1
|
Burge KY, Zhong H, Wilson AP, Chaaban H. Network-Based Bioinformatics Highlights Broad Importance of Human Milk Hyaluronan. Int J Mol Sci 2024; 25:12679. [PMID: 39684390 DOI: 10.3390/ijms252312679] [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: 10/27/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Human milk (HM) is rich in bioactive factors promoting postnatal small intestinal development and maturation of the microbiome. HM is also protective against necrotizing enterocolitis (NEC), a devastating inflammatory condition predominantly affecting preterm infants. The HM glycosaminoglycan, hyaluronan (HA), is present at high levels in colostrum and early milk. Our group has demonstrated that HA with a molecular weight of 35 kDa (HA35) promotes maturation of the murine neonatal intestine and protects against two distinct models of NEC. However, the molecular mechanisms underpinning HA35-induced changes in the developing ileum are unclear. CD-1 mouse pups were treated with HA35 or vehicle control daily, from P7 to P14, and we used network and functional analyses of bulk RNA-seq ileal transcriptomes to further characterize molecular mechanisms through which HA35 likely influences intestinal maturation. HA35-treated pups separated well by principal component analysis, and cell deconvolution revealed increases in stromal, Paneth, and mature enterocyte and progenitor cells in HA35-treated pups. Gene set enrichment and pathway analyses demonstrated upregulation in key processes related to antioxidant and growth pathways, such as nuclear factor erythroid 2-related factor-mediated oxidative stress response, hypoxia inducible factor-1 alpha, mechanistic target of rapamycin, and downregulation of apoptotic signaling. Collectively, pro-growth and differentiation signals induced by HA35 may present novel mechanisms by which this HM bioactive factor may protect against NEC.
Collapse
Affiliation(s)
- Kathryn Y Burge
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hua Zhong
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Adam P Wilson
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hala Chaaban
- Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
2
|
Fang YX, Lu EQ, Xu E, Zhang YY, Zhu M. Arf1 promotes porcine intestinal epithelial cell proliferation via the mTORC1 signaling pathway. In Vitro Cell Dev Biol Anim 2024; 60:1009-1020. [PMID: 39093368 DOI: 10.1007/s11626-024-00942-y] [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: 02/22/2024] [Accepted: 06/20/2024] [Indexed: 08/04/2024]
Abstract
The promotion of gut health, a pervasive problem in modern animal husbandry, positively affects organismal health, productivity, and economics. Porcine intestinal epithelial cells (IPEC-J2) continuously proliferate to maintain intestinal homeostasis, including barrier, immune, and absorptive functions. Gut homeostasis is fundamental to organismal health. ADP-ribosylation factor 1 (Arf1), a small GTPase, plays a crucial role in coordinating mTORC1 in response to nutrients, especially amino acid availability in the gut. mTORC1 is the central hub of proliferation. Thus, it seems likely that Arf1 promotes IPEC-J2 cell proliferation. However, the exact role of Arf1 in the porcine gut remains unclear. Therefore, we evaluated the functional role and possible mechanisms of Arf1 in the porcine intestine through Arf1 overexpression and knockdown in IPEC-J2 cells. Arf1 overexpression and knockdown significantly enhanced and inhibited, respectively, IPEC-J2 cell viability, and PCNA expression varied with Arf1 expression. Moreover, the proportion of Ki67-positive cells was significantly greater in the Arf1-overexpressing group than in the control group. These results suggest that Arf1 improves IPEC-J2 cell proliferation. The underlying mechanism was explored by Western blotting. Arf1 overexpression and knockdown significantly enhanced and suppressed, respectively, the levels of p-S6K1 and p-RPS6, which are key downstream targets of the mTORC1 signaling pathway. Collectively, our findings reveal the role of the Arf1-mTORC1 axis in IPEC-J2 cell proliferation and its potential function in regulating intestinal homeostasis and health.
Collapse
Affiliation(s)
- Yong-Xia Fang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - En-Qing Lu
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - E Xu
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Yi-Yu Zhang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Min Zhu
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou Province, China.
- Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
3
|
Deans-Fielder K, Wu T, Nguyen T, To S, Huang YZ, Bark SJ, Mills JC, Shroyer NF. Mechanisms driving fasting-induced protection from genotoxic injury in the small intestine. Am J Physiol Gastrointest Liver Physiol 2024; 326:G504-G524. [PMID: 38349111 PMCID: PMC11376978 DOI: 10.1152/ajpgi.00126.2023] [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: 06/23/2023] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 04/05/2024]
Abstract
Genotoxic agents such as doxorubicin (DXR) can cause damage to the intestines that can be ameliorated by fasting. How fasting is protective and the optimal timing of fasting and refeeding remain unclear. Here, our analysis of fasting/refeeding-induced global intestinal transcriptional changes revealed metabolic shifts and implicated the cellular energetic hub mechanistic target of rapamycin complex 1 (mTORC1) in protecting from DXR-induced DNA damage. Our analysis of specific transcripts and proteins in intestinal tissue and tissue extracts showed that fasting followed by refeeding at the time of DXR administration reduced damage and caused a spike in mTORC1 activity. However, continued fasting after DXR prevented the mTORC1 spike and damage reduction. Surprisingly, the mTORC1 inhibitor, rapamycin, did not block fasting/refeeding-induced reduction in DNA damage, suggesting that increased mTORC1 is dispensable for protection against the initial DNA damage response. In Ddit4-/- mice [DDIT4 (DNA-damage-inducible transcript 4) functions to regulate mTORC1 activity], fasting reduced DNA damage and increased intestinal crypt viability vs. ad libitum-fed Ddit4-/- mice. Fasted/refed Ddit4-/- mice maintained body weight, with increased crypt proliferation by 5 days post-DXR, whereas ad libitum-fed Ddit4-/- mice continued to lose weight and displayed limited crypt proliferation. Genes encoding epithelial stem cell and DNA repair proteins were elevated in DXR-injured, fasted vs. ad libitum Ddit4-/- intestines. Thus, fasting strongly reduced intestinal damage when normal dynamic regulation of mTORC1 was lost. Overall, the results confirm that fasting protects the intestines against DXR and suggests that fasting works by pleiotropic - including both mTORC1-dependent and independent - mechanisms across the temporally dynamic injury response.NEW & NOTEWORTHY New findings are 1) DNA damage reduction following a 24-h fast depends on the timing of postfast refeeding in relation to chemotherapy initiation; 2) fasting/refeeding-induced upregulation of mTORC1 activity is not required for early (6 h) protection against DXR-induced DNA damage; and 3) fasting increases expression of intestinal stem cell and DNA damage repair genes, even when mTORC1 is dysregulated, highlighting fasting's crucial role in regulating mTORC1-dependent and independent mechanisms in the dynamic recovery process.
Collapse
Affiliation(s)
- Kali Deans-Fielder
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Translational Biology and Molecular Medicine Graduate Program, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, United States
| | - Timothy Wu
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| | - Thanh Nguyen
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Cancer and Cell Biology Graduate Program, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States
| | - Sarah To
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Yang-Zhe Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Cancer and Cell Biology Graduate Program, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States
| | - Steven J Bark
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Jason C Mills
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, United States
| | - Noah F Shroyer
- Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
- Translational Biology and Molecular Medicine Graduate Program, Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, United States
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
4
|
Yetkin-Arik B, Jansen SA, Varderidou-Minasian S, Westendorp B, Skarp KP, Altelaar M, Lindemans CA, Lorenowicz MJ. Mesenchymal stromal/stem cells promote intestinal epithelium regeneration after chemotherapy-induced damage. Stem Cell Res Ther 2024; 15:125. [PMID: 38679715 PMCID: PMC11057078 DOI: 10.1186/s13287-024-03738-9] [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: 10/06/2023] [Accepted: 04/20/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for leukemia and a range of non-malignant disorders. The success of the therapy is hampered by occurrence of acute graft-versus-host disease (aGvHD); an inflammatory response damaging recipient organs, with gut, liver, and skin being the most susceptible. Intestinal GvHD injury is often a life-threatening complication in patients unresponsive to steroid treatment. Allogeneic mesenchymal stromal/stem cell (MSC) infusions are a promising potential treatment for steroid-resistant aGvHD. Data from our institution and others demonstrate rescue of approximately 40-50% of aGvHD patients with MSCs in Phase I, II studies and minor side effects. Although promising, better understanding of MSC mode of action and patient response to MSC-based therapy is essential to improve this lifesaving treatment. METHODS Single cell human small intestine organoids were embedded in Matrigel, grown for 5 days and treated with busulfan for 48 h. Organoids damaged by treatment with busulfan or control organoids were co-cultured with 5000, 10,000, and 50,000 MSCs for 24 h, 48 h or 7 days and the analyses such as surface area determination, proliferation and apoptosis assessment, RNA sequencing and proteomics were performed. RESULTS Here, we developed a 3D co-culture model of human small intestinal organoids and MSCs, which allows to study the regenerative effects of MSCs on intestinal epithelium in a more physiologically relevant setting than existing in vitro systems. Using this model we mimicked chemotherapy-mediated damage of the intestinal epithelium. The treatment with busulfan, the chemotherapeutic commonly used as conditioning regiment before the HSCT, affected pathways regulating epithelial to mesenchymal transition, proliferation, and apoptosis in small intestinal organoids, as shown by transcriptomic and proteomic analysis. The co-culture of busulfan-treated intestinal organoids with MSCs reversed the effects of busulfan on the transcriptome and proteome of intestinal epithelium, which we also confirmed by functional evaluation of proliferation and apoptosis. CONCLUSIONS Collectively, we demonstrate that our in vitro co-culture system is a new valuable tool to facilitate the investigation of the molecular mechanisms behind the therapeutic effects of MSCs on damaged intestinal epithelium. This could benefit further optimization of the use of MSCs in HSCT patients.
Collapse
Affiliation(s)
- B Yetkin-Arik
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
- Regenerative Medicine Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
- Centre for Living Technologies, Alliance TU/E, WUR, UU, UMC Utrecht, Princetonlaan 6, 3584 CB, Utrecht, The Netherlands
| | - S A Jansen
- Regenerative Medicine Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
- Division of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
- Pediatric Stem Cell Transplantation, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - S Varderidou-Minasian
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
- Regenerative Medicine Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - B Westendorp
- Regenerative Medicine Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
- Division Cell Biology, Metabolism and Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - K-P Skarp
- Biomedical Primate Research Center, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands
| | - M Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research, and Utrecht Institute For Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - C A Lindemans
- Regenerative Medicine Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
- Division of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands
- Pediatric Stem Cell Transplantation, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - M J Lorenowicz
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
- Regenerative Medicine Center, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands.
- Biomedical Primate Research Center, Lange Kleiweg 161, 2288 GJ, Rijswijk, The Netherlands.
| |
Collapse
|
5
|
Huang GX, Hallen NR, Lee M, Zheng K, Wang X, Mandanas MV, Djeddi S, Fernandez D, Hacker J, Ryan T, Bergmark RW, Bhattacharyya N, Lee S, Maxfield AZ, Roditi RE, Buchheit KM, Laidlaw TM, Gern JE, Hallstrand TS, Ray A, Wenzel SE, Boyce JA, Gutierrez-Arcelus M, Barrett NA. Increased epithelial mTORC1 activity in chronic rhinosinusitis with nasal polyps. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562288. [PMID: 37904989 PMCID: PMC10614789 DOI: 10.1101/2023.10.13.562288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Background The airway epithelium plays a central role in the pathogenesis of chronic respiratory diseases such as asthma and chronic rhinosinusitis with nasal polyps (CRSwNP), but the mechanisms by which airway epithelial cells (EpCs) maintain inflammation are poorly understood. Objective We hypothesized that transcriptomic assessment of sorted airway EpCs across the spectrum of differentiation would allow us to define mechanisms by which EpCs perpetuate airway inflammation. Methods Ethmoid sinus EpCs from adult patients with CRS were sorted into 3 subsets, bulk RNA sequenced, and analyzed for differentially expressed genes and pathways. Single cell RNA-seq (scRNA-seq) datasets from eosinophilic and non-eosinophilic CRSwNP and bulk RNA-seq of EpCs from mild/moderate and severe asthma were assessed. Immunofluorescent staining and ex vivo functional analysis of sinus EpCs were used to validate our findings. Results Analysis within and across purified EpC subsets revealed an enrichment in glycolytic programming in CRSwNP vs CRSsNP. Correlation analysis identified mammalian target of rapamycin complex 1 (mTORC1) as a potential regulator of the glycolytic program and identified EpC expression of cytokines and wound healing genes as potential sequelae. mTORC1 activity was upregulated in CRSwNP, and ex vivo inhibition demonstrated that mTOR is critical for EpC generation of CXCL8, IL-33, and CXCL2. Across patient samples, the degree of glycolytic activity was associated with T2 inflammation in CRSwNP, and with both T2 and non-T2 inflammation in severe asthma. Conclusions Together, these findings highlight a metabolic axis required to support epithelial generation of cytokines critical to both chronic T2 and non-T2 inflammation in CRSwNP and asthma.
Collapse
Affiliation(s)
- George X. Huang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Nils R. Hallen
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Minkyu Lee
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Kelly Zheng
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Xin Wang
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | | | - Sarah Djeddi
- Division of Immunology, Boston Children’s Hospital; Boston, MA
| | | | - Jonathan Hacker
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Tessa Ryan
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Regan W. Bergmark
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Neil Bhattacharyya
- Department of Otolaryngology, Head and Neck Surgery, Massachusetts Eye and Ear Infirmary; Boston, MA
| | - Stella Lee
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Alice Z. Maxfield
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Rachel E. Roditi
- Department of Otolaryngology, Head and Neck Surgery, Brigham and Women’s Hospital; Boston, MA
| | - Kathleen M. Buchheit
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Tanya M. Laidlaw
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - James E. Gern
- Division of Allergy, Immunology, and Rheumatology, University of Wisconsin School of Medicine and Public Health; Madison, WI
| | - Teal S. Hallstrand
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington Medical Center; Seattle, WA
| | - Anuradha Ray
- Department of Immunology, University of Pittsburgh; Pittsburgh, PA
| | - Sally E. Wenzel
- Department of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center; Pittsburgh, PA
| | - Joshua A. Boyce
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| | - Maria Gutierrez-Arcelus
- Division of Immunology, Boston Children’s Hospital; Boston, MA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard; Cambridge, MA
| | - Nora A. Barrett
- Jeff and Penny Vinik Center for Translational Immunology Research, Division of Allergy and Clinical Immunology, Brigham and Women’s Hospital; Boston, MA
- Department of Medicine, Harvard Medical School; Boston, MA
| |
Collapse
|
6
|
Solanki S, Sanchez K, Ponnusamy V, Kota V, Bell HN, Cho CS, Kowalsky AH, Green M, Lee JH, Shah YM. Dysregulated Amino Acid Sensing Drives Colorectal Cancer Growth and Metabolic Reprogramming Leading to Chemoresistance. Gastroenterology 2023; 164:376-391.e13. [PMID: 36410445 PMCID: PMC10448739 DOI: 10.1053/j.gastro.2022.11.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 11/01/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND & AIMS Colorectal cancer (CRC) is a devastating disease that is highly modulated by dietary nutrients. Mechanistic target of rapamycin complex 1 (mTORC1) contributes to tumor growth and limits therapy responses. Growth factor signaling is a major mechanism of mTORC1 activation. However, compensatory pathways exist to sustain mTORC1 activity after therapies that target oncogenic growth factor signaling. Amino acids potently activate mTORC1 via amino acid-sensing GTPase activity towards Rags (GATOR). The role of amino acid-sensing pathways in CRC is unclear. METHODS Human colon cancer cell lines, preclinical intestinal epithelial-specific GATOR1 and GATOR2 knockout mice subjected to colitis-induced or sporadic colon tumor models, small interfering RNA screening targeting regulators of mTORC1, and tissues of patients with CRC were used to assess the role of amino acid sensing in CRC. RESULTS We identified loss-of-function mutations of the GATOR1 complex in CRC and showed that altered expression of amino acid-sensing pathways predicted poor patient outcomes. We showed that dysregulated amino acid-sensing induced mTORC1 activation drives colon tumorigenesis in multiple mouse models. We found amino acid-sensing pathways to be essential in the cellular reprogramming of chemoresistance, and chemotherapeutic-resistant patients with colon cancer exhibited de-regulated amino acid sensing. Limiting amino acids in in vitro and in vivo models (low-protein diet) reverted drug resistance, revealing a metabolic vulnerability. CONCLUSIONS Our findings suggest a critical role for amino acid-sensing pathways in driving CRC and highlight the translational implications of dietary protein intervention in CRC.
Collapse
Affiliation(s)
- Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Katherine Sanchez
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Varun Ponnusamy
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Vasudha Kota
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Hannah N Bell
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Chun-Seok Cho
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Allison H Kowalsky
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Michael Green
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Departments of Radiation Oncology and Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan; Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
7
|
Mazzei A, Pagliara P, Del Vecchio G, Giampetruzzi L, Croce F, Schiavone R, Verri T, Barca A. Cytoskeletal Responses and Aif-1 Expression in Caco-2 Monolayers Exposed to Phorbol-12-Myristate-13-Acetate and Carnosine. BIOLOGY 2022; 12:biology12010036. [PMID: 36671729 PMCID: PMC9855102 DOI: 10.3390/biology12010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/01/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022]
Abstract
The dis(re)organization of the cytoskeletal actin in enterocytes mediates epithelial barrier dys(re)function, playing a key role in modulating epithelial monolayer's integrity and remodeling under transition from physiological to pathological states. Here, by fluorescence-based morphological and morphometric analyses, we detected differential responses of cytoskeletal actin in intestinal epithelial Caco-2 cell monolayers at two different stages of their spontaneous differentiation, i.e., undifferentiated cells at 7 days post-seeding (dps) and differentiated enterocyte-like cells at 21 dps, upon challenge in vitro with the inflammation-mimicking stimulus of phorbol-12-myristate-13-acetate (PMA). In addition, specific responses were found in the presence of the natural dipeptide carnosine detecting its potential counteraction against PMA-induced cytoskeletal alterations and remodeling in differentiated Caco-2 monolayers. In such an experimental context, by both immunocytochemistry and Western blot assays in Caco-2 monolayers, we identified the expression of the allograft inflammatory factor 1 (AIF-1) as protein functionally related to both inflammatory and cytoskeletal pathways. In 21 dps monolayers, particularly, we detected variations of its intracellular localization associated with the inflammatory stimulus and its mRNA/protein increase associated with the differentiated 21 dps enterocyte-like monolayer compared to the undifferentiated cells.
Collapse
Affiliation(s)
- Aurora Mazzei
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Patrizia Pagliara
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
- Correspondence: (P.P.); (A.B.); Tel.: +39-0832-298662 (A.B.)
| | - Gianmarco Del Vecchio
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Lucia Giampetruzzi
- Institute for Microelectronics and Microsystems IMM-CNR, Via per Monteroni “Campus Ecotekne”, 73100 Lecce, Italy
| | - Francesca Croce
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Roberta Schiavone
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Amilcare Barca
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
- Correspondence: (P.P.); (A.B.); Tel.: +39-0832-298662 (A.B.)
| |
Collapse
|
8
|
Kaur H, Moreau R. Raptor knockdown concurrently increases the electrical resistance and paracellular permeability of Caco-2 cell monolayers. Life Sci 2022; 308:120989. [PMID: 36152680 DOI: 10.1016/j.lfs.2022.120989] [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: 07/13/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 11/18/2022]
Abstract
AIMS As a critical regulatory point of nutrient sensing, growth and metabolism, the mechanistic target of rapamycin complex 1 (mTORC1) is poised to influence intestinal homeostasis under basal conditions and in disease state. Intestinal barrier integrity ensures tissue homeostasis by closely regulating the permeability of the epithelium to lumenal contents. The role of mTORC1 in the regulation of intestinal barrier function and permeability remains to be fully elucidated. MATERIALS AND METHODS In this study, we employed lentivirus-mediated knockdown of mTORC1 signaling-associated proteins Raptor (regulatory-associated protein of mTOR) and TSC2 (tuberin) to ascertain the effects of constitutive activation or repression of mTORC1 activity on barrier function in Caco-2 cell monolayers. KEY FINDINGS Results showed that the loss of Raptor concomitantly raised the transepithelial electrical resistance (TEER) and para/transcellular permeability leading to a cell monolayer that is leaky for dextran yet electrically resistant to the movement of ions. Paracellular permeability was linked to the downregulation of tight junction protein expression and enhanced autophagy. Raptor-depleted cells had the highest abundance of myosin binding subunit MYPT1 concomitantly with the lowest abundance of p-MYPT1 (Thr696) and phosphorylated myosin light chain (p-MLC, Ser19) implying that MLC phosphatase activity was increased resulting in MLC relaxation. Although rapamycin suppressed mTORC1 activity and decreased the abundance of tight junction proteins in control cells, rapamycin caused a modest increase of TEER compared to Raptor knockdown. SIGNIFICANCE The study showed that epithelium paracellular permeability of small molecular weight dextran is dissociated from TEER.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| |
Collapse
|
9
|
Adkins-Threats M, Mills JC. Cell plasticity in regeneration in the stomach and beyond. Curr Opin Genet Dev 2022; 75:101948. [PMID: 35809361 PMCID: PMC10378711 DOI: 10.1016/j.gde.2022.101948] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/29/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
Recent studies using cell lineage-tracing techniques, organoids, and single-cell RNA sequencing analyses have revealed: 1) adult organs use cell plasticity programs to recruit progenitor cells to regenerate tissues after injury, and 2) plasticity is far more common than previously thought, even in homeostasis. Here, we focus on the complex interplay of normal stem cell differentiation and plasticity in homeostasis and after injury, using the gastric epithelium as a touchstone. We also examine common features of regenerative programs and discuss the evolutionarily conserved, stepwise process of paligenosis which reprograms mature cells into progenitors that can repair damaged tissue. Finally, we discuss how conserved plasticity programs may help us better understand pathological processes like metaplasia.
Collapse
Affiliation(s)
- Mahliyah Adkins-Threats
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, USA. https://twitter.com/@madkinsthreats
| | - Jason C Mills
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, USA; Department of Pathology & Immunology, Baylor College of Medicine, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, USA.
| |
Collapse
|
10
|
Mercier J, Calmel C, Mésinèle J, Sutanto E, Merabtene F, Longchampt E, Sage E, Kicic A, Boëlle PY, Corvol H, Ruffin M, Guillot L. SLC6A14 Impacts Cystic Fibrosis Lung Disease Severity via mTOR and Epithelial Repair Modulation. Front Mol Biosci 2022; 9:850261. [PMID: 35372502 PMCID: PMC8965518 DOI: 10.3389/fmolb.2022.850261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/16/2022] [Indexed: 12/26/2022] Open
Abstract
Cystic fibrosis (CF), due to pathogenic variants in CFTR gene, is associated with chronic infection/inflammation responsible for airway epithelium alteration and lung function decline. Modifier genes induce phenotype variability between people with CF (pwCF) carrying the same CFTR variants. Among these, the gene encoding for the amino acid transporter SLC6A14 has been associated with lung disease severity and age of primary airway infection by the bacteria Pseudomonas aeruginosa. In this study, we investigated whether the single nucleotide polymorphism (SNP) rs3788766, located within SLC6A14 promoter, is associated with lung disease severity in a large French cohort of pwCF. We also studied the consequences of this SNP on SLC6A14 promoter activity using a luciferase reporter and the role of SLC6A14 in the mechanistic target of rapamycin kinase (mTOR) signaling pathway and airway epithelial repair. We confirm that SLC6A14 rs3788766 SNP is associated with lung disease severity in pwCF (p = 0.020; n = 3,257, pancreatic insufficient, aged 6-40 years old), with the minor allele G being deleterious. In bronchial epithelial cell lines deficient for CFTR, SLC6A14 promoter activity is reduced in the presence of the rs3788766 G allele. SLC6A14 inhibition with a specific pharmacological blocker reduced 3H-arginine transport, mTOR phosphorylation, and bronchial epithelial repair rates in wound healing assays. To conclude, our study highlights that SLC6A14 genotype might affect lung disease severity of people with cystic fibrosis via mTOR and epithelial repair mechanism modulation in the lung.
Collapse
Affiliation(s)
- Julia Mercier
- Sorbonne Université, Inserm, Centre de Recherche Saint Antoine, CRSA, Paris, France
| | - Claire Calmel
- Sorbonne Université, Inserm, Centre de Recherche Saint Antoine, CRSA, Paris, France
| | - Julie Mésinèle
- Sorbonne Université, Inserm, Centre de Recherche Saint Antoine, CRSA, Paris, France
- Sorbonne Université, Inserm, Institut Pierre Louis D'épidémiologie et de Santé Publique, IPLESP, APHP, Hôpital Saint-Antoine, Paris, France
| | - Erika Sutanto
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
| | - Fatiha Merabtene
- Sorbonne Université, Inserm, Centre de Recherche Saint Antoine, CRSA, Paris, France
| | | | - Edouard Sage
- Départment de Chirurgie Thoracique et Transplantation Pulmonaire, Hôpital Foch, Suresnes, France
- UMR 0892 UVSQ-INRAE, VIM, Université Paris-Saclay, Jouy-en-Josas, France
| | - Anthony Kicic
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
- School of Population Health, Curtin University, Bentley, WA, Australia
- Centre for Cell Therapy and Regenerative Medicine, Medical School, The University of Western Australia, Nedlands, WA, Australia
- Department of Respiratory and Sleep Medicine, Perth Children’s Hospital, Nedlands, WA, Australia
| | - Pierre-Yves Boëlle
- Sorbonne Université, Inserm, Institut Pierre Louis D'épidémiologie et de Santé Publique, IPLESP, APHP, Hôpital Saint-Antoine, Paris, France
| | - Harriet Corvol
- Sorbonne Université, Inserm, Centre de Recherche Saint Antoine, CRSA, Paris, France
- AP-HP, Hôpital Trousseau, Service de Pneumologie Pédiatrique, Paris, France
| | - Manon Ruffin
- Sorbonne Université, Inserm, Centre de Recherche Saint Antoine, CRSA, Paris, France
| | - Loïc Guillot
- Sorbonne Université, Inserm, Centre de Recherche Saint Antoine, CRSA, Paris, France
| |
Collapse
|
11
|
Kaur H, Erickson A, Moreau R. Divergent regulation of inflammatory cytokines by mTORC1 in THP-1-derived macrophages and intestinal epithelial Caco-2 cells. Life Sci 2021; 284:119920. [PMID: 34478760 DOI: 10.1016/j.lfs.2021.119920] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 08/13/2021] [Accepted: 08/21/2021] [Indexed: 12/12/2022]
Abstract
AIMS The sustained activation of intestinal mechanistic target of rapamycin complex 1 (mTORC1) brought about by repeated mucosal insult or injury has been linked to escalation of gut inflammatory response, which may progress to damage the epithelium if not controlled. This study investigated the role of mTORC1 in the response of macrophage and enterocyte to inflammatory stimuli. MATERIALS AND METHODS We genetically manipulated human THP-1 monocytes and epithelial intestinal Caco-2 cells to generate stable cell lines with baseline, low or high mTORC1 kinase activity. The effects of THP-1 macrophage secretions onto Caco-2 cells were investigated by means of conditioned media transfer experiments. KEY FINDINGS The priming of mTORC1 for activation promoted lipopolysaccharide (LPS)-mediated THP-1 macrophage immune response as evidenced by the stimulation of inflammatory mediators (TNFα, IL-6, IL-8, IL-1β and IL-10). The treatment of THP-1 macrophages with LPS more than the manipulated level of mTORC1 activity of macrophages determined whether cytokine gene expression was induced in Caco-2 cells. LPS carry over was not responsible for the stimulation of Caco-2 cells' cytokine response. Knocking down Raptor in Caco-2 cells or treating Caco-2 cells with rapamycin enhanced Caco-2 TNFα gene expression revealing the anti-inflammatory role of a functional mTORC1 in intestinal epithelial cells exposed to macrophage-derived pro-inflammatory stimuli. SIGNIFICANCE Taken together, mTORC1 differentially impacts the immune responses of THP-1-derived macrophages and Caco-2 epithelial cells when placed in a pro-inflammatory microenvironment.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Anjeza Erickson
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| |
Collapse
|
12
|
Sfera A, Osorio C, Zapata Martín del Campo CM, Pereida S, Maurer S, Maldonado JC, Kozlakidis Z. Endothelial Senescence and Chronic Fatigue Syndrome, a COVID-19 Based Hypothesis. Front Cell Neurosci 2021; 15:673217. [PMID: 34248502 PMCID: PMC8267916 DOI: 10.3389/fncel.2021.673217] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/25/2021] [Indexed: 12/14/2022] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome is a serious illness of unknown etiology, characterized by debilitating exhaustion, memory impairment, pain and sleep abnormalities. Viral infections are believed to initiate the pathogenesis of this syndrome although the definite proof remains elusive. With the unfolding of COVID-19 pandemic, the interest in this condition has resurfaced as excessive tiredness, a major complaint of patients infected with the SARS-CoV-2 virus, often lingers for a long time, resulting in disability, and poor life quality. In a previous article, we hypothesized that COVID-19-upregulated angiotensin II triggered premature endothelial cell senescence, disrupting the intestinal and blood brain barriers. Here, we hypothesize further that post-viral sequelae, including myalgic encephalomyelitis/chronic fatigue syndrome, are promoted by the gut microbes or toxin translocation from the gastrointestinal tract into other tissues, including the brain. This model is supported by the SARS-CoV-2 interaction with host proteins and bacterial lipopolysaccharide. Conversely, targeting microbial translocation and cellular senescence may ameliorate the symptoms of this disabling illness.
Collapse
Affiliation(s)
- Adonis Sfera
- Patton State Hospital, San Bernardino, CA, United States
| | | | | | | | - Steve Maurer
- Patton State Hospital, San Bernardino, CA, United States
| | - Jose Campo Maldonado
- Department of Internal Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Zisis Kozlakidis
- International Agency for Research on Cancer (IARC), Lyon, France
| |
Collapse
|
13
|
Modulation of intestinal stem cell homeostasis by nutrients: a novel therapeutic option for intestinal diseases. Nutr Res Rev 2021; 35:150-158. [PMID: 34100341 DOI: 10.1017/s0954422421000172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Intestinal stem cells, which are capable of both self-renewal and differentiation to mature cell types, are responsible for maintaining intestinal epithelial homeostasis. Recent evidence indicates that these processes are mediated, in part, through nutritional status in response to diet. Diverse dietary patterns including caloric restriction, fasting, high-fat diets, ketogenic diets and high-carbohydrate diets as well as other nutrients control intestinal stem cell self-renewal and differentiation through nutrient-sensing pathways such as mammalian target of rapamycin and AMP-activated kinase. Herein, we summarise the current understanding of how intestinal stem cells contribute to intestinal epithelial homeostasis and diseases. We also discuss the effects of diet and nutrient-sensing pathways on intestinal stem cell self-renewal and differentiation, as well as their potential application in the prevention and treatment of intestinal diseases.
Collapse
|
14
|
Wang X, Undi RB, Ali N, Huycke MM. It takes a village: microbiota, parainflammation, paligenosis and bystander effects in colorectal cancer initiation. Dis Model Mech 2021; 14:dmm048793. [PMID: 33969420 PMCID: PMC10621663 DOI: 10.1242/dmm.048793] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sporadic colorectal cancer (CRC) is a leading cause of worldwide cancer mortality. It arises from a complex milieu of host and environmental factors, including genetic and epigenetic changes in colon epithelial cells that undergo mutation, selection, clonal expansion, and transformation. The gut microbiota has recently gained increasing recognition as an additional important factor contributing to CRC. Several gut bacteria are known to initiate CRC in animal models and have been associated with human CRC. In this Review, we discuss the factors that contribute to CRC and the role of the gut microbiota, focusing on a recently described mechanism for cancer initiation, the so-called microbiota-induced bystander effect (MIBE). In this cancer mechanism, microbiota-driven parainflammation is believed to act as a source of endogenous mutation, epigenetic change and induced pluripotency, leading to the cancerous transformation of colon epithelial cells. This theory links the gut microbiota to key risk factors and common histologic features of sporadic CRC. MIBE is analogous to the well-characterized radiation-induced bystander effect. Both phenomena drive DNA damage, chromosomal instability, stress response signaling, altered gene expression, epigenetic modification and cellular proliferation in bystander cells. Myeloid-derived cells are important effectors in both phenomena. A better understanding of the interactions between the gut microbiota and mucosal immune effector cells that generate bystander effects can potentially identify triggers for parainflammation, and gain new insights into CRC prevention.
Collapse
Affiliation(s)
- Xingmin Wang
- Nantong Institute of Genetics and Reproductive Medicine, Nantong Maternity and Child Healthcare Hospital, Nantong University, Nantong, Jiangsu 226018, China
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ram Babu Undi
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Naushad Ali
- Department of Internal Medicine, Section of Digestive Diseases and Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Mark M. Huycke
- Department of Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
15
|
Chen H, Gao S, Liu W, Wong CC, Wu J, Wu J, Liu D, Gou H, Kang W, Zhai J, Li C, Su H, Wang S, Soares F, Han J, He HH, Yu J. RNA N 6-Methyladenosine Methyltransferase METTL3 Facilitates Colorectal Cancer by Activating the m 6A-GLUT1-mTORC1 Axis and Is a Therapeutic Target. Gastroenterology 2021; 160:1284-1300.e16. [PMID: 33217448 DOI: 10.1053/j.gastro.2020.11.013] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 10/21/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS RNA N6-methyladenosine (m6A) modification has recently emerged as a new regulatory mechanism in cancer progression. We aimed to explore the role of the m6A regulatory enzyme METTL3 in colorectal cancer (CRC) pathogenesis and its potential as a therapeutic target. METHODS The expression and clinical implication of METTL3 were investigated in multiple human CRC cohorts. The underlying mechanisms of METTL3 in CRC were investigated by integrative m6A sequencing, RNA sequencing, and ribosome profiling analyses. The efficacy of targeting METTL3 in CRC treatment was elucidated in CRC cell lines, patient-derived CRC organoids, and Mettl3-knockout mouse models. RESULTS Using targeted clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 dropout screening, we identified METTL3 as the top essential m6A regulatory enzyme in CRC. METTL3 was overexpressed in 62.2% (79/127) and 88.0% (44/50) of primary CRCs from 2 independent cohorts. High METTL3 expression predicted poor survival in patients with CRC (n = 374, P < .01). Functionally, silencing METTL3 suppressed tumorigenesis in CRC cells, human-derived primary CRC organoids, and Mettl3-knockout mouse models. We discovered the novel functional m6A methyltransferase domain of METTL3 in CRC cells by domain-focused CRISPR screening and mutagenesis assays. Mechanistically, METTL3 directly induced the m6A-GLUT1-mTORC1 axis as identified by integrated m6A sequencing, RNA sequencing, ribosome sequencing, and functional validation. METTL3 induced GLUT1 translation in an m6A-dependent manner, which subsequently promoted glucose uptake and lactate production, leading to the activation of mTORC1 signaling and CRC development. Furthermore, inhibition of mTORC1 potentiated the anticancer effect of METTL3 silencing in CRC patient-derived organoids and METTL3 transgenic mouse models. CONCLUSIONS METTL3 promotes CRC by activating the m6A-GLUT1-mTORC1 axis. METTL3 is a promising therapeutic target for the treatment of CRC.
Collapse
Affiliation(s)
- Huarong Chen
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Shanshan Gao
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Weixin Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Chi-Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology and School of Life Sciences, Xiamen University, Xiamen, China
| | - Jingtong Wu
- State Key Laboratory of Cellular Stress Biology and School of Life Sciences, Xiamen University, Xiamen, China
| | - Dabin Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Hongyan Gou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Jianning Zhai
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Chuangen Li
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Hao Su
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Shiyan Wang
- Princess Margaret Cancer Centre, University Health Network, Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Fraser Soares
- Princess Margaret Cancer Centre, University Health Network, Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology and School of Life Sciences, Xiamen University, Xiamen, China
| | - Housheng Hansen He
- Princess Margaret Cancer Centre, University Health Network, Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
16
|
Abstract
Blood is generated throughout life by continued proliferation and differentiation of hematopoietic progenitors, while at the top of the hierarchy, hematopoietic stem cells (HSCs) remain largely quiescent. This way HSCs avoid senescence and preserve their capacity to repopulate the hematopoietic system. But HSCs are not always quiescent, proliferating extensively in conditions such as those found in the fetal liver. Understanding the elusive mechanisms that regulate HSC fate would enable us to comprehend a crucial piece of HSC biology and pave the way for ex-vivo HSC expansion with clear clinical benefit. Here we review how metabolism, endoplasmic reticulum stress and oxidative stress condition impact HSCs decision to self-renew or differentiate and how these signals integrate into the mammalian target of rapamycin (mTOR) pathway. We argue that the bone marrow microenvironment continuously favors differentiation through the activation of the mTOR complex (mTORC)1 signaling, while the fetal liver microenvironment favors self-renewal through the inverse mechanism. In addition, we also postulate that strategies that have successfully achieved HSC expansion, directly or indirectly, lead to the inactivation of mTORC1. Finally, we propose a mechanism by which mTOR signaling, during cell division, conditions HSC fate. This mechanism has already been demonstrated in mature hematopoietic cells (T-cells), that face a similar decision after activation, either undergoing clonal expansion or differentiation.
Collapse
|
17
|
Kaur H, Moreau R. mTORC1 silencing during intestinal epithelial Caco-2 cell differentiation is mediated by the activation of the AMPK/TSC2 pathway. Biochem Biophys Res Commun 2021; 545:183-188. [PMID: 33561653 DOI: 10.1016/j.bbrc.2021.01.070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/20/2021] [Indexed: 12/22/2022]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) signaling is the prototypical pathway regulating protein synthesis and cell proliferation. The level of mTORC1 activity is high in intestinal stem cells located at the base of the crypts and thought to gradually decrease as transit-amplifying cells migrate out of the crypts and differentiate into enterocytes, goblet cells or enteroendocrine cells along the epithelium. The unknown mechanism responsible for the silencing of intestinal epithelium mTORC1 during cell differentiation was investigated in Caco-2 cells, which spontaneously differentiate into enterocytes in standard growth medium. The results show that TSC2, an upstream negative regulator of mTORC1 was central to mTORC1 silencing in differentiated Caco-2 cells. AMPK-mediated activation of TSC2 (Ser1387) and repression of Raptor (Ser792), an essential component of mTORC1, were stimulated in differentiated Caco-2 cells. ERK1/2-mediated repression of TSC2 (Ser664) seen in undifferentiated Caco-2 cells was lifted in differentiated cells. IRS-1-mediated activation of AKT (Thr308) phosphorylation was stimulated in differentiated Caco-2 cells and may be involved in cross-pathway repression of ERK1/2. Additionally, PRAS40 (Thr246) phosphorylation was decreased in differentiated Caco-2 cells compared to undifferentiated cells allowing dephosphorylated PRAS40 to displace Raptor thereby repressing mTORC1 kinase activity.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA.
| |
Collapse
|
18
|
Kaur H, Moreau R. Curcumin steers THP-1 cells under LPS and mTORC1 challenges toward phenotypically resting, low cytokine-producing macrophages. J Nutr Biochem 2020; 88:108553. [PMID: 33220404 DOI: 10.1016/j.jnutbio.2020.108553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/07/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022]
Abstract
The persistent activation of intestinal mechanistic target of rapamycin complex 1 (mTORC1) triggered by mucosal stress has been linked to deregulation of the gut immune response resulting in intestinal inflammation and cell death. The present study investigated the regulatory properties of food-derived mTORC1 modulators, curcumin, and piperine, toward the polarization of stimulated macrophages and the differentiation of monocytes at two mTORC1 activity levels (baseline and elevated). To that end, we created stable human THP-1 monocytes exhibiting normal or constitutively active mTORC1. Curcumin or its combination with piperine, but not piperine alone, suppressed mTORC1 kinase activity, curtailed lipopolysaccharide-mediated inflammatory response of THP-1 macrophages, and repressed macrophage activation by inhibiting signaling pathways involved in M1 (mTORC1) and M2 (mTORC2 and cAMP response element binding protein) polarization. The effects of piperine in the curcumin/piperine combination were modest overall, indicating it was curcumin that modulated differentiating monocytes into acquiring a M0 macrophage phenotype characterized by low inflammatory cytokine output.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.
| |
Collapse
|
19
|
Kaur H, Moreau R. Curcumin represses mTORC1 signaling in Caco-2 cells by a two-sided mechanism involving the loss of IRS-1 and activation of AMPK. Cell Signal 2020; 78:109842. [PMID: 33234350 DOI: 10.1016/j.cellsig.2020.109842] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 01/09/2023]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a central modulator of inflammation and tumorigenesis in the gastrointestinal tract. Growth factors upregulate mTORC1 via the PI3K/AKT and/or Ras/MAPK signal pathways. Curcumin (CUR), a polyphenol found in turmeric roots (Curcuma longa) can repress mTORC1 kinase activity in colon cancer cell lines; however, key aspects of CUR mechanism of action remain to be elucidated including its primary cellular target. We investigated the molecular effects of physiologically attainable concentration of CUR (20 μM) in the intestinal lumen on mTORC1 signaling in Caco-2 cells. CUR markedly inhibited mTORC1 kinase activity as determined by the decreased phosphorylation of p70S6K (Thr389, -99%, P < 0.0001) and S6 (Ser235/236, -92%, P < 0.0001). Mechanistically, CUR decreased IRS-1 protein abundance (-80%, P < 0.0001) thereby downregulating AKT phosphorylation (Ser473, -94%, P < 0.0001) and in turn PRAS40 phosphorylation (Thr246, -99%, P < 0.0001) while total PRAS40 abundance was unchanged. The use of proteasome inhibitor MG132 showed that CUR-mediated loss of IRS-1 involved proteasomal degradation. CUR lowered Raptor protein abundance, which combined with PRAS40 hypophosphorylation, suggests CUR repressed mTORC1 activity by inducing compositional changes that hinder the complex assembly. In addition, CUR activated AMPK (Thr172 phosphorylation, P < 0.0001), a recognized repressor of mTORC1, and AMPK upstream regulator LKB1. Although cargo adapter protein p62 was decreased by CUR (-49%, P < 0.004), CUR did not significantly induce autophagy. Inhibition of AKT/mTORC1 signaling by CUR may have lifted the cross-inhibition onto MAPK signaling, which became induced; p-ERK1/2 (+670%, P < 0.0001), p-p38 (+1433%, P < 0.0001). By concomitantly targeting IRS-1 and AMPK, CUR's mechanism of mTORC1 inhibition is distinct from that of rapamycin.
Collapse
Affiliation(s)
- Harleen Kaur
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Régis Moreau
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA.
| |
Collapse
|
20
|
Kwon O, Han TS, Son MY. Intestinal Morphogenesis in Development, Regeneration, and Disease: The Potential Utility of Intestinal Organoids for Studying Compartmentalization of the Crypt-Villus Structure. Front Cell Dev Biol 2020; 8:593969. [PMID: 33195268 PMCID: PMC7644937 DOI: 10.3389/fcell.2020.593969] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022] Open
Abstract
The morphology and structure of the intestinal epithelium are rearranged dynamically during development, tissue regeneration, and disease progression. The most important characteristic of intestinal epithelial morphogenesis is the repetitive compartmentalized structures of crypt-villus units, which are crucial for maintaining intestinal homeostasis and functions. Abnormal structures are known to be closely associated with disease development and progression. Therefore, understanding how intestinal crypt-villus structures are formed and grown is essential for elucidating the physiological and pathophysiological roles of the intestinal epithelium. However, a critical knowledge gap in understanding the compartmentalization of the crypt-villus axis remains when using animal models, due to obvious inter-species differences and difficulty in real-time monitoring. Recently, emerging technologies such as organoid culture, lineage tracing, and single cell sequencing have enabled the assessment of the intrinsic mechanisms of intestinal epithelial morphogenesis. In this review, we discuss the latest research on the regulatory factors and signaling pathways that play a central role in the formation, maintenance, and regeneration of crypt-villus structures in the intestinal epithelium. Furthermore, we discuss how these factors and pathways play a role in development, tissue regeneration, and disease. We further explore how the current technology of three-dimensional intestinal organoids has contributed to the understanding of crypt-villus compartmentalization, highlighting new findings related to the self-organizing-process-driven initiation and propagation of crypt-villus structures. We also discuss intestinal diseases featuring abnormalities of the crypt-villus structure to provide insights for the development of novel therapeutic strategies targeting intestinal morphogenesis and crypt-villus formation.
Collapse
Affiliation(s)
- Ohman Kwon
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Tae-Su Han
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Mi-Young Son
- Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
- KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
21
|
Li J, Cai Q, Ge H, Xue C, Fu Q, Cheng B. WITHDRAWN: RNA binding protein QKI5 accelerates tenogenic differentiation and promotes tendon healing through AKT-mTOR signalling by stabilizing MALAT1. Life Sci 2019:117236. [PMID: 31887297 DOI: 10.1016/j.lfs.2019.117236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/17/2019] [Accepted: 12/25/2019] [Indexed: 10/25/2022]
Abstract
This article has been withdrawn at the request of the authors. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Jun Li
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Qiuchen Cai
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Heng'an Ge
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Chao Xue
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China
| | - Qiang Fu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Biao Cheng
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai 200072, China.
| |
Collapse
|
22
|
Li H, Fu X, Yao F, Tian T, Wang C, Yang A. MTHFD1L-Mediated Redox Homeostasis Promotes Tumor Progression in Tongue Squamous Cell Carcinoma. Front Oncol 2019; 9:1278. [PMID: 31867267 PMCID: PMC6906156 DOI: 10.3389/fonc.2019.01278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 11/04/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Routine changes in cell metabolism can drive tumor development, as the cellular program develops to promote glycolysis and redox homeostasis during tumor progression; however, the associated mechanisms in tongue squamous cell carcinoma (TSCC) remain unclear. Methods: We investigated methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L) expression, its clinical relevance, redox modification, and molecular mechanisms using TSCC cells and tissues. The anti-tumor effects of MTHFD1L knockdown on TSCC tumorigenesis were evaluated in vitro and in vivo. Kaplan-Meier curves and the log-rank test were used to analyze disease-free survival and overall survival. Results: TSCC patients with high expression levels of MTHFD1L had shorter overall survival (P < 0.05) and disease-free survival (P < 0.05). Knockdown of MTHFD1L reduced nicotinamide adenine dinucleotide phosphate (NADPH) levels and increased reactive oxygen species (ROS), which accelerated cell death under oxidative stress, such as hypoxia or glucose deprivation. Additionally, inhibition of MTHFD1L suppressed TSCC cell growth and delayed the cell cycle, including in xenograft experiments. Conclusions: MTHFD1L confers redox homeostasis and promotes TSCC cell growth, which provides a great opportunity to study tumor metabolism in head and neck cancer. The mTORC1-4EBP1-eIF4E axis may affect the expression of MTHFD1L in TSCC. Inhibition of the expression of MTHFD1L may be an actionable and effective therapeutic target in TSCC.
Collapse
Affiliation(s)
- Hao Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaoyan Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Fan Yao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Tian Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chunyang Wang
- Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ankui Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|