ,Digestive Diseases ,Animals ,Humans ,Paneth Cells ,Flagellin ,Anti-Infective Agents ,Bacteria ,Flagella ,Mammals ,Immunology ,Medical Microbiology ,Virology ,Medical microbiology - Abstract
In the mammalian intestine, flagellar motility can provide microbes competitive advantage, but also threatens the spatial segregation established by the host at the epithelial surface. Unlike microbicidal defensins, previous studies indicated that the protective activities of human α-defensin 6 (HD6), a peptide secreted by Paneth cells of the small intestine, resides in its remarkable ability to bind microbial surface proteins and self-assemble into protective fibers and nets. Given its ability to bind flagellin, we proposed that HD6 might be an effective inhibitor of bacterial motility. Here, we utilized advanced automated live cell fluorescence imaging to assess the effects of HD6 on actively swimming Salmonella enterica in real time. We found that HD6 was able to effectively restrict flagellar motility of individual bacteria. Flagellin-specific antibody, a classic inhibitor of flagellar motility that utilizes a mechanism of agglutination, lost its activity at low bacterial densities, whereas HD6 activity was not diminished. A single amino acid variant of HD6 that was able to bind flagellin, but not self-assemble, lost ability to inhibit flagellar motility. Together, these results suggest a specialized role of HD6 self-assembly into polymers in targeting and restricting flagellar motility.
- Published
- 2023
17. Paneth cell proteins DEFA6 and GUCA2A as tissue markers in necrotizing enterocolitis
- Author
-
Hoffsten, Alice, Lilja, Helene Engstrand, Mobini-Far, Hamid, Sindelar, Richard, and Markasz, Laszlo
- Published
- 2023
- Full Text
- View/download PDF
18. LAT1 expression influences Paneth cell number and tumor development in ApcMin/+ mice
- Author
-
Sui, Yunlong, Hoshi, Namiko, Ohgaki, Ryuichi, Kong, Lingling, Yoshida, Ryutaro, Okamoto, Norihiro, Kinoshita, Masato, Miyazaki, Haruka, Ku, Yuna, Tokunaga, Eri, Ito, Yuki, Watanabe, Daisuke, Ooi, Makoto, Shinohara, Masakazu, Sasaki, Kengo, Zen, Yoh, Kotani, Takenori, Matozaki, Takashi, Tian, Zibin, Kanai, Yoshikatsu, and Kodama, Yuzo
- Published
- 2023
- Full Text
- View/download PDF
19. Paneth cell-derived iNOS is required to maintain homeostasis in the intestinal stem cell niche
- Author
-
Huang, Lingxiao, Xu, Zhenni, Lei, Xudan, Huang, Yujun, Tu, Siyu, Xu, Lu, Xia, Jieying, and Liu, Dengqun
- Published
- 2023
- Full Text
- View/download PDF
20. Paneth cells in farm animals: current status and future direction
- Author
-
Cui, Chenbin, Li, Lindeng, Wu, Lin, Wang, Xinru, Zheng, Yao, Wang, Fangke, Wei, Hongkui, and Peng, Jian
- Published
- 2023
- Full Text
- View/download PDF
21. Treatment of Patients With Teduglutide (GLP-2) for GVHD and Analysis of Paneth Cells of GVHD Patients
- Author
-
Robert Zeiser, Director of the Division of Tumor Immunology
- Published
- 2023
22. Human intelectin‐2 (ITLN2) is selectively expressed by secretory Paneth cells
- Author
-
Nonnecke, Eric B, Castillo, Patricia A, Johansson, Malin EV, Hollox, Edward J, Shen, Bo, Lönnerdal, Bo, and Bevins, Charles L
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,Crohn's Disease ,Autoimmune Disease ,Inflammatory Bowel Disease ,Prevention ,Digestive Diseases ,Aetiology ,2.1 Biological and endogenous factors ,Oral and gastrointestinal ,Autophagy-Related Proteins ,Crohn Disease ,Humans ,Lectins ,Nod2 Signaling Adaptor Protein ,Paneth Cells ,RNA ,Messenger ,Secretory Vesicles ,alpha-defensin ,Crohn's disease ,innate immunity ,intelectin ,lectin ,metaplasia ,omentin ,Paneth cell ,small intestine ,ulcerative colitis ,Physiology ,Medical Physiology ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical physiology - Abstract
Intelectins (intestinal lectins) are highly conserved across chordate evolution and have been implicated in various human diseases, including Crohn's disease (CD). The human genome encodes two intelectin genes, intelectin-1 (ITLN1) and intelectin-2 (ITLN2). Other than its high sequence similarity with ITLN1, little is known about ITLN2. To address this void in knowledge, we report that ITLN2 exhibits discrete, yet notable differences from ITLN1 in primary structure, including a unique amino terminus, as well as changes in amino acid residues associated with the glycan-binding activity of ITLN1. We identified that ITLN2 is a highly abundant Paneth cell-specific product, which localizes to secretory granules, and is expressed as a multimeric protein in the small intestine. In surgical specimens of ileal CD, ITLN2 mRNA levels were reduced approximately five-fold compared to control specimens. The ileal expression of ITLN2 was unaffected by previously reported disease-associated variants in ITLN2 and CD-associated variants in neighboring ITLN1 as well as NOD2 and ATG16L1. ITLN2 mRNA expression was undetectable in control colon tissue; however, in both ulcerative colitis (UC) and colonic CD, metaplastic Paneth cells were found to express ITLN2. Together, the data reported establish the groundwork for understanding ITLN2 function(s) in the intestine, including its possible role in CD.
- Published
- 2022
23. Intestinal commensal microbiota and cytokines regulate Fut2+ Paneth cells for gut defense
- Author
-
Kamioka, Mariko, Goto, Yoshiyuki, Nakamura, Kiminori, Yokoi, Yuki, Sugimoto, Rina, Ohira, Shuya, Kurashima, Yosuke, Umemoto, Shingo, Sato, Shintaro, Kunisawa, Jun, Takahashi, Yu, Domino, Steven E, Renauld, Jean-Christophe, Nakae, Susumu, Iwakura, Yoichiro, Ernst, Peter B, Ayabe, Tokiyoshi, and Kiyono, Hiroshi
- Subjects
Microbiology ,Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Stem Cell Research ,Prevention ,Vaccine Related ,Digestive Diseases ,Biodefense ,Aetiology ,2.1 Biological and endogenous factors ,Oral and gastrointestinal ,Animals ,Cytokines ,Fucosyltransferases ,Gastrointestinal Microbiome ,Ileum ,Interleukin-17 ,Interleukins ,Mice ,Paneth Cells ,Symbiosis ,alpha-Defensins ,Interleukin-22 ,Galactoside 2-alpha-L-fucosyltransferase ,Paneth cell ,alpha-defensin ,fucosyltransferase 2 ,IL-22 ,IL-17a ,α-defensin - Abstract
Paneth cells are intestinal epithelial cells that release antimicrobial peptides, such as α-defensin as part of host defense. Together with mesenchymal cells, Paneth cells provide niche factors for epithelial stem cell homeostasis. Here, we report two subtypes of murine Paneth cells, differentiated by their production and utilization of fucosyltransferase 2 (Fut2), which regulates α(1,2)fucosylation to create cohabitation niches for commensal bacteria and prevent invasion of the intestine by pathogenic bacteria. The majority of Fut2- Paneth cells were localized in the duodenum, whereas the majority of Fut2+ Paneth cells were in the ileum. Fut2+ Paneth cells showed higher granularity and structural complexity than did Fut2- Paneth cells, suggesting that Fut2+ Paneth cells are involved in host defense. Signaling by the commensal bacteria, together with interleukin 22 (IL-22), induced the development of Fut2+ Paneth cells. IL-22 was found to affect the α-defensin secretion system via modulation of Fut2 expression, and IL-17a was found to increase the production of α-defensin in the intestinal tract. Thus, these intestinal cytokines regulate the development and function of Fut2+ Paneth cells as part of gut defense.
- Published
- 2022
24. Paneth cell-derived iNOS is required to maintain homeostasis in the intestinal stem cell niche
- Author
-
Lingxiao Huang, Zhenni Xu, Xudan Lei, Yujun Huang, Siyu Tu, Lu Xu, Jieying Xia, and Dengqun Liu
- Subjects
iNOS ,Paneth cell ,Intestinal stem cell ,Proliferation ,Differentiation ,Medicine - Abstract
Abstract Background Mammalian intestinal epithelium constantly undergoes rapid self-renewal and regeneration sustained by intestinal stem cells (ISCs) within crypts. Inducible nitric oxide synthase (iNOS) is an important regulator in tissue homeostasis and inflammation. However, the functions of iNOS on ISCs have not been clarified. Here, we aimed to investigate the expression pattern of inducible nitric oxide synthase (iNOS) within crypts and explore its function in the homeostatic maintenance of the ISC niche. Methods Expression of iNOS was determined by tissue staining and qPCR. iNOS−/− and Lgr5 transgenic mice were used to explore the influence of iNOS ablation on ISC proliferation and differentiation. Enteroids were cultured to study the effect of iNOS on ISCs in vitro. Ileum samples from wild-type and iNOS−/− mice were collected for RNA-Seq to explore the molecular mechanisms by which iNOS regulates ISCs. Results iNOS was physiologically expressed in Paneth cells. Knockout of iNOS led to apparent morphological changes in the intestine, including a decrease in the small intestine length and in the heights of both villi and crypts. Knockout of iNOS decreased the number of Ki67+ or BrdU+ proliferative cells in crypts. Loss of iNOS increased the number of Olfm4+ ISCs but inhibited the differentiation and migration of Lgr5+ ISCs in vivo. iNOS depletion also inhibited enteroid formation and the budding efficiency of crypts in vitro. Moreover, iNOS deficiency altered gluconeogenesis and the adaptive immune response in the ileum transcriptome. Conclusion Paneth cell-derived iNOS is required to maintain a healthy ISC niche, and Knockout of iNOS hinders ISC function in mice. Therefore, iNOS represents a potential target for the development of new drugs and other therapeutic interventions for intestinal disorders.
- Published
- 2023
- Full Text
- View/download PDF
25. To sense or not to sense, Paneth cell regulation of mucosal immunity.
- Author
-
Weis, Sebastian, King, Irah L., and Vivas, Wolfgang
- Abstract
Paneth cells located within intestinal crypts support epithelial stem cells and immunity through growth factors and antimicrobial peptides. In this issue of Cell Host & Microbe , Wallaeys et al. report that TNF sensing by Paneth cells disrupts the unfolded protein response and decreases antimicrobial peptides, causing bacterial translocation and sepsis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Identification and Characterization of Multiple Paneth Cell Types in the Mouse Small Intestine
- Author
-
Steven Timmermans, Charlotte Wallaeys, Natalia Garcia-Gonzalez, Lotte Pollaris, Yvan Saeys, and Claude Libert
- Subjects
Paneth cells ,single-cell RNA-seq ,small intestine ,Cytology ,QH573-671 - Abstract
The small intestinal crypts harbor secretory Paneth cells (PCs) which express bactericidal peptides that are crucial for maintaining intestinal homeostasis. Considering the diverse environmental conditions throughout the course of the small intestine, multiple subtypes of PCs are expected to exist. We applied single-cell RNA-sequencing of PCs combined with deep bulk RNA-sequencing on PC populations of different small intestinal locations and discovered several expression-based PC clusters. Some of these are discrete and resemble tuft cell-like PCs, goblet cell (GC)-like PCs, PCs expressing stem cell markers, and atypical PCs. Other clusters are less discrete but appear to be derived from different locations along the intestinal tract and have environment-dictated functions such as food digestion and antimicrobial peptide production. A comprehensive spatial analysis using Resolve Bioscience was conducted, leading to the identification of different PC’s transcriptomic identities along the different compartments of the intestine, but not between PCs in the crypts themselves.
- Published
- 2024
- Full Text
- View/download PDF
27. Nur77 as a novel regulator of Paneth cell differentiation and function
- Author
-
Cui, Chenbin, Wang, Xinru, Zheng, Yao, Wu, Lin, Li, Lindeng, Wei, Hongkui, and Peng, Jian
- Published
- 2024
- Full Text
- View/download PDF
28. Paneth cells in farm animals: current status and future direction
- Author
-
Chenbin Cui, Lindeng Li, Lin Wu, Xinru Wang, Yao Zheng, Fangke Wang, Hongkui Wei, and Jian Peng
- Subjects
Antimicrobial peptide ,Farm animal ,Intestinal organoid ,Intestine ,Paneth cell ,Animal culture ,SF1-1100 ,Veterinary medicine ,SF600-1100 - Abstract
Abstract A healthy intestine plays an important role in the growth and development of farm animals. In small intestine, Paneth cells are well known for their regulation of intestinal microbiota and intestinal stem cells (ISCs). Although there has been a lot of studies and reviews on human and murine Paneth cells under intestinal homeostasis or disorders, little is known about Paneth cells in farm animals. Most farm animals possess Paneth cells in their small intestine, as identified by various staining methods, and Paneth cells of various livestock species exhibit noticeable differences in cell shape, granule number, and intestinal distribution. Paneth cells in farm animals and their antimicrobial peptides (AMPs) are susceptible to multiple factors such as dietary nutrients and intestinal infection. Thus, the comprehensive understanding of Paneth cells in different livestock species will contribute to the improvement of intestinal health. This review first summarizes the current status of Paneth cells in pig, cattle, sheep, horse, chicken and rabbit, and points out future directions for the investigation of Paneth cells in the reviewed animals.
- Published
- 2023
- Full Text
- View/download PDF
29. Paneth cell ontogeny in term and preterm ovine models
- Author
-
Geoanna M. Bautista, Anjali J. Cera, Rebecca J. Schoenauer, Michele Persiani, Satyan Lakshminrusimha, Praveen Chandrasekharan, Sylvia F Gugino, Mark A. Underwood, and Steven J. McElroy
- Subjects
Paneth cell ,immature intestine ,perinatal bradycardic stress ,preterm ,ovine ,Veterinary medicine ,SF600-1100 - Abstract
IntroductionPaneth cells are critically important to intestinal health, including protecting intestinal stem cells, shaping the intestinal microbiome, and regulating host immunity. Understanding Paneth cell biology in the immature intestine is often modeled in rodents with little information in larger mammals such as sheep. Previous studies have only established the distribution pattern of Paneth cells in healthy adult sheep. Our study aimed to examine the ontogeny, quantification, and localization of Paneth cells in fetal and newborn lambs at different gestational ages and with perinatal transient asphyxia. We hypothesized that ovine Paneth cell distribution at birth resembles the pattern seen in humans (highest concentrations in the ileum) and that ovine Paneth cell density is gestation-dependent.MethodsIntestinal samples were obtained from 126–127 (preterm, with and without perinatal transient asphyxia) and 140–141 (term) days gestation sheep. Samples were quantified per crypt in at least 100 crypts per animal and confirmed as Paneth cells through in immunohistochemistry.ResultsPaneth cells had significantly higher density in the ileum compared to the jejunum and were absent in the colon.DiscussionExposure to perinatal transient asphyxia acutely decreased Paneth cell numbers. These novel data support the possibility of utilizing ovine models for understanding Paneth cell biology in the fetus and neonate.
- Published
- 2024
- Full Text
- View/download PDF
30. Development of a deep learning algorithm for Paneth cell density quantification for inflammatory bowel diseaseResearch in context
- Author
-
Liang-I Kang, Kathryn Sarullo, Jon N. Marsh, Liang Lu, Pooja Khonde, Changqing Ma, Talin Haritunians, Angela Mujukian, Emebet Mengesha, Dermot P.B. McGovern, Thaddeus S. Stappenbeck, S. Joshua Swamidass, and Ta-Chiang Liu
- Subjects
Whole slide images ,Crohn's disease ,Pathology ,Prognosis ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Alterations in ileal Paneth cell (PC) density have been described in gut inflammatory diseases such as Crohn's disease (CD) and could be used as a biomarker for disease prognosis. However, quantifying PCs is time-intensive, a barrier for clinical workflow. Deep learning (DL) has transformed the development of robust and accurate tools for complex image evaluation. Our aim was to use DL to quantify PCs for use as a quantitative biomarker. Methods: A retrospective cohort of whole slide images (WSI) of ileal tissue samples from patients with/without inflammatory bowel disease (IBD) was used for the study. A pathologist-annotated training set of WSI were used to train a U-net two-stage DL model to quantify PC number, crypt number, and PC density. For validation, a cohort of 48 WSIs were manually quantified by study pathologists and compared to the DL algorithm, using root mean square error (RMSE) and the coefficient of determination (r2) as metrics. To test the value of PC quantification as a biomarker, resection specimens from patients with CD (n = 142) and without IBD (n = 48) patients were analysed with the DL model. Finally, we compared time to disease recurrence in patients with CD with low versus high DL-quantified PC density using Log-rank test. Findings: Initial one-stage DL model showed moderate accuracy in predicting PC density in cross-validation tests (RMSE = 1.880, r2 = 0.641), but adding a second stage significantly improved accuracy (RMSE = 0.802, r2 = 0.748). In the validation of the two-stage model compared to expert pathologists, the algorithm showed good performance up to RMSE = 1.148, r2 = 0.708. The retrospective cross-sectional cohort had mean ages of 62.1 years in the patients without IBD and 38.6 years for the patients with CD. In the non-IBD cohort, 43.75% of the patients were male, compared to 49.3% of the patients with CD. Analysis by the DL model showed significantly higher PC density in non-IBD controls compared to the patients with CD (4.04 versus 2.99 PC/crypt). Finally, the algorithm quantification of PCs density in patients with CD showed patients with the lowest 25% PC density (Quartile 1) have significantly shorter recurrence-free interval (p = 0.0399). Interpretation: The current model performance demonstrates the feasibility of developing a DL-based tool to measure PC density as a predictive biomarker for future clinical practice. Funding: This study was funded by the National Institutes of Health (NIH).
- Published
- 2024
- Full Text
- View/download PDF
31. Macrophage LRRK2 hyperactivity impairs autophagy and induces Paneth cell dysfunction.
- Author
-
Sun, Shengxiang, Hodel, Miki, Wang, Xiang, De Vicente, Javier, Haritunians, Talin, Debebe, Anketse, Hung, Chen-Ting, Ma, Changqing, Malique, Atika, Nguyen, Hoang N., Agam, Maayan, Maloney, Michael T., Goo, Marisa S., Kluss, Jillian H., Mishra, Richa, Frein, Jennifer, Foster, Amanda, Ballentine, Samuel, Pandey, Uday, and Kern, Justin
- Abstract
LRRK2 polymorphisms (G2019S/N2081D) that increase susceptibility to Parkinson's disease and Crohn's disease (CD) lead to LRRK2 kinase hyperactivity and suppress autophagy. This connection suggests that LRRK2 kinase inhibition, a therapeutic strategy being explored for Parkinson's disease, may also benefit patients with CD. Paneth cell homeostasis is tightly regulated by autophagy, and their dysfunction is a precursor to gut inflammation in CD. Here, we found that patients with CD and mice carrying hyperactive LRRK2 polymorphisms developed Paneth cell dysfunction. We also found that LRRK2 kinase can be activated in the context of interactions between genes (genetic autophagy deficiency) and the environment (cigarette smoking). Unexpectedly, lamina propria immune cells were the main intestinal cell types that express LRRK2, instead of Paneth cells as previously suggested. We showed that LRRK2-mediated pro-inflammatory cytokine release from phagocytes impaired Paneth cell function, which was rescued by LRRK2 kinase inhibition through activation of autophagy. Together, these data suggest that LRRK2 kinase inhibitors maintain Paneth cell homeostasis by restoring autophagy and may represent a therapeutic strategy for CD. Editor's summary: Specific variants in the leucine-rich repeat kinase 2 (LRRK2) gene are associated with increased susceptibility to Crohn's disease (CD), but how LRRK2 contributes to the pathogenesis of CD is not fully understood. Using mice harboring LRRK2 risk alleles, Sun et al. found that hyperactive LRRK2 in macrophages led to impaired autophagy and Paneth cell dysfunction. Pathogenic LRRK2 kinase activity could also be triggered by deficiency of the autophagy gene ATG16L1 in combination with cigarette smoke and rescued by an LRRK2 inhibitor. Together, these findings identify a Paneth cell–extrinsic pathway by which hyperactive LRRK2 interferes with autophagy and intestinal epithelial homeostasis. —Claire Olingy [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
32. Paneth cell maturation is related to epigenetic modification during neonatal–weaning transition
- Author
-
Baba, Ryoko, Kokubu, Keiji, Nakamura, Kenta, Fujita, Mamoru, and Morimoto, Hiroyuki
- Published
- 2022
- Full Text
- View/download PDF
33. Azathioprine promotes intestinal epithelial cell differentiation into Paneth cells and alleviates ileal Crohn’s disease severity
- Author
-
Mohab Ragab, Heidi Schlichting, Maren Hicken, Patricia Mester, Misa Hirose, Larissa N. Almeida, Lea Christiansen, Saleh Ibrahim, Hauke Christian Tews, Senad Divanovic, Christian Sina, and Stefanie Derer
- Subjects
Medicine ,Science - Abstract
Abstract Paneth cells (PCs), a subset of intestinal epithelial cells (IECs) found at the base of small intestinal crypts, play an essential role in maintaining intestinal homeostasis. Altered PCs function is associated with diverse intestinal pathologies, including ileal Crohn’s disease (CD). CD patients with ileal involvement have been previously demonstrated to display impairment in PCs and decreased levels of anti-microbial peptides. Although the immunosuppressive drug Azathioprine (AZA) is widely used in CD therapy, the impact of AZA on IEC differentiation remains largely elusive. In the present study, we hypothesized that the orally administered drug AZA also exerts its effect through modulation of the intestinal epithelium and specifically via modulation of PC function. AZA-treated CD patients exhibited an ileal upregulation of AMPs on both mRNA and protein levels compared to non-AZA treated patients. Upon in vitro AZA stimulation, intestinal epithelial cell line MODE-K exhibited heightened expression levels of PC marker in concert with diminished cell proliferation but boosted mitochondrial OXPHOS activity. Moreover, differentiation of IECs, including PCs differentiation, was boosted in AZA-treated murine small intestinal organoids and was associated with decreased D-glucose consumption and decreased growth rates. Of note, AZA treatment strongly decreased Lgr5 mRNA expression as well as Ki67 positive cells. Further, AZA restored dysregulated PCs associated with mitochondrial dysfunction. AZA-dependent inhibition of IEC proliferation is accompanied by boosted mitochondria function and IEC differentiation into PC.
- Published
- 2024
- Full Text
- View/download PDF
34. The Paneth Cell: The Curator and Defender of the Immature Small Intestine
- Author
-
Lueschow, Shiloh R and McElroy, Steven J
- Subjects
Pediatric ,Perinatal Period - Conditions Originating in Perinatal Period ,Preterm ,Low Birth Weight and Health of the Newborn ,Prevention ,Digestive Diseases ,Infant Mortality ,2.1 Biological and endogenous factors ,Aetiology ,Reproductive health and childbirth ,Oral and gastrointestinal ,Animals ,Enterocolitis ,Necrotizing ,Humans ,Infant ,Newborn ,Infant ,Premature ,Intestine ,Small ,Paneth Cells ,paneth cell ,necrotizing enterocolitis ,immature intestine ,defensins ,cathelicidin ,cell death ,Immunology ,Medical Microbiology - Abstract
Paneth cells were first described in the late 19th century by Gustav Schwalbe and Josef Paneth as columnar epithelial cells possessing prominent eosinophilic granules in their cytoplasm. Decades later there is continued interest in Paneth cells as they play an integral role in maintaining intestinal homeostasis and modulating the physiology of the small intestine and its associated microbial flora. Paneth cells are highly specialized secretory epithelial cells located in the small intestinal crypts of Lieberkühn. The dense granules produced by Paneth cells contain an abundance of antimicrobial peptides and immunomodulating proteins that function to regulate the composition of the intestinal flora. This in turn plays a significant role in secondary regulation of the host microvasculature, the normal injury and repair mechanisms of the intestinal epithelial layer, and the levels of intestinal inflammation. These critical functions may have even more importance in the immature intestine of premature infants. While Paneth cells begin to develop in the middle of human gestation, they do not become immune competent or reach their adult density until closer to term gestation. This leaves preterm infants deficient in normal Paneth cell biology during the greatest window of susceptibility to develop intestinal pathology such as necrotizing enterocolitis (NEC). As 10% of infants worldwide are currently born prematurely, there is a significant population of infants contending with an inadequate cohort of Paneth cells. Infants who have developed NEC have decreased Paneth cell numbers compared to age-matched controls, and ablation of murine Paneth cells results in a NEC-like phenotype suggesting again that Paneth cell function is critical to homeostasis to the immature intestine. This review will provide an up to date and comprehensive look at Paneth cell ontogeny, the impact Paneth cells have on the host-microbial axis in the immature intestine, and the repercussions of Paneth cell dysfunction or loss on injury and repair mechanisms in the immature gut.
- Published
- 2020
35. Paneth-like cells disruption and intestinal dysbiosis in the development of enterocolitis in an iatrogenic rectosigmoid hypoganglionosis rat model
- Author
-
Iskandar Rahardjo Budianto, Kusmardi Kusmardi, Andi Muh. Maulana, Somasundaram Arumugam, Rejina Afrin, and Vivian Soetikno
- Subjects
Hirschsprung disease ,hypoganglionosis ,inflammation ,antimicrobial peptide ,enterocolitis ,Surgery ,RD1-811 - Abstract
BackgroundHypoganglionosis resembles Hirschsprung disease (HSCR) which is characterized by severe constipation. Enterocolitis due to hypoganglionosis or Hirschsprung-associated enterocolitis (HAEC) is a life-threatening complication of both diseases. This study investigated the role of Paneth-like cells (PLCs) and gut microbiota in the development of enterocolitis in an iatrogenic rectosigmoid hypoganglionosis rat model.MethodsThe rectosigmoid serosa of male Sprague-Dawley rats were exposed to 0.1% benzalkonium chloride (BAC). The rats were then sacrificed after 1, 3, 5, 8, and 12 weeks. A sham group was sacrificed on Week 12. With hematoxylin-eosin staining, the ganglionic cells were quantified, the degree of enterocolitis was analyzed, and the PLCs was identified. Intestinal barrier function was assessed for the anti-peripherin, occludin, and acetylcholinesterase (AChE)/butyrylcholinesterase (BChE) ratio. qRT-PCR was used as reference for the evaluation of antimicrobial peptide (AMP) of PLCs using cryptdins, secretory Phospholipase A2, and lysozyme levels. 16S rRNA high-throughput sequencing on fecal samples was performed to analyze the changes in the intestinal microbiota diversity in each group.ResultsAfter 1 week of intervention, the ganglion cells were fewer in all sacrificial 0.1% BAC groups at varying times than those in the sham group. Occludin and peripherin were decreased, while the AChE/BChE ratio was increased. At Week 5 postintervention, the number of α-defensins-positive PLCs increased in the sigmoid colon tissues from BAC-treated rats. Conversely, PLCs-produced AMP decreased from Week 5 to Week 12. The sham group demonstrated increased Lactobacillus and decreased Bacteroides, while the 0.1% BAC group exhibited reciprocal changes, indicating dysbiosis. Enterocolitis occurred from Week 1 postintervention.ConclusionApplication with BAC influences the disruption of PLCs in Week 5 postintervention, and dysbiosis exacerbate the occurrence of enterocolitis. Further research on Paneth cells involvement in HAEC development is warranted.
- Published
- 2024
- Full Text
- View/download PDF
36. Paneth cells protect intestinal stem cell niche to alleviate deoxynivalenol-induced intestinal injury
- Author
-
Chenbin Cui, Xinru Wang, Yao Zheng, Lindeng Li, Fangke Wang, Hongkui Wei, and Jian Peng
- Subjects
Paneth cell ,Deoxynivalenol ,Intestinal microbiota ,Intestinal stem cell ,Lysozyme ,Wnt/β-catenin signaling ,Environmental pollution ,TD172-193.5 ,Environmental sciences ,GE1-350 - 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.
- Published
- 2023
- Full Text
- View/download PDF
37. Paneth cell: The missing link between obesity, MASH and portal hypertension
- Author
-
Kumar, Minu Sajeev
- Published
- 2024
- Full Text
- View/download PDF
38. Investigators at Washington University Report Findings in Crohn's Disease (Macrophage Lrrk2 Hyperactivity Impairs Autophagy and Induces Paneth Cell Dysfunction)
- Subjects
Medical research -- Reports ,Medicine, Experimental -- Reports ,Macrophages -- Reports -- Research ,Gastrointestinal diseases -- Research ,Physical fitness -- Research -- Reports ,Health - Abstract
2024 DEC 14 (NewsRx) -- By a News Reporter-Staff News Editor at Obesity, Fitness & Wellness Week -- Research findings on Digestive System Diseases and Conditions - Crohn's Disease are [...]
- Published
- 2024
39. Metastasis of colon cancer requires Dickkopf-2 to generate cancer cells with Paneth cell properties (Updated September 25, 2024)
- Subjects
Colon cancer ,Metastasis ,Stem cells ,Cancer cells ,Health - Abstract
2024 OCT 7 (NewsRx) -- By a News Reporter-Staff News Editor at Stem Cell Week -- According to news reporting based on a preprint abstract, our journalists obtained the following [...]
- Published
- 2024
40. Feeding Formula Eliminates the Necessity of Bacterial Dysbiosis and Induces Inflammation and Injury in the Paneth Cell Disruption Murine NEC Model in an Osmolality-Dependent Manner.
- Author
-
Lueschow, Shiloh R, Kern, Stacy L, Gong, Huiyu, Grobe, Justin L, Segar, Jeffrey L, Carlson, Susan J, and McElroy, Steven J
- Subjects
Paneth Cells ,Intestine ,Small ,Animals ,Mice ,Inbred C57BL ,Animals ,Newborn ,Humans ,Mice ,Enterocolitis ,Necrotizing ,Infant ,Newborn ,Diseases ,Disease Models ,Animal ,Inflammation ,Osmolar Concentration ,Infant Formula ,Infant ,Newborn ,Dysbiosis ,Gastrointestinal Microbiome ,formula ,immature intestine ,inflammation ,microbiome ,necrotizing enterocolitis ,osmolality ,Food Sciences ,Nutrition and Dietetics - Abstract
Necrotizing enterocolitis (NEC) remains a significant cause of morbidity and mortality in preterm infants. Formula feeding is a risk factor for NEC and osmolality, which is increased by the fortification that is required for adequate growth of the infant, has been suggested as a potential cause. Our laboratory has shown that Paneth cell disruption followed by induction of dysbiosis can induce NEC-like pathology in the absence of feeds. We hypothesized adding formula feeds to the model would exacerbate intestinal injury and inflammation in an osmolality-dependent manner. NEC-like injury was induced in 14-16 day-old C57Bl/6J mice by Paneth cell disruption with dithizone or diphtheria toxin, followed by feeding rodent milk substitute with varying osmolality (250-1491 mOsm/kg H2O). Animal weight, serum cytokines and osmolality, small intestinal injury, and cecal microbial composition were quantified. Paneth cell-disrupted mice fed formula had significant NEC scores compared to controls and no longer required induction of bacterial dysbiosis. Significant increases in serum inflammatory markers, small intestinal damage, and overall mortality were osmolality-dependent and not related to microbial changes. Overall, formula feeding in combination with Paneth cell disruption induced NEC-like injury in an osmolality-dependent manner, emphasizing the importance of vigilance in designing preterm infant feeds.
- Published
- 2020
41. Paneth cells as the cornerstones of intestinal and organismal health: a primer
- Author
-
Wallaeys, Charlotte, Garcia‐Gonzalez, Natalia, and Libert, Claude
- Published
- 2023
- Full Text
- View/download PDF
42. HNF4α Acts as Upstream Functional Regulator of Intestinal Wnt3 and Paneth Cell FateSummary
- Author
-
Christine Jones, Mariano Avino, Véronique Giroux, and Francois Boudreau
- Subjects
Enteroids ,HNF4α ,Paneth Cells ,Wnt3 ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Background & Aims: The intestinal epithelium intrinsically renews itself ex vivo via the proliferation of Lgr5+ intestinal stem cells, which is sustained by the establishment of an epithelial stem cell niche. Differentiated Paneth cells are the main source of epithelial-derived niche factor supplies and produce Wnt3 as an essential factor in supporting Lgr5+ stem cell activity in the absence of redundant mesenchymal Wnts. Maturation of Paneth cells depends on canonical Wnt signaling, but few transcriptional regulators have been identified to this end. The role of HNF4α in intestinal epithelial cell differentiation is considered redundant with its paralog HNF4γ. However, it is unclear whether HNF4α alone controls intrinsic intestinal epithelial cell growth and fate in the absence of a mesenchymal niche. Methods: We used transcriptomic analyses to dissect the role of HNF4α in the maintenance of jejunal epithelial culture when cultured ex vivo as enteroids in the presence or absence of compensatory mesenchymal cells. Results: HNF4α plays a crucial role in supporting the growth and survival of jejunal enteroids. Transcriptomic analyses revealed an autonomous function of HNF4α in Wnt3 transcriptional regulation and Paneth cell differentiation. We showed that Wnt3a supplementation or co-culture with intestinal subepithelial mesenchymal cells reversed cell death and transcriptional changes caused by the deletion of Hnf4a in jejunal enteroids. Conclusions: Our results support the intrinsic epithelial role of HNF4α in regulating Paneth cell homeostasis and intestinal epithelium renewal in the absence of compensatory Wnt signaling.
- Published
- 2023
- Full Text
- View/download PDF
43. Paneth-like cells produced from OLFM4+ stem cells support OLFM4+ stem cell growth in advanced colorectal cancer
- Author
-
Mizuho Sakahara, Takuya Okamoto, Upasna Srivastava, Yasuko Natsume, Hitomi Yamanaka, Yutaka Suzuki, Kazutaka Obama, Satoshi Nagayama, and Ryoji Yao
- Subjects
Biology (General) ,QH301-705.5 - Abstract
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.
- Published
- 2024
- Full Text
- View/download PDF
44. The RNA helicase Dhx15 mediates Wnt-induced antimicrobial protein expression in Paneth cells
- Author
-
Wang, Yalong, He, Kaixin, Sheng, Baifa, Lei, Xuqiu, Tao, Wanyin, Zhu, Xiaoliang, Wei, Zheng, Fu, Rongjie, Wang, Anlei, Bai, Shengdan, Zhang, Zhao, Hong, Na, Ye, Chao, Tian, Ye, Wang, Jun, Li, Mingsong, Zhang, Kaiguang, Li, Lin, Yang, Hua, Li, Hua-Bing, Flavell, Richard, and Zhu, Shu
- Published
- 2021
45. Identification and Characterization of Multiple Paneth Cell Types in the Mouse Small Intestine.
- Author
-
Timmermans, Steven, Wallaeys, Charlotte, Garcia-Gonzalez, Natalia, Pollaris, Lotte, Saeys, Yvan, and Libert, Claude
- Subjects
- *
ANTIMICROBIAL peptides , *LIFE sciences , *RNA sequencing , *STEM cells , *INTESTINES - Abstract
The small intestinal crypts harbor secretory Paneth cells (PCs) which express bactericidal peptides that are crucial for maintaining intestinal homeostasis. Considering the diverse environmental conditions throughout the course of the small intestine, multiple subtypes of PCs are expected to exist. We applied single-cell RNA-sequencing of PCs combined with deep bulk RNA-sequencing on PC populations of different small intestinal locations and discovered several expression-based PC clusters. Some of these are discrete and resemble tuft cell-like PCs, goblet cell (GC)-like PCs, PCs expressing stem cell markers, and atypical PCs. Other clusters are less discrete but appear to be derived from different locations along the intestinal tract and have environment-dictated functions such as food digestion and antimicrobial peptide production. A comprehensive spatial analysis using Resolve Bioscience was conducted, leading to the identification of different PC's transcriptomic identities along the different compartments of the intestine, but not between PCs in the crypts themselves. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
46. Paneth cells as the cornerstones of intestinal and organismal health: a primer
- Author
-
Charlotte Wallaeys, Natalia Garcia‐Gonzalez, and Claude Libert
- Subjects
paneth cells ,gut homeostasis ,infection ,antimicrobial peptides ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Paneth cells are versatile secretory cells located in the crypts of Lieberkühn of the small intestine. In normal conditions, they function as the cornerstones of intestinal health by preserving homeostasis. They perform this function by providing niche factors to the intestinal stem cell compartment, regulating the composition of the microbiome through the production and secretion of antimicrobial peptides, performing phagocytosis and efferocytosis, taking up heavy metals, and preserving barrier integrity. Disturbances in one or more of these functions can lead to intestinal as well as systemic inflammatory and infectious diseases. This review discusses the multiple functions of Paneth cells, and the mechanisms and consequences of Paneth cell dysfunction. It also provides an overview of the tools available for studying Paneth cells.
- Published
- 2022
- Full Text
- View/download PDF
47. Paneth cells protect intestinal stem cell niche to alleviate deoxynivalenol-induced intestinal injury
- Author
-
Cui, Chenbin, Wang, Xinru, Zheng, Yao, Li, Lindeng, Wang, Fangke, Wei, Hongkui, and Peng, Jian
- Published
- 2023
- Full Text
- View/download PDF
48. Interleukin-9 production by type 2 innate lymphoid cells induces Paneth cell metaplasia and small intestinal remodeling
- Author
-
Yuan, Chengyin, Rayasam, Aditya, Moe, Alison, Hayward, Michael, Wells, Clive, Szabo, Aniko, Mackenzie, Andrew, Salzman, Nita, and Drobyski, William R.
- Published
- 2023
- Full Text
- View/download PDF
49. Trans-Golgi protein TVP23B regulates host-microbe interactions via Paneth cell homeostasis and Goblet cell glycosylation
- Author
-
Song, Ran, McAlpine, William, Fond, Aaron M., Nair-Gill, Evan, Choi, Jin Huk, Nyström, Elisabeth E. L., Arike, Liisa, Field, Sydney, Li, Xiaohong, SoRelle, Jeffrey A., Moresco, James J., Moresco, Eva Marie Y., Yates, 3rd, John R., Azadi, Parastoo, Ni, Josephine, Birchenough, George M. H., Beutler, Bruce, and Turer, Emre E.
- Published
- 2023
- Full Text
- View/download PDF
50. Decreased Paneth cell α-defensins promote fibrosis in a choline-deficient L-amino acid-defined high-fat diet-induced mouse model of nonalcoholic steatohepatitis via disrupting intestinal microbiota
- Author
-
Nakamura, Shunta, Nakamura, Kiminori, Yokoi, Yuki, Shimizu, Yu, Ohira, Shuya, Hagiwara, Mizu, Song, Zihao, Gan, Li, Aizawa, Tomoyasu, Hashimoto, Daigo, Teshima, Takanori, Ouellette, Andre J., and Ayabe, Tokiyoshi
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.