1
|
Wu C, Gao Y, Jin Z, Huang Z, Wang H, Lu S, Guo S, Zhang F, Zhang J, Huang J, Tao X, Liu X, Zhang X, You L, Li Q, Wu J. PTPRG-AS1 regulates the KITLG/KIT pathway through the ceRNA axis to promote the malignant progression of gastric cancer and the intervention effect of Compound Kushen injection on it. Pharmacol Res 2025; 215:107743. [PMID: 40250508 DOI: 10.1016/j.phrs.2025.107743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/12/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Gastric cancer (GC) is a common malignant tumor with high mortality, recurrence, and metastasis rates. Compound Kushen injection (CKI) combination chemotherapy has been clinically used for the treatment of GC in China for many years, but its underlying mechanisms of action remain unclear. Recent reports have highlighted the important role of the competing endogenous RNA (ceRNA) mechanism of noncoding RNA (ncRNA) and messenger RNA (mRNA) formation in GC and other tumors. This study aimed to investigate the effects of CKI on GC from the ceRNA perspective. We confirmed the inhibitory effect of CKI on GC in mouse models and cell lines. By examining the GC cell lines sensitive to CKI treatment, we developed the CNScore method to analyze the ceRNA network, revealing that the CKI-GC ceRNA network promotes GC proliferation and metastasis through the PTPRG-AS1/hsa-miR-421/KITLG axis. Finally, we constructed GC cell models with PTPRG-AS1 overexpression or knockdown and GC liver metastasis models and found that PTPRG-AS1 can sponge hsa-miR-421, releasing KITLG and promoting GC proliferation and metastasis through the KITLG/KIT pathway. Taken together, CKI can suppress these malignant phenotypes by regulating the PTPRG-AS1/hsa-miR-421/KITLG axis.
Collapse
Affiliation(s)
- Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing 100053, China
| | - Yifei Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhengsen Jin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Zhihong Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Haojia Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Lu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Siyu Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fanqin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jingyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiaqi Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiaoyu Tao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinkui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Xiaomeng Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Leiming You
- School of Life Science, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Qinglin Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang Province 310022, China.
| | - Jiarui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
2
|
Zhang Y, Choi EL, Hayashi Y. Cellular and molecular mechanisms underlying aging-related gastric neuromuscular dysfunction. J Smooth Muscle Res 2025; 61:43-50. [PMID: 40204454 PMCID: PMC11996696 DOI: 10.1540/jsmr.61.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/03/2025] [Indexed: 04/11/2025] Open
Abstract
Aging is linked to a gradual decline in the gastric motor function, contributing to reduced food intake, and its association with frailty and sarcopenia. A key cellular change in the gastric neuromuscular apparatus is the loss of interstitial cells of Cajal (ICC), pacemaker cells of the gut. The ICC function as pacemakers that generate electrical slow waves and mediate enteric neurotransmission, playing a critical role in gastric motility. Aging-related ICC depletion leads to impaired gastric compliance and reduced slow wave activity, which contributes to early satiety and reduced food intake. Recent studies have elucidated the molecular and epigenetic mechanisms underlying aging-related ICC decline, highlighting the roles of ICC stem/precursor cells (ICC-SCs), transformation-related protein 53 (TRP53), extracellular signal-regulated kinase (ERK), and insulin-like growth factor 1 (IGF1) pathways, and epigenetic regulation mediated by the histone methyltransferase enhancer of zeste 2 (EZH2). By synthesizing the current findings, this review aims to provide a comprehensive understanding of the mechanisms driving ICC decline and to explore potential therapeutic strategies for preserving gastric motility in aging populations. Future research should aim to translate these discoveries into clinical applications to improve the gastric motor function and overall health in the aging population. Identifying effective interventions targeting ICC maintenance may ultimately help to alleviate age-related gastric motor dysfunction and its associated health burdens, including frailty, malnutrition, and impaired quality of life.
Collapse
Affiliation(s)
- Yuebo Zhang
- Enteric NeuroScience Program and Department of Physiology and
Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Egan L. Choi
- Enteric NeuroScience Program and Department of Physiology and
Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Yujiro Hayashi
- Enteric NeuroScience Program and Department of Physiology and
Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Ro S. Improving Gastric Motility in Aging Through EZH2 Inhibition and Preservation of Interstitial Cells of Cajal. Cell Mol Gastroenterol Hepatol 2024; 18:101382. [PMID: 39127454 PMCID: PMC11519705 DOI: 10.1016/j.jcmgh.2024.101382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Affiliation(s)
- Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada.
| |
Collapse
|
4
|
Taheri N, Choi EL, Nguyen VTT, Zhang Y, Huynh NM, Kellogg TA, van Wijnen AJ, Ordog T, Hayashi Y. Inhibition of EZH2 Reduces Aging-Related Decline in Interstitial Cells of Cajal of the Mouse Stomach. Cell Mol Gastroenterol Hepatol 2024; 18:101376. [PMID: 38969206 PMCID: PMC11359770 DOI: 10.1016/j.jcmgh.2024.101376] [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: 01/24/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND & AIMS Restricted gastric motor functions contribute to aging-associated undernutrition, sarcopenia, and frailty. We previously identified a decline in interstitial cells of Cajal (ICC; gastrointestinal pacemaker and neuromodulator cells) and their stem cells (ICC-SC) as a key factor of gastric aging. Altered functionality of the histone methyltransferase enhancer of zeste homolog 2 (EZH2) is central to organismal aging. Here, we investigated the role of EZH2 in the aging-related loss of ICC/ICC-SC. METHODS klotho mice, a model of accelerated aging, were treated with the most clinically advanced EZH2 inhibitor, EPZ6438 (tazemetostat; 160 mg/kg intraperitoneally twice a day for 3 weeks). Gastric ICC were analyzed by Western blotting and immunohistochemistry. ICC and ICC-SC were quantified by flow cytometry. Gastric slow wave activity was assessed by intracellular electrophysiology. Ezh2 was deactivated in ICC by treating KitcreERT2/+;Ezh2fl/fl mice with tamoxifen. TRP53, a key mediator of aging-related ICC loss, was induced with nutlin 3a in gastric muscle organotypic cultures and an ICC-SC line. RESULTS In klotho mice, EPZ6438 treatment mitigated the decline in the ICC growth factor KIT ligand/stem cell factor and gastric ICC. EPZ6438 also improved gastric slow wave activity and mitigated the reduced food intake and impaired body weight gain characteristic of this strain. Conditional genomic deletion of Ezh2 in Kit-expressing cells also prevented ICC loss. In organotypic cultures and ICC-SC, EZH2 inhibition prevented the aging-like effects of TRP53 stabilization on ICC/ICC-SC. CONCLUSIONS Inhibition of EZH2 with EPZ6438 mitigates aging-related ICC/ICC-SC loss and gastric motor dysfunction, improving slow wave activity and food intake in klotho mice.
Collapse
Affiliation(s)
- Negar Taheri
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Egan L Choi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Vy Truong Thuy Nguyen
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Yuebo Zhang
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Nick M Huynh
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Todd A Kellogg
- Department of Surgery, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | | | - Tamas Ordog
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Yujiro Hayashi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota.
| |
Collapse
|
5
|
Taheri N, Choi EL, Zhang Y, Hayashi Y. Protocol for gene knockdown using siRNA in organotypic cultures of murine gastric muscle. J Smooth Muscle Res 2024; 60:64-71. [PMID: 39675996 PMCID: PMC11638714 DOI: 10.1540/jsmr.60.64] [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: 09/03/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
Understanding the molecular interactions within the neuromuscular apparatus in the stomach is crucial for understanding their role in maintaining interstitial cells, such as the interstitial cells of Cajal (ICC), smooth muscle, and enteric neurons, as well as their contribution to gastric motility in both healthy and diseased states. Disruptions of these systems can lead to various gastric motor disorders and diseases, making it essential to explore their functions in detail. We herein present a protocol for gene knockdown using small interfering RNA (siRNAs) in organotypic culture. This ex vivo approach allows the precise manipulation of the gene expression in a tissue environment that closely mimics in vivo conditions, providing valuable insights into the gene function and its effects on gastric physiology. The protocol includes detailed steps for tissue preparation to ensure the preservation of the gastric muscles and the associated neuromuscular apparatus. We then describe the process of siRNA-mediated gene knockdown, offering tips for optimizing transfection efficiency and gene silencing. Additionally, we outline methods for analyzing the effectiveness of knockdown, including both quantitative and qualitative methods for the evaluation of the target gene expression. This protocol is adaptable to various research needs, allowing researchers to focus on specific genes of interest within the neuromuscular system of the stomach. By applying this approach, investigators can deepen their understanding of the molecular mechanisms underlying gastric motility and contribute to the development of new therapeutic strategies for treating gastric motor disorders and diseases.
Collapse
Affiliation(s)
- Negar Taheri
- Department of Physiology and Biomedical Engineering, 200
First Street SW, Rochester, MN 55905, USA
| | - Egan L. Choi
- Department of Physiology and Biomedical Engineering, 200
First Street SW, Rochester, MN 55905, USA
| | - Yuebo Zhang
- Department of Physiology and Biomedical Engineering, 200
First Street SW, Rochester, MN 55905, USA
| | - Yujiro Hayashi
- Department of Physiology and Biomedical Engineering, 200
First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
6
|
Choi EL, Taheri N, Tan E, Matsumoto K, Hayashi Y. The Crucial Role of the Interstitial Cells of Cajal in Neurointestinal Diseases. Biomolecules 2023; 13:1358. [PMID: 37759758 PMCID: PMC10526372 DOI: 10.3390/biom13091358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Neurointestinal diseases result from dysregulated interactions between the nervous system and the gastrointestinal (GI) tract, leading to conditions such as Hirschsprung's disease and irritable bowel syndrome. These disorders affect many people, significantly diminishing their quality of life and overall health. Central to GI motility are the interstitial cells of Cajal (ICC), which play a key role in muscle contractions and neuromuscular transmission. This review highlights the role of ICC in neurointestinal diseases, revealing their association with various GI ailments. Understanding the functions of the ICC could lead to innovative perspectives on the modulation of GI motility and introduce new therapeutic paradigms. These insights have the potential to enhance efforts to combat neurointestinal diseases and may lead to interventions that could alleviate or even reverse these conditions.
Collapse
Affiliation(s)
- Egan L. Choi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Guggenheim 10, 200 1st Street SW, Rochester, MN 55905, USA; (E.L.C.); (N.T.)
- Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Negar Taheri
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Guggenheim 10, 200 1st Street SW, Rochester, MN 55905, USA; (E.L.C.); (N.T.)
- Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Elijah Tan
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Guggenheim 10, 200 1st Street SW, Rochester, MN 55905, USA; (E.L.C.); (N.T.)
- Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Kenjiro Matsumoto
- Laboratory of Pathophysiology, Faculty of Pharmaceutical Sciences, Doshisha Women’s College of Liberal Arts, Kyoto 610-0395, Japan;
| | - Yujiro Hayashi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Guggenheim 10, 200 1st Street SW, Rochester, MN 55905, USA; (E.L.C.); (N.T.)
- Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
7
|
Tieniber AD, Rossi F, Hanna AN, Liu M, Etherington MS, Loo JK, Param N, Zeng S, Do K, Wang L, DeMatteo RP. Multiple intratumoral sources of kit ligand promote gastrointestinal stromal tumor. Oncogene 2023; 42:2578-2588. [PMID: 37468679 DOI: 10.1038/s41388-023-02777-5] [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: 01/16/2023] [Revised: 06/22/2023] [Accepted: 07/05/2023] [Indexed: 07/21/2023]
Abstract
Gastrointestinal stromal tumor (GIST) is the most common human sarcoma and is typically driven by a single mutation in the Kit or PDGFRA receptor. While highly effective, tyrosine kinase inhibitors (TKIs) are not curative. The natural ligand for the Kit receptor is Kit ligand (KitL), which exists in both soluble and membrane-bound forms. While KitL is known to stimulate human GIST cell lines in vitro, we used a genetically engineered mouse model of GIST containing a common human KIT mutation to investigate the intratumoral sources of KitL, importance of KitL during GIST oncogenesis, and contribution of soluble KitL to tumor growth in vivo. We discovered that in addition to tumor cells, endothelia and smooth muscle cells produced KitL in KitV558Δ/+ tumors, even after imatinib therapy. Genetic reduction of total KitL in tumor cells of KitV558Δ/+ mice impaired tumor growth in vivo. Similarly, genetic reduction of tumor cell soluble KitL in KitV558Δ/+ mice decreased tumor size. By RNA sequencing, quantitative PCR, and immunohistochemistry, KitL expression was heterogeneous in human GIST specimens. In particular, PDGFRA-mutant tumors had much higher KitL expression than Kit-mutant tumors, suggesting the benefit of Kit activation in the absence of mutant KIT. Serum KitL was higher in GIST patients with tumors resistant to imatinib and in those with tumors expressing more KitL RNA. Overall, KitL supports the growth of GIST at baseline and after imatinib therapy and remains a potential biomarker and therapeutic target.
Collapse
Affiliation(s)
- Andrew D Tieniber
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ferdinando Rossi
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew N Hanna
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Marion Liu
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Mark S Etherington
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer K Loo
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Nesteene Param
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Shan Zeng
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin Do
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Laura Wang
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ronald P DeMatteo
- Department of Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Truong Thuy Nguyen V, Taheri N, Choi EL, Kellogg TA, Linden DR, Hayashi Y. Insulin-Like Growth Factor1 Preserves Gastric Pacemaker Cells and Motor Function in Aging via ERK1/2 Activation. Cell Mol Gastroenterol Hepatol 2023; 16:369-383. [PMID: 37301443 PMCID: PMC10372898 DOI: 10.1016/j.jcmgh.2023.06.002] [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: 11/29/2022] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND & AIMS Impaired gastric motor function in the elderly causes reduced food intake leading to frailty and sarcopenia. We previously found that aging-related impaired gastric compliance was mainly owing to depletion of interstitial cells of Cajal (ICC), pacemaker cells, and neuromodulator cells. These changes were associated with reduced food intake. Transformation-related protein 53-induced suppression of extracellular signal-regulated protein kinase (ERK)1/2 in ICC stem cell (ICC-SC) cell-cycle arrest is a key process for ICC depletion and gastric dysfunction during aging. Here, we investigated whether insulin-like growth factor 1 (IGF1), which can activate ERK in gastric smooth muscles and invariably is reduced with age, could mitigate ICC-SC/ICC loss and gastric dysfunction in klotho mice, a model of accelerated aging. METHODS Klotho mice were treated with the stable IGF1 analog LONG R3 recombinant human (rh) IGF1 (150 μg/kg intraperitoneally twice daily for 3 weeks). Gastric ICC/ICC-SC and signaling pathways were studied by flow cytometry, Western blot, and immunohistochemistry. Gastric compliance was assessed in ex vivo systems. Transformation-related protein 53 was induced with nutlin 3a and ERK1/2 signaling was activated by rhIGF-1 in the ICC-SC line. RESULTS LONG R3 rhIGF1 treatment prevented reduced ERK1/2 phosphorylation and gastric ICC/ICC-SC decrease. LONG R3 rhIGF1 also mitigated the reduced food intake and impaired body weight gain. Improved gastric function by LONG R3 rhIGF1 was verified by in vivo systems. In ICC-SC cultures, rhIGF1 mitigated nutlin 3a-induced reduced ERK1/2 phosphorylation and cell growth arrest. CONCLUSIONS IGF1 can mitigate age-related ICC/ICC-SC loss by activating ERK1/2 signaling, leading to improved gastric compliance and increased food intake in klotho mice.
Collapse
Affiliation(s)
- Vy Truong Thuy Nguyen
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Negar Taheri
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Egan L Choi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Todd A Kellogg
- Department of Surgery, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - David R Linden
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Yujiro Hayashi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic College of Medicine and Science, Rochester, Minnesota.
| |
Collapse
|
9
|
Hayashi Y, Asuzu DT, Bardsley MR, Gajdos GB, Kvasha SM, Linden DR, Nagy RA, Saravanaperumal SA, Syed SA, Toyomasu Y, Yan H, Chini EN, Gibbons SJ, Kellogg TA, Khazaie K, Kuro-o M, Machado Espindola Netto J, Singh MP, Tidball JG, Wehling-Henricks M, Farrugia G, Ordog T. Wnt-induced, TRP53-mediated Cell Cycle Arrest of Precursors Underlies Interstitial Cell of Cajal Depletion During Aging. Cell Mol Gastroenterol Hepatol 2020; 11:117-145. [PMID: 32771388 PMCID: PMC7672319 DOI: 10.1016/j.jcmgh.2020.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Gastric dysfunction in the elderly may cause reduced food intake, frailty, and increased mortality. The pacemaker and neuromodulator cells interstitial cells of Cajal (ICC) decline with age in humans, and their loss contributes to gastric dysfunction in progeric klotho mice hypomorphic for the anti-aging Klotho protein. The mechanisms of ICC depletion remain unclear. Klotho attenuates Wnt (wingless-type MMTV integration site) signaling. Here, we examined whether unopposed Wnt signaling could underlie aging-associated ICC loss by up-regulating transformation related protein TRP53 in ICC stem cells (ICC-SC). METHODS Mice aged 1-107 weeks, klotho mice, APCΔ468 mice with overactive Wnt signaling, mouse ICC-SC, and human gastric smooth muscles were studied by RNA sequencing, reverse transcription-polymerase chain reaction, immunoblots, immunofluorescence, histochemistry, flow cytometry, and methyltetrazolium, ethynyl/bromodeoxyuridine incorporation, and ex-vivo gastric compliance assays. Cells were manipulated pharmacologically and by gene overexpression and RNA interference. RESULTS The klotho and aged mice showed similar ICC loss and impaired gastric compliance. ICC-SC decline preceded ICC depletion. Canonical Wnt signaling and TRP53 increased in gastric muscles of klotho and aged mice and middle-aged humans. Overstimulated canonical Wnt signaling increased DNA damage response and TRP53 and reduced ICC-SC self-renewal and gastric ICC. TRP53 induction persistently inhibited G1/S and G2/M cell cycle phase transitions without activating apoptosis, autophagy, cellular quiescence, or canonical markers/mediators of senescence. G1/S block reflected increased cyclin-dependent kinase inhibitor 1B and reduced cyclin D1 from reduced extracellular signal-regulated kinase activity. CONCLUSIONS Increased Wnt signaling causes age-related ICC loss by up-regulating TRP53, which induces persistent ICC-SC cell cycle arrest without up-regulating canonical senescence markers.
Collapse
Affiliation(s)
- Yujiro Hayashi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota,Yujiro Hayashi, PhD, Mayo Clinic, Guggenheim 10, 200 First Street SW, Rochester, Minnesota 55906. fax: (507) 255-6318.
| | - David T. Asuzu
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Michael R. Bardsley
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Gabriella B. Gajdos
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sergiy M. Kvasha
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - David R. Linden
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Rea A. Nagy
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Siva Arumugam Saravanaperumal
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sabriya A. Syed
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yoshitaka Toyomasu
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Huihuang Yan
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | - Eduardo N. Chini
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center and Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - Simon J. Gibbons
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | | | | | - Makoto Kuro-o
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas,Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Jair Machado Espindola Netto
- Signal Transduction and Molecular Nutrition Laboratory, Kogod Aging Center and Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - James G. Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California
| | | | - Gianrico Farrugia
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Tamas Ordog
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota,Correspondence Address correspondence to: Tamas Ordog, MD, Mayo Clinic, Guggenheim 10, 200 First Street SW, Rochester, Minnesota 55906. fax: (507) 255-6318.
| |
Collapse
|
10
|
von Mehren M, George S, Heinrich MC, Schuetze SM, Yap JT, Yu JQ, Abbott A, Litwin S, Crowley J, Belinsky M, Janeway KA, Hornick JL, Flieder DB, Chugh R, Rink L, Van den Abbeele AD. Linsitinib (OSI-906) for the Treatment of Adult and Pediatric Wild-Type Gastrointestinal Stromal Tumors, a SARC Phase II Study. Clin Cancer Res 2020; 26:1837-1845. [PMID: 31792037 PMCID: PMC7856429 DOI: 10.1158/1078-0432.ccr-19-1069] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 07/03/2019] [Accepted: 11/22/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE Most gastrointestinal stromal tumors (GIST) have activating mutations of KIT, PDGFRA, or uncommonly BRAF. Fifteen percent of adult and 85% of pediatric GISTs are wild type (WT), commonly having high expression of IGF-1R and loss of succinate dehydrogenase (SDH) complex function. We tested the efficacy of linsitinib, an oral TKI IGF-1R inhibitor, in patients with WT GIST. PATIENTS AND METHODS A multicenter phase II trial of linsitinib was conducted. The primary endpoint was objective response rate. Secondary endpoints were clinical benefit rate: complete response, partial response, and stable disease (SD) ≥ 9 months, and quantitative 2[18F]fluoro-2-deoxy-D-glucose (FDG) metabolic response (MR) at week 8. Serum levels for glucose, insulin, IGF-1R ligand IGF1, and binding proteins were obtained to explore correlations to patient outcomes and FDG-PET results. RESULTS Twenty patients were accrued in a 6-month period. Grade 3-4 toxicities possibly related to linsitinib were uncommon (8.5%). No objective responses were seen. Clinical benefit rate (CBR) at 9 months was 40%. Intense FDG uptake was observed at baseline, with partial MR of 12% and stable metabolic disease of 65% at week 8; these patients had RECIST 1.1 SD as their best response. Progression-free survival (PFS) and overall survival Kaplan-Meier estimates at 9 months were 52% and 80%, respectively. SDHA/B loss determined by IHC was seen in 35% and 88% of cases, respectively. CONCLUSIONS Linsitinib is well tolerated in patients with WT GIST. Although the 9-month CBR was 40%, and PFS at 9 months was 52%, no objective responses were observed. Rapid accrual to this study demonstrates that clinical trials of experimental agents in selected subtypes of GIST are feasible.
Collapse
Affiliation(s)
| | | | - Michael C Heinrich
- Portland VA Health Care System and OHSU Knight Cancer Institute, Portland, Oregon
| | | | - Jeffrey T Yap
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Jain Q Yu
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | | | | | - John Crowley
- Cancer Research and Biostatistics, Seattle, Washington
| | | | | | - Jason L Hornick
- Harvard Medical School, Boston, Massachusetts
- Brigham Health, Boston, Massachusetts
| | | | - Rashmi Chugh
- University of Michigan, Ann Arbor, Michigan
- Sarcoma Alliance for Research through Collaboration, Ann Arbor, Michigan
| | - Lori Rink
- Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Annick D Van den Abbeele
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
- Brigham Health, Boston, Massachusetts
| |
Collapse
|
11
|
Hedgehog pathway dysregulation contributes to the pathogenesis of human gastrointestinal stromal tumors via GLI-mediated activation of KIT expression. Oncotarget 2018; 7:78226-78241. [PMID: 27793025 PMCID: PMC5346634 DOI: 10.18632/oncotarget.12909] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 10/13/2016] [Indexed: 12/20/2022] Open
Abstract
Gastrointestinal stromal tumors (GIST) arise within the interstitial cell of Cajal (ICC) lineage due to activating KIT/PDGFRA mutations. Both ICC and GIST possess primary cilia (PC), which coordinate PDGFRA and Hedgehog signaling, regulators of gastrointestinal mesenchymal development. Therefore, we hypothesized that Hedgehog signaling may be altered in human GIST and controls KIT expression. Quantitative RT-PCR, microarrays, and next generation sequencing were used to describe Hedgehog/PC-related genes in purified human ICC and GIST. Genetic and pharmacologic approaches were employed to investigate the effects of GLI manipulation on KIT expression and GIST cell viability. We report that Hedgehog pathway and PC components are expressed in ICC and GIST and subject to dysregulation during GIST oncogenesis, irrespective of KIT/PDGFRA mutation status. Using genomic profiling, 10.2% of 186 GIST studied had potentially deleterious genomic alterations in 5 Hedgehog-related genes analyzed, including in the PTCH1 tumor suppressor (1.6%). Expression of the predominantly repressive GLI isoform, GLI3, was inversely correlated with KIT mRNA levels in GIST cells and non-KIT/non-PDGFRA mutant GIST. Overexpression of the 83-kDa repressive form of GLI3 or small interfering RNA-mediated knockdown of the activating isoforms GLI1/2 reduced KIT mRNA. Treatment with GLI1/2 inhibitors, including arsenic trioxide, significantly increased GLI3 binding to the KIT promoter, decreased KIT expression, and reduced viability in imatinib-sensitive and imatinib-resistant GIST cells. These data offer new evidence that genes necessary for Hedgehog signaling and PC function in ICC are dysregulated in GIST. Hedgehog signaling activates KIT expression irrespective of mutation status, offering a novel approach to treat imatinib-resistant GIST.
Collapse
|
12
|
Yuan J, Yin Z, Tao K, Wang G, Gao J. Function of insulin-like growth factor 1 receptor in cancer resistance to chemotherapy. Oncol Lett 2017; 15:41-47. [PMID: 29285186 DOI: 10.3892/ol.2017.7276] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/28/2017] [Indexed: 02/07/2023] Open
Abstract
Drug resistance is a primary cause of chemotherapeutic failure; however, how this resistance develops is complex. A comprehensive understanding of chemotherapeutic resistance mechanisms may aid in identifying more effective drugs and improve the survival rates of patients with cancer. Insulin-like growth factor 1 receptor (IGF1R), a member of the insulin receptor family, has been extensively assessed for biological activity, and its putative contribution to tumor cell development and progression. Furthermore, researchers have attended to drugs that target IGF1R since IGF1R functions as a membrane receptor. However, how IGF1R participates in chemotherapeutic resistance remains unclear. Therefore, the present study described the IGF1R gene and its associated signaling pathways, and offered details of IGF1R-induced tumor chemoresistance associated with promoting cell proliferation, inhibition of apoptosis, regulation of ATP-binding cassette transporter proteins and interactions with the extracellular matrix. The present study offered additional explanations for tumor chemotherapy resistance and provided a theoretical basis of IGF1R and its downstream pathways for future possible chemotherapy treatment options.
Collapse
Affiliation(s)
- Jingsheng Yuan
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zhijie Yin
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Guobing Wang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jinbo Gao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
13
|
Hayashi Y, Toyomasu Y, Saravanaperumal SA, Bardsley MR, Smestad JA, Lorincz A, Eisenman ST, Cipriani G, Nelson Holte MH, Al Khazal FJ, Syed SA, Gajdos GB, Choi KM, Stoltz GJ, Miller KE, Kendrick ML, Rubin BP, Gibbons SJ, Bharucha AE, Linden DR, Maher LJ, Farrugia G, Ordog T. Hyperglycemia Increases Interstitial Cells of Cajal via MAPK1 and MAPK3 Signaling to ETV1 and KIT, Leading to Rapid Gastric Emptying. Gastroenterology 2017; 153:521-535.e20. [PMID: 28438610 PMCID: PMC5526732 DOI: 10.1053/j.gastro.2017.04.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Depletion of interstitial cells of Cajal (ICCs) is common in diabetic gastroparesis. However, in approximately 20% of patients with diabetes, gastric emptying (GE) is accelerated. GE also occurs faster in obese individuals, and is associated with increased blood levels of glucose in patients with type 2 diabetes. To understand the fate of ICCs in hyperinsulinemic, hyperglycemic states characterized by rapid GE, we studied mice with mutation of the leptin receptor (Leprdb/db), which in our colony had accelerated GE. We also investigated hyperglycemia-induced signaling in the ICC lineage and ICC dependence on glucose oxidative metabolism in mice with disruption of the succinate dehydrogenase complex, subunit C gene (Sdhc). METHODS Mice were given breath tests to analyze GE of solids. ICCs were studied by flow cytometry, intracellular electrophysiology, isometric contractility measurement, reverse-transcription polymerase chain reaction, immunoblot, immunohistochemistry, enzyme-linked immunosorbent assays, and metabolite assays; cells and tissues were manipulated pharmacologically and by RNA interference. Viable cell counts, proliferation, and apoptosis were determined by methyltetrazolium, Ki-67, proliferating cell nuclear antigen, bromodeoxyuridine, and caspase-Glo 3/7 assays. Sdhc was disrupted in 2 different strains of mice via cre recombinase. RESULTS In obese, hyperglycemic, hyperinsulinemic female Leprdb/db mice, GE was accelerated and gastric ICC and phasic cholinergic responses were increased. Female KitK641E/+ mice, which have genetically induced hyperplasia of ICCs, also had accelerated GE. In isolated cells of the ICC lineage and gastric organotypic cultures, hyperglycemia stimulated proliferation by mitogen-activated protein kinase 1 (MAPK1)- and MAPK3-dependent stabilization of ets variant 1-a master transcription factor for ICCs-and consequent up-regulation of v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) receptor tyrosine kinase. Opposite changes occurred in mice with disruption of Sdhc. CONCLUSIONS Hyperglycemia increases ICCs via oxidative metabolism-dependent, MAPK1- and MAPK3-mediated stabilization of ets variant 1 and increased expression of KIT, causing rapid GE. Increases in ICCs might contribute to the acceleration in GE observed in some patients with diabetes.
Collapse
Affiliation(s)
- Yujiro Hayashi
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Yoshitaka Toyomasu
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Siva Arumugam Saravanaperumal
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Michael R. Bardsley
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - John A. Smestad
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Andrea Lorincz
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Fatimah J. Al Khazal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Sabriya A. Syed
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Gabriella B. Gajdos
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Kyoung Moo Choi
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota
| | - Gary J. Stoltz
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Katie E. Miller
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | | | - Brian P. Rubin
- Departments of Anatomic Pathology and Cancer Biology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Lerner Research Institute and Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Adil E. Bharucha
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - David R. Linden
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Louis James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | | | - Tamas Ordog
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota; Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
14
|
Huizinga JD. Commentary: Phase-amplitude coupling at the organism level: The amplitude of spontaneous alpha rhythm fluctuations varies with the phase of the infra-slow gastric basal rhythm. Front Neurosci 2017; 11:102. [PMID: 28303088 PMCID: PMC5332408 DOI: 10.3389/fnins.2017.00102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 02/17/2017] [Indexed: 01/09/2023] Open
Affiliation(s)
- Jan D Huizinga
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University Hamilton, ON, Canada
| |
Collapse
|
15
|
Chen J, Toyomasu Y, Hayashi Y, Linden DR, Szurszewski JH, Nelson H, Farrugia G, Kashyap PC, Chia N, Ordog T. Altered gut microbiota in female mice with persistent low body weights following removal of post-weaning chronic dietary restriction. Genome Med 2016; 8:103. [PMID: 27716401 PMCID: PMC5048651 DOI: 10.1186/s13073-016-0357-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/15/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Nutritional interventions often fail to prevent growth failure in childhood and adolescent malnutrition and the mechanisms remain unclear. Recent studies revealed altered microbiota in malnourished children and anorexia nervosa. To facilitate mechanistic studies under physiologically relevant conditions, we established a mouse model of growth failure following chronic dietary restriction and examined microbiota in relation to age, diet, body weight, and anabolic treatment. METHODS Four-week-old female BALB/c mice (n = 12/group) were fed ad libitum (AL) or offered limited food to abolish weight gain (LF). A subset of restricted mice was treated with an insulin-like growth factor 1 (IGF1) analog. Food access was restored in a subset of untreated LF (LF-RF) and IGF1-treated LF mice (TLF-RF) on day 97. Gut microbiota were determined on days 69, 96-99 and 120 by next generation sequencing of the V3-5 region of the 16S rRNA gene. Microbiota-host factor associations were analyzed by distance-based PERMANOVA and quantified by the coefficient of determination R2 for age, diet, and normalized body weight change (Δbwt). Microbial taxa on day 120 were compared following fitting with an overdispersed Poisson regression model. The machine learning algorithm Random Forests was used to predict age based on the microbiota. RESULTS On day 120, Δbwt in AL, LF, LF-RF, and TLF-RF mice was 52 ± 3, -6 ± 1*, 40 ± 3*, and 46 ± 2 % (*, P < 0.05 versus AL). Age and diet, but not Δbwt, were associated with gut microbiota composition. Age explained a larger proportion of the microbiota variability than diet or Δbwt. Random Forests predicted chronological age based on the microbiota and indicated microbiota immaturity in the LF mice before, but not after, refeeding. However, on day 120, the microbiota community structure of LF-RF mice was significantly different from that of both AL and LF mice. IGF1 mitigated the difference from the AL group. Refed groups had a higher abundance of Bacteroidetes and Proteobacteria and a lower abundance of Firmicutes than AL mice. CONCLUSIONS Persistent growth failure can be induced by 97-day dietary restriction in young female mice and is associated with microbiota changes seen in lean mice and individuals and anorexia nervosa. IGF1 facilitates recovery of body weights and microbiota.
Collapse
Affiliation(s)
- Jun Chen
- Division of Biomedical Statistics and Bioinformatics, Department of Health Sciences Research, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Yoshitaka Toyomasu
- Enteric NeuroScience Program, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Yujiro Hayashi
- Enteric NeuroScience Program, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - David R Linden
- Enteric NeuroScience Program, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Joseph H Szurszewski
- Enteric NeuroScience Program, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Heidi Nelson
- Department of Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Purna C Kashyap
- Enteric NeuroScience Program, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Nicholas Chia
- Division of Biomedical Statistics and Bioinformatics, Department of Health Sciences Research, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Department of Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.,Center for Individualized Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Tamas Ordog
- Enteric NeuroScience Program, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA. .,Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA. .,Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA. .,Center for Individualized Medicine, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
16
|
Hayashi Y, Bardsley MR, Toyomasu Y, Milosavljevic S, Gajdos GB, Choi KM, Reid-Lombardo KM, Kendrick ML, Bingener-Casey J, Tang CM, Sicklick JK, Gibbons SJ, Farrugia G, Taguchi T, Gupta A, Rubin BP, Fletcher JA, Ramachandran A, Ordog T. Platelet-Derived Growth Factor Receptor-α Regulates Proliferation of Gastrointestinal Stromal Tumor Cells With Mutations in KIT by Stabilizing ETV1. Gastroenterology 2015; 149:420-32.e16. [PMID: 25865047 PMCID: PMC4516576 DOI: 10.1053/j.gastro.2015.04.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 04/02/2015] [Accepted: 04/06/2015] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS In gastrointestinal muscles, v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) is predominantly expressed by interstitial cells of Cajal (ICC) and platelet-derived growth factor receptor-α (PDGFRA) polypeptide is expressed by so-called fibroblast-like cells. KIT and PDGFRA have been reported to be coexpressed in ICC precursors and gastrointestinal stromal tumors (GISTs), which originate from the ICC lineage. PDGFRA signaling has been proposed to stimulate growth of GISTs that express mutant KIT, but the effects and mechanisms of selective blockade of PDGFRA are unclear. We investigated whether inhibiting PDGFRA could reduce proliferation of GIST cells with mutant KIT via effects on the KIT-dependent transcription factor ETV1. METHODS We studied 53 gastric, small intestinal, rectal, or abdominal GISTs collected immediately after surgery or archived as fixed blocks at the Mayo Clinic and University of California, San Diego. In human GIST cells carrying imatinib-sensitive and imatinib-resistant mutations in KIT, PDGFRA was reduced by RNA interference (knockdown) or inhibited with crenolanib besylate (a selective inhibitor of PDGFRA and PDGFRB). Mouse ICC precursors were retrovirally transduced to overexpress wild-type Kit. Cell proliferation was analyzed by methyltetrazolium, 5-ethynyl-2'-deoxyuridine incorporation, and Ki-67 immunofluorescence assays; we also analyzed growth of xenograft tumors in mice. Gastric ICC and ICC precursors, and their PDGFRA(+) subsets, were analyzed by flow cytometry and immunohistochemistry in wild-type, Kit(+/copGFP), Pdgfra(+/eGFP), and NOD/ShiLtJ mice. Immunoblots were used to quantify protein expression and phosphorylation. RESULTS KIT and PDGFRA were coexpressed in 3%-5% of mouse ICC, 35%-44% of ICC precursors, and most human GIST samples and cell lines. PDGFRA knockdown or inhibition with crenolanib efficiently reduced proliferation of imatinib-sensitive and imatinib-resistant KIT(+)ETV1(+)PDGFRA(+) GIST cells (50% maximal inhibitory concentration = 5-32 nM), but not of cells lacking KIT, ETV1, or PDGFRA (50% maximal inhibitory concentration >230 nM). Crenolanib inhibited phosphorylation of PDGFRA and PDGFRB, but not KIT. However, Kit overexpression sensitized mouse ICC precursors to crenolanib. ETV1 knockdown reduced KIT expression and GIST proliferation. Crenolanib down-regulated ETV1 by inhibiting extracellular-signal-regulated kinase (ERK)-dependent stabilization of ETV1 protein and also reduced expression of KIT and PDGFRA. CONCLUSIONS In KIT-mutant GIST, inhibition of PDGFRA disrupts a KIT-ERK-ETV1-KIT signaling loop by inhibiting ERK activation. The PDGFRA inhibitor crenolanib might be used to treat patients with imatinib-resistant, KIT-mutant GIST.
Collapse
Affiliation(s)
- Yujiro Hayashi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Michael R. Bardsley
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Yoshitaka Toyomasu
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Srdjan Milosavljevic
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Gabriella B. Gajdos
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota
| | - Kyoung Moo Choi
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Chih-Min Tang
- Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, California
| | - Jason K. Sicklick
- Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, California
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Takahiro Taguchi
- Division of Human Health and Medical Science, Graduate School of Kuroshio Science, Kochi University, Kochi, Japan
| | - Anu Gupta
- Departments of Pathology and Molecular Genetics, Lerner Research Institute and Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Brian P. Rubin
- Departments of Pathology and Molecular Genetics, Lerner Research Institute and Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Jonathan A. Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Tamas Ordog
- Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota; Gastroenterology Research Unit, Mayo Clinic, Rochester, Minnesota; Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
17
|
Ordog T, Zörnig M, Hayashi Y. Targeting Disease Persistence in Gastrointestinal Stromal Tumors. Stem Cells Transl Med 2015; 4:702-7. [PMID: 25934947 DOI: 10.5966/sctm.2014-0298] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 03/16/2015] [Indexed: 01/11/2023] Open
Abstract
UNLABELLED SummaryGastrointestinal stromal tumors (GISTs) represent 20%-40% of human sarcomas. Although approximately half of GISTs are cured by surgery, prognosis of advanced disease used to be poor due to the high resistance of these tumors to conventional chemo- and radiotherapy. The introduction of molecularly targeted therapy (e.g., with imatinib mesylate) following the discovery of the role of oncogenic mutations in the receptor tyrosine kinases KIT and platelet-derived growth factor α (PDGFRA) significantly increased patient survival. However, GIST cells persist in 95%-97% of imatinib-treated patients who eventually progress and die of the disease because of the emergence of clones with drug-resistant mutations. Because these secondary mutations are highly heterogeneous, even second- and third-line drugs that are effective against certain genotypes have only moderately increased progression-free survival. Consequently, alternative strategies such as targeting molecular mechanisms underlying disease persistence should be considered. We reviewed recently discovered cell-autonomous and microenvironmental mechanisms that could promote the survival of GIST cells in the presence of tyrosine kinase inhibitor therapy. We particularly focused on the potential role of adult precursors for interstitial cells of Cajal (ICCs), the normal counterpart of GISTs. ICC precursors share phenotypic characteristics with cells that emerge in a subset of patients treated with imatinib and in young patients with GIST characterized by loss of succinate dehydrogenase complex proteins and lack of KIT or PDGFRA mutations. Eradication of residual GIST cells and cure of GIST will likely require individualized combinations of several approaches tailored to tumor genotype and phenotype. SIGNIFICANCE Gastrointestinal stromal tumors (GISTs) are one of the most common connective tissue cancers. Most GISTs that cannot be cured by surgery respond to molecularly targeted therapy (e.g., with imatinib); however, tumor cells persist in almost all patients and eventually acquire drug-resistant mutations. Several mechanisms contribute to the survival of GIST cells in the presence of imatinib, including the activation of "escape" mechanisms and the selection of stem-like cells that are not dependent on the expression of the drug targets for survival. Eradication of residual GIST cells and cure of GIST will likely require individualized combinations of several approaches tailored to the genetic makeup and other characteristics of the tumors.
Collapse
Affiliation(s)
- Tamas Ordog
- Center for Individualized Medicine, Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, and Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA;
| | - Martin Zörnig
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Yujiro Hayashi
- Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, and Enteric Neuroscience Program, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|