1
|
Liu CL, Ren T, Ruan PC, Huang YF, Ceccobelli S, Huang DJ, Zhang LP, E GX. Genome-Wide Association Integrating a Transcriptomic Meta-Analysis Suggests That Genes Related to Fat Deposition and Muscle Development Are Closely Associated with Growth in Huaxi Cattle. Vet Sci 2025; 12:109. [PMID: 40005876 PMCID: PMC11860805 DOI: 10.3390/vetsci12020109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Growth traits are among the most important economic phenotypes targeted in the genetic improvement of beef cattle. To understand the genetic basis of growth traits in Huaxi cattle, we performed a genome-wide association study (GWAS) on body weight, eye muscle area, and back fat thickness across five developmental stages in a population of 202 Huaxi cattle. Additionally, publicly available RNA-seq data from the longissimus dorsi muscle of both young and adult cattle were analyzed to identify key genes and genetic markers associated with growth in Huaxi cattle. In total, 7.19 million high-quality variant loci (SNPs and INDELs) were identified across all samples. In the GWAS, the three multilocus models (FarmCPU, MLMM, and BLINK) outperformed the conventional single-locus models (CMLM, GLM, and MLM). Consequently, GWAS analysis was conducted using multilocus models, which identified 99 variant loci significantly associated with growth traits and annotated a total of 83 candidate genes (CDGs). Additionally, 23 of the 83 CDGs overlapped with significantly differentially expressed genes identified from public RNA-seq datasets of longissimus dorsi muscle between young and adult cattle. Furthermore, gene functional enrichment (KEGG and GO) analyses revealed that over 30% of the pathways and GO terms were associated with muscle development and fat deposition, crucial factors for beef production. Specifically, key genes identified included MGLL, SGMS1, SNX29 and AKAP6, which are implicated in lipid metabolism, adipogenesis, and muscle growth. In summary, this study provides new insights into the genetic mechanisms underlying growth traits in Huaxi cattle and presents promising markers for future breeding improvements.
Collapse
Affiliation(s)
- Cheng-Li Liu
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (C.-L.L.); (T.R.); (P.-C.R.); (Y.-F.H.)
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Tao Ren
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (C.-L.L.); (T.R.); (P.-C.R.); (Y.-F.H.)
| | - Peng-Cheng Ruan
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (C.-L.L.); (T.R.); (P.-C.R.); (Y.-F.H.)
| | - Yong-Fu Huang
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (C.-L.L.); (T.R.); (P.-C.R.); (Y.-F.H.)
| | - Simone Ceccobelli
- Department of Agricultural, Food and Environmental Sciences, Università Politecnica delle Marche, 60131 Ancona, Italy;
| | - De-Jun Huang
- Chongqing Academy of Animal Science, Chongqing 402460, China;
| | - Lu-Pei Zhang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100006, China
| | - Guang-Xin E
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; (C.-L.L.); (T.R.); (P.-C.R.); (Y.-F.H.)
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| |
Collapse
|
2
|
Yu LE, Yang WC, Liang YC. Crosstalk Within the Intestinal Epithelium: Aspects of Intestinal Absorption, Homeostasis, and Immunity. Biomedicines 2024; 12:2771. [PMID: 39767678 PMCID: PMC11673925 DOI: 10.3390/biomedicines12122771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Gut health is crucial in many ways, such as in improving human health in general and enhancing production in agricultural animals. To maximize the effect of a healthy gastrointestinal tract (GIT), an understanding of the regulation of intestinal functions is needed. Proper intestinal functions depend on the activity, composition, and behavior of intestinal epithelial cells (IECs). There are various types of IECs, including enterocytes, Paneth cells, enteroendocrine cells (EECs), goblet cells, tuft cells, M cells, and intestinal epithelial stem cells (IESCs), each with unique 3D structures and IEC distributions. Although the communication between IECs and other cell types, such as immune cells and neurons, has been intensively reviewed, communication between different IECs has rarely been addressed. The present paper overviews the networks among IECs that influence intestinal functions. Intestinal absorption is regulated by incretins derived from EECs that induce nutrient transporter activity in enterocytes. EECs, Paneth cells, tuft cells, and enterocytes release signals to activate Notch signaling, which modulates IESC activity and intestinal homeostasis, including proliferation and differentiation. Intestinal immunity can be altered via EECs, goblet cells, tuft cells, and cytokines derived from IECs. Finally, tools for investigating IEC communication have been discussed, including the novel 3D intestinal cell model utilizing enteroids that can be considered a powerful tool for IEC communication research. Overall, the importance of IEC communication, especially EECs and Paneth cells, which cover most intestinal functional regulating pathways, are overviewed in this paper. Such a compilation will be helpful in developing strategies for maintaining gut health.
Collapse
Affiliation(s)
| | | | - Yu-Chaun Liang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115201, Taiwan; (L.-E.Y.); (W.-C.Y.)
| |
Collapse
|
3
|
Angelis N, Baulies A, Hubl F, Kucharska A, Kelly G, Llorian M, Boeing S, Li VSW. Loss of ARID3A perturbs intestinal epithelial proliferation-differentiation ratio and regeneration. J Exp Med 2024; 221:e20232279. [PMID: 39150450 PMCID: PMC11329776 DOI: 10.1084/jem.20232279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/08/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024] Open
Abstract
Intestinal stem cells at the crypt divide and give rise to progenitor cells that proliferate and differentiate into various mature cell types in the transit-amplifying (TA) zone. Here, we showed that the transcription factor ARID3A regulates intestinal epithelial cell proliferation and differentiation at the TA progenitors. ARID3A forms an expression gradient from the villus tip to the upper crypt mediated by TGF-β and WNT. Intestinal-specific deletion of Arid3a reduces crypt proliferation, predominantly in TA cells. Bulk and single-cell transcriptomic analysis shows increased enterocyte and reduced secretory differentiation in the Arid3a cKO intestine, accompanied by enriched upper-villus gene signatures of both cell lineages. We find that the enhanced epithelial differentiation in the Arid3a-deficient intestine is caused by increased binding and transcription of HNF1 and HNF4. Finally, we show that loss of Arid3a impairs irradiation-induced regeneration with sustained cell death and reprogramming. Our findings imply that Arid3a functions to fine-tune the proliferation-differentiation dynamics at the TA progenitors, which are essential for injury-induced regeneration.
Collapse
Affiliation(s)
- Nikolaos Angelis
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute , London, UK
| | - Anna Baulies
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute , London, UK
| | - Florian Hubl
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute , London, UK
| | - Anna Kucharska
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute , London, UK
| | - Gavin Kelly
- Bioinformatics and Biostatistics Science Technology Platform, The Francis Crick Institute , London, UK
| | - Miriam Llorian
- Bioinformatics and Biostatistics Science Technology Platform, The Francis Crick Institute , London, UK
| | - Stefan Boeing
- Bioinformatics and Biostatistics Science Technology Platform, The Francis Crick Institute , London, UK
| | - Vivian S W Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute , London, UK
| |
Collapse
|
4
|
I Kutyavin V, Korn LL, Medzhitov R. Nutrient-derived signals regulate eosinophil adaptation to the small intestine. Proc Natl Acad Sci U S A 2024; 121:e2316446121. [PMID: 38271336 PMCID: PMC10835075 DOI: 10.1073/pnas.2316446121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/05/2023] [Indexed: 01/27/2024] Open
Abstract
Eosinophils are well recognized as effector cells of type 2 immunity, yet they also accumulate in many tissues under homeostatic conditions. However, the processes that govern homeostatic eosinophil accumulation and tissue-specific adaptation, and their functional significance, remain poorly defined. Here, we investigated how eosinophils adapt to the small intestine (SI) microenvironment and the local signals that regulate this process. We observed that eosinophils gradually migrate along the crypt-villus axis, giving rise to a villus-resident subpopulation with a distinct transcriptional signature. Retinoic acid signaling was specifically required for maintenance of this subpopulation, while IL-5 was largely dispensable outside of its canonical role in eosinophil production. Surprisingly, we found that a high-protein diet suppressed the accumulation of villus-resident eosinophils. Purified amino acids were sufficient for this effect, which was a consequence of accelerated eosinophil turnover within the tissue microenvironment and was not due to altered development in the bone marrow. Our study provides insight into the process of eosinophil adaptation to the SI, highlighting its reliance on nutrient-derived signals.
Collapse
Affiliation(s)
- Vassily I Kutyavin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Lisa L Korn
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
- Department of Medicine (Rheumatology), Yale University School of Medicine, New Haven, CT 06510
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
- HHMI, Yale University School of Medicine, New Haven, CT 06510
| |
Collapse
|
5
|
Burclaff J. Transcriptional regulation of metabolism in the intestinal epithelium. Am J Physiol Gastrointest Liver Physiol 2023; 325:G501-G507. [PMID: 37786942 PMCID: PMC10894668 DOI: 10.1152/ajpgi.00147.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Epithelial metabolism in the intestine is increasingly known to be important for stem cell maintenance and activity while also affecting weight gain and diseases. This review compiles studies from recent years which describe major transcription factors controlling metabolic activity across the intestinal epithelium as well as transcriptional and epigenetic networks controlling the factors themselves. Recent studies show that transcriptional regulators serve as the link between signals from the microbiota and diet and epithelial metabolism. Studies have advanced this paradigm to identify druggable targets to block weight gain or disease progression in mice. As such, there is great potential that a better understanding of these regulatory networks will improve our knowledge of intestinal physiology and promote discoveries to benefit human health.
Collapse
Affiliation(s)
- Joseph Burclaff
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, North Carolina, United States
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States
| |
Collapse
|
6
|
Fujino M, Ojima M, Takahashi S. Exploring Large MAF Transcription Factors: Functions, Pathology, and Mouse Models with Point Mutations. Genes (Basel) 2023; 14:1883. [PMID: 37895232 PMCID: PMC10606904 DOI: 10.3390/genes14101883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/23/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Large musculoaponeurotic fibrosarcoma (MAF) transcription factors contain acidic, basic, and leucine zipper regions. Four types of MAF have been elucidated in mice and humans, namely c-MAF, MAFA, MAFB, and NRL. This review aimed to elaborate on the functions of MAF transcription factors that have been studied in vivo so far, as well as describe the pathology of human patients and corresponding mouse models with c-MAF, MAFA, and MAFB point mutations. To identify the functions of MAF transcription factors in vivo, we generated genetically modified mice lacking c-MAF, MAFA, and MAFB and analyzed their phenotypes. Further, in recent years, c-MAF, MAFA, and MAFB have been identified as causative genes underpinning many rare diseases. Careful observation of human patients and animal models is important to examine the pathophysiological mechanisms underlying these conditions for targeted therapies. Murine models exhibit phenotypes similar to those of human patients with c-MAF, MAFA, and MAFB mutations. Therefore, generating these animal models emphasizes their usefulness for research uncovering the pathophysiology of point mutations in MAF transcription factors and the development of etiology-based therapies.
Collapse
Affiliation(s)
- Mitsunori Fujino
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan; (M.F.); (M.O.)
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| | - Masami Ojima
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan; (M.F.); (M.O.)
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan; (M.F.); (M.O.)
- Laboratory Animal Resource Center, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
- Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Ibaraki, Japan
| |
Collapse
|
7
|
Berková L, Fazilaty H, Yang Q, Kubovčiak J, Stastna M, Hrckulak D, Vojtechova M, Dalessi T, Brügger MD, Hausmann G, Liberali P, Korinek V, Basler K, Valenta T. Terminal differentiation of villus tip enterocytes is governed by distinct Tgfβ superfamily members. EMBO Rep 2023; 24:e56454. [PMID: 37493498 PMCID: PMC10481656 DOI: 10.15252/embr.202256454] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023] Open
Abstract
The protective and absorptive functions of the intestinal epithelium rely on differentiated enterocytes in the villi. The differentiation of enterocytes is orchestrated by sub-epithelial mesenchymal cells producing distinct ligands along the villus axis, in particular Bmps and Tgfβ. Here, we show that individual Bmp ligands and Tgfβ drive distinct enterocytic programs specific to villus zonation. Bmp4 is expressed from the centre to the upper part of the villus and activates preferentially genes connected to lipid uptake and metabolism. In contrast, Bmp2 is produced by villus tip mesenchymal cells and it influences the adhesive properties of villus tip epithelial cells and the expression of immunomodulators. Additionally, Tgfβ induces epithelial gene expression programs similar to those triggered by Bmp2. Bmp2-driven villus tip program is activated by a canonical Bmp receptor type I/Smad-dependent mechanism. Finally, we establish an organoid cultivation system that enriches villus tip enterocytes and thereby better mimics the cellular composition of the intestinal epithelium. Our data suggest that not only a Bmp gradient but also the activity of individual Bmp drives specific enterocytic programs.
Collapse
Affiliation(s)
- Linda Berková
- Laboratory of Cell and Developmental BiologyInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Hassan Fazilaty
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | - Qiutan Yang
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of ZoologyChinese Academy of SciencesBeijingChina
- Institute for Stem Cell and Regeneration, Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and Regenerative MedicineBeijingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jan Kubovčiak
- Laboratory of Genomics and BioinformaticsInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Monika Stastna
- Laboratory of Cell and Developmental BiologyInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Dusan Hrckulak
- Laboratory of Cell and Developmental BiologyInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Martina Vojtechova
- Laboratory of Cell and Developmental BiologyInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Tosca Dalessi
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | | | - George Hausmann
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI)BaselSwitzerland
| | - Vladimir Korinek
- Laboratory of Cell and Developmental BiologyInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Konrad Basler
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| | - Tomas Valenta
- Laboratory of Cell and Developmental BiologyInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
- Department of Molecular Life SciencesUniversity of ZurichZurichSwitzerland
| |
Collapse
|
8
|
Fujino M, Ojima M, Ishibashi S, Mizuno S, Takahashi S. Generation and mutational analysis of a transgenic murine model of the human MAF mutation. Am J Med Genet A 2023. [PMID: 37186330 DOI: 10.1002/ajmg.a.63220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 03/29/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023]
Abstract
Aymé-Gripp syndrome is an autosomal dominant multisystem disorder. The major clinical features of this syndrome include congenital cataracts, sensorineural hearing loss, intellectual disability, and a distinctive flat facial appearance. MAF has been identified as a causative gene of the syndrome, and heterozygous variants owing to impairment in glycogen synthase kinase 3 (GSK3)-mediated MAF phosphorylation shows related disorders. However, the underlying mechanisms of these types of disorders in affected individuals remain poorly understood. To explore the underlying mechanisms and discover new phenotypes, a murine model with a Maf mutation on a GSK3 phosphorylation motif, p.Thr58Ile, was generated using CRISPR-Cas9 gene editing. This is a homologous mutation to that in human patients. Our murine model exhibited similar phenotypes to those in humans, such as lens abnormalities, short stature, growth retardation, and abnormal skull morphology. The murine model showed decreased brain volume and malocclusion. Considering the sequencing and genotyping data, our models were successfully generated for the first time (to the best of our knowledge). Therefore, this study offers new and unique functional insights into human and murine MAF and novel clinical values of MAF pathogenic variants associated with changes in the functions of several organs based on a viable murine model.
Collapse
Affiliation(s)
- Mitsunori Fujino
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
- Human Biology, School of Integrative and Global Majors, University of Tsukuba, Ibaraki, Japan
| | - Masami Ojima
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
| | - Shun Ishibashi
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
- Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
- Laboratory Animal Resource Center, University of Tsukuba, Ibaraki, Japan
- Life Science Center, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Ibaraki, Japan
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
9
|
Fujino M, Morito N, Hayashi T, Ojima M, Ishibashi S, Kuno A, Koshiba S, Yamagata K, Takahashi S. Transcription factor c-Maf deletion improves streptozotocin-induced diabetic nephropathy by directly regulating Sglt2 and Glut2. JCI Insight 2023; 8:163306. [PMID: 36787192 PMCID: PMC10070115 DOI: 10.1172/jci.insight.163306] [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: 07/06/2022] [Accepted: 02/01/2023] [Indexed: 02/15/2023] Open
Abstract
The transcription factor c-Maf has been widely studied and has been reported to play a critical role in embryonic kidney development; however, the postnatal functions of c-Maf in adult kidneys remain unknown as c-Maf-null C57BL/6J mice exhibit embryonic lethality. In this study, we investigated the role of c-Maf in adult mouse kidneys by comparing the phenotypes of tamoxifen-inducible (TAM-inducible) c-Maf-knockout mice (c-Maffl/fl; CAG-Cre-ERTM mice named "c-MafΔTAM") with those of c-Maffl/fl control mice, 10 days after TAM injection [TAM(10d)]. In addition, we examined the effects of c-Maf deletion on diabetic conditions by injecting the mice with streptozotocin, 4 weeks before TAM injection. c-MafΔTAM mice displayed primary glycosuria caused by sodium-glucose cotransporter 2 (Sglt2) and glucose transporter 2 (Glut2) downregulation in the kidneys without diabetes, as well as morphological changes and life-threatening injuries in the kidneys on TAM(10d). Under diabetic conditions, c-Maf deletion promoted recovery from hyperglycemia and suppressed albuminuria and diabetic nephropathy by causing similar effects as did Sglt2 knockout and SGLT2 inhibitors. In addition to demonstrating the potentially unique gene regulation of c-Maf, these findings highlight the renoprotective effects of c-Maf deficiency under diabetic conditions and suggest that c-Maf could be a novel therapeutic target gene for treating diabetic nephropathy.
Collapse
Affiliation(s)
- Mitsunori Fujino
- Department of Anatomy and Embryology, Faculty of Medicine
- PhD Program in Human Biology, School of Integrative and Global Majors
| | - Naoki Morito
- Department of Nephrology, Faculty of Medicine; and
| | - Takuto Hayashi
- Department of Anatomy and Embryology, Faculty of Medicine
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Masami Ojima
- Department of Anatomy and Embryology, Faculty of Medicine
| | - Shun Ishibashi
- Department of Anatomy and Embryology, Faculty of Medicine
- Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Akihiro Kuno
- Department of Anatomy and Embryology, Faculty of Medicine
| | - Seizo Koshiba
- Tohoku Medical Megabank Organization and
- Advanced Research Center for Innovations in Next-Generation Medicine (INGEM), Tohoku University, Sendai, Japan
| | | | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine
- Laboratory Animal Resource Center
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA)
- International Institute for Integrative Sleep Medicine (WPI-IIIS), and
- Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
10
|
Kotas ME, O'Leary CE, Locksley RM. Tuft Cells: Context- and Tissue-Specific Programming for a Conserved Cell Lineage. ANNUAL REVIEW OF PATHOLOGY 2023; 18:311-335. [PMID: 36351364 PMCID: PMC10443898 DOI: 10.1146/annurev-pathol-042320-112212] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tuft cells are found in tissues with distinct stem cell compartments, tissue architecture, and luminal exposures but converge on a shared transcriptional program, including expression of taste transduction signaling pathways. Here, we summarize seminal and recent findings on tuft cells, focusing on major categories of function-instigation of type 2 cytokine responses, orchestration of antimicrobial responses, and emerging roles in tissue repair-and describe tuft cell-derived molecules used to affect these functional programs. We review what is known about the development of tuft cells from epithelial progenitors under homeostatic conditions and during disease. Finally, we discuss evidence that immature, or nascent, tuft cells with potential for diverse functions are driven toward dominant effector programs by tissue- or perturbation-specific contextual cues, which may result in heterogeneous mature tuft cell phenotypes both within and between tissues.
Collapse
Affiliation(s)
- Maya E Kotas
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, University of California, San Francisco, California, USA
- Department of Medicine, University of California, San Francisco, California, USA
| | - Claire E O'Leary
- Department of Medicine, University of California, San Francisco, California, USA
- Current affiliation: Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA;
- Howard Hughes Medical Institute, University of California, San Francisco, California, USA
| |
Collapse
|
11
|
Cosovanu C, Resch P, Jordan S, Lehmann A, Ralser M, Farztdinov V, Spranger J, Mülleder M, Brachs S, Neumann C. Intestinal epithelial c-Maf expression determines enterocyte differentiation and nutrient uptake in mice. J Exp Med 2022; 219:213479. [PMID: 36121416 PMCID: PMC9486084 DOI: 10.1084/jem.20220233] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 06/20/2022] [Accepted: 08/24/2022] [Indexed: 11/04/2022] Open
Abstract
The primary function of the small intestine (SI) is to absorb nutrients to maintain whole-body energy homeostasis. Enterocytes are the major epithelial cell type facilitating nutrient sensing and uptake. However, the molecular regulators governing enterocytes have remained undefined. Here, we identify c-Maf as an enterocyte-specific transcription factor within the SI epithelium. c-Maf expression was determined by opposing Noggin/BMP signals and overlapped with the zonated enrichment of nutrient transporters in the mid-villus region. Functionally, enterocytes required c-Maf to appropriately differentiate along the villus axis. Specifically, gene programs controlling carbohydrate and protein absorption were c-Maf-dependent. Consequently, epithelial cell-specific c-Maf deletion resulted in impaired enterocyte maturation and nutrient uptake, including defects in the adaptation to different nutrient availability. Concomitantly, intraepithelial lymphocytes were less abundant, while commensal epithelial cell-attaching SFB overgrew in a c-Maf-deficient environment, highlighting the close interdependence between the intestinal epithelium, immune system, and microbiota. Collectively, our data identified c-Maf as a key regulator of SI enterocyte differentiation and function, essential for nutrient, immune, and microbial homeostasis.
Collapse
Affiliation(s)
- Catalina Cosovanu
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Resch
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Jordan
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Lehmann
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Ralser
- Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany.,The Francis Crick Institute, Molecular Biology of Metabolism Laboratory, London, UK
| | - Vadim Farztdinov
- Core Facility - High-Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, partner site Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility - High-Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Brachs
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Centre for Cardiovascular Research, partner site Berlin, Berlin, Germany
| | - Christian Neumann
- Department of Microbiology, Infectious Diseases and Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
12
|
Bara AM, Chen L, Ma C, Underwood J, Moreci RS, Sumigray K, Sun T, Diao Y, Verzi M, Lechler T. Maf family transcription factors are required for nutrient uptake in the mouse neonatal gut. Development 2022; 149:285915. [PMID: 36504079 PMCID: PMC10112929 DOI: 10.1242/dev.201251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
There are fundamental differences in how neonatal and adult intestines absorb nutrients. In adults, macromolecules are broken down into simpler molecular components in the lumen of the small intestine, then absorbed. In contrast, neonates are thought to rely on internalization of whole macromolecules and subsequent degradation in the lysosome. Here, we identify the Maf family transcription factors MAFB and c-MAF as markers of terminally differentiated intestinal enterocytes throughout life. The expression of these factors is regulated by HNF4α and HNF4γ, master regulators of enterocyte cell fate. Loss of Maf factors results in a neonatal-specific failure to thrive and loss of macromolecular nutrient uptake. RNA-Seq and CUT&RUN analyses defined an endo-lysosomal program as being downstream of these transcription factors. We demonstrate major transcriptional changes in metabolic pathways, including fatty acid oxidation and increases in peroxisome number, in response to loss of Maf proteins. Finally, we show that loss of BLIMP1, a repressor of adult enterocyte genes, shows highly overlapping changes in gene expression and similar defects in macromolecular uptake. This work defines transcriptional regulators that are necessary for nutrient uptake in neonatal enterocytes.
Collapse
Affiliation(s)
- Anne M Bara
- Department of Dermatology, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Lei Chen
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA
| | - Celina Ma
- Department of Dermatology, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Julie Underwood
- Department of Dermatology, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Rebecca S Moreci
- Department of Dermatology, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Kaelyn Sumigray
- Department of Dermatology, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Tongyu Sun
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Yarui Diao
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Michael Verzi
- Department of Genetics, Rutgers University, Piscataway, NJ 08854, USA
| | - Terry Lechler
- Department of Dermatology, Duke University, Durham, NC 27710, USA.,Department of Cell Biology, Duke University, Durham, NC 27710, USA
| |
Collapse
|