1
|
Yang N, Pang YS, Zheng Y, Gong YJ, Ding WJ. Nobiletin restores the intestinal barrier of HFD-induced obese mice by promoting MHC-II expression and lipid metabolism. Mol Med 2025; 31:26. [PMID: 39865231 PMCID: PMC11770984 DOI: 10.1186/s10020-025-01072-1] [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: 10/27/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025] Open
Abstract
The incidence of obesity is increasing annually worldwide. A high-fat diet (HFD) causes intestinal barrier damage, but effective interventions are currently unavailable. Our previous work demonstrated the therapeutic effect of nobiletin on obese mice; thus, we hypothesized that nobiletin could reverse HFD-induced damage to the intestinal barrier. Male C57BL/6 J mice were orally administered nobiletin for 14 d. After identification, the obese mice were equally divided into three groups: the HFD group, the low-dose (NOL, 100 mg/kg/d) group and the high-dose nobiletin (NOH, 200 mg/kg/d) group. A normal control group (CON) was also included. Hematoxylin and eosin (HE) staining and immunofluorescence were used to observe the intestinal barrier. RT-qPCR was used to determine the transcriptomic levels of genes involved in intestinal barrier integrity and lipid metabolism. The results revealed that intestinal tight proteins, including ZO-1 and Occludin, were significantly reduced in HFD-fed mice but markedly restored after nobiletin intervention, particularly in NOH mice. Improvements in the intestinal barrier and lipid metabolism associated with major histocompatibility complex class II (MHC-II) and relevant elements were revealed after nobiletin intervention. Enrichment analysis revealed that MHC-II plays an important role in the restoration of the intestinal barrier. Taken together, nobiletin restored intestinal barrier integrity and lipid metabolism by regulating MHC-II expression.
Collapse
Affiliation(s)
- Ni Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yue-Shan Pang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yali Zheng
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Yan-Ju Gong
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Wei-Jun Ding
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China.
| |
Collapse
|
2
|
Perez RE, Eckerdt F, Platanias LC. Schlafens: Emerging Therapeutic Targets. Cancers (Basel) 2024; 16:1805. [PMID: 38791884 PMCID: PMC11119473 DOI: 10.3390/cancers16101805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
The interferon (IFN) family of immunomodulatory cytokines has been a focus of cancer research for over 50 years with direct and indirect implications in cancer therapy due to their properties to inhibit malignant cell proliferation and modulate immune responses. Among the transcriptional targets of the IFNs is a family of genes referred to as Schlafens. The products of these genes, Schlafen proteins, exert important roles in modulating cellular proliferation, differentiation, immune responses, viral replication, and chemosensitivity of malignant cells. Studies have demonstrated that abnormal expression of various Schlafens contributes to the pathophysiology of various cancers. Schlafens are now emerging as promising biomarkers and potentially attractive targets for drug development in cancer research. Here, we highlight research suggesting the use of Schlafens as cancer biomarkers and the rationale for the development of specific drugs targeting Schlafen proteins.
Collapse
Affiliation(s)
- Ricardo E. Perez
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (R.E.P.); (F.E.)
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Frank Eckerdt
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (R.E.P.); (F.E.)
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611, USA; (R.E.P.); (F.E.)
- Division of Hematology-Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
3
|
Su X, Jin M, Xu C, Gao Y, Yang Y, Qi H, Zhang Q, Yang X, Ya W, Zhang Y, Yang R. FABP4 in Paneth cells regulates antimicrobial protein expression to reprogram gut microbiota. Gut Microbes 2022; 14:2139978. [PMID: 36519446 PMCID: PMC9635462 DOI: 10.1080/19490976.2022.2139978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Antimicrobial proteins possess a broad spectrum of bactericidal activity and play an important role in shaping the composition of gut microbiota, which is related to multiple diseases such as metabolic syndrome. However, it is incompletely known for the regulation of defensin expression in the gut Paneth cells. Here, we found that FABP4 in the Paneth cells of gut epithelial cells and organoids can downregulate the expression of defensins. FABP4fl/flpvillinCreT mice were highly resistance to Salmonella Typhimurium (S.T) infection and had increased bactericidal ability to pathogens. The FABP4-mediated downregulation of defensins is through degrading PPARγ after K48 ubiquitination. We also demonstrate that high-fat diet (HFD)-mediated downregulation of defensins is through inducing a robust FABP4 in Paneth cells. Firmicutes/Bacteroidetes (F/B) ratio in FABP4fl/flpvillinCreT mice is lower than control mice, which is opposite to that in mice fed HFD, indicating that FABP4 in the Paneth cells could reprogram gut microbiota. Interestingly, FABP4-mediated downregulation of defensins in Paneth cells not only happens in mice but also in human. A better understanding of the regulation of defensins, especially HFD-mediated downregulation of defensin in Paneth cells will provide insights into factor(s) underlying modern diseases.Abbreviations: FABP4: Fatty acid binding protein 4; S. T: Salmonella Typhimurium; HFD: High-fat diet; Defa: α-defensin; 930 HD5: Human α-defensin 5; HD6: Human α-defensin 6; F/B: Firmicutes/Bacteroidetes; SFB: Segmental filamentous bacteria; AMPs: Antimicrobial peptides; PPARγ: Peroxisome proliferator-activated receptor γ; P-PPAR: Phosphorylated PPAR; Dhx15: DEAD-box helicase 15; 935 EGF: Epidermal growth factor; ENR: Noggin and R-spondin 1; CFU: Colony forming unit; Lyz1: Lysozyme 1; Saa1: Serum amyoid A 1; Pla2g2a: Phospholipase A2, group IIA; MMP-7: Matrix metalloproteinase; AU-PAGE: Acid-urea polyacrylamide gel electrophoresis; PA: Palmitic 940 acid; GPR40: G-protein-coupled receptor; GF: Germ-free; EGF: Epidermal growth factor; LP: Lamina propria; KO: Knock out; WT: Wild-type.
Collapse
Affiliation(s)
- Xiaomin Su
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China
| | - Mengli Jin
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China,Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Chen Xu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Yunhuan Gao
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China,Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Yazheng Yang
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China,Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Houbao Qi
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China,Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Qianjing Zhang
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China,Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Xiaorong Yang
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China,Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Wang Ya
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China
| | - Yuan Zhang
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China,Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China
| | - Rongcun Yang
- Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin, China,Translational Medicine Institute, Affiliated Tianjin Union Medical Center of Nankai University, Tianjin, China,State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, China,CONTACT Rongcun Yang Department of Immunology, Nankai University School of Medicine; Nankai University, Tianjin300071, China
| |
Collapse
|
4
|
Harper JA, Morrow EH. Systematic review reveals sexually antagonistic knockouts in model organisms. Ecol Evol 2022; 12:e9671. [PMID: 36619711 PMCID: PMC9798040 DOI: 10.1002/ece3.9671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 12/10/2022] [Indexed: 12/30/2022] Open
Abstract
Sexual antagonism is thought to be an important selective force in multiple evolutionary processes, but very few examples of the genes involved are known. Such a deficit of loci could partially be explained by the lack of overlap in terminology between scientific disciplines. Following a similar review in humans, we searched systematically for studies that described genes with sexually antagonistic or sex-opposite effects in any taxa, using terms designed to capture alternative descriptions of sexual antagonism. Despite drawing on a potentially very large pool of studies we found only eight articles, which between them described seven candidate variants, five of these were gene knockouts. In every case, the variants had net negative effects on the focal trait. One locus was independently validated between studies, but in comparison to previous data on variants in humans and the fruit-fly, the studies generally suffered from small sample sizes, with concomitant high variance. Our review highlights the radically different effects that gene deletions can have on males and females, where the beneficial effects seen in one sex may facilitate the evolution of gene loss. We searched systematically for genetic variants with sexually antagonistic or sex-opposite effects in any taxa. Of 2116 articles, we found seven candidate variants, five of which were gene knockouts. Our review highlights the radically different effects that gene deletions can have on males and females, where the beneficial effects seen in one sex may facilitate the evolution of gene loss.
Collapse
Affiliation(s)
- Jon Alexander Harper
- Evolution, Behaviour and Environment Group, School of Life SciencesJohn Maynard Smith Building, University of SussexBrightonUK
| | - Edward H. Morrow
- Department of Environmental and Life SciencesKarlstad UniversityKarlstadSweden
| |
Collapse
|
5
|
Vomhof-DeKrey EE, Singhal S, Singhal SK, Stover AD, Rajpathy O, Preszler E, Garcia L, Basson MD. RNA Sequencing of Intestinal Enterocytes Pre- and Post-Roux-en-Y Gastric Bypass Reveals Alteration in Gene Expression Related to Enterocyte Differentiation, Restitution, and Obesity with Regulation by Schlafen 12. Cells 2022; 11:3283. [PMID: 36291149 PMCID: PMC9601224 DOI: 10.3390/cells11203283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The intestinal lining renews itself in a programmed fashion that can be affected by adaptation to surgical procedures such as gastric bypass. METHODS To assess adaptive mechanisms in the human intestine after Roux-en-Y gastric bypass (RYGB), we biopsied proximal jejunum at the anastomotic site during surgery to establish a baseline and endoscopically re-biopsied the same area 6-9 months after bypass for comparison. Laser microdissection was performed on pre- and post-RYGB biopsies to isolate enterocytes for RNA sequencing. RESULTS RNA sequencing suggested significant decreases in gene expression associated with G2/M DNA damage checkpoint regulation of the cell cycle pathway, and significant increases in gene expression associated with the CDP-diacylglycerol biosynthesis pathway TCA cycle II pathway, and pyrimidine ribonucleotide salvage pathway after RYGB. Since Schlafen 12 (SLFN12) is reported to influence enterocytic differentiation, we stained mucosa for SLFN12 and observed increased SLFN12 immunoreactivity. We investigated SLFN12 overexpression in HIEC-6 and FHs 74 Int intestinal epithelial cells and observed similar increased expression of the following genes that were also increased after RYGB: HES2, CARD9, SLC19A2, FBXW7, STXBP4, SPARCL1, and UTS. CONCLUSIONS Our data suggest that RYGB promotes SLFN12 protein expression, cellular mechanism and replication pathways, and genes associated with differentiation and restitution (HES2, CARD9, SLC19A2), as well as obesity-related genes (FBXW7, STXBP4, SPARCL1, UTS).
Collapse
Affiliation(s)
- Emilie E. Vomhof-DeKrey
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Sonalika Singhal
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Sandeep K. Singhal
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Allie D. Stover
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Odele Rajpathy
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Elizabeth Preszler
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Luis Garcia
- Sanford Health Clinic, Sioux Falls, ND 57117, USA
| | - Marc D. Basson
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| |
Collapse
|
6
|
Boulund U, Bastos DM, Ferwerda B, van den Born BJ, Pinto-Sietsma SJ, Galenkamp H, Levin E, Groen AK, Zwinderman AH, Nieuwdorp M. Gut microbiome associations with host genotype vary across ethnicities and potentially influence cardiometabolic traits. Cell Host Microbe 2022; 30:1464-1480.e6. [PMID: 36099924 DOI: 10.1016/j.chom.2022.08.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/16/2022] [Accepted: 08/17/2022] [Indexed: 12/13/2022]
Abstract
Previous studies in mainly European populations have reported that the gut microbiome composition is associated with the human genome. However, the genotype-microbiome interaction in different ethnicities is largely unknown. We performed a large fecal microbiome genome-wide association study of a single multiethnic cohort, the Healthy Life in an Urban Setting (HELIUS) cohort (N = 4,117). Mendelian randomization was performed using the multiethnic Pan-UK Biobank (N = 460,000) to dissect potential causality. We identified ethnicity-specific associations between host genomes and gut microbiota. Certain microbes were associated with genotype in multiple ethnicities. Several of the microbe-associated loci were found to be related to immune functions, interact with glutamate and the mucus layer, or be expressed in the gut or brain. Additionally, we found that gut microbes potentially influence cardiometabolic health factors such as BMI, cholesterol, and blood pressure. This provides insight into the relationship of ethnicity and gut microbiota and into the possible causal effects of gut microbes on cardiometabolic traits.
Collapse
Affiliation(s)
- Ulrika Boulund
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands
| | - Diogo M Bastos
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands
| | - Bart Ferwerda
- Department of Clinical Epidemiology and Biostatistics, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands
| | - Bert-Jan van den Born
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands; Department of Public and Occupational Health, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands
| | - Sara-Joan Pinto-Sietsma
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands; Department of Clinical Epidemiology and Biostatistics, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands
| | - Henrike Galenkamp
- Department of Public and Occupational Health, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands
| | - Evgeni Levin
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands; HorAIzon BV, 2645 LT Delfgauw, the Netherlands
| | - Albert K Groen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology and Biostatistics, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Centers, location AMC, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
7
|
Yumi Noronha N, da Silva Rodrigues G, Harumi Yonehara Noma I, Fernanda Cunha Brandao C, Pereira Rodrigues K, Colello Bruno A, Sae-Lee C, Moriguchi Watanabe L, Augusta de Souza Pinhel M, Mello Schineider I, Luciano de Almeida M, Barbosa Júnior F, Araújo Morais D, Tavares de Sousa Júnior W, Plösch T, Roberto Bueno Junior C, Barbosa Nonino C. 14-weeks combined exercise epigenetically modulated 118 genes of menopausal women with prediabetes. Front Endocrinol (Lausanne) 2022; 13:895489. [PMID: 36046788 PMCID: PMC9423096 DOI: 10.3389/fendo.2022.895489] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 07/19/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Pre-diabetes precedes Diabetes Mellitus (DM) disease and is a critical period for hyperglycemia treatment, especially for menopausal women, considering all metabolic alterations due to hormonal changes. Recently, the literature has demonstrated the role of physical exercise in epigenetic reprogramming to modulate the gene expression patterns of metabolic conditions, such as hyperglycemia, and prevent DM development. In the present study, we hypothesized that physical exercise training could modify the epigenetic patterns of women with poor glycemic control. METHODS 48 post-menopause women aged 60.3 ± 4.5 years were divided according to their fasting blood glucose levels into two groups: Prediabetes Group, PG (n=24), and Normal Glucose Group, NGG (n=24). All participants performed 14 weeks of physical exercise three times a week. The Infinium Methylation EPIC BeadChip measured the participants' Different Methylated Regions (DMRs). RESULTS Before the intervention, the PG group had 12 DMRs compared to NGG. After the intervention, five DMRs remained different. Interestingly, when comparing the PG group before and after training, 118 DMRs were found. The enrichment analysis revealed that the genes were related to different biological functions such as energy metabolism, cell differentiation, and tumor suppression. CONCLUSION Physical exercise is a relevant alternative in treating hyperglycemia and preventing DM in post-menopause women with poor glycemic control.
Collapse
Affiliation(s)
- Natália Yumi Noronha
- Department of Internal Medicine, Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Guilherme da Silva Rodrigues
- Department of Internal Medicine, Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- *Correspondence: Guilherme da Silva Rodrigues,
| | - Isabella Harumi Yonehara Noma
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Camila Fernanda Cunha Brandao
- Department of Internal Medicine, Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Physical Education School, Minas Gerais State University, Divinópolis, Minas Gerais, Brazil
| | - Karine Pereira Rodrigues
- Department of Internal Medicine, Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Alexandre Colello Bruno
- Department of Radiotherapy, Ribeirão Preto Medical School Hospital and Clinics, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Chanachai Sae-Lee
- Research Division, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Marcela Augusta de Souza Pinhel
- Department of Internal Medicine, Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Department of Molecular Biology, São José do Rio Preto Medical School, São José do Rio Preto, SP, Brazil
| | | | | | - Fernando Barbosa Júnior
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Déborah Araújo Morais
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Wellington Tavares de Sousa Júnior
- Department of Clinical Analysis, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Torsten Plösch
- University Medical Center Groningen, Groningen, Netherlands
| | - Carlos Roberto Bueno Junior
- Department of Internal Medicine, Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Ribeirão Preto School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Carla Barbosa Nonino
- Department of Internal Medicine, Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Department of Health Sciences, Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
8
|
Dey P, Chaudhuri SR, Efferth T, Pal S. The intestinal 3M (microbiota, metabolism, metabolome) zeitgeist - from fundamentals to future challenges. Free Radic Biol Med 2021; 176:265-285. [PMID: 34610364 DOI: 10.1016/j.freeradbiomed.2021.09.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
The role of the intestine in human health and disease has historically been neglected and was mostly attributed to digestive and absorptive functions. In the past two decades, however, discoveries related to human nutrition and intestinal host-microbe reciprocal interaction have established the essential role of intestinal health in the pathogenesis of chronic diseases and the overall wellbeing. That transfer of gut microbiota could be a means of disease phenotype transfer has revolutionized our understanding of chronic disease pathogenesis. This narrative review highlights the major concepts related to intestinal microbiota, metabolism, and metabolome (3M) that have facilitated our fundamental understanding of the association between the intestine, and human health and disease. In line with increased interest of microbiota-dependent modulation of human health by dietary phytochemicals, we have also discussed the emerging concepts beyond the phytochemical bioactivities which emphasizes the integral role of microbial metabolites of parent phytochemicals at extraintestinal tissues. Finally, this review concludes with challenges and future prospects in defining the 3M interactions and has emphasized the fact that, it takes 'guts' to stay healthy.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India.
| | - Saumya Ray Chaudhuri
- Council of Scientific and Industrial Research (CSIR), Institute of Microbial Technology, Chandigarh, India
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Sirshendu Pal
- Mukherjee Hospital, Mitra's Clinic and Nursing Home, Siliguri, West Bengal, India
| |
Collapse
|
9
|
Vomhof-DeKrey EE, Stover AD, Labuhn M, Osman MR, Basson MD. Vil-Cre specific Schlafen 3 knockout mice exhibit sex-specific differences in intestinal differentiation markers and Schlafen family members expression levels. PLoS One 2021; 16:e0259195. [PMID: 34710177 PMCID: PMC8553116 DOI: 10.1371/journal.pone.0259195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/14/2021] [Indexed: 11/25/2022] Open
Abstract
The intestinal epithelium requires self-renewal and differentiation in order to function and adapt to pathological diseases such as inflammatory bowel disease, short gut syndrome, and ulcers. The rodent Slfn3 protein and the human Slfn12 analog are known to regulate intestinal epithelial differentiation. Previous work utilizing a pan-Slfn3 knockout (KO) mouse model revealed sex-dependent gene expression disturbances in intestinal differentiation markers, metabolic pathways, Slfn family member mRNA expression, adaptive immune cell proliferation/functioning genes, and phenotypically less weight gain and sex-dependent changes in villus length and crypt depth. We have now created a Vil-Cre specific Slfn3KO (VC-Slfn3KO) mouse to further evaluate its role in intestinal differentiation. There were increases in Slfn1, Slfn2, Slfn4, and Slfn8 and decreases in Slfn5 and Slfn9 mRNA expression that were intestinal region and sex-specific. Differentiation markers, sucrase isomaltase (SI), villin 1, and dipeptidyl peptidase 4 and glucose transporters, glucose transporter 1 (Glut1), Glut2, and sodium glucose transporter 1 (SGLT1), were increased in expression in VC-Slfn3KO mice based on intestinal region and were also highly female sex-biased, except for SI in the ileum was also increased for male VC-Slfn3KO mice and SGLT1 was decreased for both sexes. Overall, the variations that we observed in these VC-Slfn3KO mice indicate a complex regulation of intestinal gene expression that is sex-dependent.
Collapse
Affiliation(s)
- Emilie E. Vomhof-DeKrey
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Allie D. Stover
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Mary Labuhn
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Marcus R. Osman
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Marc D. Basson
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| |
Collapse
|
10
|
Thomas PA, Schafler ED, Ruff SE, Voisin M, Ha S, Logan SK. UXT in Sertoli cells is required for blood-testis barrier integrity†. Biol Reprod 2021; 103:880-891. [PMID: 32678429 DOI: 10.1093/biolre/ioaa121] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/12/2020] [Accepted: 07/16/2020] [Indexed: 01/01/2023] Open
Abstract
Spermatogenesis is a complex process that establishes male fertility and involves proper communication between the germline (spermatozoa) and the somatic tissue (Sertoli cells). Many factors that are important for spermatozoa production are also required for Sertoli cell function. Recently, we showed that the transcriptional cofactor ubiquitously expressed transcript (UXT) encodes a protein that is essential in germ cells for spermatogenesis and fertility. However, the role of UXT within Sertoli cells and how it affects Sertoli cell function was still unclear. Here we describe a novel role for UXT in the Sertoli cell's ability to support spermatogenesis. We find that the conditional deletion of Uxt in Sertoli cells results in smaller testis size and weight, which coincided with a loss of germ cells in a subset of seminiferous tubules. In addition, the deletion of Uxt has no impact on Sertoli cell abundance or maturity, as they express markers of mature Sertoli cells. Gene expression analysis reveals that the deletion of Uxt in Sertoli cells reduces the transcription of genes involved in the tight junctions of the blood-testis barrier (BTB). Furthermore, tracer experiments and electron microscopy reveal that the BTB is permeable in UXT KO animals. These findings broaden our understanding of UXT's role in Sertoli cells and its contribution to the structural integrity of the BTB.
Collapse
Affiliation(s)
- Phillip A Thomas
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Eric D Schafler
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Sophie E Ruff
- Department of Urology, New York University School of Medicine, New York, NY, USA.,Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Maud Voisin
- Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Susan Ha
- Department of Urology, New York University School of Medicine, New York, NY, USA
| | - Susan K Logan
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA.,Department of Urology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
11
|
Al-Marsoummi S, Vomhof-DeKrey EE, Basson MD. Schlafens: Emerging Proteins in Cancer Cell Biology. Cells 2021; 10:2238. [PMID: 34571887 PMCID: PMC8465726 DOI: 10.3390/cells10092238] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/29/2022] Open
Abstract
Schlafens (SLFN) are a family of genes widely expressed in mammals, including humans and rodents. These intriguing proteins play different roles in regulating cell proliferation, cell differentiation, immune cell growth and maturation, and inhibiting viral replication. The emerging evidence is implicating Schlafens in cancer biology and chemosensitivity. Although Schlafens share common domains and a high degree of homology, different Schlafens act differently. In particular, they show specific and occasionally opposing effects in some cancer types. This review will briefly summarize the history, structure, and non-malignant biological functions of Schlafens. The roles of human and mouse Schlafens in different cancer types will then be outlined. Finally, we will discuss the implication of Schlafens in the anti-tumor effect of interferons and the use of Schlafens as predictors of chemosensitivity.
Collapse
Affiliation(s)
- Sarmad Al-Marsoummi
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (E.E.V.-D.)
| | - Emilie E. Vomhof-DeKrey
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (E.E.V.-D.)
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Marc D. Basson
- Department of Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (E.E.V.-D.)
- Department of Surgery, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
- Department of Pathology, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| |
Collapse
|
12
|
Vomhof-DeKrey EE, Lansing JT, Darland DC, Umthun J, Stover AD, Brown C, Basson MD. Loss of Slfn3 induces a sex-dependent repair vulnerability after 50% bowel resection. Am J Physiol Gastrointest Liver Physiol 2021; 320:G136-G152. [PMID: 33237796 PMCID: PMC7864235 DOI: 10.1152/ajpgi.00344.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/13/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023]
Abstract
Bowel resection accelerates enterocyte proliferation in the remaining gut with suboptimal absorptive and digestive capacity because of a proliferation-associated decrease in functional differentiation markers. We hypothesized that although schlafen 3 (Slfn3) is an important regulator of enterocytic differentiation, Slfn3 would have less impact on bowel resection adaptation, where accelerated proliferation takes priority over differentiation. We assessed proliferation, cell shedding, and enterocyte differentiation markers from resected and postoperative bowel of wild-type (WT) and Slfn3-knockout (Slfn3KO) mice. Villus length and crypt depth were increased in WT mice and were even longer in Slfn3KO mice. Mitotic marker, Phh3+, and the proliferation markers Lgr5, FoxL1, and platelet-derived growth factor-α (PDGFRα) were increased after resection in male WT, but this was blunted in male Slfn3KO mice. Cell-shedding regulators Villin1 and TNFα were downregulated in female mice and male WT mice only, whereas Gelsolin and EGFR increased expression in all mice. Slfn3 expression increased after resection in WT mice, whereas other Slfn family members 1, 2, 5, 8, and 9 had varied expressions that were affected also by sex difference and loss of Slfn3. Differentiation markers sucrase isomaltase, Dpp4, Glut2, and SGLT1 were all decreased, suggesting that enterocytic differentiation effort is incompatible with rapid proliferation shift in intestinal adaptation. Slfn3 absence potentiates villus length and crypt depth, suggesting that the differentiating stimulus of Slfn3 signaling may restrain mucosal mass increase through regulating Villin1, Gelsolin, EGFR, TNFα, and proliferation markers. Therefore, Slfn3 may be an important regulator not only of "normal" enterocytic differentiation but also in response to bowel resection.NEW & NOTEWORTHY The differentiating stimulus of Slfn3 signaling restrains an increase in mucosal mass after bowel resection, and there is a Slfn3-sex interaction regulating differentiation gene expression and intestinal adaptation. This current study highlights the combinatory effects of gender and Slfn3 genotype on the gene expression changes that contribute to the adaptation in intestinal cellular milleu (i.e. villus and crypt structure) which are utilized to compensate for the stress-healing response that the animals display in intestinal adaptation.
Collapse
Affiliation(s)
- Emilie E Vomhof-DeKrey
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Jack T Lansing
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
- Department of Biology, University of North Dakota, Grand Forks, North Dakota
| | - Diane C Darland
- Department of Biology, University of North Dakota, Grand Forks, North Dakota
| | - Josey Umthun
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
- Department of Biology, University of North Dakota, Grand Forks, North Dakota
| | - Allie D Stover
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Christopher Brown
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
| | - Marc D Basson
- Departments of Surgery, Pathology, and Biomedical Sciences, School of Medicine and the Health Sciences, University of North Dakota, Grand Forks, North Dakota
| |
Collapse
|
13
|
Krieger NS, Chen L, Becker J, Chan MR, Bushinsky DA. Deletion of the proton receptor OGR1 in mouse osteoclasts impairs metabolic acidosis-induced bone resorption. Kidney Int 2020; 99:609-619. [PMID: 33159961 DOI: 10.1016/j.kint.2020.10.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/30/2020] [Accepted: 10/09/2020] [Indexed: 01/03/2023]
Abstract
Metabolic acidosis induces osteoclastic bone resorption and inhibits osteoblastic bone formation. Previously we found that mice with a global deletion of the proton receptor OGR1 had increased bone density although both osteoblast and osteoclast activity were increased. To test whether direct effects on osteoclast OGR1 are critical for metabolic acidosis stimulated bone resorption, we generated knockout mice with an osteoclast-specific deletion of OGR1 (knockout mice). We studied bones from three-month old female mice and the differentiated osteoclasts derived from bone marrow of femurs from these knockout and wild type mice. MicroCT demonstrated increased density in tibiae and femurs but not in vertebrae of the knockout mice. Tartrate resistant acid phosphatase staining of tibia indicated a decrease in osteoclast number and surface area/bone surface from knockout compared to wild type mice. Osteoclasts derived from the marrow of knockout mice demonstrated decreased pit formation, osteoclast staining and osteoclast-specific gene expression compared to those from wild type mice. In response to metabolic acidosis, osteoclasts from knockout mice had decreased nuclear translocation of NFATc1, a transcriptional regulator of differentiation, and no increase in size or number compared to osteoclasts from wild type mice. Thus, loss of osteoclast OGR1 decreased both basal and metabolic acidosis-induced osteoclast activity indicating osteoclast OGR1 is important in mediating metabolic acidosis-induced bone resorption. Understanding the role of OGR1 in metabolic acidosis-induced bone resorption will provide insight into bone loss in acidotic patients with chronic kidney disease.
Collapse
Affiliation(s)
- Nancy S Krieger
- Division of Nephrology, Department of Medicine, University of Rochester School of Medicine, Rochester, New York, USA.
| | - Luojing Chen
- Division of Nephrology, Department of Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - Jennifer Becker
- Division of Nephrology, Department of Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - Michaela R Chan
- Division of Nephrology, Department of Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| | - David A Bushinsky
- Division of Nephrology, Department of Medicine, University of Rochester School of Medicine, Rochester, New York, USA
| |
Collapse
|
14
|
Sex differences in behavioral and metabolic effects of gene inactivation: The neuropeptide Y and Y receptors in the brain. Neurosci Biobehav Rev 2020; 119:333-347. [PMID: 33045245 DOI: 10.1016/j.neubiorev.2020.09.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
Brain and gonadal hormones interplay controls metabolic and behavioral functions in a sex-related manner. However, most translational neuroscience research related to animal models of endocrine and psychiatric disorders are often carried out in male animals only. The Neuropeptide Y (NPY) system shows sex-dependent differences and is sensitive to gonadal steroids. Based on published data from our and other laboratories, in this review we will discuss the sex related differences of NPY action on energy balance, bone homeostasis and behavior in rodents with the genetic manipulation of genes encoding NPY and its Y1, Y2 and Y5 cognate receptors. Comparative analyses of the phenotype of transgenic and knockout NPY and Y receptor rodents unravels sex dependent differences in the functions of this neurotransmission system, potentially helping to develop therapeutics for a variety of sex-related disorders including metabolic syndrome, osteoporosis and ethanol addiction.
Collapse
|
15
|
Bertocchi I, Oberto A, Longo A, Palanza P, Eva C. Conditional inactivation of Npy1r gene in mice induces sex-related differences of metabolic and behavioral functions. Horm Behav 2020; 125:104824. [PMID: 32755609 DOI: 10.1016/j.yhbeh.2020.104824] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/15/2020] [Accepted: 07/23/2020] [Indexed: 02/07/2023]
Abstract
Sex hormone-driven differences in gene expression have been identified in experimental animals, highlighting brain neuronal populations implicated in dimorphism of metabolic and behavioral functions. Neuropeptide Y-Y1 receptor (NPY-Y1R) system is sexually dimorphic and sensitive to gonadal steroids. In the present study we compared the phenotype of male and female conditional knockout mice (Npy1rrfb mice), carrying the inactivation of Npy1r gene in excitatory neurons of the brain limbic system. Compared to their male control (Npy1r2lox) littermates, male Npy1rrfb mice exhibited hyperactivation of the hypothalamic-pituitary-adrenal (HPA) axis that is associated with anxiety and executive dysfunction, reduced body weight growth, after-fasting refeeding, white adipose tissue (WAT) mass and plasma leptin levels. Conversely, female Npy1rrfb mice displayed an anxious-like behavior but no differences in HPA axis activity, executive function and body weight, compared to control females. Moreover, conditional inactivation of Npy1r gene induced an increase of subcutaneous and gonadal WAT weight and plasma leptin levels and a compensatory decrease of Agouti-related protein immunoreactivity in the hypothalamic arcuate (ARC) nucleus in females, compared to their respective control littermates. Interestingly, Npy1r mRNA expression was reduced in the ARC and in the paraventricular hypothalamic nuclei of female, but not male mice. These results demonstrated that female mice are resilient to hormonal and metabolic effects of limbic Npy1r gene inactivation, suggesting the existence of an estrogen-dependent relay necessary to ensure the maintenance of the homeostasis, that can be mediated by hypothalamic Y1R.
Collapse
Affiliation(s)
- Ilaria Bertocchi
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043 Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126 Turin, Italy; Neuroscience Institute of Turin, Italy
| | - Alessandra Oberto
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043 Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126 Turin, Italy; Neuroscience Institute of Turin, Italy
| | - Angela Longo
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043 Orbassano, Turin, Italy
| | - Paola Palanza
- Department of Medicine and Surgery, University of Parma, 43100 Parma, Italy
| | - Carola Eva
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043 Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126 Turin, Italy; Neuroscience Institute of Turin, Italy.
| |
Collapse
|
16
|
Vomhof-DeKrey EE, Umthun J, Basson MD. Loss of Schlafen3 influences the expression levels of Schlafen family members in ileum, thymus, and spleen tissue. PeerJ 2020; 8:e8461. [PMID: 32025381 PMCID: PMC6993753 DOI: 10.7717/peerj.8461] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/26/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The Schlafen (Slfn) family proteins are important for regulation of cell growth, cell differentiation and cell cycle progression. We sought to distinguish Slfn family expression in Slfn3 knockout (KO) mice after RNA sequencing analysis of Slfn3KO vs. wildtype (WT) mice revealed varying expressions of Slfn family in ileal mucosa. METHODS Quantitative PCR analysis of Slfn members was evaluated in ileal mucosa, thymus and spleen tissue since Slfn family members have roles in differentiating intestinal and immune cells. RESULTS Ileal mucosa of Slfn3KO mice displayed a decrease in Slfn3, 4, 8 and 9 while Slfn1 and 5 increased in mRNA expression vs. WT mice. Thymic tissue had a Slfn9 increase and a Slfn4 decrease while splenic tissue had a Slfn8 and Slfn9 increase in Slfn3KO mice vs. WT mice. These differential expressions of Slfn members could indicate a feedback regulatory mechanism within the Slfn family. Indeed, MATCH™ tool from geneXplain predicted that all Slfn members have regions in their promoters for the Kruppel-like factor-6 transcription factor. In addition, NFAT related factors, ING4, ZNF333 and KLF4 are also predicted to bind in up to 6 of the 8 Slfn promoters. This study further describes a possible autoregulatory mechanism amongst the Slfn family members which could be important in how they regulate the differentiation of various cell types.
Collapse
Affiliation(s)
- Emilie E. Vomhof-DeKrey
- Departments of Surgery, Pathology, and Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Josey Umthun
- Departments of Surgery, Pathology, and Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Marc D. Basson
- Departments of Surgery, Pathology, and Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| |
Collapse
|