1
|
AlAsfoor S, Jessen E, Pullapantula SR, Voisin JR, Hsi LC, Pavelko KD, Farwana S, Patraw JA, Chai XY, Ji S, Strausbauch MA, Cipriani G, Wei L, Linden DR, Hou R, Myers R, Bhattarai Y, Wykosky J, Burns AJ, Dasari S, Farrugia G, Grover M. Mass cytometric analysis of circulating monocyte subsets in a murine model of diabetic gastroparesis. Am J Physiol Gastrointest Liver Physiol 2025; 328:G323-G341. [PMID: 39947648 DOI: 10.1152/ajpgi.00229.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/06/2024] [Accepted: 12/23/2024] [Indexed: 03/14/2025]
Abstract
Circulating monocytes (Mo) are precursors to a subset of gastric resident muscularis macrophages. Changes in muscularis macrophages (MMs) result in delayed gastric emptying (DGE) in diabetic gastroparesis. However, the dynamics of Mo in the development of DGE in an animal model are unknown. Using cytometry by time-of-flight and computational approaches, we show a high heterogeneity within the Mo population. In DGE mice, via unbiased clustering, we identified two reduced Mo clusters that exhibit migratory phenotype (Ly6ChiCCR2hi-intCD62LhiLy6GhiCD45RhiMERTKhiintLGALS3intCD14intCX3CR1lowSiglec-Hint-low) resembling classical Mo (CMo-like). All markers enriched in these clusters are known to regulate cell differentiation, proliferation, adhesion, and migration. Trajectory inference analysis predicted these Mo as precursors to subsequent Mo lineages. In gastric muscle tissue, we demonstrated an increase in the gene expression levels of chemokine receptor C-C chemokine receptor type 2 (Ccr2) and its C-C motif ligand 2 (Ccl2), suggesting increased trafficking of classical-Mo. These findings establish a link between two CMo-like clusters and the development of the DGE phenotype and contribute to a better understanding of the heterogenicity of the Mo population.NEW & NOTEWORTHY Using 32 immune cell surface markers, we identified 23 monocyte clusters in murine blood. Diabetic gastroparesis was associated with a significant decrease in two circulating classical monocyte-like clusters and an upregulation of the Ccr2-Ccl2 axis in the gastric muscularis propria, suggesting increased tissue monocyte migration. This study offers new targets by pointing to a possible role for two classical monocyte subsets connected to the Ccr2-Ccl2 axis.
Collapse
Affiliation(s)
- Shefaa AlAsfoor
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
| | - Erik Jessen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, United States
| | | | - Jennifer R Voisin
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
| | - Linda C Hsi
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
| | - Kevin D Pavelko
- Immune Monitoring Core, Office of Core Shared Services, Mayo Clinic, Rochester, Minnesota, United States
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States
| | - Samera Farwana
- Immune Monitoring Core, Office of Core Shared Services, Mayo Clinic, Rochester, Minnesota, United States
| | - Jack A Patraw
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
| | - Xin-Yi Chai
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
| | - Sihan Ji
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Neuroendocrine Pharmacology, School of Pharmacy, China Medical University, Shenyang, People's Republic of China
| | - Michael A Strausbauch
- Immune Monitoring Core, Office of Core Shared Services, Mayo Clinic, Rochester, Minnesota, United States
| | - Gianluca Cipriani
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
| | - Lai Wei
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
| | - David R Linden
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Ruixue Hou
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States
| | - Richard Myers
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., San Diego, California, United States
| | - Yogesh Bhattarai
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., San Diego, California, United States
| | - Jill Wykosky
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States
| | - Alan J Burns
- Gastrointestinal Drug Discovery Unit, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, United States
| | - Surendra Dasari
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, United States
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
| | - Madhusudan Grover
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
2
|
Ohishi K, Dora D, Han CY, Guyer RA, Ohkura T, Kazimierczyk S, Picard N, Leavitt AR, Ott LC, Rahman AA, Mueller JL, Shpigel NY, Jain N, Nagy N, Hotta R, Goldstein AM, Stavely R. Resolving Resident Colonic Muscularis Macrophage Diversity and Plasticity During Colitis. Inflamm Bowel Dis 2025; 31:151-168. [PMID: 39102823 PMCID: PMC11701110 DOI: 10.1093/ibd/izae155] [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: 12/21/2023] [Indexed: 08/07/2024]
Abstract
BACKGROUND Immune cell populations in the intestinal muscularis propria during colitis are poorly resolved. Maintaining homeostasis in this niche is critical, highlighted by the poorer prognosis of inflammatory bowel disease associated with muscularis propria inflammation. METHODS This study utilizes single-cell RNA sequencing to survey the immune cell populations within the muscularis propria of normal colon and dextran sodium sulfate-induced colitis. Findings are validated by immunohistochemistry, flow cytometry and cell-lineage tracing in vivo, and in vitro assays with muscularis macrophages (MMφ). RESULTS In naïve conditions, transcriptional duality is observed in MMφs with 2 major subpopulations: conventional resident Cx3cr1+ MMφs and Lyve1+ MMφs. The Lyve1+ population is phagocytic and expresses several known MMφ markers in mouse and human, confirming their identity as a bona fide MMφ subset. Single-cell transcriptomics indicate that resident MMφs are retained during colitis and exhibit plasticity toward an inflammatory profile. Lyve1+ MMφs, which express anti-inflammatory marker CD163, are absent during colitis, as confirmed by flow cytometry. In contrast, lineage tracing finds that resident Cx3cr1+ MMφs remain during colitis and are not completely replaced by the inflammatory infiltrating monocytes. In vitro studies provide biological evidence of the plasticity of resident Cx3cr1+ MMφs in response to lipopolysaccharide (LPS), mirroring transcriptional observations in vivo of their inflammatory plasticity. Potential markers for colitic MMφs, validated in animal models and in individuals with ulcerative colitis, are identified. CONCLUSIONS Our findings contribute to the understanding of the immune system in the muscularis propria niche during colitis by resolving the heterogeneity and origins of colitic MMφs.
Collapse
Affiliation(s)
- Kensuke Ohishi
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co., Ltd., Akitakata, Hiroshima, Japan
| | - David Dora
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Christopher Y Han
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Richard A Guyer
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Takahiro Ohkura
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon Kazimierczyk
- Mucosal Immunology and Biology Research Center, Mass General Hospital for Children, Charlestown, MA, USA
| | - Nicole Picard
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abigail R Leavitt
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leah C Ott
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ahmed A Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica L Mueller
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nahum Y Shpigel
- Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Nitya Jain
- Mucosal Immunology and Biology Research Center, Mass General Hospital for Children, Charlestown, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Nandor Nagy
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Ryo Hotta
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
Lou M, Heuckeroth RO, Tjaden NEB. Neuroimmune Crossroads: The Interplay of the Enteric Nervous System and Intestinal Macrophages in Gut Homeostasis and Disease. Biomolecules 2024; 14:1103. [PMID: 39334870 PMCID: PMC11430413 DOI: 10.3390/biom14091103] [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: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
A defining unique characteristic of the gut immune system is its ability to respond effectively to foreign pathogens while mitigating unnecessary inflammation. Intestinal macrophages serve as the cornerstone of this balancing act, acting uniquely as both the sword and shield in the gut microenvironment. The GI tract is densely innervated by the enteric nervous system (ENS), the intrinsic nervous system of the gut. Recent advances in sequencing technology have increasingly suggested neuroimmune crosstalk as a critical component for homeostasis both within the gut and in other tissues. Here, we systematically review the ENS-macrophage axis. We focus on the pertinent molecules produced by the ENS, spotlight the mechanistic contributions of intestinal macrophages to gut homeostasis and inflammation, and discuss both existing and potential strategies that intestinal macrophages use to integrate signals from the ENS. This review aims to elucidate the complex molecular basis governing ENS-macrophage signaling, highlighting their cooperative roles in sustaining intestinal health and immune equilibrium.
Collapse
Affiliation(s)
- Meng Lou
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute, Abramson Research Center and Department of Pediatrics, Pearlman School of Medicine at the University of Pennsylvania, 3615 Civic Center Blvd, Philadelphia, PA 19004, USA; (R.O.H.); (N.E.B.T.)
| | - Robert O. Heuckeroth
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute, Abramson Research Center and Department of Pediatrics, Pearlman School of Medicine at the University of Pennsylvania, 3615 Civic Center Blvd, Philadelphia, PA 19004, USA; (R.O.H.); (N.E.B.T.)
- Division of Gastroenterology, Nutrition and Hepatology, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19004, USA
| | - Naomi E. Butler Tjaden
- Department of Pediatrics, The Children’s Hospital of Philadelphia Research Institute, Abramson Research Center and Department of Pediatrics, Pearlman School of Medicine at the University of Pennsylvania, 3615 Civic Center Blvd, Philadelphia, PA 19004, USA; (R.O.H.); (N.E.B.T.)
- Division of Gastroenterology, Nutrition and Hepatology, The Children’s Hospital of Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19004, USA
| |
Collapse
|
4
|
Cingolani F, Balasubramaniam A, Srinivasan S. Molecular mechanisms of enteric neuropathies in high-fat diet feeding and diabetes. Neurogastroenterol Motil 2024:e14897. [PMID: 39119749 PMCID: PMC11807233 DOI: 10.1111/nmo.14897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/12/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Obesity and diabetes are associated with altered gastrointestinal function and with the development of abdominal pain, nausea, diarrhea, and constipation among other symptoms. The enteric nervous system (ENS) regulates gastrointestinal motility. Enteric neuropathies defined as damage or loss of enteric neurons can lead to motility disorders. PURPOSE Here, we review the molecular mechanisms that drive enteric neurodegeneration in diabetes and obesity, including signaling pathways leading to neuronal cell death, oxidative stress, and microbiota alteration. We also highlight potential approaches to treat enteric neuropathies including antioxidant therapy to prevent oxidative stress-induced damage and the use of stem cells.
Collapse
Affiliation(s)
- Francesca Cingolani
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA, United States
- Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Arun Balasubramaniam
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA, United States
- Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA, United States
- Atlanta Veterans Affairs Health Care System, Decatur, GA, United States
| |
Collapse
|
5
|
Maalouf E, Khasawneh H, Karbhari A, AlAsfoor S, Breen-Lyles M, Bernard C, Rajan E, Farrugia G, Lowe V, Goenka A, Grover M. Preliminary study on the dynamic positron emission tomography imaging with 11C-ER176 to delineate macrophage activation in diabetic gastroparesis. Neurogastroenterol Motil 2024; 36:e14762. [PMID: 38376247 DOI: 10.1111/nmo.14762] [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: 11/16/2023] [Revised: 01/24/2024] [Accepted: 02/01/2024] [Indexed: 02/21/2024]
Abstract
BACKGROUND Animal models and human data have suggested macrophage-driven immune dysregulation in diabetic gastroparesis (DG). Translocator protein (TSPO) upregulation has been suggested to indicate activated state of macrophages and ER176 is a high affinity third generation TSPO-specific radioligand. The aim of this study was to determine feasibility of dynamic 11C-ER 176 PET to identify macrophage activation in DG. METHODS Twelve patients, all females, were recruited (4 DG, 4 diabetics, and 4 healthy volunteers) for 11C-ER 176 PET/CT scanning. The standardized uptake value (SUVmax) in the gastric fundus, body, pylorus, and descending part of the duodenum were compared between three groups using Kruskal-Wallis test to perform the comparisons, and a p-value of 0.05 was considered statistically significant. KEY RESULTS Age was comparable among the three groups with a median of 53 years. The uptake was higher in pylorus in diabetics compared to DG and healthy (SUVmax healthy 4.6 ± 0.2, diabetics 8.4 ± 4.1, DG 5.5 ± 1.0, p = 0.04). The uptake was similar in gastric fundus (9.0 ± 1.6, 13.1 ± 8.3, 7.8 ± 1.9 respectively, p = 0.3), body (7.7 ± 1.9, 13 ± 9.2, 7.8 ± 1.9 respectively, p = 0.8), and duodenum (6.2 ± 2.1, 9.5 ± 6.8, 7.0 ± 1.8 respectively, p = 0.6). No correlation was observed between SUVmax uptake and either HbA1C or fasting blood glucose. CONCLUSIONS AND INFERENCES Female diabetic gastroparesis patients did not demonstrate increased TSPO ligand 11C-ER 176 uptake in the stomach. Possible explanations include lack of specificity of ligand for specific macrophage phenotypes in DG, sex effect, or small sample size. Further studies investigating non-invasive ways of analyzing immune dysregulation in neurogastrointestinal disorders are warranted.
Collapse
Affiliation(s)
- Elisia Maalouf
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hala Khasawneh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Radiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Aashna Karbhari
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Shefaa AlAsfoor
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Margaret Breen-Lyles
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Cheryl Bernard
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth Rajan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Val Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ajit Goenka
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
6
|
Zhang YX, Zhang YJ, Li M, Tian JX, Tong XL. Common Pathophysiological Mechanisms and Treatment of Diabetic Gastroparesis. J Neurogastroenterol Motil 2024; 30:143-155. [PMID: 38576367 PMCID: PMC10999838 DOI: 10.5056/jnm23100] [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: 07/06/2023] [Revised: 10/29/2023] [Accepted: 11/06/2023] [Indexed: 04/06/2024] Open
Abstract
Diabetic gastroparesis (DGP) is a common complication of diabetes mellitus, marked by gastrointestinal motility disorder, a delayed gastric emptying present in the absence of mechanical obstruction. Clinical manifestations include postprandial fullness and epigastric discomfort, bloating, nausea, and vomiting. DGP may significantly affect the quality of life and productivity of patients. Research on the relationship between gastrointestinal dynamics and DGP has received much attention because of the increasing prevalence of DGP. Gastrointestinal motility disorders are closely related to a variety of factors including the absence and destruction of interstitial cells of Cajal, abnormalities in the neuro-endocrine system and hormone levels. Therefore, this study will review recent literature on the mechanisms of DGP and gastrointestinal motility disorders as well as the development of prokinetic treatment of gastrointestinal motility disorders in order to give future research directions and identify treatment strategies for DGP.
Collapse
Affiliation(s)
- Yu-Xin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yan-Jiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Li
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia-Xing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao-Lin Tong
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Wang L, Hong W, Zhu H, He Q, Yang B, Wang J, Weng Q. Macrophage senescence in health and diseases. Acta Pharm Sin B 2024; 14:1508-1524. [PMID: 38572110 PMCID: PMC10985037 DOI: 10.1016/j.apsb.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/16/2023] [Accepted: 12/06/2023] [Indexed: 04/05/2024] Open
Abstract
Macrophage senescence, manifested by the special form of durable cell cycle arrest and chronic low-grade inflammation like senescence-associated secretory phenotype, has long been considered harmful. Persistent senescence of macrophages may lead to maladaptation, immune dysfunction, and finally the development of age-related diseases, infections, autoimmune diseases, and malignancies. However, it is a ubiquitous, multi-factorial, and dynamic complex phenomenon that also plays roles in remodeled processes, including wound repair and embryogenesis. In this review, we summarize some general molecular changes and several specific biomarkers during macrophage senescence, which may bring new sight to recognize senescent macrophages in different conditions. Also, we take an in-depth look at the functional changes in senescent macrophages, including metabolism, autophagy, polarization, phagocytosis, antigen presentation, and infiltration or recruitment. Furthermore, some degenerations and diseases associated with senescent macrophages as well as the mechanisms or relevant genetic regulations of senescent macrophages are integrated, not only emphasizing the possibility of regulating macrophage senescence to benefit age-associated diseases but also has an implication on the finding of potential targets or drugs clinically.
Collapse
Affiliation(s)
- Longling Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Wenxiang Hong
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong Zhu
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Taizhou Institute of Zhejiang University, Taizhou 318000, China
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Taizhou Institute of Zhejiang University, Taizhou 318000, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
8
|
Chikkamenahalli LL, Jessen E, Bernard CE, Ip WE, Breen-Lyles M, Cipriani G, Pullapantula SR, Li Y, AlAsfoor S, Wilson L, Koch KL, Kuo B, Shulman RJ, Chumpitazi BP, McKenzie TJ, Kellogg TA, Tonascia J, Hamilton FA, Sarosiek I, McCallum R, Parkman HP, Pasricha PJ, Abell TL, Farrugia G, Dasari S, Grover M. Single cell atlas of human gastric muscle immune cells and macrophage-driven changes in idiopathic gastroparesis. iScience 2024; 27:108991. [PMID: 38384852 PMCID: PMC10879712 DOI: 10.1016/j.isci.2024.108991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 11/17/2023] [Accepted: 01/17/2024] [Indexed: 02/23/2024] Open
Abstract
Gastrointestinal immune cells, particularly muscularis macrophages (MM) interact with the enteric nervous system and influence gastrointestinal motility. Here we determine the human gastric muscle immunome and its changes in patients with idiopathic gastroparesis (IG). Single cell sequencing was performed on 26,000 CD45+ cells obtained from the gastric tissue of 20 subjects. We demonstrate 11 immune cell clusters with T cells being most abundant followed by myeloid cells. The proportions of cells belonging to the 11 clusters were similar between IG and controls. However, 9/11 clusters showed 578-11,429 differentially expressed genes. In IG, MM had decreased expression of tissue-protective and microglial genes and increased the expression of monocyte trafficking and stromal activating genes. Furthermore, in IG, IL12 mediated JAK-STAT signaling involved in the activation of tissue-resident macrophages and Eph-ephrin signaling involved in monocyte chemotaxis were upregulated. Patients with IG had a greater abundance of monocyte-like cells. These data further link immune dysregulation to the pathophysiology of gastroparesis.
Collapse
Affiliation(s)
| | - Erik Jessen
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Cheryl E. Bernard
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - W.K. Eddie Ip
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Margaret Breen-Lyles
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - Gianluca Cipriani
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - Suraj R. Pullapantula
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - Ying Li
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Shefaa AlAsfoor
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - Laura Wilson
- Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | | | - Braden Kuo
- Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | - James Tonascia
- Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Frank A. Hamilton
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Irene Sarosiek
- Texas Tech University Health Sciences Center, El Paso, TX, USA
| | | | | | | | | | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
| | - the NIDDK Gastroparesis Clinical Research Consortium (GpCRC)
- Division of Gastroenterology and Hepatology, Enteric Neuroscience Program, Mayo Clinic, Rochester, MN, USA
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
- Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Wake Forest University, Winston-Salem, NC, USA
- Massachusetts General Hospital, Boston, MA, USA
- Baylor College of Medicine, Houston, TX, USA
- Duke University, Durham, NC, USA
- Department of Surgery, Mayo Clinic, Rochester, MN, USA
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
- Texas Tech University Health Sciences Center, El Paso, TX, USA
- Temple University, Philadelphia, PA, USA
- Mayo Clinic, Scottsdale, AZ, USA
- University of Louisville, Louisville, KY, USA
| |
Collapse
|
9
|
Zhou L, Lian H, Yin Y, Zheng YS, Han YX, Liu GQ, Wang ZY. New insights into muscularis macrophages in the gut: from their origin to therapeutic targeting. Immunol Res 2023; 71:785-799. [PMID: 37219708 DOI: 10.1007/s12026-023-09397-x] [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: 02/10/2023] [Accepted: 05/15/2023] [Indexed: 05/24/2023]
Abstract
Muscularis macrophages, as the most abundant immune cells in the intestinal muscularis externa, exhibit tissue protective phenotype in the steady state. Owing to tremendous advances in technology, we now know the fact that muscularis macrophages are a heterogeneous population of cells which could be divided into different functional subsets depending on their anatomic niches. There is emerging evidence showing that these subsets, through molecular interactions with their neighbours, take part in a wide range of physiological and pathophysiological processes in the gut. In this review, we summarize recent progress (particularly over the past 4 years) on distribution, morphology, origin and functions of muscularis macrophages and, where possible, the characteristics of specific subsets in response to the microenvironment they occupy, with particular emphasis on their role in muscular inflammation. Furthermore, we also integrate their role in inflammation-related gastrointestinal disorders, such as post-operative ileus and diabetic gastroparesis, in order to propose future therapeutic strategies.
Collapse
Affiliation(s)
- Li Zhou
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Hui Lian
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yue Yin
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yuan-Sheng Zheng
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yu-Xin Han
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Gao-Qi Liu
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Zhi-Yong Wang
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
- Xinxiang Key Laboratory of Molecular Neurology, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
10
|
Xu S, Liang S, Pei Y, Wang R, Zhang Y, Xu Y, Huang B, Li H, Li J, Tan B, Cao H, Guo S. TRPV1 Dysfunction Impairs Gastric Nitrergic Neuromuscular Relaxation in High-Fat Diet-Induced Diabetic Gastroparesis Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:548-557. [PMID: 36740184 DOI: 10.1016/j.ajpath.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023]
Abstract
Diabetic gastroparesis (DGP) is characterized by delayed gastric emptying of solid food. Nitrergic neuron-mediated fundus relaxation and intragastric peristalsis are pivotal for gastric emptying and are impaired in DGP. Transient receptor potential vanilloid 1 (TRPV1) ion channels are expressed in gastrointestinal vagal afferent nerves and have a potential role in relevant gastrointestinal disorders. In this study, mice with high-fat diet (HFD)-induced type 2 diabetes mellitus (T2DM), associated with gastroparesis, were used to determine the role of TRPV1 in DGP. After feeding with HFD, mice exhibited obesity, hyperglycemia, insulin resistance, and delayed gastric emptying. Cholinergic- and nitrergic neuron-mediated neuromuscular contractions and relaxation were impaired. The antral tone of the DGP mice was attenuated. Interestingly, activating or suppressing TRPV1 facilitated or inhibited gastric fundus relaxation in normal mice. These effects were neutralized by using a nitric oxide synthase (NOS) inhibitor. Activation or suppression of TRPV1 also increased or reduced NO release. TRPV1 was specifically localized with neuronal NOS in the gastric fundus. These data suggest that TRPV1 activation facilitates gastric fundus relaxation by regulating neuronal NOS and promoting NO release. However, these effects and mechanisms disappeared in mice with DGP induced by HFD diet. TRPV1 expression was only marginally decreased in the fundus of DGP mice. TRPV1 dysfunction may be a potential mechanism underlying the dysfunction of DGP gastric nitrergic neuromuscular relaxation.
Collapse
Affiliation(s)
- Siyuan Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China; Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Shaochan Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Pei
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yao Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yifei Xu
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bin Huang
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Haiwen Li
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Juanjuan Li
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bo Tan
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongying Cao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Shaoju Guo
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China.
| |
Collapse
|
11
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
12
|
Chavero-Pieres M, Viola MF, Appeltans I, Abdurahiman S, Gsell W, Matteoli G, Himmelreich U, Boeckxstaens G. Magnetic resonance imaging as a non-invasive tool to assess gastric emptying in mice. Neurogastroenterol Motil 2023; 35:e14490. [PMID: 36371706 PMCID: PMC10078537 DOI: 10.1111/nmo.14490] [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: 07/19/2022] [Revised: 10/04/2022] [Accepted: 10/18/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Methods to study gastric emptying in rodents are time consuming or terminal, preventing repetitive assessment in the same animal. Magnetic resonance imaging (MRI) is a non-invasive technique increasingly used to investigate gastrointestinal function devoid of these shortcomings. Here, we evaluated MRI to measure gastric emptying in control animals and in two different models of gastroparesis. METHODS Mice were scanned using a 9.4 Tesla MR scanner. Gastric volume was measured by delineating the stomach lumen area. Control mice were scanned every 30 min after ingestion of a 0.2 g meal and stomach volume was quantified. The ability of MRI to detect delayed gastric emptying was evaluated in models of morphine-induced gastroparesis and streptozotocin-induced diabetes. KEY RESULTS Magnetic resonance imaging reproducibly detected increased gastric volume following ingestion of a standard meal and progressively decreased with a half emptying time of 59 ± 5 min. Morphine significantly increased gastric volume measured at t = 120 min (saline: 20 ± 2 vs morphine: 34 ± 5 mm3 ; n = 8-10; p < 0.001) and increased half emptying time using the breath test (saline: 85 ± 22 vs morphine: 161 ± 46 min; n = 10; p < 0.001). In diabetic mice, gastric volume assessed by MRI at t = 60 min (control: 23 ± 2 mm3 ; n = 14 vs diabetic: 26 ± 5 mm3 ; n = 18; p = 0.014) but not at t = 120 min (control: 21 ± 3 mm3 ; n = 13 vs diabetic: 18 ± 5 mm3 ; n = 18; p = 0.115) was significantly increased compared to nondiabetic mice. CONCLUSIONS AND INFERENCES Our data indicate that MRI is a reliable and reproducible tool to assess gastric emptying in mice and represents a useful technique to study gastroparesis in disease models or for evaluation of pharmacological compounds.
Collapse
Affiliation(s)
- Marta Chavero-Pieres
- Laboratory for Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | - Maria Francesca Viola
- Laboratory for Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | - Iris Appeltans
- Laboratory for Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | - Saeed Abdurahiman
- Laboratory for Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI Unit, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | - Gianluca Matteoli
- Laboratory for Mucosal Immunology, Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI Unit, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Laboratory for Neuro-Immune Interaction, Translational Research Center for Gastrointestinal Disorders (TARGID), Department of Chronic Diseases and Metabolism (ChroMeta), KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Bishop ES, Namkoong H, Aurelian L, McCarthy M, Nallagatla P, Zhou W, Neshatian L, Gurland B, Habtezion A, Becker L. Age-dependent Microglial Disease Phenotype Results in Functional Decline in Gut Macrophages. GASTRO HEP ADVANCES 2022; 2:261-276. [PMID: 36908772 PMCID: PMC10003669 DOI: 10.1016/j.gastha.2022.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/15/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND AND AIMS Muscularis macrophages (MMs) are tissue-resident macrophages in the gut muscularis externa which play a supportive role to the enteric nervous system. We have previously shown that age-dependent MM alterations drive low-grade enteric nervous system inflammation, resulting in neuronal loss and disruption of gut motility. The current studies were designed to identify the MM genetic signature involved in these changes, with particular emphasis on comparison to genes in microglia, the central nervous system macrophage population involved in age-dependent cognitive decline. METHODS Young (3 months) and old (16-24 months) C57BL/6 mice and human tissue were studied. Immune cells from mouse small intestine, colon, and spinal cord and human colon were dissociated, immunophenotyped by flow cytometry, and examined for gene expression by single-cell RNA sequencing and quantitative real-time PCR. Phagocytosis was assessed by in vivo injections of pHrodo beads (Invitrogen). Macrophage counts were performed by immunostaining of muscularis whole mounts. RESULTS MMs from young and old mice express homeostatic microglial genes, including Gpr34, C1qc, Trem2, and P2ry12. An MM subpopulation that becomes more abundant with age assumes a geriatric state (GS) phenotype characterized by increased expression of disease-associated microglia genes including Cd9, Clec7a, Itgax (CD11c), Bhlhe40, Lgals3, IL-1β, and Trem2 and diminished phagocytic activity. Acquisition of the GS phenotype is associated with clearance of α-synuclein aggregates. Human MMs demonstrate a similar age-dependent acquisition of the GS phenotype associated with intracellular α-synuclein accumulation. CONCLUSION MMs demonstrate age-dependent genetic changes that mirror the microglial disease-associated microglia phenotype and result in functional decline.
Collapse
Affiliation(s)
- Estelle Spear Bishop
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California
| | - Hong Namkoong
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California
| | - Laure Aurelian
- Stanford University School of Medicine OFDD, Stanford, California
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Madison McCarthy
- Department of Surgery, Stanford University, Stanford, California
| | - Pratima Nallagatla
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Stanford, California
| | - Wenyu Zhou
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Stanford, California
- Department of Genetics, Stanford University, Stanford, California
| | - Leila Neshatian
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California
| | - Brooke Gurland
- Department of Surgery, Stanford University, Stanford, California
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California
| | - Laren Becker
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Stanford, California
| |
Collapse
|
14
|
Delfini M, Stakenborg N, Viola MF, Boeckxstaens G. Macrophages in the gut: Masters in multitasking. Immunity 2022; 55:1530-1548. [PMID: 36103851 DOI: 10.1016/j.immuni.2022.08.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/17/2022] [Accepted: 08/09/2022] [Indexed: 11/05/2022]
Abstract
The gastrointestinal tract has the important task of absorbing nutrients, a complex process that requires an intact barrier allowing the passage of nutrients but that simultaneously protects the host against invading microorganisms. To maintain and regulate intestinal homeostasis, the gut is equipped with one of the largest populations of macrophages in the body. Here, we will discuss our current understanding of intestinal macrophage heterogeneity and describe their main functions in the different anatomical niches of the gut during steady state. In addition, their role in inflammatory conditions such as infection, inflammatory bowel disease, and postoperative ileus are discussed, highlighting the roles of macrophages in immune defense. To conclude, we describe the interaction between macrophages and the enteric nervous system during development and adulthood and highlight their contribution to neurodegeneration in the context of aging and diabetes.
Collapse
Affiliation(s)
- Marcello Delfini
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Maria Francesca Viola
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium.
| |
Collapse
|
15
|
Mischopoulou M, D'Ambrosio M, Bigagli E, Luceri C, Farrugia G, Cipriani G. Role of Macrophages and Mast Cells as Key Players in the Maintenance of Gastrointestinal Smooth Muscle Homeostasis and Disease. Cell Mol Gastroenterol Hepatol 2022; 13:1849-1862. [PMID: 35245688 PMCID: PMC9123576 DOI: 10.1016/j.jcmgh.2022.02.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 12/18/2022]
Abstract
The gut contains the largest macrophage pool in the body, with populations of macrophages residing in the mucosa and muscularis propria of the gastrointestinal (GI) tract. Muscularis macrophages (MMs), which are located within the muscularis propria, interact with cells essential for GI function, such as interstitial cells of Cajal, enteric neurons, smooth muscle cells, enteric glia, and fibroblast-like cells, suggesting that these immune cells contribute to several aspects of GI function. This review focuses on the latest insights on the factors contributing to MM heterogeneity and the functional interaction of MMs with other cell types essential for GI function. This review integrates the latest findings on macrophages in other organs with increasing knowledge of MMs to better understand their role in a healthy and diseased gut. We describe the factors that contribute to (muscularis macrophage) MM heterogeneity, and the nature of MM interactions with cells regulating GI function. Finally, we also describe the increasing evidence suggesting a critical role of another immune cell type, the mast cell, in normal and diseased GI physiology.
Collapse
Affiliation(s)
| | - Mario D'Ambrosio
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Elisabetta Bigagli
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Cristina Luceri
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | | | | |
Collapse
|
16
|
Viola MF, Boeckxstaens G. Niche-specific functional heterogeneity of intestinal resident macrophages. Gut 2021; 70:1383-1395. [PMID: 33384336 PMCID: PMC8223647 DOI: 10.1136/gutjnl-2020-323121] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/22/2022]
Abstract
Intestinal resident macrophages are at the front line of host defence at the mucosal barrier within the gastrointestinal tract and have long been known to play a crucial role in the response to food antigens and bacteria that are able to penetrate the mucosal barrier. However, recent advances in single-cell RNA sequencing technology have revealed that resident macrophages throughout the gut are functionally specialised to carry out specific roles in the niche they occupy, leading to an unprecedented understanding of the heterogeneity and potential biological functions of these cells. This review aims to integrate these novel findings with long-standing knowledge, to provide an updated overview on our understanding of macrophage function in the gastrointestinal tract and to speculate on the role of specialised subsets in the context of homoeostasis and disease.
Collapse
Affiliation(s)
- Maria Francesca Viola
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Flanders, Belgium
| | - Guy Boeckxstaens
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (Chrometa), KU Leuven, Leuven, Flanders, Belgium
| |
Collapse
|
17
|
Song S, An J, Liu S. Electroacupuncture accelerates the delayed intestinal transit in POI by suppressing M1 like muscularis macrophages and IL6 secretion. Neurogastroenterol Motil 2021; 33:e14066. [PMID: 33483984 DOI: 10.1111/nmo.14066] [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: 08/10/2020] [Revised: 10/22/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Electroacupuncture (EA) at ST-36 could accelerate the delayed gastrointestinal (GI) motility in many GI motility dysfunction models, but the definite effect and mechanisms are unclear. In this study, we intended to investigate the effects of EA on intestinal manipulation (IM) mice model and involved mechanisms. METHODS Male C57BL/6 mice were randomized into five groups: normal control, intestinal manipulation (IM), IM with sham EA (SEA), IM with high-frequency EA (HEA), and IM with low-frequency EA (LEA). The GI transit was evaluated. The infiltration of muscularis macrophages (MMφ) and its phenotype were analyzed with flow cytometry. Magnetic-activated cell sorting was applied to isolate MMφ, and the relationship between the MMφ and interstitial cells of Cajal (ICCs) was further investigated. RESULTS (1) Compared with the IM group, HEA and LEA attenuated the delayed intestinal transit. (2) Both the HEA and LEA obviously reduced the MMφ and suppressed the M1 activation of the MMφ in the ileum. (3) EA restored the disrupted ICC networks through inhibiting the release of IL6 by the MMφ. CONCLUSION (1) Electroacupuncture at acupoint ST-36 could accelerate the delayed intestinal transit in the IM murine model by restoring the ICC networks. (2) EA protected the ICCs through reducing the MMφ, inhibiting its M1 polarization and its IL6 secretion.
Collapse
Affiliation(s)
- Shuangning Song
- Division of Gastroenterology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing An
- Division of Gastroenterology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shi Liu
- Division of Gastroenterology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Gottfried-Blackmore A, Namkoong H, Adler E, Martin B, Gubatan J, Fernandez-Becker N, Clarke JO, Idoyaga J, Nguyen L, Habtezion A. Gastric Mucosal Immune Profiling and Dysregulation in Idiopathic Gastroparesis. Clin Transl Gastroenterol 2021; 12:e00349. [PMID: 33979305 PMCID: PMC8132986 DOI: 10.14309/ctg.0000000000000349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/05/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION It is unclear how immune perturbations may influence the pathogenesis of idiopathic gastroparesis, a prevalent functional disorder of the stomach which lacks animal models. Several studies have noted altered immune characteristics in the deep gastric muscle layer associated with gastroparesis, but data are lacking for the mucosal layer, which is endoscopically accessible. We hypothesized that immune dysregulation is present in the gastroduodenal mucosa in idiopathic gastroparesis and that specific immune profiles are associated with gastroparesis clinical parameters. METHODS In this cross-sectional prospective case-control study, routine endoscopic biopsies were used for comprehensive immune profiling by flow cytometry, multicytokine array, and gene expression in 3 segments of the stomach and the duodenal bulb. Associations of immune endpoints with clinical parameters of gastroparesis were also explored. RESULTS The gastric mucosa displayed large regional variation of distinct immune profiles. Furthermore, several-fold increases in innate and adaptive immune cells were found in gastroparesis. Various immune cell types showed positive correlations with duration of disease, proton pump inhibitor dosing, and delayed gastric emptying. DISCUSSION This initial observational study showed immune compartmentalization of the human stomach mucosa and significant immune dysregulation at the level of leukocyte infiltration in idiopathic gastroparesis patients that extends to the duodenum. Select immune cells, such as macrophages, may correlate with clinicopathological traits of gastroparesis. This work supports further mucosal studies to advance our understanding of gastroparesis pathophysiology.
Collapse
Affiliation(s)
| | - Hong Namkoong
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - Emerald Adler
- Northwestern University Feinberg School of Medicine,
Division of Gastroenterology and Hepatology, Chicago, Illinois, USA
| | - Brock Martin
- Department of Pathology, Stanford University,
Stanford, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - Nielsen Fernandez-Becker
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - John O. Clarke
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, Stanford
University School of Medicine, Stanford, USA
| | - Linda Nguyen
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| | - Aida Habtezion
- Division of Gastroenterology and Hepatology,
Department of Medicine, Stanford University, Stanford, USA
| |
Collapse
|
19
|
Wang H, Zhao K, Ba Y, Gao T, Shi N, Niu Q, Liu C, Chen Y. Gastric Electrical Pacing Reduces Apoptosis of Interstitial Cells of Cajal via Antioxidative Stress Effect Attributing to Phenotypic Polarization of M2 Macrophages in Diabetic Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1298657. [PMID: 33728017 PMCID: PMC7937455 DOI: 10.1155/2021/1298657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 02/08/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Gastric electrical pacing (GEP) could restore interstitial cells of Cajal in diabetic rats. M2 macrophages contribute to the repair of interstitial cells of Cajal injury though secreting heme oxygenase-1 (HO-1). The aim of the study is to investigate the effects and mechanisms of gastric electrical pacing on M2 macrophages in diabetic models. METHODS Sixty male Sprague-Dawley rats were randomized into control, diabetic (DM), diabetic with the sham GEP (DM+SGEP), diabetic with GEP1 (5.5 cpm, 100 ms, 4 mA) (DM+GEP1), diabetic with GEP2 (5.5 cpm, 300 ms, 4 mA) (DM+GEP2), and diabetic with GEP3 (5.5 cpm, 550 ms, 4 mA) (DM+GEP3) groups. The apoptosis of interstitial cells of Cajal and the expression of macrophages were detected by immunofluorescence technique. The expression levels of the Nrf2/HO-1 and NF-κB pathway were evaluated using western blot analysis or immunohistochemical method. Malonaldehyde, superoxide dismutase, and reactive oxygen species were tested to reflect the level of oxidative stress. RESULTS Apoptosis of interstitial cells of Cajal was increased in the DM group but significantly decreased in the DM+GEP groups. The total number of macrophages was almost the same in each group. In the DM group, M1 macrophages were increased and M2 macrophages were decreased. However, M2 macrophages were dramatically increased and M1 macrophages were reduced in the DM+GEP groups. Gastric electrical pacing improved the Nrf2/HO-1 pathway and downregulated the phosphorylation of NF-κB. In the DM group, the levels of malonaldehyde and reactive oxygen species were elevated and superoxide dismutase was lowered, while gastric electrical pacing reduced the levels of malonaldehyde and reactive oxygen species and improved superoxide dismutase. CONCLUSION Gastric electrical pacing reduces apoptosis of interstitial cells of Cajal though promoting M2 macrophages polarization to play an antioxidative stress effect in diabetic rats, which associates with the activated Nrf2/HO-1 pathway and the phosphorylation of NF-κB pathway.
Collapse
Affiliation(s)
- Hongcai Wang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Kaile Zhao
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ying Ba
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Tao Gao
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ning Shi
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Qiong Niu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Chengxia Liu
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yan Chen
- Department of Gastroenterology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| |
Collapse
|
20
|
Chikkamenahalli LL, Pasricha PJ, Farrugia G, Grover M. Gastric Biopsies in Gastroparesis: Insights into Gastric Neuromuscular Disorders to Aid Treatment. Gastroenterol Clin North Am 2020; 49:557-570. [PMID: 32718570 PMCID: PMC7387746 DOI: 10.1016/j.gtc.2020.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The cellular and molecular understanding of human gastroparesis has markedly improved due to studies on full-thickness gastric biopsies. A decrease in the number of interstitial cells of Cajal (ICC) and functional changes in ICC constitutes the hallmark cellular feature of gastroparesis. More recently, in animal models, macrophages have also been identified to play a central role in development of delayed gastric emptying. Activation of macrophages leads to loss of ICC. In human gastroparesis, loss of anti-inflammatory macrophages in gastric muscle has been shown. Deeper molecular characterization using transcriptomics and proteomics has identified macrophage-based immune dysregulation in human gastroparesis.
Collapse
Affiliation(s)
- Lakshmikanth L. Chikkamenahalli
- Enteric NeuroScience Program, Mayo clinic, Division of Gastroenterology & Hepatology, Physiology & Biomedical Engineering Mayo Clinic, 200 1 Street SW, Rochester, MN 55905, Tel: +1 507-538-0337
| | - Pankaj J. Pasricha
- Center for Neurogastroenterology, Division of Gastroenterology & Hepatology Johns Hopkins School of Medicine, Ross 958, 720 Rutland Avenue, Baltimore, MD 21205, Tel: +1 443-613-8152
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology & Hepatology, Physiology & Biomedical Engineering Mayo Clinic, 200 1 Street SW, Rochester, MN 55905, Tel: +1 507-284-4695
| | - Madhusudan Grover
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA; Division of Physiology and Biomedical Engineering, Mayo Clinic, 200 1st Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
21
|
Viola MF, Boeckxstaens G. Intestinal resident macrophages: Multitaskers of the gut. Neurogastroenterol Motil 2020; 32:e13843. [PMID: 32222060 PMCID: PMC7757264 DOI: 10.1111/nmo.13843] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Intestinal resident macrophages play a crucial role in homeostasis and have been implicated in numerous gastrointestinal diseases. While historically believed to be largely of hematopoietic origin, recent advances in fate-mapping technology have unveiled the existence of long-lived, self-maintaining populations located in specific niches throughout the gut wall. Furthermore, the advent of single-cell technology has enabled an unprecedented characterization of the functional specialization of tissue-resident macrophages throughout the gastrointestinal tract. PURPOSE The purpose of this review was to provide a panorama on intestinal resident macrophages, with particular focus to the recent advances in the field. Here, we discuss the functions and phenotype of intestinal resident macrophages and, where possible, the functional specialization of these cells in response to the niche they occupy. Furthermore, we will discuss their role in gastrointestinal diseases.
Collapse
Affiliation(s)
- Maria Francesca Viola
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA)Laboratory for Neuro Immune InteractionTranslational Research in GastroIntestinal Disorders (TARGID)KU LeuvenLeuvenBelgium
| | - Guy Boeckxstaens
- Department of Chronic Diseases, Metabolism and Ageing (CHROMETA)Laboratory for Neuro Immune InteractionTranslational Research in GastroIntestinal Disorders (TARGID)KU LeuvenLeuvenBelgium
| |
Collapse
|
22
|
Pervez M, Ratcliffe E, Parsons SP, Chen JH, Huizinga JD. The cyclic motor patterns in the human colon. Neurogastroenterol Motil 2020; 32:e13807. [PMID: 32124528 DOI: 10.1111/nmo.13807] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/31/2019] [Accepted: 01/03/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND High-resolution colonic manometry gives an unprecedented window into motor patterns of the human colon. Our objective was to characterize motor activities throughout the entire colon that possessed persistent rhythmicity and spanning at least 5 cm. METHODS High-resolution colonic manometry using an 84-channel water-perfused catheter was performed in 19 healthy volunteers. Rhythmic activity was assessed during baseline, proximal balloon distention, meal, and bisacodyl administration. KEY RESULTS Throughout the entire colon, a cyclic motor pattern occurred either in isolation or following a high-amplitude propagating pressure wave (HAPW), consisting of clusters of pressure waves at a frequency centered on 11-13 cycles/min, unrelated to breathing. The cluster duration was 1-6 minutes; the pressure waves traveled for 8-27 cm, lasting 5-8 seconds. The clusters itself could be rhythmic at 0.5-2 cpm. The propagation direction of the individual pressure waves was mixed with >50% occurring simultaneous. This high-frequency cyclic motor pattern co-existed with the well-known low-frequency cyclic motor pattern centered on 3-4 cpm. In the rectum, the low-frequency cyclic motor pattern dominated, propagating predominantly in retrograde direction. Proximal balloon distention, a meal and bisacodyl administration induced HAPWs followed by cyclic motor patterns. CONCLUSIONS AND INFERENCES Within cyclic motor patterns, retrograde propagating, low-frequency pressure waves dominate in the rectum, likely keeping the rectum empty; and mixed propagation, high-frequency pressure waves dominate in the colon, likely promoting absorption and storage, hence contributing to continence. Propagation and frequency characteristics are likely determined by network properties of the interstitial cells of Cajal.
Collapse
Affiliation(s)
- Maham Pervez
- Department of Medicine, Division of Gastroenterology, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | | | - Sean P Parsons
- Department of Medicine, Division of Gastroenterology, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Ji-Hong Chen
- Department of Medicine, Division of Gastroenterology, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Jan D Huizinga
- Department of Medicine, Division of Gastroenterology, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW This review highlights recent work that will lead to near-term advances in the understanding and treatment of gastroparesis (Gp). RECENT FINDINGS Major current advancements in the pathophysiology of Gp, include recognition of the SIP syncytium as the pacemaking unit rather than ICC alone and that Gp may be part of a pan-enteric autoimmune and/or autonomic disorder with macrophage imbalance. The development of newer techniques to assess gastric emptying (gastric emptying breath test and wireless motility capsule) and pyloric distensibility (EndoFLIP®) are allowing clinicians better characterization of their patients. In addition to pharmaceutical compounds in the pipeline, neuromodulation and endosurgical techniques, such as G-POEM, may help address refractory Gp. We expect that the 2020 decade will witness exciting developments. Treatments targeting gastrointestinal motility, immunological dysfunction, and inflammatory mediators will be evaluated. We anticipate future studies will be guided by biomarkers correlated with patient outcomes and therapeutic efficacy to establish new paradigms in the management of Gp.
Collapse
|
24
|
Grover M, Farrugia G, Stanghellini V. Gastroparesis: a turning point in understanding and treatment. Gut 2019; 68:2238-2250. [PMID: 31563877 PMCID: PMC6874806 DOI: 10.1136/gutjnl-2019-318712] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 07/29/2019] [Accepted: 08/22/2019] [Indexed: 12/21/2022]
Abstract
Gastroparesis is defined by delayed gastric emptying (GE) and symptoms of nausea, vomiting, bloating, postprandial fullness, early satiety and abdominal pain. Most common aetiologies include diabetes, postsurgical and postinfectious, but in many cases it is idiopathic. Clinical presentation and natural history vary by the aetiology. There is significant morbidity and healthcare utilisation associated with gastroparesis. Mechanistic studies from diabetic animal models of delayed GE as well as human full-thickness biopsies have significantly advanced our understanding of this disorder. An innate immune dysregulation and injury to the interstitial cells of Cajal and other components of the enteric nervous system through paracrine and oxidative stress mediators is likely central to the pathogenesis of gastroparesis. Scintigraphy and 13C breath testing provide the most validated assessment of GE. The stagnant gastroparesis therapeutic landscape is likely to soon see significant changes. Relatively newer treatment strategies include antiemetics (aprepitant), prokinetics (prucalopride, relamorelin) and fundic relaxants (acotiamide, buspirone). Endoscopic pyloromyotomy appears promising over the short term, especially for symptoms of nausea and vomiting. Further controlled trials and identification of the appropriate subgroup with pyloric dysfunction and assessment of long-term outcomes are essential. This review highlights the clinical presentation, diagnosis, mechanisms and treatment advancements for gastroparesis.
Collapse
Affiliation(s)
- Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vincenzo Stanghellini
- Department of Digestive Diseases and Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
25
|
Grover M, Dasari S, Bernard CE, Chikkamenahalli LL, Yates KP, Pasricha PJ, Sarosiek I, McCallum R, Koch KL, Abell TL, Kuo B, Shulman RJ, Gibbons SJ, McKenzie TJ, Kellogg TA, Kendrick ML, Tonascia J, Hamilton FA, Parkman HP, Farrugia G. Proteomics in gastroparesis: unique and overlapping protein signatures in diabetic and idiopathic gastroparesis. Am J Physiol Gastrointest Liver Physiol 2019; 317:G716-G726. [PMID: 31482734 PMCID: PMC6879892 DOI: 10.1152/ajpgi.00115.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Macrophage-based immune dysregulation plays a critical role in development of delayed gastric emptying in diabetic mice. Loss of anti-inflammatory macrophages and increased expression of genes associated with pro-inflammatory macrophages has been reported in full-thickness gastric biopsies from gastroparesis patients. We aimed to determine broader protein expression (proteomics) and protein-based signaling pathways in gastric biopsies of diabetic (DG) and idiopathic gastroparesis (IG) patients. Additionally, we determined correlations between protein expressions, gastric emptying, and symptoms. Full-thickness gastric antrum biopsies were obtained from nine DG patients, seven IG patients, and five nondiabetic controls. Aptamer-based SomaLogic tissue scan that quantitatively identifies 1,305 human proteins was used. Protein fold changes were computed, and differential expressions were calculated using Limma. Ingenuity pathway analysis and correlations were carried out. Multiple-testing corrected P < 0.05 was considered statistically significant. Seventy-three proteins were differentially expressed in DG, 132 proteins were differentially expressed in IG, and 40 proteins were common to DG and IG. In both DG and IG, "Role of Macrophages, Fibroblasts and Endothelial Cells" was the most statistically significant altered pathway [DG false discovery rate (FDR) = 7.9 × 10-9; IG FDR = 6.3 × 10-12]. In DG, properdin expression correlated with GCSI bloating (r = -0.99, FDR = 0.02) and expressions of prostaglandin G/H synthase 2, protein kinase C-ζ type, and complement C2 correlated with 4 h gastric retention (r = -0.97, FDR = 0.03 for all). No correlations were found between proteins and symptoms or gastric emptying in IG. Protein expression changes suggest a central role of macrophage-driven immune dysregulation in gastroparesis, specifically, complement activation in diabetic gastroparesis.NEW & NOTEWORTHY This study uses SOMAscan, a novel proteomics assay for determination of altered proteins and associated molecular pathways in human gastroparesis. Seventy-three proteins were changed in diabetic gastroparesis, 132 in idiopathic gastroparesis compared with controls. Forty proteins were common in both. Macrophage-based immune dysregulation pathway was most significantly affected in both diabetic and idiopathic gastroparesis. Proteins involved in the complement and prostaglandin synthesis pathway were associated with symptoms and gastric emptying delay in diabetic gastroparesis.
Collapse
Affiliation(s)
| | - Surendra Dasari
- 2Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | | | | | - Katherine P. Yates
- 3Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | | | - Irene Sarosiek
- 5Texas Tech University Health Sciences Center, El Paso, Texas
| | | | | | | | - Braden Kuo
- 8Massachusetts General Hospital, Boston, Massachusetts
| | | | - Simon J. Gibbons
- 1Enteric NeuroScience Program, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - James Tonascia
- 3Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Frank A. Hamilton
- 11National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | | | | |
Collapse
|
26
|
Abstract
This review covers the epidemiology, pathophysiology, clinical features, diagnosis, and management of diabetic gastroparesis, and more broadly diabetic gastroenteropathy, which encompasses all the gastrointestinal manifestations of diabetes mellitus. Up to 50% of patients with type 1 and type 2 DM and suboptimal glycemic control have delayed gastric emptying (GE), which can be documented with scintigraphy, 13C breath tests, or a wireless motility capsule; the remainder have normal or rapid GE. Many patients with delayed GE are asymptomatic; others have dyspepsia (i.e., mild to moderate indigestion, with or without a mild delay in GE) or gastroparesis, which is a syndrome characterized by moderate to severe upper gastrointestinal symptoms and delayed GE that suggest, but are not accompanied by, gastric outlet obstruction. Gastroparesis can markedly impair quality of life, and up to 50% of patients have significant anxiety and/or depression. Often the distinction between dyspepsia and gastroparesis is based on clinical judgement rather than established criteria. Hyperglycemia, autonomic neuropathy, and enteric neuromuscular inflammation and injury are implicated in the pathogenesis of delayed GE. Alternatively, there are limited data to suggest that delayed GE may affect glycemic control. The management of diabetic gastroparesis is guided by the severity of symptoms, the magnitude of delayed GE, and the nutritional status. Initial options include dietary modifications, supplemental oral nutrition, and antiemetic and prokinetic medications. Patients with more severe symptoms may require a venting gastrostomy or jejunostomy and/or gastric electrical stimulation. Promising newer therapeutic approaches include ghrelin receptor agonists and selective 5-hydroxytryptamine receptor agonists.
Collapse
Affiliation(s)
- Adil E Bharucha
- Clinical Enteric Neuroscience Translational and Epidemiological Research Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Yogish C Kudva
- Division of Endocrinology. Mayo Clinic, Rochester, Minnesota
| | - David O Prichard
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
27
|
Herring BP, Chen M, Mihaylov P, Hoggatt AM, Gupta A, Nakeeb A, Choi JN, Wo JM. Transcriptome profiling reveals significant changes in the gastric muscularis externa with obesity that partially overlap those that occur with idiopathic gastroparesis. BMC Med Genomics 2019; 12:89. [PMID: 31221130 PMCID: PMC6587273 DOI: 10.1186/s12920-019-0550-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 06/07/2019] [Indexed: 12/13/2022] Open
Abstract
Background Gastric emptying is impaired in patients with gastroparesis whereas it is either unchanged or accelerated in obese individuals. The goal of the current study was to identify changes in gene expression in the stomach muscularis that may be contributing to altered gastric motility in idiopathic gastroparesis and obesity. Methods Quantitative real time RT-PCR and whole transcriptome sequencing were used to compare the transcriptomes of lean individuals, obese individuals and either lean or obese individuals with idiopathic gastroparesis. Results Obesity leads to an increase in mRNAs associated with muscle contractility whereas idiopathic gastroparesis leads to a decrease in mRNAs associated with PDGF BB signaling. Both obesity and idiopathic gastroparesis were also associated with similar alterations in pathways associated with inflammation. Conclusions Our findings show that obesity and idiopathic gastroparesis result in overlapping but distinct changes in the gastric muscularis transcriptome. Increased expression of mRNAs encoding smooth muscle contractile proteins may be contributing to the increased gastric motility observed in obese subjects, whereas decreased PDGF BB signaling may be contributing to the impaired motility seen in subjects with idiopathic gastroparesis. Electronic supplementary material The online version of this article (10.1186/s12920-019-0550-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- B Paul Herring
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA.
| | - Meng Chen
- Department of Oncology Research for Biologics and Immunotherapy Translation (ORBIT), MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Plamen Mihaylov
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - April M Hoggatt
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN, 46202, USA
| | - Anita Gupta
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Attila Nakeeb
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jennifer N Choi
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - John M Wo
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
28
|
Kong MM, Kong MM, Huang P, Jia CH. Therapeutic effect of Huangqi Jianzhong decoction on diabetic gastroparesis in rats: Impact on SCF-Kit signaling pathway in the gastric antrum. Shijie Huaren Xiaohua Zazhi 2019; 27:87-93. [DOI: 10.11569/wcjd.v27.i2.87] [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] [Indexed: 02/06/2023] Open
Abstract
AIM To analyze the therapeutic effect of Huangqi Jianzhong decoction on diabetic gastroparesis (DGP) in rats and its impact on the stem cell factor (SCF)-Kit signaling pathway in the gastric antrum.
METHODS Seventy-five male Wistar rats of SPF grade were randomly divided into five groups: model group, normal group, positive control group, low-dose Huangqi Jianzhong decoction group, and high-dose Huangqi Jianzhong decoction group (15 rats in each group). Except the normal group, DGP was induced in other groups. After DGP was induced, the rats in the positive control group were intra-gastrically administered with 0.78 g/mL of morphine suspension, and the two Huangqi Jianzhong decoction groups were intra-gastrically administered with 0.78 g/mL and 3.12 g/mL of Huangqi Jianzhong decoction. The normal group and the model group were given normal saline. The dose of each group was 10 mL/kg. After continuous administration for 6 wk, the general condition, body weight, gastric emptying rate, serum soluble SCF (sSCF), gastrin (GAS), motilin (MLT), and interstitial cells of Cajal (ICC) in the gastric antrum were observed. C-kit and membrane-bound SCF (mSCF) protein expression was detected.
RESULTS Compared with the normal group, the blood glucose, GAS, and MLT levels in the model group increased, and the sSCF content and gastric emptying rate decreased. Compared with the model group, positive control group, and low dose Huangqi Jianzhong decoction group, the contents of blood glucose, GAS, and MLT significantly decreased, and the sSCF content and gastric emptying rate significantly increased in the high-dose group (P < 0.05). Compared with the normal group, the expression of c-kit and mSCF proteins in the gastric antrum decreased in the model group. Compared with the model group, the expression of c-Kit and mSCF proteins in the positive control group, low-dose and high-dose Huangqi Jianzhong decoction groups significantly increased (P < 0.05). Compared with the positive control group, the expression of c-Kit and mSCF proteins in the low-dose and high-dose Huangqi Jianzhong decoction groups increased, but the difference was not statistically significant (P > 0.05).
CONCLUSION Huangqi Jianzhong decoction enhances the expression of mSCF protein and up-regulates the SCF-Kit signaling pathway, which increases the number of ICC in the gastric antrum of DGP rats, improves gastric pacing function, and enhances gastric motility.
Collapse
Affiliation(s)
- Meng-Meng Kong
- Zhejiang University of Traditional Chinese Medicine, Hangzhou Zhejiang Province, China
| | - Meng-Meng Kong
- Community Health Service Center, Gulou Street, Haishu District, Ningbo 315000, Zhejiang Province, China
| | - Ping Huang
- Second Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, Hangzhou 310053, Zhejiang Province, China
| | - Cai-Hua Jia
- Department of Pediatrics, Ninghe District Hospital, Tianjin 301500, China
| |
Collapse
|
29
|
Shen S, Xu J, Lamm V, Vachaparambil CT, Chen H, Cai Q. Diabetic Gastroparesis and Nondiabetic Gastroparesis. Gastrointest Endosc Clin N Am 2019; 29:15-25. [PMID: 30396524 DOI: 10.1016/j.giec.2018.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Gastroparesis can be divided into diabetic and nondiabetic, and the 3 main causes of gastroparesis are diabetic, postsurgical, and idiopathic. Delayed gastric emptying is the main manifestation of motility disorders for gastroparesis. Symptoms of gastroparesis are nonspecific and severity can vary. Nausea and vomiting are more common in diabetic gastroparesis whereas abdominal pain and early satiety are more frequent in idiopathic gastroparesis. Medication is still the mainstay of treatment of gastroparesis; however, the development of gastric electric stimulation and gastric peroral endoscopic pyloromyotomy brings more options for the treatment of diabetic and nondiabetic gastroparesis.
Collapse
Affiliation(s)
- Shanshan Shen
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Jennifer Xu
- Division of Digestive Diseases, Emory University, 1365 Clifton Road, Atlanta, GA 30322, USA
| | - Vladimir Lamm
- Division of Digestive Diseases, Emory University, 1365 Clifton Road, Atlanta, GA 30322, USA
| | - Cicily T Vachaparambil
- Division of Digestive Diseases, Emory University, 1365 Clifton Road, Atlanta, GA 30322, USA
| | - Huimin Chen
- Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai 200127, China
| | - Qiang Cai
- Division of Digestive Diseases, Emory University, 1365 Clifton Road, Atlanta, GA 30322, USA.
| |
Collapse
|
30
|
Grover M, Gibbons SJ, Nair AA, Bernard CE, Zubair AS, Eisenman ST, Wilson LA, Miriel L, Pasricha PJ, Parkman HP, Sarosiek I, McCallum RW, Koch KL, Abell TL, Snape WJ, Kuo B, Shulman RJ, McKenzie TJ, Kellogg TA, Kendrick ML, Tonascia J, Hamilton FA, Farrugia G. Transcriptomic signatures reveal immune dysregulation in human diabetic and idiopathic gastroparesis. BMC Med Genomics 2018; 11:62. [PMID: 30086735 PMCID: PMC6081936 DOI: 10.1186/s12920-018-0379-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/23/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cellular changes described in human gastroparesis have revealed a role for immune dysregulation, however, a mechanistic understanding of human gastroparesis and the signaling pathways involved are still unclear. METHODS Diabetic gastroparetics, diabetic non-gastroparetic controls, idiopathic gastroparetics and non-diabetic non-gastroparetic controls underwent full-thickness gastric body biopsies. Deep RNA sequencing was performed and pathway analysis of differentially expressed transcripts was done using Ingenuity®. A subset of differentially expressed genes in diabetic gastroparesis was validated in a separate cohort using QT-PCR. RESULTS 111 genes were differentially expressed in diabetic gastroparesis and 181 in idiopathic gastroparesis with a log2fold difference of | ≥ 2| and false detection rate (FDR) < 5%. Top canonical pathways in diabetic gastroparesis included genes involved with macrophages, fibroblasts and endothelial cells in rheumatoid arthritis, osteoarthritis pathway and differential regulation of cytokine production in macrophages and T helper cells by IL-17A and IL-17F. Top canonical pathways in idiopathic gastroparesis included genes involved in granulocyte adhesion and diapedesis, agranulocyte adhesion and diapedesis, and role of macrophages, fibroblasts and endothelial cells in rheumatoid arthritis. Sixty-five differentially expressed genes (log2fold difference | ≥ 2|, FDR < 5%) were common in both diabetic and idiopathic gastroparesis with genes in the top 5 canonical pathways associated with immune signaling. 4/5 highly differentially expressed genes (SGK1, APOLD1, CXCR4, CXCL2, and FOS) in diabetic gastroparesis were validated in a separate cohort of patients using RT-PCR. Immune profile analysis revealed that genes associated with M1 (pro inflammatory) macrophages were enriched in tissues from idiopathic gastroparesis tissues compared to controls (p < 0.05). CONCLUSIONS Diabetic and idiopathic gastroparesis have both unique and overlapping transcriptomic signatures. Innate immune signaling likely plays a central role in pathogenesis of human gastroparesis.
Collapse
Affiliation(s)
- Madhusudan Grover
- Enteric NeuroScience Program, Division of Gastroenterology & Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905 USA
| | - Simon J. Gibbons
- Enteric NeuroScience Program, Division of Gastroenterology & Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905 USA
| | - Asha A. Nair
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN USA
| | - Cheryl E. Bernard
- Enteric NeuroScience Program, Division of Gastroenterology & Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905 USA
| | - Adeel S. Zubair
- Enteric NeuroScience Program, Division of Gastroenterology & Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905 USA
| | - Seth T. Eisenman
- Enteric NeuroScience Program, Division of Gastroenterology & Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905 USA
| | - Laura A. Wilson
- Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD USA
| | - Laura Miriel
- Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD USA
| | | | | | | | | | | | | | | | - Braden Kuo
- Massachusetts General Hospital, Boston, MA USA
| | | | | | | | | | - James Tonascia
- Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD USA
| | - Frank A. Hamilton
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD USA
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology & Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905 USA
| |
Collapse
|
31
|
Cipriani G, Gibbons SJ, Miller KE, Yang DS, Terhaar ML, Eisenman ST, Ördög T, Linden DR, Gajdos GB, Szurszewski JH, Farrugia G. Change in Populations of Macrophages Promotes Development of Delayed Gastric Emptying in Mice. Gastroenterology 2018; 154:2122-2136.e12. [PMID: 29501441 PMCID: PMC5985210 DOI: 10.1053/j.gastro.2018.02.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS Muscularis propria macrophages lie close to cells that regulate gastrointestinal motor function, including interstitial cells of Cajal (ICC) and myenteric neurons. In animal models of diabetic gastroparesis, development of delayed gastric emptying has been associated with loss of macrophages that express cytoprotective markers and reduced networks of ICC. Mice with long-term diabetes and normal gastric emptying have macrophages that express anti-inflammatory markers and have normal gastric ICC. Mice homozygous for the osteopetrosis spontaneous mutation in the colony-stimulating factor 1 gene (Csf1op/op) do not have macrophages; when they are given streptozotocin to induce diabetes, they do not develop delayed gastric emptying. We investigated whether population of the gastric muscularis propria of diabetic Csf1op/op mice with macrophages is necessary to change gastric emptying, ICC, and myenteric neurons and investigated the macrophage-derived factors that determine whether diabetic mice do or do not develop delayed gastric emptying. METHODS Wild-type and Csf1op/op mice were given streptozotocin to induce diabetes. Some Csf1op/op mice were given daily intraperitoneal injections of CSF1 for 7 weeks; gastric tissues were collected and cellular distributions were analyzed by immunohistochemistry. CD45+, CD11b+, F4/80+ macrophages were dissociated from gastric muscularis propria, isolated by flow cytometry and analyzed by quantitative real-time polymerase chain reaction. Cultured gastric muscularis propria from Csf1op/op mice was exposed to medium that was conditioned by culture with bone marrow-derived macrophages from wild-type mice. RESULTS Gastric muscularis propria from Csf1op/op mice given CSF1 contained macrophages; 11 of 15 diabetic mice given CSF1 developed delayed gastric emptying and had damaged ICC. In non-diabetic Csf1op/op mice, administration of CSF1 reduced numbers of gastric myenteric neurons but did not affect the proportion of nitrergic neurons or ICC. In diabetic Csf1op/op mice given CSF1 that developed delayed gastric emptying, the proportion of nitrergic neurons was the same as in non-diabetic wild-type controls. Medium conditioned by macrophages previously exposed to oxidative injury caused damage to ICC in cultured gastric muscularis propria from Csf1op/op mice; neutralizing antibodies against IL6R or TNF prevented this damage to ICC. CD45+, CD11b+, and F4/80+ macrophages isolated from diabetic wild-type mice with delayed gastric emptying expressed higher levels of messenger RNAs encoding inflammatory markers (IL6 and inducible nitric oxide synthase) and lower levels of messenger RNAs encoding markers of anti-inflammatory cells (heme oxygenase 1, arginase 1, and FIZZ1) than macrophages isolated from diabetic mice with normal gastric emptying. CONCLUSIONS In studies of Csf1op/op and wild-type mice with diabetes, we found delayed gastric emptying to be associated with increased production of inflammatory factors, and reduced production of anti-inflammatory factors, by macrophages, leading to loss of ICC.
Collapse
|
32
|
The Investigation and Treatment of Diabetic Gastroparesis. Clin Ther 2018; 40:850-861. [PMID: 29748143 DOI: 10.1016/j.clinthera.2018.04.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/05/2018] [Accepted: 04/17/2018] [Indexed: 12/17/2022]
Abstract
PURPOSE This review provides an update on the investigations and treatment options for gastroparesis. METHODS A comprehensive literature search of Medline, PubMed, Embase and OVID was conducted which included all systematic reviews and research articles that focused on the diagnosis, investigations and management diabetic gastroparesis. FINDINGS Dietary modifications and pharmacologic treatment with prokinetics to increase gastric motility form the mainstay of treatment. However, the use of prokinetics is limited by adverse effects and serious adverse effects, leaving metoclopramide as the only drug approved by the US Food and Drug Administration for the treatment of gastroparesis. Newer therapies, including motilin receptor agonists, ghrelin receptor agonists, and neurokinin receptor antagonists, are currently being investigated. Transpyloric stenting, gastric electrical stimulation, and gastric per-oral endoscopic myotomy provide mechanical options for intervention, and surgical interventions in severe intractable gastroparesis include laparoscopic pyloroplasty or gastrectomy. IMPLICATIONS Advances to better understand the pathophysiology and management of diabetic gastroparesis have been limited, especially with discordance between symptoms and severity of delay in gastric emptying. Established treatment options are limited; however, recent pharmacologic and surgical interventions show promise.
Collapse
|
33
|
Becker L, Nguyen L, Gill J, Kulkarni S, Pasricha PJ, Habtezion A. Age-dependent shift in macrophage polarisation causes inflammation-mediated degeneration of enteric nervous system. Gut 2018; 67:827-836. [PMID: 28228489 PMCID: PMC5565713 DOI: 10.1136/gutjnl-2016-312940] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/21/2016] [Accepted: 01/27/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The enteric nervous system (ENS) undergoes neuronal loss and degenerative changes with age. The cause of this neurodegeneration is poorly understood. Muscularis macrophages residing in close proximity to enteric ganglia maintain neuromuscular function via direct crosstalk with enteric neurons and have been implicated in the pathogenesis of GI motility disorders like gastroparesis and postoperative ileus. The aim of this study was to assess whether ageing causes alterations in macrophage phenotype that contributes to age-related degeneration of the ENS. DESIGN Longitudinal muscle and myenteric plexus from small intestine of young, mid-aged and old mice were dissected and prepared for whole mount immunostaining, flow cytometry, Luminex immunoassays, western blot analysis, enteric neural stem cell (ENSC) isolation or conditioned media. Bone marrow derived macrophages were prepared and polarised to classic (M1) or alternative (M2) activation states. Markers for macrophage phenotype were measured using quantitative RT-PCR. RESULTS Ageing causes a shift in macrophage polarisation from anti-inflammatory 'M2' to proinflammatory 'M1' that is associated with a rise in cytokines and immune cells in the ENS. This phenotypic shift is associated with a neural response to inflammatory signals, increase in apoptosis and loss of enteric neurons and ENSCs, and delayed intestinal transit. An age-dependent decrease in expression of the transcription factor FoxO3, a known longevity gene, contributes to the loss of anti-inflammatory behaviour in macrophages of old mice, and FoxO3-deficient mice demonstrate signs of premature ageing of the ENS. CONCLUSIONS A shift by macrophages towards a proinflammatory phenotype with ageing causes inflammation-mediated degeneration of the ENS.
Collapse
Affiliation(s)
- Laren Becker
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California,Corresponding Authors Contact Information: Laren Becker, MD, PhD, Stanford University School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, 300 Pasteur Drive, Stanford, CA 94305, Tel: 650 721 1264, Fax: 650 723 5488, . Aida Habtezion, MD MSc, Stanford University School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, 300 Pasteur Drive, Stanford, CA 94305, Tel: 650 725 6511, Fax: 650 723 5488,
| | - Linh Nguyen
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California
| | - Jaspreet Gill
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California
| | - Subhash Kulkarni
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, Maryland
| | - Pankaj Jay Pasricha
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, Maryland
| | - Aida Habtezion
- Department of Medicine, Division of Gastroenterology and Hepatology, Stanford University, Stanford, California,Corresponding Authors Contact Information: Laren Becker, MD, PhD, Stanford University School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, 300 Pasteur Drive, Stanford, CA 94305, Tel: 650 721 1264, Fax: 650 723 5488, . Aida Habtezion, MD MSc, Stanford University School of Medicine, Department of Medicine, Division of Gastroenterology and Hepatology, 300 Pasteur Drive, Stanford, CA 94305, Tel: 650 725 6511, Fax: 650 723 5488,
| |
Collapse
|
34
|
Miller KE, Bajzer Ž, Hein SS, Phillips JE, Syed S, Wright AM, Cipriani G, Gibbons SJ, Szurszewski JH, Farrugia G, Ordog T, Linden DR. High temporal resolution gastric emptying breath tests in mice. Neurogastroenterol Motil 2018; 30:e13333. [PMID: 29575442 PMCID: PMC6157017 DOI: 10.1111/nmo.13333] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/11/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gastric emptying is a complex physiological process regulating the division of a meal into smaller partitions for the small intestine. Disrupted gastric emptying contributes to digestive disease, yet current measures may not reflect different mechanisms by which the process can be altered. METHODS We have developed high temporal resolution solid and liquid gastric emptying breath tests in mice using [13 C]-octanoic acid and off axis- integrated cavity output spectroscopy (OA-ICOS). Stretched gamma variate and 2-component stretched gamma variate models fit measured breath excretion data. KEY RESULTS These assays detect acceleration and delay using pharmacological (7.5 mg/kg atropine) or physiological (nutrients, cold exposure stress, diabetes) manipulations and remain stable over time. High temporal resolution resolved complex excretion curves with 2 components, which was more prevalent in mice with delayed gastric emptying following streptozotocin-induced diabetes. There were differences in the gastric emptying of Balb/c vs C57Bl6 mice, with slower gastric emptying and a greater occurrence of two-phase gastric emptying curves in the latter strain. Gastric emptying of C57Bl6 could be accelerated by halving the meal size, but with no effect on the occurrence of two-phase gastric emptying curves. A greater proportion of two-phase gastric emptying was induced in Balb/c mice with the administration of PYY (8-80 nmol) 60 min following meal ingestion. CONCLUSIONS AND INFERENCES Collectively, these results demonstrate the utility of high temporal resolution gastric emptying assays. Two-phase gastric emptying is more prevalent than previously reported, likely involves intestinal feedback, but contributes little to the overall rate of gastric emptying.
Collapse
Affiliation(s)
- Katie E. Miller
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
| | - Željko Bajzer
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic
College of Medicine, Rochester, MN 55905 USA
| | - Stephanie S. Hein
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
| | - Jessica E. Phillips
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
| | - Sabriya Syed
- Biochemistry and Molecular Biology Graduate Program, Mayo Clinic
College of Medicine, Rochester, MN 55905 USA
| | - Alec M. Wright
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
| | - Gianluca Cipriani
- Division of Gastroenterology and Hepatology, Department of Medicine,
Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - Simon J. Gibbons
- Division of Gastroenterology and Hepatology, Department of Medicine,
Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - Joseph H. Szurszewski
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
- Division of Gastroenterology and Hepatology, Department of Medicine,
Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - Gianrico Farrugia
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
- Division of Gastroenterology and Hepatology, Department of Medicine,
Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - Tamas Ordog
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
- Division of Gastroenterology and Hepatology, Department of Medicine,
Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - David R. Linden
- Department of Physiology and Biomedical Engineering and Enteric
NeuroScience Program, Mayo Clinic College of Medicine, Rochester MN 55905 USA
| |
Collapse
|
35
|
De Schepper S, Stakenborg N, Matteoli G, Verheijden S, Boeckxstaens GE. Muscularis macrophages: Key players in intestinal homeostasis and disease. Cell Immunol 2017; 330:142-150. [PMID: 29291892 PMCID: PMC6108422 DOI: 10.1016/j.cellimm.2017.12.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022]
Abstract
Muscularis macrophages densily colonize the outermost layer of the gastrointestinal tract. Muscularis macrophages communicate with enteric neurons in a bidirectional matter. Muscularis macrophages are tissue-protective but can contribute to disease. Current challenges are to decipher therapeutic potentials of muscularis macrophages.
Macrophages residing in the muscularis externa of the gastrointestinal tract are highly specialized cells that are essential for tissue homeostasis during steady-state conditions as well as during disease. They are characterized by their unique protective functional phenotype that is undoubtedly a consequence of the reciprocal interaction with their environment, including the enteric nervous system. This muscularis macrophage-neuron interaction dictates intestinal motility and promotes tissue-protection during injury and infection, but can also contribute to tissue damage in gastrointestinal disorders such as post-operative ileus and gastroparesis. Although the importance of muscularis macrophages is clearly recognized, different aspects of these cells remain largely unexplored such their origin, longevity and instructive signals that determine their function and phenotype. In this review, we will discuss the phenotype, functions and origin of muscularis macrophages during steady-state and disease conditions. We will highlight the bidirectional crosstalk with neurons and potential therapeutic strategies that target and manipulate muscularis macrophages to restore their protective signature as a treatment for disease.
Collapse
Affiliation(s)
- Sebastiaan De Schepper
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Nathalie Stakenborg
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Gianluca Matteoli
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Laboratory for Mucosal Immunology, University of Leuven, Leuven, Belgium.
| | - Simon Verheijden
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| | - Guy E Boeckxstaens
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID), Intestinal Neuro-immune Interactions, University of Leuven, Leuven, Belgium.
| |
Collapse
|
36
|
Grover M, Bernard CE, Pasricha PJ, Parkman HP, Gibbons SJ, Tonascia J, Koch KL, McCallum RW, Sarosiek I, Hasler WL, Nguyen LAB, Abell TL, Snape WJ, Kendrick ML, Kellogg TA, Hamilton FA, Farrugia G. Diabetic and idiopathic gastroparesis is associated with loss of CD206-positive macrophages in the gastric antrum. Neurogastroenterol Motil 2017; 29:10.1111/nmo.13018. [PMID: 28066953 PMCID: PMC5423829 DOI: 10.1111/nmo.13018] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/30/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Animal studies have increasingly highlighted the role of macrophages in the development of delayed gastric emptying. However, their role in the pathophysiology of human gastroparesis is unclear. Our aim was to determine changes in macrophages and other cell types in the gastric antrum muscularis propria of patients with diabetic and idiopathic gastroparesis. METHODS Full thickness gastric antrum biopsies were obtained from patients enrolled in the Gastroparesis Clinical Research Consortium (11 diabetic, 6 idiopathic) and 5 controls. Immunolabeling and quantitative assessment was done for interstitial cells of Cajal (ICC) (Kit), enteric nerves protein gene product 9.5, neuronal nitric oxide synthase, vasoactive intestinal peptide, substance P, tyrosine hydroxylase), overall immune cells (CD45) and anti-inflammatory macrophages (CD206). Gastric emptying was assessed using nuclear medicine scintigraphy and symptom severity using the Gastroparesis Cardinal Symptom Index. RESULTS Both diabetic and idiopathic gastroparesis patients showed loss of ICC as compared to controls (Mean [standard error of mean]/hpf: diabetic, 2.28 [0.16]; idiopathic, 2.53 [0.47]; controls, 6.05 [0.62]; P=.004). Overall immune cell population (CD45) was unchanged but there was a loss of anti-inflammatory macrophages (CD206) in circular muscle (diabetic, 3.87 [0.32]; idiopathic, 4.16 [0.52]; controls, 6.59 [1.09]; P=.04) and myenteric plexus (diabetic, 3.83 [0.27]; idiopathic, 3.59 [0.68]; controls, 7.46 [0.51]; P=.004). There was correlation between the number of ICC and CD206-positive cells (r=.55, P=.008). Enteric nerves (PGP9.5) were unchanged: diabetic, 33.64 (3.45); idiopathic, 41.26 (6.40); controls, 46.80 (6.04). CONCLUSION Loss of antral CD206-positive anti-inflammatory macrophages is a key feature in human gastroparesis and it is associates with ICC loss.
Collapse
Affiliation(s)
- M Grover
- Mayo Clinic, Enteric NeuroScience Program, Rochester, MN, USA
| | - CE Bernard
- Mayo Clinic, Enteric NeuroScience Program, Rochester, MN, USA
| | - PJ Pasricha
- Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - HP Parkman
- Temple University, Philadelphia, PA, USA
| | - SJ Gibbons
- Mayo Clinic, Enteric NeuroScience Program, Rochester, MN, USA
| | - J Tonascia
- Johns Hopkins University Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - KL Koch
- Wake Forest University, Winston-Salem, NC, USA
| | | | | | - WL Hasler
- University of Michigan, Ann Arbor, MI, USA
| | | | - TL Abell
- University of Louisville, Louisville, KY, USA
| | - WJ Snape
- California Pacific Medical Center, San Francisco, CA, USA
| | - ML Kendrick
- Mayo Clinic, Enteric NeuroScience Program, Rochester, MN, USA
| | - TA Kellogg
- Mayo Clinic, Enteric NeuroScience Program, Rochester, MN, USA
| | - FA Hamilton
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - G Farrugia
- Mayo Clinic, Enteric NeuroScience Program, Rochester, MN, USA
| | | | | |
Collapse
|
37
|
Eisenman ST, Gibbons SJ, Verhulst PJ, Cipriani G, Saur D, Farrugia G. Tumor necrosis factor alpha derived from classically activated "M1" macrophages reduces interstitial cell of Cajal numbers. Neurogastroenterol Motil 2017; 29:10.1111/nmo.12984. [PMID: 27781339 PMCID: PMC5367986 DOI: 10.1111/nmo.12984] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/20/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Delayed gastric emptying in diabetic mice and humans is associated with changes in macrophage phenotype and loss of interstitial cells of Cajal (ICC) in the gastric muscle layers. In diabetic mice, classically activated M1 macrophages are associated with delayed gastric emptying, whereas alternatively activated M2 macrophages are associated with normal gastric emptying. This study aimed to determine if secreted factors from M1 macrophages could injure mouse ICC in primary culture. METHODS Cultures of gastric ICC were treated with conditioned medium (CM) from activated bone marrow-derived macrophages (BMDMs) and the effect of CM was quantified by counting ICC per high-powered field. KEY RESULTS Bone marrow-derived macrophages were activated to a M1 or M2 phenotype confirmed by qRT-PCR. Conditioned medium from M1 macrophages reduced ICC numbers by 41.1%, whereas M2-CM had no effect as compared to unconditioned, control media. Immunoblot analysis of 40 chemokines/cytokines found 12 that were significantly increased in M1-CM, including tumor necrosis factor alpha (TNF-α). ELISA detected 0.697±0.03 ng mL-1 TNF-α in M1-CM. Recombinant mouse TNF-α reduced Kit expression and ICC numbers in a concentration-dependent manner (EC50 = 0.817 ng mL-1 ). Blocking M1-CM TNF-α with a neutralizing antibody preserved ICC numbers. The caspase inhibitor Z-VAD.fmk partly preserved ICC numbers (cells/field; 6.63±1.04, 9.82±1.80 w/Z-VAD.fmk, n=6, P<.05). CONCLUSIONS & INFERENCES This work demonstrates that TNF-α secreted from M1 macrophages can result in Kit loss and directly injure ICC in vitro partly through caspase-dependent apoptosis and may play an important role in ICC depletion in diabetic gastroparesis.
Collapse
Affiliation(s)
| | | | | | | | - Dieter Saur
- Department of Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | | |
Collapse
|
38
|
Choi KM, Gibbons SJ, Sha L, Beyder A, Verhulst PJ, Cipriani G, Phillips JE, Bauer AJ, Ordog T, Camp JJ, Ge X, Bharucha AE, Linden DR, Szurszewski JH, Kashyap PC, Farrugia G. Interleukin 10 Restores Gastric Emptying, Electrical Activity, and Interstitial Cells of Cajal Networks in Diabetic Mice. Cell Mol Gastroenterol Hepatol 2016; 2:454-467. [PMID: 27795979 PMCID: PMC5042607 DOI: 10.1016/j.jcmgh.2016.04.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 04/17/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Gastroparesis is a complication of diabetes characterized by delayed emptying of stomach contents and accompanied by early satiety, nausea, vomiting, and pain. No safe and reliable treatments are available. Interleukin 10 (IL10) activates the M2 cytoprotective phenotype of macrophages and induces expression of heme oxygenase 1 (HO1) protein. We investigated whether IL10 administration could improve gastric emptying and reverse the associated cellular and electrical abnormalities in diabetic mice. METHODS Nonobese diabetic mice with delayed gastric emptying were given either IL10 (0.1-1 μg, twice/day) or vehicle (controls). Stomach tissues were isolated, and sharp microelectrode recordings were made of the electrical activity in the gastric muscle layers. Changes to interstitial cells of Cajal (ICC), reduced nicotinamide adenine dinucleotide phosphate diaphorase, and levels and distribution of HO1 protein were determined by histochemical and imaging analyses of the same tissues. RESULTS Gastric emptying remained delayed in vehicle-treated diabetic mice but returned to normal in mice given IL10 (n = 10 mice; P < .05). In mice given IL10, normalization of gastric emptying was associated with a membrane potential difference between the proximal and distal stomach, and lower irregularity and higher frequency of slow-wave activity, particularly in the distal stomach. Levels of HO1 protein were higher in stomach tissues from mice given IL10, and ICC networks were more organized, better connected, and more evenly distributed compared with controls. CONCLUSIONS IL10 increases gastric emptying in diabetic mice and has therapeutic potential for patients with diabetic gastroparesis. This response is associated with up-regulation of HO1 and repair of connectivity of ICC networks.
Collapse
Affiliation(s)
- Kyoung Moo Choi
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Simon J. Gibbons
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Lei Sha
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Arthur Beyder
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Pieter-Jan Verhulst
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Gianluca Cipriani
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Jessica E. Phillips
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Anthony J. Bauer
- Department of Integrative Physiology and Pharmacology, Liberty University College of Osteopathic Medicine, Lynchburg, Virginia
| | - Tamas Ordog
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Jon J. Camp
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Xin Ge
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Adil E. Bharucha
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - David R. Linden
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | | | - Purna C. Kashyap
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Mayo Clinic School of Medicine, Rochester, Minnesota,Correspondence Address correspondence to: Gianrico Farrugia, MD, Enteric NeuroScience Program, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905. fax: (507) 284–0266.Enteric NeuroScience ProgramMayo Clinic200 First Street SWRochesterMinnesota 55905
| |
Collapse
|
39
|
Cipriani G, Gibbons SJ, Kashyap PC, Farrugia G. Intrinsic Gastrointestinal Macrophages: Their Phenotype and Role in Gastrointestinal Motility. Cell Mol Gastroenterol Hepatol 2016; 2:120-130.e1. [PMID: 27047989 PMCID: PMC4817106 DOI: 10.1016/j.jcmgh.2016.01.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
There is an increasing awareness of the role of macrophages in the regulation and maintenance of gastrointestinal function in health and disease. This work has proceeded in the context of an increased understanding of the complex phenotypic variation in macrophages throughout the body and has revealed previously un-identified roles for macrophages in diseases like gastroparesis, post-operative ileus and inflammatory bowel disease. Opportunities for exploiting the phenotypic modulation of tissue resident macrophages have been identified as possible therapies for some of these diseases. In addition, macrophages are an established component of the innate immune system that can respond to variations and changes in the intestinal microbiome and potentially mediate part of the impact of the microbiota on intestinal health. We reviewed the latest work on novel concepts in defining macrophage phenotype, discuss possible mechanisms of action for tissue-resident macrophages in the gut, address the significance of microbiome effects on macrophage phenotype and review the known and possible roles of macrophages in motility disorders of the gastrointestinal tract.
Collapse
Affiliation(s)
- Gianluca Cipriani
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, 55905, USA
| | - Simon J Gibbons
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, 55905, USA
| | - Purna C Kashyap
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, 55905, USA
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, 55905, USA
| |
Collapse
|
40
|
Affiliation(s)
- Shanthi Srinivasan
- Correspondence Address correspondence to: Shanthi Srinivasan, MD, Department of Digestive Diseases, Emory University School of Medicine, RM 1C-174, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, Georgia 30033.Department of Digestive DiseasesEmory University School of MedicineRM 1C-174, Atlanta VA Medical Center1670 Clairmont RoadDecaturGeorgia 30033
| |
Collapse
|