1
|
Van Doren VE, Ackerley CG, Arthur RA, Murray PM, Smith SA, Hu YJ, Kelley CF. Rectal mucosal inflammation, microbiome, and wound healing in men who have sex with men who engage in receptive anal intercourse. Sci Rep 2024; 14:31598. [PMID: 39738273 DOI: 10.1038/s41598-024-80074-1] [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: 08/06/2024] [Accepted: 11/14/2024] [Indexed: 01/01/2025] Open
Abstract
Mucosal injury is common during consensual intercourse and induces an inflammatory response that could contribute to pathogen transmission including HIV. Here, we compared mucosal immune and microbiome responses to experimentally induced mucosal injury between men who have sex with men engaging in receptive anal intercourse (MSM-RAI) and men who do not engage in RAI (controls), all without HIV. Rectal mucosal secretions were collected from adult MSM-RAI (n = 19) and controls (n = 6) via anoscopy before and up to eight days after experimentally induced injury. Mucosal healing was evaluated by repeated injury surface area measurements with digital imaging. MSM-RAI demonstrated overall significantly higher concentrations of pro-inflammatory cytokines and a distinct rectal microbiome compared with controls. Wound healing was numerically faster in MSM-RAI but did not meet statistical significance (p = 0.09). Different cytokine injury response patterns were observed between MSM-RAI and controls; however, IL-6 and IP-10 were important mediators in both groups. Microbial guilds, particularly from the Lachnospiraceae and Prevotellaceae families, were associated with rectal mucosal inflammation. This work is the first experimental study of rectal mucosal injury and the immune environment in healthy humans and provides a more nuanced understanding of rectal mucosal inflammation after injury, which can inform our understanding of HIV transmission.
Collapse
Affiliation(s)
- Vanessa E Van Doren
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court #200, 30030, Decatur, Georgia, United States.
| | - Cassie G Ackerley
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court #200, 30030, Decatur, Georgia, United States
| | - Robert A Arthur
- Emory Integrated Computational Core, Emory University, Woodruff Memorial Research Building, Suite 7110, 101 Woodruff Circle, 30322, Atlanta, Georgia, United States
| | - Phillip M Murray
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court #200, 30030, Decatur, Georgia, United States
| | - S Abigail Smith
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court #200, 30030, Decatur, Georgia, United States
| | - Yi-Juan Hu
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, 1518 Clifton Road, 30322, Atlanta, Georgia, United States
| | - Colleen F Kelley
- The Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, 500 Irvin Court #200, 30030, Decatur, Georgia, United States
- Grady Health System, 80 Jesse Hill Jr Drive, 30303, Atlanta, Georgia, United States
| |
Collapse
|
2
|
Zhang W, Pan L, Wu X, Slivano OJ, Dong K, Long X. Functional characterization of human IL-8 in vascular stenosis using a novel humanized transgenic mouse model. Vascul Pharmacol 2024; 157:107438. [PMID: 39486776 DOI: 10.1016/j.vph.2024.107438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
IL-8 (aka interleukin 8, CXCL8) is a prototypic cytokine that is highly expressed in the diseased vessel wall and its plasma concentration is strongly associated with cardiovascular events. However, whether IL-8 plays a causative role in cardiovascular diseases remains largely unknown. In this study we used a human IL-8 transgenic (Tg) mouse strain with a bacterial artificial chromosome (BAC) integrated into its genome. This BAC encompasses 166 kb of sequence encompassing the human IL-8 gene locus as well as upstream and downstream DNA sequences containing regulatory elements. This BAC ensured a pathophysiologically regulated, rather than forced constitutive, expression of human IL-8 in the mouse. Tg mice were subjected to complete carotid ligation injury. IL-8 was highly expressed in the ligation-injured carotid artery from 3 days until 2 weeks after injury. As a result, exacerbated neointimal hyperplasia and increased Mac2 and PCNA positive cells were observed in Tg mice. To further confirm its role in promoting neointimal formation, IL-8 was neutralized by anti-IL8 treatment at the ligation site. Consequently, the size of neointima was significantly reduced. Our results provided new insights into the regulation and function of IL-8 in response to vascular insult and during neointima formation.
Collapse
Affiliation(s)
- Wei Zhang
- Lemole Center for Integrated Lymphatic and Vascular Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Lihua Pan
- Lemole Center for Integrated Lymphatic and Vascular Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA; Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoliang Wu
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Orazio J Slivano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Kunzhe Dong
- Immunology Center of Georgia, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
3
|
Chen Q, Chen T, Xiao H, Wang F, Li C, Hu N, Bao L, Tong X, Feng Y, Xu Y, Li C, Zhu JW, Wang D, Li MX. APEX1 in intestinal epithelium triggers neutrophil infiltration and intestinal barrier damage in ulcerative colitis. Free Radic Biol Med 2024; 225:359-373. [PMID: 39389211 DOI: 10.1016/j.freeradbiomed.2024.10.260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Ulcerative colitis (UC) can lead to the generation of large amounts of reactive oxygen species and DNA damage. DNA repair caused by base excision repair (BER) enzymes is an important mechanism for maintaining genomic integrity. However, the specific relationship between the function of BER enzymes and UC remains unclear. To address this, we conducted a study on non-cancerous colon tissue from patients with UC, focusing on the role of apurinic/apyrimidinic endonuclease 1 (APEX1) in BER to explore its significance in the progression of UC. Our research found that the expression of APEX1 in epithelium cells was significantly correlated to the severity of inflammatory bowel disease (IBD) and the infiltration and function of neutrophils in human UC and mouse models, particularly in relation to neutrophil extracellular traps (NETs) and the degranulation processes. APEX1 deficiency resulted in decreased production of the chemokines CXCL1 by the NF-κB pathway in epithelium cells, leading to reduced accumulation and activation of neutrophils associated with colitis in colon tissue, as well as decreased levels of IL-1β. Furthermore, APEX1 deficiency reduced symptoms of colitis by decreasing epithelial cell apoptosis and altering the gut microbiome. Studies related to the redox activity of APEX1 have shown that the combination of the redox inhibitor E3330 with 5-aminosalicylic acid (5-ASA) can effectively alleviate colitis, indicating that APEX1 has promising prospects for clinical treatment of IBD. APEX1 is required for interactions between neutrophil and intestinal epithelial cells. This study provided a mechanism demonstrating that APEX1 protein triggered the risk of UC by promoting neutrophil infiltration and compromising intestinal epithelial barrier function.
Collapse
Affiliation(s)
- Qian Chen
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China.
| | - TianYi Chen
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - He Xiao
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Fangjie Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Army Medical Center (Daping Hospital), Army Medical University, Chongqing, 400042, China
| | - ChaoFan Li
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Nana Hu
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China; Yu-Yue Pathology Scientific Research Center, Choongqing, China
| | - Lingbo Bao
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Xueling Tong
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Yan Feng
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Yu Xu
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - ChunXue Li
- Department of General Surgery, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Jian Wu Zhu
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China
| | - Dong Wang
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China.
| | - Meng Xia Li
- Cancer Center of Daping Hospital Army Medical University, Chongqing 400042, China.
| |
Collapse
|
4
|
Yao Y, Shang W, Bao L, Peng Z, Wu C. Epithelial-immune cell crosstalk for intestinal barrier homeostasis. Eur J Immunol 2024; 54:e2350631. [PMID: 38556632 DOI: 10.1002/eji.202350631] [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: 12/19/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
The intestinal barrier is mainly formed by a monolayer of epithelial cells, which forms a physical barrier to protect the gut tissues from external insults and provides a microenvironment for commensal bacteria to colonize while ensuring immune tolerance. Moreover, various immune cells are known to significantly contribute to intestinal barrier function by either directly interacting with epithelial cells or by producing immune mediators. Fulfilling this function of the gut barrier for mucosal homeostasis requires not only the intrinsic regulation of intestinal epithelial cells (IECs) but also constant communication with immune cells and gut microbes. The reciprocal interactions between IECs and immune cells modulate mucosal barrier integrity. Dysregulation of barrier function could lead to dysbiosis, inflammation, and tumorigenesis. In this overview, we provide an update on the characteristics and functions of IECs, and how they integrate their functions with tissue immune cells and gut microbiota to establish gut homeostasis.
Collapse
Affiliation(s)
- Yikun Yao
- Shanghai Institute of Nutrition & Health, Chinese Academy of Science, Shanghai, China
| | - Wanjing Shang
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lingyu Bao
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Zhaoyi Peng
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Koduri MA, Pingali T, Prasad D, Singh V, Singh S, Shanbhag SS, Basu S, Singh V. Neutrophil-driven and interleukin-36γ-associated ocular surface inflammation in chronic Stevens-Johnson syndrome. Allergy 2024. [PMID: 38682250 DOI: 10.1111/all.16126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 05/01/2024]
Abstract
PURPOSE This study aims to elucidate the tear proteome and understand the underlying molecular mechanisms involved in the ocular complications following Stevens-Johnson syndrome/toxic epidermal necrolysis (SJS/TEN). METHODS Mass spectrometry (MS) was performed to quantify the tear fluid proteins from chronic SJS/TEN patients (n = 22 eyes) and age- and gender-matched controls (n = 22 eyes). The candidate proteins were validated using ELISA (n = 80 eyes) in tear samples and immunohistochemistry (IHC; n = 12) in eyelid margin specimens. These proteins were compared for significant differences based on age, gender, disease duration, and ocular severity. RESULTS A total of 1692 tear fluid proteins were identified, of which 470 were significantly differentially regulated in chronic SJS/TEN. The top 10 significantly upregulated proteins were neutrophil secretions including neutrophil elastase (p < .0001), defensin (p < .0001), and matrix metalloproteinase 8 (p < .0001). The presence of neutrophils was confirmed by the upregulation of IL-8 (p < .001) in tears, a key cytokine known for recruiting neutrophils. Additionally, positive expression of myeloperoxidase was observed in the keratinized eyelid margins of SJS/TEN to validate the presence of neutrophils. Among 41 unique proteins identified by MS, IL-36γ (p < .01) was expressed in three SJS/TEN patients and was confirmed in SJS/TEN tears and eyelid margins by ELISA and IHC, respectively. IL-36γ was specifically expressed in the superficial layers of eyelid margin keratinized conjunctiva. The majority of the significantly downregulated proteins were lacrimal gland secretions such as lacritin (p < .0001) and opiorphin (p < .002). Neutrophil elastase (p < .02) was significantly elevated in patients with severe eyelid margin keratinization. CONCLUSION Our observations indicate a clear correlation between eyelid margin keratinization and the expression of IL-36γ, potentially mediated by neutrophils recruited via IL-8. Future experimental studies are needed to test the role of therapies targeting IL-8 and/or IL-36γ in reducing eyelid margin keratinization and its associated ocular complications in SJS/TEN.
Collapse
Affiliation(s)
- Madhuri Amulya Koduri
- Centre for Ocular Regeneration (CORE), Prof. Brien Holden Eye Research Centre (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Tejaswini Pingali
- Centre for Ocular Regeneration (CORE), Prof. Brien Holden Eye Research Centre (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Deeksha Prasad
- Centre for Ocular Regeneration (CORE), Prof. Brien Holden Eye Research Centre (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Vijay Singh
- Centre for Ocular Regeneration (CORE), Prof. Brien Holden Eye Research Centre (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Swati Singh
- Centre for Ocular Regeneration (CORE), Prof. Brien Holden Eye Research Centre (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Swapna S Shanbhag
- The Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Sayan Basu
- Centre for Ocular Regeneration (CORE), Prof. Brien Holden Eye Research Centre (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
- The Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Vivek Singh
- Centre for Ocular Regeneration (CORE), Prof. Brien Holden Eye Research Centre (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
- The Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
| |
Collapse
|
6
|
Song YJ, Liu HX, Yang XG. The deposition of lanthanum carbonate may activate macrophages to induce gastrointestinal mucosal injury in patients with chronic kidney disease: an in vitro caco-2/THP-1 macrophage coculture model study. J Biol Inorg Chem 2024; 29:101-112. [PMID: 38148422 DOI: 10.1007/s00775-023-02033-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: 03/02/2023] [Accepted: 09/11/2023] [Indexed: 12/28/2023]
Abstract
The aim of this study was to investigate the effect and possible underlying mechanism of La2(CO3)3 deposition on GI mucosal inflammation. Our results showed that La2(CO3)3 can dissolve in artificial gastric fluids and form lanthanum phosphate (LaPO4) precipitates with an average size of about 1 μm. To mimic the intestinal mucosa and epithelial barrier, we established a Caco-2/THP-1 macrophage coculture model and a Caco-2 monoculture model, respectively. Our findings demonstrated that the medium of THP-1 macrophages stimulated by LaPO4 particles can damage the Caco-2 monolayer integrity in the coculture model, while the particles themselves had no direct impact on the Caco-2 monolayer integrity in the monoculture model. We measured values of trans-epithelial electrical resistance and detected images using a laser scanning confocal microscope. These results indicate that continuous stimulation of LaPO4 particles on macrophages can lead to a disruption of intestinal epithelium integrity. In addition, LaPO4 particles could stimulate THP-1 macrophages to secrete both IL-1β and IL-8. Although LaPO4 particles can also promote Caco-2 cells to secrete IL-8, the secretion was much lower than that produced by THP-1 macrophages. In summary, the deposition of La2(CO3)3 has been shown to activate macrophages and induce damage to intestinal epithelial cells, which may exacerbate inflammation in patients with chronic kidney disease. Therefore, patients taking lanthanum carbonate, especially those with gastrointestinal mucosal inflammation, should be mindful of the potential for drug deposition in the GI system.
Collapse
Affiliation(s)
- Ya-Ju Song
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Hui-Xue Liu
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Xiao-Gai Yang
- Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
7
|
Park SW, Choi YH, Gho JY, Kang GA, Kang SS. Synergistic Inhibitory Effect of Lactobacillus Cell Lysates and Butyrate on Poly I:C-Induced IL-8 Production in Human Intestinal Epithelial Cells. Probiotics Antimicrob Proteins 2024; 16:1-12. [PMID: 36720771 DOI: 10.1007/s12602-023-10042-0] [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] [Accepted: 11/22/2022] [Indexed: 02/02/2023]
Abstract
Postbiotics include cell lysates (CLs), enzymes, cell wall fragments, and heat-killed bacteria derived from probiotics. Although postbiotics are increasingly being considered for their potential health-promoting properties, the effects of postbiotics on virus-mediated inflammatory responses in the intestine have not been elucidated. Hence, the present study aimed to examine whether CLs of Lactipantibacillus plantarum (LP CL) and Lacticaseibacillus rhamnosus GG (LR CL) could inhibit virus-mediated inflammatory responses in the human intestinal epithelial cell line HT-29 in vitro. Pretreatment with LP CL and LR CL significantly inhibited interleukin (IL)-8 production, which was induced by poly I:C, a synthetic analog of double-stranded RNA (dsRNA) viruses, at the mRNA and protein levels in HT-29 cells. However, peptidoglycans and heat-killed L. plantarum and L. rhamnosus GG did not effectively inhibit IL-8 production. LP CL and LR CL attenuated the poly I:C-induced phosphorylation of ERK and JNK and the activation of NF-κB, suggesting that these CLs could inhibit poly I:C-induced IL-8 production by regulating intracellular signaling pathways in HT-29 cells. Furthermore, among the short-chain fatty acids, butyrate enhanced the inhibitory effect of CLs on poly I:C-induced IL-8 production at the mRNA and protein levels in HT-29 cells, while acetate and propionate did not. Taken together, these results suggest that both LP CL and LR CL could act as potent effector molecules that can inhibit virus-mediated inflammatory responses and confer synergistic inhibitory effects with butyrate in human intestinal epithelial cells.
Collapse
Affiliation(s)
- Sun Woo Park
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, 10326, Republic of Korea
| | - Young Hyeon Choi
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, 10326, Republic of Korea
| | - Ju Young Gho
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, 10326, Republic of Korea
| | - Gweon Ah Kang
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, 10326, Republic of Korea
| | - Seok-Seong Kang
- Department of Food Science and Biotechnology, College of Life Science and Biotechnology, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, 10326, Republic of Korea.
| |
Collapse
|
8
|
Li Y, Wu S, Zhao Y, Dinh T, Jiang D, Selfridge JE, Myers G, Wang Y, Zhao X, Tomchuck S, Dubyak G, Lee RT, Estfan B, Shapiro M, Kamath S, Mohamed A, Huang SCC, Huang AY, Conlon R, Krishnamurthi S, Eads J, Willis JE, Khorana AA, Bajor D, Wang Z. Neutrophil extracellular traps induced by chemotherapy inhibit tumor growth in murine models of colorectal cancer. J Clin Invest 2024; 134:e175031. [PMID: 38194275 PMCID: PMC10904055 DOI: 10.1172/jci175031] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Neutrophil extracellular traps (NETs), a web-like structure of cytosolic and granule proteins assembled on decondensed chromatin, kill pathogens and cause tissue damage in diseases. Whether NETs can kill cancer cells is unexplored. Here, we report that a combination of glutaminase inhibitor CB-839 and 5-FU inhibited the growth of PIK3CA-mutant colorectal cancers (CRCs) in xenograft, syngeneic, and genetically engineered mouse models in part through NETs. Disruption of NETs by either DNase I treatment or depletion of neutrophils in CRCs attenuated the efficacy of the drug combination. Moreover, NETs were present in tumor biopsies from patients treated with the drug combination in a phase II clinical trial. Increased NET levels in tumors were associated with longer progression-free survival. Mechanistically, the drug combination induced the expression of IL-8 preferentially in PIK3CA-mutant CRCs to attract neutrophils into the tumors. Further, the drug combination increased the levels of ROS in neutrophils, thereby inducing NETs. Cathepsin G (CTSG), a serine protease localized in NETs, entered CRC cells through the RAGE cell surface protein. The internalized CTSG cleaved 14-3-3 proteins, released BAX, and triggered apoptosis in CRC cells. Thus, our studies illuminate a previously unrecognized mechanism by which chemotherapy-induced NETs kill cancer cells.
Collapse
Affiliation(s)
- Yamu Li
- Department of Genetics and Genome Sciences
- Case Comprehensive Cancer Center
| | - Sulin Wu
- Department of Genetics and Genome Sciences
- Department of Internal Medicine
- Department of Medical Genetics, Case Western Reserve University, Cleveland, Ohio. USA
| | - Yiqing Zhao
- Department of Genetics and Genome Sciences
- Case Comprehensive Cancer Center
| | - Trang Dinh
- Department of Genetics and Genome Sciences
- Case Comprehensive Cancer Center
| | - Dongxu Jiang
- Department of Genetics and Genome Sciences
- Case Comprehensive Cancer Center
| | - J. Eva Selfridge
- Department of Genetics and Genome Sciences
- Case Comprehensive Cancer Center
- Department of Internal Medicine
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | | | - Yuxiang Wang
- Department of Genetics and Genome Sciences
- Case Comprehensive Cancer Center
| | - Xuan Zhao
- Department of Genetics and Genome Sciences
- Case Comprehensive Cancer Center
| | | | - George Dubyak
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio. USA
| | - Richard T. Lee
- Department of Internal Medicine
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Bassam Estfan
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Marc Shapiro
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Suneel Kamath
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Amr Mohamed
- Department of Internal Medicine
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | | | | | | | | | - Jennifer Eads
- Department of Internal Medicine
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Alok A. Khorana
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - David Bajor
- Department of Internal Medicine
- Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Zhenghe Wang
- Department of Genetics and Genome Sciences
- Case Comprehensive Cancer Center
| |
Collapse
|
9
|
Zhu F, Jing D, Zhou H, Hu Z, Wang Y, Jin G, Yang Y, Zhou G. Blockade of Syk modulates neutrophil immune-responses via the mTOR/RUBCNL-dependent autophagy pathway to alleviate intestinal inflammation in ulcerative colitis. PRECISION CLINICAL MEDICINE 2023; 6:pbad025. [PMID: 37941642 PMCID: PMC10628969 DOI: 10.1093/pcmedi/pbad025] [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: 08/21/2023] [Accepted: 09/26/2023] [Indexed: 11/10/2023] Open
Abstract
Background Ulcerative colitis (UC) is a progressive chronic inflammatory disorder. Neutrophils play a critical role in regulating intestinal mucosal homeostasis in UC. Spleen tyrosine kinase (Syk) is involved in several inflammatory diseases. Here, we evaluated the effects and underlying mechanisms of Syk on neutrophil immune-responses in UC. Methods Syk expression in the colonic tissues of patients with UC was determined using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting, and immunohistochemistry. Colonic biopsies from patients with UC were obtained for single-cell RNA-sequencing. Neutrophils isolated from peripheral blood were pre-treated with R788 (a Syk inhibitor) and gene differences were determined using RNA sequencing. Neutrophil functions were analyzed using qRT-PCR, flow cytometry, and Transwell assay. R788 was administered daily to mice with dextran sulfate sodium (DSS)-induced colitis to verify the effects of Syk on intestinal inflammation. Results Syk expression was increased in inflamed mucosa and neutrophils of patients with UC and positively correlated with disease activity. Pharmacological inhibition of Syk in neutrophils decreased the production of pro-inflammatory cytokines, chemokines, neutrophil extracellular traps, reactive oxygen species, and myeloperoxidase. Apoptosis and migration of neutrophils were suppressed by Syk blockade. Syk blockade ameliorated mucosal inflammation in DSS-induced murine colitis by inhibiting neutrophil-associated immune responses. Mechanistically, Syk regulated neutrophil immune-responses via the mammalian target of rapamycin kinase/rubicon-like autophagy enhancer-dependent autophagy pathway. Conclusions Our findings indicate that Syk facilitates specific neutrophil functional responses to mucosal inflammation in UC, and its inhibition ameliorates mucosal inflammation in DSS-induced murine colitis, suggesting its potential as a novel therapeutic target for UC treatment.
Collapse
Affiliation(s)
- Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Dehuai Jing
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Huihui Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Zongjing Hu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Yan Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Guiyuan Jin
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Yonghong Yang
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| |
Collapse
|
10
|
Pike CM, Zwarycz B, McQueen BE, Castillo M, Barron C, Morowitz JM, Levi JA, Phadke D, Balik-Meisner M, Mav D, Shah R, Glasspoole DLC, Laetham R, Thelin W, Bunger MK, Boazak EM. Characterization and optimization of variability in a human colonic epithelium culture model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.559007. [PMID: 37790345 PMCID: PMC10542543 DOI: 10.1101/2023.09.22.559007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Animal models have historically been poor preclinical predictors of gastrointestinal (GI) directed therapeutic efficacy and drug-induced GI toxicity. Human stem and primary cell-derived culture systems are a major focus of efforts to create biologically relevant models that enhance preclinical predictive value of intestinal efficacy and toxicity. The inherent variability in stem-cell-based complex cultures makes development of useful models a challenge; the stochastic nature of stem-cell differentiation interferes with the ability to build and validate robust, reproducible assays that query drug responses and pharmacokinetics. In this study, we aimed to characterize and reduce potential sources of variability in a complex stem cell-derived intestinal epithelium model, termed RepliGut® Planar, across cells from multiple human donors, cell lots, and passage numbers. Assessment criteria included barrier formation and integrity, gene expression, and cytokine responses. Gene expression and culture metric analyses revealed that controlling for stem/progenitor-cell passage number reduces variability and maximizes physiological relevance of the model. After optimizing passage number, donor-specific differences in cytokine responses were observed in a case study, suggesting biologic variability is observable in cell cultures derived from multiple human sources. Our findings highlight key considerations for designing assays that can be applied to additional primary-cell derived systems, as well as establish utility of the RepliGut® Planar platform for robust development of human-predictive drug-response assays.
Collapse
|
11
|
McGhee ML, Acosta JP, Stein HH. Weanling pigs consume more feed if hybrid rye replaces corn in diets, but average daily gain and fecal scores are not impacted by hybrid rye. Transl Anim Sci 2023; 7:txad022. [PMID: 36911552 PMCID: PMC9997771 DOI: 10.1093/tas/txad022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/15/2023] [Indexed: 02/21/2023] Open
Abstract
An experiment was conducted to test the hypothesis that growth performance and health status of pigs will not be reduced if hybrid rye is included in diets at the expense of corn during the initial 5 wk post-weaning. A total of 128 weanling pigs (5.6 ± 0.5 kg) were randomly allotted to 32 pens and 4 dietary treatments. Pigs were fed experimental diets for 35 d in three phases with days 1 to 7 being phase 1, days 8 to 21 being phase 2, and days 22 to 35 being phase 3. Within each phase, a control diet primarily based on corn and soybean meal was formulated, and three additional diets were formulated by including 8.0, 16.0, or 24.0% (phase 1), 16.0, 32.0, or 48.0% (phase 2), and 20.0, 40.0, or 60.3% (phase 3) hybrid rye in the diet at the expense of corn. Pig weights were recorded at the start and conclusion of each phase, fecal scores were visually assessed every other day on a pen basis, and blood samples were obtained from 1 pig per pen on days 21 and 35. Results indicated that average daily gain (ADG) in phase 1 increased (linear, P < 0.05) as the inclusion of hybrid rye increased, but no other differences in ADG were observed. Average daily feed intake linearly increased in phase 1, phase 3, and overall (P < 0.05) as hybrid rye inclusion increased in the diets, and gain:feed was negatively impacted by the inclusion of hybrid rye in the diet (phase 1, linear, P < 0.05; phases 2, 3, and overall, quadratic, P < 0.05). No differences in average fecal scores or diarrhea incidence were observed. On days 21 and 35, blood urea N increased (linear, P < 0.05) as hybrid rye increased in the diets; and on day 21, serum total protein also increased (linear, P < 0.05) with increasing hybrid rye inclusion in the diet. Mean blood hemoglobin concentration on day 35 increased and then decreased as hybrid rye inclusion increased (quadratic, P < 0.05). On day 21, interleukin (IL) 2 and IL 10 decreased and then increased (quadratic, P < 0.05) as hybrid rye inclusion increased. On day 35, IL 8 and IL 12 increased and then decreased (quadratic, P < 0.05) and interferon-gamma decreased and then increased (quadratic, P < 0.01) as hybrid rye inclusion increased. In conclusion, the ADG of pigs was not different among treatments, but at the highest hybrid rye inclusion level, pigs consumed more feed than if corn was fed and gain:feed was reduced with increasing hybrid rye in diets. Differences in blood serum cytokines indicate the immune system was affected differently when hybrid rye instead of corn was fed.
Collapse
Affiliation(s)
- Molly L McGhee
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Jessica P Acosta
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Hans H Stein
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA.,Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
12
|
Tang L, Bai X, Xie X, Chen G, Jia X, Lei M, Li C, Lai S. Negative effects of heat stress on ovarian tissue in female rabbit. Front Vet Sci 2022; 9:1009182. [PMID: 36452142 PMCID: PMC9704112 DOI: 10.3389/fvets.2022.1009182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/27/2022] [Indexed: 07/30/2023] Open
Abstract
Numerous studies have highlighted the role of miRNA in the deformation and necrosis of cells of ovarian tissue caused by heat stress (HS), which ultimately affects ovarian function. Although the role of small RNAs has been investigated in alterations in ovarian tissue functioning in response to HS, the expression profile of ovarian miRNA has been explored to a lesser extent. In this study, female rabbits were subject to HS treatment by using electrical heater. The current work demonstrated that HS could significantly change physiological performance of female rabbits including body weight, rectal temperature and relative ovary weight, and significantly reduce serum IL-2, IL-8, CAT, and GSH-Px concentrations by enzyme-linked immunosorbent assay (ELISA) technique. As a result, an increase in apoptosis in ovarian cells, as well as unhealthy follicles, were observed by Hematoxylin-eosin (HE) and TUNEL staining. Additionally, small RNA-seq revealed changes in the miRNA expression profile of rabbit ovaries under HS. Five hundred fourteen miRNAs were obtained including known miRNAs 442 and novel miRNAs 72. Among these miRNAs, 23 miRNAs were significantly expressed under HS. Eleven differentially expressed miRNAs (DE miRNAs) and 9 their predicted targets were confirmed by qPCR, which were expected miRNA-mRNA negative regulation pattern. Among the DE miRNAs and targets, miR-141-39 may target COQ6, miR-449a-5p and miR-34c-5p may control RFC5 and RTN2 together, miR-449a-5p may target ACADVL, miR-34c-5p potentially targets Bcl-2 and miR-196b-5p potentially regulates CASK and HOXB6. Thus, the current work suggested the negative effects of HS on the ovarian tissue of female rabbits, and in conclusion these changes could be caused by decreased serum IL-2, IL-8, CAT and GSH-Px levels, increased ovarian apoptosis, and changed the expression of miRNAs.
Collapse
Affiliation(s)
- Lipeng Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xue Bai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xiaohong Xie
- Sichuan Provincial Key Laboratory of Animal Genetics and Breeding, Sichuan Academy of Animal Science, Chengdu, China
| | - Guanhe Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Xianbo Jia
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ming Lei
- Sichuan Provincial Key Laboratory of Animal Genetics and Breeding, Sichuan Academy of Animal Science, Chengdu, China
| | - Congyan Li
- Sichuan Provincial Key Laboratory of Animal Genetics and Breeding, Sichuan Academy of Animal Science, Chengdu, China
| | - Songjia Lai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
13
|
Izadparast F, Riahi-Zajani B, Yarmohammadi F, Hayes AW, Karimi G. Protective effect of berberine against LPS-induced injury in the intestine: a review. Cell Cycle 2022; 21:2365-2378. [PMID: 35852392 PMCID: PMC9645259 DOI: 10.1080/15384101.2022.2100682] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 07/03/2022] [Accepted: 07/08/2022] [Indexed: 12/11/2022] Open
Abstract
Sepsis is a systemic inflammatory condition caused by an unbalanced immunological response to infection, which affects numerous organs, including the intestines. Lipopolysaccharide (LPS; also known as endotoxin), a substance found in Gram-negative bacteria, plays a major role in sepsis and is mostly responsible for the disease's morbidity and mortality. Berberine is an isoquinoline alkaloid found in a variety of plant species that has anti-inflammatory properties. For many years, berberine has been used to treat intestinal inflammation and infection. Berberine has been reported to reduce LPS-induced intestinal damage. The potential pathways through which berberine protects against LPS-induced intestinal damage by inhibiting NF-κB, suppressing MAPK, modulating ApoM/S1P pathway, inhibiting COX-2, modulating Wnt/Beta-Catenin signaling pathway, and/or increasing ZIP14 expression are reviewed.Abbreviations: LPS, lipopolysaccharide; TLR, Toll-like receptor; MD-2, myeloid differentiation factor 2; CD14, cluster of differentiation 14; LBP, lipopolysaccharide-binding protein; MYD88, myeloid differentiation primary response 88; NF-κB, nuclear factor kappa light-chain enhancer of activated B cells; MAPK, mitogen-activated protein kinase; IL, interleukin; TNFα, tumor necrosis factor-alpha; Caco-2, cyanocobalamin uptake by human colon adenocarcinoma cell line; MLCK, myosin light-chain kinase; TJ, tight junction; IκBα, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha; IBS, irritable bowel syndrome; ERK, extracellular signal-regulated kinase; JNK, c-Jun N-terminal kinase (JNK; GVB, gut-vascular barrier; ApoM, apolipoprotein M; S1P, sphingosine-1-phosphate; VE-cadherin, vascular endothelial cadherin; AJ, adherens junction; PV1, plasmalemma vesicle-associated protein-1; HDL, high-density lipoprotein; Wnt, wingless-related integration site; Fzd, 7-span transmembrane protein Frizzled; LRP, low-density lipoprotein receptor-related protein; TEER, transendothelial/transepithelial electrical resistance; COX-2, cyclooxygenase-2; iNOS, inducible nitric oxide synthase; IGF, insulin-like growth factor; IGFBP, insulin-like growth factor-binding protein; ZIP, Zrt-Irt-like protein; PPAR, peroxisome proliferator-activated receptors; p-PPAR, phosphorylated-peroxisome proliferator-activated receptors; ATF, activating transcription factors; SOD, superoxide dismutase; GSH-Px, glutathione peroxidase; SARA, subacute ruminal acidosis; IPEC-J2, porcine intestinal epithelial cells; ALI, acute lung injury; ARDS, acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Faezeh Izadparast
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bamdad Riahi-Zajani
- Medical Toxicology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Yarmohammadi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A. Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
14
|
BORGONETTI V, COCETTA V, BIAGI M, CARNEVALI I, GOVERNA P, MONTOPOLI M. Anti-inflammatory activity of a fixed combination of probiotics and herbal extract in an in-vitro model of intestinal inflammation by stimulating Caco-2 cells with LPS-conditioned THP-1 cells medium. Minerva Pediatr (Torino) 2022; 74:511-518. [DOI: 10.23736/s2724-5276.20.05765-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
15
|
Neutrophil-Epithelial Crosstalk During Intestinal Inflammation. Cell Mol Gastroenterol Hepatol 2022; 14:1257-1267. [PMID: 36089244 PMCID: PMC9583449 DOI: 10.1016/j.jcmgh.2022.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 01/31/2023]
Abstract
Neutrophils are the most abundant leukocyte population in the human circulatory system and are rapidly recruited to sites of inflammation. Neutrophils play a multifaceted role in intestinal inflammation, as they contribute to the elimination of invading pathogens. Recently, their role in epithelial restitution has been widely recognized; however, they are also associated with bystander tissue damage. The intestinal epithelium provides a physical barrier to prevent direct contact of luminal contents with subepithelial tissues, which is extremely important for the maintenance of intestinal homeostasis. Numerous studies have demonstrated that transepithelial migration of neutrophils is closely related to disease symptoms and disruption of crypt architecture in inflammatory bowel disease and experimental colitis. There has been growing interest in how neutrophils interact with the epithelium under inflammatory conditions. Most studies focus on the effects of neutrophils on intestinal epithelial cells; however, the effects of intestinal epithelial cells on neutrophils during intestinal inflammation need to be well-established. Based on these data, we have summarized recent articles on the role of neutrophil-epithelial interactions in intestinal inflammation, particularly highlighting the epithelium-derived molecular regulators that mediate neutrophil recruitment, transepithelial migration, and detachment from the epithelium, as well as the functional consequences of their crosstalk. A better understanding of these molecular events may help develop novel therapeutic targets for mitigating the deleterious effects of neutrophils in inflammatory bowel disease.
Collapse
|
16
|
Sui C, Tao L, Bai C, Shao L, Miao J, Chen K, Wang M, Hu Q, Wang F. Molecular and cellular mechanisms underlying postoperative paralytic ileus by various immune cell types. Front Pharmacol 2022; 13:929901. [PMID: 35991871 PMCID: PMC9385171 DOI: 10.3389/fphar.2022.929901] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Postoperative ileus (POI) is a well-known complication following gut manipulation or surgical trauma, leading to an impaired gut motility and prolonged postoperative recovery time. Few current therapeutic strategies can prevent POI, and this disorder remains to be a major clinical challenge for patients undergoing surgery. Comprehensive understanding of cellular and molecular mechanisms related to the pathogenesis of POI stimulates the discovery of more promising targets for treatment. POI is closely associated with a series of inflammatory events within the bowel wall, and as key components of inflammatory mechanisms, different types of immune cells, including macrophages, dendritic cells, and T lymphocytes, play significant roles during the development of POI. A variety of immune cells are recruited into the manipulation sites after surgery, contributing to early inflammatory events or impaired gut motility. Our review intends to summarize the specific relationship between different immune cells and POI, mainly focusing on the relevant mechanisms underlying this disorder.
Collapse
Affiliation(s)
- Chao Sui
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Liang Tao
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Chunhua Bai
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Lihua Shao
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ji Miao
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Kai Chen
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
| | - Meng Wang
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Meng Wang, ; Qiongyuan Hu, ; Feng Wang,
| | - Qiongyuan Hu
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Medical School of Nanjing University, Nanjing, China
- *Correspondence: Meng Wang, ; Qiongyuan Hu, ; Feng Wang,
| | - Feng Wang
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Meng Wang, ; Qiongyuan Hu, ; Feng Wang,
| |
Collapse
|
17
|
Epithelial chemerin-CMKLR1 signaling restricts microbiota-driven colonic neutrophilia and tumorigenesis by up-regulating lactoperoxidase. Proc Natl Acad Sci U S A 2022; 119:e2205574119. [PMID: 35858331 PMCID: PMC9304024 DOI: 10.1073/pnas.2205574119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Intestinal barrier immunity is essential for controlling gut microbiota without eliciting harmful immune responses, while its defect contributes to the breakdown of intestinal homeostasis and colitis development. Chemerin, which is abundantly expressed in barrier tissues, has been demonstrated to regulate tissue inflammation via CMKLR1, its functional receptor. Several studies have reported the association between increased expression of chemerin-CMKLR1 and disease severity and immunotherapy resistance in inflammatory bowel disease (IBD) patients. However, the pathophysiological role of endogenous chemerin-CMKLR1 signaling in intestinal homeostasis remains elusive. We herein demonstrated that deficiency of chemerin or intestinal epithelial cell (IEC)-specific CMKLR1 conferred high susceptibility to microbiota-driven neutrophilic colon inflammation and subsequent tumorigenesis in mice following epithelial injury. Unexpectedly, we found that lack of chemerin-CMKLR1 signaling specifically reduced expression of lactoperoxidase (LPO), a peroxidase that is predominantly expressed in colonic ECs and utilizes H2O2 to oxidize thiocyanates to the antibiotic compound, thereby leading to the outgrowth and mucosal invasion of gram-negative bacteria and dysregulated CXCL1/2-mediated neutrophilia. Importantly, decreased LPO expression was causally linked to aggravated microbiota-driven colitis and associated tumorigenesis, as LPO supplementation could completely rescue such phenotypes in mice deficient in epithelial chemerin-CMKLR1 signaling. Moreover, epithelial chemerin-CMKLR1 signaling is necessary for early host defense against bacterial infection in an LPO-dependent manner. Collectively, our study reveals that the chemerin-CMKLR1/LPO axis represents an unrecognized immune mechanism that potentiates epithelial antimicrobial defense and restricts harmful colonic neutrophilia and suggests that LPO supplementation may be beneficial for microbiota dysbiosis in IBD patients with a defective innate antimicrobial mechanism.
Collapse
|
18
|
Sharma A, Raman V, Lee J, Forbes NS. Microbial Imbalance Induces Inflammation by Promoting Salmonella Penetration through the Mucosal Barrier. ACS Infect Dis 2022; 8:969-981. [PMID: 35404574 DOI: 10.1021/acsinfecdis.1c00530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The balance of microbial species in the intestine must be maintained to prevent inflammation and disease. Healthy bacteria suppress infection by pathogens and prevent disorders such as inflammatory bowel diseases (IBDs). The role of mucus in the relation between pathogens and the intestinal microbiota is poorly understood. Here, we hypothesized that healthy bacteria inhibit infection by preventing pathogens from penetrating the mucus layer and that microbial imbalance leads to inflammation by promoting the penetration of the mucosal barrier. We tested this hypothesis with an in vitro model that contains mucus, an epithelial cell layer, and resident immune cells. We found that, unlike probiotic VSL#3 bacteria, Salmonella penetrated the mucosal layers and induced the production of interleukin-8 (IL-8) and tumor necrosis factor (TNF)-α. At ratios greater than 104:1, probiotic bacteria suppressed the growth and penetration of Salmonella and reduced the production of inflammatory cytokines. Counterintuitively, low densities of healthy bacteria increased both pathogen penetration and cytokine production. In all cases, mucus increased Salmonella penetration and the production of cytokines. These results suggest that mucus lessens the protective effect of probiotic bacteria by promoting barrier penetration. In this model, a more imbalanced microbial population caused infection and inflammation by selecting pathogens that are more invasive and immunogenic. Combined, the results suggest that the depletion of commensal bacteria or an insufficient dosage of probiotics could worsen an infection and cause increased inflammation. A better understanding of the interactions between pathogens, healthy microbes, and the mucosal barrier will improve the treatment of infections and inflammatory diseases.
Collapse
Affiliation(s)
- Abhinav Sharma
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts 01003, United States
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
19
|
Müller I, Kym U, Galati V, Tharakan S, Subotic U, Krebs T, Stathopoulos E, Schmittenbecher P, Cholewa D, Romero P, Reingruber B, Holland-Cunz S, Keck S. Cholinergic Signaling Attenuates Pro-Inflammatory Interleukin-8 Response in Colonic Epithelial Cells. Front Immunol 2022; 12:781147. [PMID: 35069554 PMCID: PMC8770536 DOI: 10.3389/fimmu.2021.781147] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
Infants affected by Hirschsprung disease (HSCR), a neurodevelopmental congenital disorder, lack ganglia of the intrinsic enteric nervous system (aganglionosis) in a variable length of the colon, and are prone to developing severe Hirschsprung-associated enterocolitis (HAEC). HSCR patients typically show abnormal dense innervation of extrinsic cholinergic nerve fibers throughout the aganglionic rectosigmoid. Cholinergic signaling has been reported to reduce inflammatory response. Consequently, a sparse extrinsic cholinergic innervation in the mucosa of the rectosigmoid correlates with increased inflammatory immune cell frequencies and higher incidence of HAEC in HSCR patients. However, whether cholinergic signals influence the pro-inflammatory immune response of intestinal epithelial cells (IEC) is unknown. Here, we analyzed colonic IEC isolated from 43 HSCR patients with either a low or high mucosal cholinergic innervation density (fiber-low versus fiber-high) as well as from control tissue. Compared to fiber-high samples, IEC purified from fiber-low rectosigmoid expressed significantly higher levels of IL-8 but not TNF-α, IL-10, TGF-β1, Muc-2 or tight junction proteins. IEC from fiber-low rectosigmoid showed higher IL-8 protein concentrations in cell lysates as well as prominent IL-8 immunoreactivity compared to IEC from fiber-high tissue. Using the human colonic IEC cell line SW480 we demonstrated that cholinergic signals suppress lipopolysaccharide-induced IL-8 secretion via the alpha 7 nicotinic acetylcholine receptor (a7nAChR). In conclusion, we showed for the first time that the presence of a dense mucosal cholinergic innervation is associated with decreased secretion of IEC-derived pro-inflammatory IL-8 in the rectosigmoid of HSCR patients likely dependent on a7nAChR activation. Owing to the association between IL-8 and enterocolitis-prone, fiber-low HSCR patients, targeted therapies against IL-8 might be a promising immunotherapy candidate for HAEC treatment.
Collapse
Affiliation(s)
- Isabelle Müller
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| | - Urs Kym
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| | - Virginie Galati
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| | - Sasha Tharakan
- Department of Pediatric Surgery, University Children's Hospital Zürich, Zürich, Switzerland
| | - Ulrike Subotic
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland.,Department of Pediatric Surgery, University Children's Hospital Zürich, Zürich, Switzerland
| | - Thomas Krebs
- Department of Pediatric Surgery, Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Eleuthere Stathopoulos
- Department of Pediatric Surgery, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | | | - Dietmar Cholewa
- Department of Pediatric Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Philipp Romero
- Department of Pediatric Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Bertram Reingruber
- Department of Pediatric Surgery, Florence Nightingale Hospital, Düsseldorf, Germany
| | | | - Stefan Holland-Cunz
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| | - Simone Keck
- Department of Pediatric Surgery, University Children's Hospital Basel (UKBB) and University of Basel, Basel, Switzerland
| |
Collapse
|
20
|
A Computational Platform Integrating a Mechanistic Model of Crohn's Disease for Predicting Temporal Progression of Mucosal Damage and Healing. Adv Ther 2022; 39:3225-3247. [PMID: 35581423 PMCID: PMC9239932 DOI: 10.1007/s12325-022-02144-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/24/2022] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Physicians are often required to make treatment decisions for patients with Crohn's disease on the basis of limited objective information about the state of the patient's gastrointestinal tissue while aiming to achieve mucosal healing. Tools to predict changes in mucosal health with treatment are needed. We evaluated a computational approach integrating a mechanistic model of Crohn's disease with a responder classifier to predict temporal changes in mucosal health. METHODS A hybrid mechanistic-statistical platform was developed to predict biomarker and tissue health time courses in patients with Crohn's disease. Eligible patients from the VERSIFY study (n = 69) were classified into archetypical response cohorts using a decision tree based on early treatment data and baseline characteristics. A virtual patient matching algorithm assigned a digital twin to each patient from their corresponding response cohort. The digital twin was used to forecast response to treatment using the mechanistic model. RESULTS The responder classifier predicted endoscopic remission and mucosal healing for treatment with vedolizumab over 26 weeks, with overall sensitivities of 80% and 75% and overall specificities of 69% and 70%, respectively. Predictions for changes in tissue damage over time in the validation set (n = 31), a measure of the overall performance of the platform, were considered good (at least 70% of data points matched), fair (at least 50%), and poor (less than 50%) for 71%, 23%, and 6% of patients, respectively. CONCLUSION Hybrid computational tools including mechanistic components represent a promising form of decision support that can predict outcomes and patient progress in Crohn's disease.
Collapse
|
21
|
Toll-Like Receptors as Drug Targets in the Intestinal Epithelium. Handb Exp Pharmacol 2021; 276:291-314. [PMID: 34783909 DOI: 10.1007/164_2021_563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs) receptors are responsible for initiation of inflammatory responses by their recognition of molecular patterns present in invading microorganisms (such as bacteria, viruses or fungi) or in molecules released following tissue damage in disease states. Expressed in the intestinal epithelium, they initiate an intracellular signalling cascade in response to molecular patterns resulting in the activation of transcription factors and the release of cytokines, chemokines and vasoactive molecules. Intestinal epithelial cells are exposed to microorganisms on a daily basis and form part of the primary defence against pathogens by using TLRs. TLRs and their accessory molecules are subject to tight regulation in these cells so as to not overreact or react in unnecessary circumstances. TLRs have more recently been associated with chronic inflammatory diseases as a result of inappropriate regulation, this can be damaging and lead to chronic inflammatory diseases such as inflammatory bowel disease (IBD). Targeting Toll-like receptors offers a potential therapeutic approach for IBD. In this review, the current knowledge on the TLRs is reviewed along with their association with intestinal diseases. Finally, compounds that target TLRs in animal models of IBD, clinic trials and their future merit as targets are discussed.
Collapse
|
22
|
Chen H, Wu X, Xu C, Lin J, Liu Z. Dichotomous roles of neutrophils in modulating pathogenic and repair processes of inflammatory bowel diseases. PRECISION CLINICAL MEDICINE 2021; 4:246-257. [PMID: 35692862 PMCID: PMC8982532 DOI: 10.1093/pcmedi/pbab025] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023] Open
Abstract
Neutrophils are considered as complex innate immune cells and play a critical role in maintaining intestinal mucosal homeostasis. They exert robust pro-inflammatory effects and recruit other immune cells in the acute phase of pathogen infection and intestinal inflammation, but paradoxically, they also limit exogenous microbial invasion and facilitate mucosal restoration. Hyperactivation or dysfunction of neutrophils results in abnormal immune responses, leading to multiple autoimmune and inflammatory diseases including systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel diseases (IBD). As a refractory intestinal inflammatory disease, the pathogenesis and progression of IBD are associated with complicated immune response processes in which neutrophils are profoundly involved. However, the consensus on potential roles of neutrophils in modulating pathogenic and repair processes of IBD remains not fully understood. Accumulated infiltrating neutrophils cross the epithelial barrier and contribute to microbial dysbiosis, aggravated intestinal architectural damage, compromised resolution of intestinal inflammation and increased risk of thrombosis during IBD. Paradoxically, activated neutrophils are also associated with effective elimination of invaded microbiota, promoted angiogenesis and tissue restoration of gut mucosa in IBD. Here, we discuss the beneficial and detrimental roles of neutrophils in the onset and resolution of intestinal mucosal inflammation, hoping to provide a precise overview of neutrophil functions in the pathogenesis of IBD.
Collapse
Affiliation(s)
- Huimin Chen
- Center for Inflammatory Bowel Disease Research, the Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaohan Wu
- Center for Inflammatory Bowel Disease Research, the Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Chunjin Xu
- Department of Gastroenterology, the First People's Hospital of Shangqiu City Affiliated to Xinxiang Medical University, Shangqiu 476100, China
| | - Jian Lin
- Department of Gastroenterology, Affiliated Hospital of Putian University, Putian 351106, China
| | - Zhanju Liu
- Center for Inflammatory Bowel Disease Research, the Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
23
|
Spijkerman R, Hesselink L, Bertinetto C, Bongers CCWG, Hietbrink F, Vrisekoop N, Leenen LPH, Hopman MTE, Jansen JJ, Koenderman L. Refractory neutrophils and monocytes in patients with inflammatory bowel disease after repeated bouts of prolonged exercise. CYTOMETRY PART B-CLINICAL CYTOMETRY 2021; 100:676-682. [PMID: 33683008 PMCID: PMC9291995 DOI: 10.1002/cyto.b.21996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/09/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022]
Abstract
Background Neutrophils and monocytes are key immune effector cells in inflammatory bowel disease (IBD) that is associated with chronic inflammation in the gut. Patients with stable IBD who perform exercise have significantly fewer flare‐ups of the disease, but no underlying mechanism has been identified. Therefore, the aim of this study was to compare the responsiveness/refractoriness of these innate immune cells after repeated bouts of prolonged exercise in IBD patients and controls. Methods Patients with IBD and age‐ and gender‐matched healthy controls were recruited from a cohort of walkers participating in a 4‐day walking event. Blood analysis was performed at baseline and after 3 days of walking. Responsiveness to the bacterial/mitochondrial‐stimulus N‐Formylmethionine‐leucyl‐phenylalanine (fMLF) was tested in granulocytes and monocytes by measuring the expression of activation markers after adding this stimulus to whole blood. Results In total 38 participants (54 ± 12 years) were included in this study: 19 walkers with and 19 walkers without IBD. After 3 days of prolonged exercise, a significant increase in responsiveness to fMLF was observed in all participants irrespective of disease. However, IBD patients showed significantly less responsiveness in neutrophils and monocytes, compared with non‐IBD walkers. Conclusions Increased responsiveness of neutrophils and monocyte to fMLF was demonstrated after repetitive bouts of prolonged exercise. Interestingly, this exercise was associated with relative refractoriness of both neutrophils and monocytes in IBD patients. These refractory cells might create a lower inflammatory state in the intestine providing a putative mechanism for the decrease in flare‐ups in IBD patients after repeated exercise.
Collapse
Affiliation(s)
- Roy Spijkerman
- Department of Trauma SurgeryUniversity Medical Center UtrechtUtrechtCXThe Netherlands
- Center for Translational Immunology (CTI)University Medical Center UtrechtUtrechtCXThe Netherlands
- Department of Respiratory MedicineUniversity Medical Center UtrechtUtrechtCXThe Netherlands
| | - Lillian Hesselink
- Department of Trauma SurgeryUniversity Medical Center UtrechtUtrechtCXThe Netherlands
- Center for Translational Immunology (CTI)University Medical Center UtrechtUtrechtCXThe Netherlands
| | - Carlo Bertinetto
- Institute for Molecules and Materials (Analytical Chemistry)Radboud UniversityNijmegenAJThe Netherlands
| | - Coen C. W. G. Bongers
- Department of Physiology Radboud Institute for Health Sciences (RIHS)Radboud university medical centerNijmegenGAThe Netherlands
| | - Falco Hietbrink
- Department of Trauma SurgeryUniversity Medical Center UtrechtUtrechtCXThe Netherlands
| | - Nienke Vrisekoop
- Center for Translational Immunology (CTI)University Medical Center UtrechtUtrechtCXThe Netherlands
- Department of Respiratory MedicineUniversity Medical Center UtrechtUtrechtCXThe Netherlands
| | - Luke P. H. Leenen
- Department of Trauma SurgeryUniversity Medical Center UtrechtUtrechtCXThe Netherlands
| | - Maria T. E. Hopman
- Department of Physiology Radboud Institute for Health Sciences (RIHS)Radboud university medical centerNijmegenGAThe Netherlands
| | - Jeroen J. Jansen
- Institute for Molecules and Materials (Analytical Chemistry)Radboud UniversityNijmegenAJThe Netherlands
| | - Leo Koenderman
- Center for Translational Immunology (CTI)University Medical Center UtrechtUtrechtCXThe Netherlands
- Department of Respiratory MedicineUniversity Medical Center UtrechtUtrechtCXThe Netherlands
| |
Collapse
|
24
|
Induction of IL-10-producing type 2 innate lymphoid cells by allergen immunotherapy is associated with clinical response. Immunity 2021; 54:291-307.e7. [PMID: 33450188 DOI: 10.1016/j.immuni.2020.12.013] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 11/04/2020] [Accepted: 12/17/2020] [Indexed: 12/12/2022]
Abstract
The role of innate immune cells in allergen immunotherapy that confers immune tolerance to the sensitizing allergen is unclear. Here, we report a role of interleukin-10-producing type 2 innate lymphoid cells (IL-10+ ILC2s) in modulating grass-pollen allergy. We demonstrate that KLRG1+ but not KLRG1- ILC2 produced IL-10 upon activation with IL-33 and retinoic acid. These cells attenuated Th responses and maintained epithelial cell integrity. IL-10+ KLRG1+ ILC2s were lower in patients with grass-pollen allergy when compared to healthy subjects. In a prospective, double-blind, placebo-controlled trial, we demonstrated that the competence of ILC2 to produce IL-10 was restored in patients who received grass-pollen sublingual immunotherapy. The underpinning mechanisms were associated with the modification of retinol metabolic pathway, cytokine-cytokine receptor interaction, and JAK-STAT signaling pathways in the ILCs. Altogether, our findings underscore the contribution of IL-10+ ILC2s in the disease-modifying effect by allergen immunotherapy.
Collapse
|
25
|
Jang KB, Purvis JM, Kim SW. Dose-response and functional role of whey permeate as a source of lactose and milk oligosaccharides on intestinal health and growth of nursery pigs. J Anim Sci 2021; 99:skab008. [PMID: 33521816 PMCID: PMC7849970 DOI: 10.1093/jas/skab008] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/09/2021] [Indexed: 12/14/2022] Open
Abstract
Two experiments were conducted to evaluate dose-response and supplemental effects of whey permeate on growth performance and intestinal health of nursery pigs. In experiment (exp.) 1, 1,080 pigs weaned at 6.24 kg body weight (BW) were allotted to five treatments (eight pens/treatment) with increasing levels of whey permeate in three phases (from 10% to 30%, 3% to 23%, and 0% to 9% for phase 1, 2, and 3, respectively) fed until 11 kg BW and then fed a common phase 4 diet (0% whey permeate) until 25 kg BW in a 48-d feeding trial. Feed intake and BW were measured at the end of each phase. In exp. 2, 1,200 nursery pigs at 7.50 kg BW were allotted to six treatments (10 pens/treatment) with increasing levels of whey permeate from 0% to 18.75% fed until 11 kg BW. Feed intake and BW were measured during 11 d. Six pigs per treatment (1 per pens) were euthanized to collect the jejunum to evaluate tumor necrosis factor-alpha, interleukin-8 (IL-8), transforming growth factor-beta 1, mucin 2, histomorphology, digestive enzyme activity, crypt cell proliferation rate, and jejunal mucosa-associated microbiota. Data were analyzed using contrasts in the MIXED procedure and a broken-line analysis using the NLIN procedure of SAS. In exp. 1, increasing whey permeate had a quadratic effect (P < 0.05) on feed efficiency (G:F; maximum: 1.35 at 18.3%) in phase 1. Increasing whey permeate linearly increased (P < 0.05) average daily gain (ADG; 292 to 327 g/d) and G:F (0.96 to 1.04) of pigs in phase 2. In exp. 2, increasing whey permeate linearly increased (P < 0.05) ADG (349 to 414 g/d) and G:F (0.78 to 0.85) and linearly increased (P < 0.05) crypt cell proliferation rate (27.8% to 37.0%). The breakpoint from a broken-line analysis was obtained at 13.6% whey permeate for maximal G:F. Increasing whey permeate tended to change IL-8 (quadratic, P = 0.052; maximum: 223 pg/mg at 10.9%), to decrease Firmicutes:Bacteroidetes (P = 0.073, 1.59 to 1.13), to increase (P = 0.089) Bifidobacteriaceae (0.73% to 1.11%), and to decrease Enterobacteriaceae (P = 0.091, 1.04% to 0.52%) and Streptococcaceae (P = 0.094, 1.50% to 0.71%) in the jejunal mucosa. In conclusion, dietary inclusion of whey permeate increased the growth of nursery pigs from 7 to 11 kg BW. Pigs grew most efficiently with 13.6% whey permeate. Improvement in growth performance is partly attributed to stimulating intestinal immune response and enterocyte proliferation with positive changes in jejunal mucosa-associated microbiota in nursery pigs.
Collapse
Affiliation(s)
- Ki Beom Jang
- Department of Animal Science, North Carolina State University, Raleigh, NC
| | | | - Sung W Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC
| |
Collapse
|
26
|
Zheng S, Zhang H, Liu R, Huang CL, Li H, Deng ZY, Tsao R. Do short chain fatty acids and phenolic metabolites of the gut have synergistic anti-inflammatory effects? – New insights from a TNF-α-induced Caco-2 cell model. Food Res Int 2021; 139:109833. [DOI: 10.1016/j.foodres.2020.109833] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/23/2022]
|
27
|
Musson R, Szukała W, Jura J. MCPIP1 RNase and Its Multifaceted Role. Int J Mol Sci 2020; 21:ijms21197183. [PMID: 33003343 PMCID: PMC7582464 DOI: 10.3390/ijms21197183] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammation is an organism’s physiological response to harmful septic and aseptic stimuli. This process begins locally through the influx of immune system cells to the damaged tissue and the subsequent activation and secretion of inflammatory mediators to restore homeostasis in the organism. Inflammation is regulated at many levels, and one of these levels is post-transcriptional regulation, which controls the half-life of transcripts that encode inflammatory mediators. One of the proteins responsible for controlling the amount of mRNA in a cell is the RNase monocyte chemoattractant protein-induced protein 1 (MCPIP1). The studies conducted so far have shown that MCPIP1 is involved not only in the regulation of inflammation but also in many other physiological and pathological processes. This paper provides a summary of the information on the role of MCPIP1 in adipogenesis, angiogenesis, cell differentiation, cancer, and skin inflammation obtained to date.
Collapse
|
28
|
Sharma A, Raman V, Lee J, Forbes NS. Mucus blocks probiotics but increases penetration of motile pathogens and induces TNF-α and IL-8 secretion. Biotechnol Bioeng 2020; 117:2540-2555. [PMID: 32396232 PMCID: PMC7806204 DOI: 10.1002/bit.27383] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 12/29/2022]
Abstract
The mucosal barrier in combination with innate immune system are the first line of defense against luminal bacteria at the intestinal mucosa. Dysfunction of the mucus layer and bacterial infiltration are linked to tissue inflammation and disease. To study host-bacterial interactions at the mucosal interface, we created an experimental model that contains luminal space, a mucus layer, an epithelial layer, and suspended immune cells. Reconstituted porcine small intestinal mucus formed an 880 ± 230 µm thick gel layer and had a porous structure. In the presence of mucus, sevenfold less probiotic and nonmotile VSL#3 bacteria transmigrated across the epithelial barrier compared to no mucus. The higher bacterial transmigration caused immune cell differentiation and increased the concentration of interleukin-8 (IL-8) and tumor necrosis factor-alpha (TNF-α; p < .01). Surprisingly, the mucus layer increased transmigration of pathogenic Salmonella and increased secretion of TNF-α and IL-8 (p < .05). Nonmotile, flagella knockout Salmonella had lower transmigration and caused lower IL-8 and TNF-α secretion (p < .05). These results demonstrate that motility enables pathogenic bacteria to cross the mucus and epithelial layers, which could lead to infection. Using an in vitro coculture platform to understand the interactions of bacteria with the intestinal mucosa has the potential to improve the treatment of intestinal diseases.
Collapse
Affiliation(s)
- Abhinav Sharma
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Vishnu Raman
- Department of Chemical Engineering, University of Massachusetts, Amherst
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| | - Neil S. Forbes
- Department of Chemical Engineering, University of Massachusetts, Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst
- Institute for Applied Life Sciences, University of Massachusetts, Amherst
| |
Collapse
|
29
|
Hosic S, Lake W, Stas E, Koppes R, Breault DT, Murthy SK, Koppes AN. Cholinergic Activation of Primary Human Derived Intestinal Epithelium Does Not Ameliorate TNF-α Induced Injury. Cell Mol Bioeng 2020; 13:487-505. [PMID: 33184579 DOI: 10.1007/s12195-020-00633-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/19/2020] [Indexed: 02/08/2023] Open
Abstract
Introduction The intestinal epithelium contains specialized cells including enterocytes, goblet, Paneth, enteroendocrine, and stem cells. Impaired barrier integrity in Inflammatory Bowel Disease is characterized by elevated levels of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α). Prior studies in immortalized lines such as Caco-2, without native epithelial heterogeneity, demonstrate the amelioration of TNF-α compromised barrier integrity via nicotinic (nAChR) or muscarinic (mAChR) acetylcholine receptor activation. Methods A tissue-engineered model of primary human small intestinal epithelium was derived from dissociated organoids cultured on collagen-coated Transwells. Differentiation was accomplished with serum-containing media and compared to Caco-2 and HT-29 regarding alkaline phosphatase expression, transepithelial electrical resistance (TEER), and IL-8 secretion. Inflammation was modeled via basal stimulation with TNF-α (25 ng/mL) with or without nicotine (nAChR agonist) or bethanechol (mAChR agonist). Apoptosis, density (cells/cm2), TEER, lucifer yellow permeability, 70 kDa dextran transport, cell morphology, and IL-8 secretion were characterized. Results Primary intestinal epithelium demonstrates significant functional differences compared to immortalized cells, including increased barrier integrity, IL-8 expression, mucus production, and the presence of absorptive and secretory cells. Exposure to TNF-α impaired barrier integrity, increased apoptosis, altered morphology, and increased secretion of IL-8. Stimulation of nAChR with nicotine did not ameliorate TNF-α induced permeability nor alter 70 kDa dextran transport. However, stimulation of mAChR with bethanechol decreased transport of 70 kDa dextran but did not ameliorate TNF-α induced paracellular permeability. Conclusions A primary model of intestinal inflammation was evaluated, demonstrating nAChR or mAChR activation does not have the same protective effects compared to immortalized epithelium. Inclusion of other native stromal support cells are underway.
Collapse
Affiliation(s)
- Sanjin Hosic
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
| | - Will Lake
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
| | - Eric Stas
- Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA
| | - Ryan Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115 USA.,Department of Pediatrics, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115 USA.,Harvard Stem Cell Institute, 7 Divinity Ave, Cambridge, MA 02138 USA
| | - Shashi K Murthy
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA.,Department of Biology, Northeastern University, 360 Huntington Ave., 313 Snell Engineering, Boston, MA 02115 USA
| |
Collapse
|
30
|
Xu K, Guo Y, Ping L, Qiu Y, Liu Q, Li Z, Wang Z. Protective Effects of SIRT6 Overexpression against DSS-Induced Colitis in Mice. Cells 2020; 9:cells9061513. [PMID: 32580272 PMCID: PMC7348883 DOI: 10.3390/cells9061513] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/21/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
Sirtuin 6 (SIRT6), as a NAD + -dependent deacetylase, plays an indispensable role in the regulation of health and physiology. Loss of SIRT6 causes spontaneous colitis in mice and makes intestinal epithelial cells prone to stress. However, whether SIRT6 overexpression increases resistance to colitis remains unknown. Here, in vivo results demonstrated that SIRT6 overexpression attenuates DSS-induced colitis in terms of clinical manifestations, histopathological damage, loss of tight junction function and imbalanced intestinal microenvironment. Additionally, we also found that the activation of NF-κB and c-Jun induced by DSS is diminished by SIRT6 overexpression. Furthermore, SIRT6 may regulate TAK1 to inhibit NF-κB and c-Jun signaling. Thus, our findings highlight the protective effect of SIRT6 on colon, further supporting the perspective that SIRT6 may be a therapeutic target for intestine injury under stress.
Collapse
Affiliation(s)
- Kang Xu
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; (K.X.); (Y.G.); (Q.L.)
| | - Yannan Guo
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; (K.X.); (Y.G.); (Q.L.)
| | - Lu Ping
- 8-year MD Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China;
| | - Ying Qiu
- School of Medicine, Tsinghua University, Beijing 100084, China;
| | - Qingfei Liu
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; (K.X.); (Y.G.); (Q.L.)
| | - Zhongchi Li
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; (K.X.); (Y.G.); (Q.L.)
- Correspondence: (Z.L.); (Z.W.); Tel.: +86-10-62772241 (Z.W.)
| | - Zhao Wang
- Protein Science Key Laboratory of the Ministry of Education, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; (K.X.); (Y.G.); (Q.L.)
- Correspondence: (Z.L.); (Z.W.); Tel.: +86-10-62772241 (Z.W.)
| |
Collapse
|
31
|
Schofield ZV, Wu MCL, Hansbro PM, Cooper MA, Woodruff TM. Acetate protects against intestinal ischemia‐reperfusion injury independent of its cognate free fatty acid 2 receptor. FASEB J 2020; 34:10418-10430. [DOI: 10.1096/fj.202000960r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Zoe V. Schofield
- School of Biomedical Sciences The University of Queensland Brisbane QLD Australia
- The Institute for Molecular Bioscience The University of Queensland Brisbane QLD Australia
| | - Mike C. L. Wu
- School of Biomedical Sciences The University of Queensland Brisbane QLD Australia
| | - Philip M. Hansbro
- Centre for Inflammation Centenary Institute Sydney NSW Australia
- Faculty of Science University of Technology Sydney Ultimo NSW Australia
| | - Matthew A. Cooper
- The Institute for Molecular Bioscience The University of Queensland Brisbane QLD Australia
| | - Trent M. Woodruff
- School of Biomedical Sciences The University of Queensland Brisbane QLD Australia
| |
Collapse
|
32
|
Vašíček O, Hájek J, Bláhová L, Hrouzek P, Babica P, Kubala L, Šindlerová L. Cyanobacterial lipopeptides puwainaphycins and minutissamides induce disruptive and pro-inflammatory processes in Caco-2 human intestinal barrier model. HARMFUL ALGAE 2020; 96:101849. [PMID: 32560836 DOI: 10.1016/j.hal.2020.101849] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 05/25/2020] [Accepted: 05/27/2020] [Indexed: 05/24/2023]
Abstract
Puwainaphycins (PUW) and minutissamides (MIN) are cyanobacterial lipopeptides found in various cyanobacterial species. The first possible target of human exposure to them is intestinal epithelium but effect of PUW/MIN on enterocytes is not known at all. Using differentiated Caco-2 cells, PUW F was found to be cytotoxic from 5 µM concentration based on lactate dehydrogenase release assay and total protein concentration. However, it is also able to induce production of interleukin 8 in non-cytotoxic concentrations 1 and 2.5 µM detected by ELISA. Effects of MIN A and C were similar but less pronounced compared to PUW F. On the other hand, MIN D was the least toxic compound with no significant pro-inflammatory effects. Surprisingly, pro-inflammatory activation of the cells by PUW F and MIN C resulted in an increase in tight junction (TJ) protein claudin 4 expression determined by western blot analysis and confirmed by confocal microscopy. Furthermore, decrease in expression of zonula occludens 3, another TJ protein, was observed after the exposure to PUW F. Taken together, these cytotoxic lipopeptides, especially PUW F, are to be studied more deeply due to their capability to activate and/or deregulate human enterocytes in low concentrations.
Collapse
Affiliation(s)
- Ondřej Vašíček
- Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno 612 65, Czech Republic.
| | - Jan Hájek
- Institute of Microbiology, Centre Algatech, The Czech Academy of Sciences, Novohradska 237, Trebon 379 80, Czech Republic.
| | - Lucie Bláhová
- RECETOX, Faculty of Science, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic.
| | - Pavel Hrouzek
- Institute of Microbiology, Centre Algatech, The Czech Academy of Sciences, Novohradska 237, Trebon 379 80, Czech Republic.
| | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic; Institute of Botany, The Czech Academy of Sciences, Lidická 25/27, Brno 602 00, Czech Republic.
| | - Lukáš Kubala
- Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno 612 65, Czech Republic.
| | - Lenka Šindlerová
- Institute of Biophysics of the Czech Academy of Sciences, Kralovopolska 135, Brno 612 65, Czech Republic.
| |
Collapse
|
33
|
Probiotic Propionibacterium freudenreichii requires SlpB protein to mitigate mucositis induced by chemotherapy. Oncotarget 2019; 10:7198-7219. [PMID: 31921383 PMCID: PMC6944450 DOI: 10.18632/oncotarget.27319] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/21/2019] [Indexed: 02/07/2023] Open
Abstract
Propionibacterium freudenreichii CIRM-BIA 129 (P. freudenreichii wild type, WT) is a probiotic bacterium, which exerts immunomodulatory effects. This strain possesses extractable surface proteins, including SlpB, which are involved in anti-inflammatory effect and in adhesion to epithelial cells. We decided to investigate the impact of slpB gene mutation on immunomodulation in vitro and in vivo. In an in vitro assay, P. freudenreichii WT reduced expression of IL-8 (p<0.0001) and TNF-α (p<0.0001) cytokines in LPS-stimulated HT-29 cells. P. freudenreichii ΔslpB, lacking the SlpB protein, failed to do so. Subsequently, both strains were investigated in vivo in a 5-FU-induced mucositis mice model. Mucositis is a common side effect of cytotoxic chemotherapy with 5-FU, characterized by mucosal injury, inflammation, diarrhea, and weight loss. The WT strain prevented weight loss, reduced inflammation and consequently histopathological scores. Furthermore, it regulated key markers, including Claudin-1 (cld1, p<0.0005) and IL-17a (Il17a, p<0.0001) genes, as well as IL-12 (p<0.0001) and IL-1β (p<0.0429) cytokines levels. Mutant strain displayed opposite regulatory effect on cld1 expression and on IL-12 levels. This work emphasizes the importance of SlpB in P. freudenreichii ability to reduce mucositis inflammation. It opens perspectives for the development of probiotic products to decrease side effects of chemotherapy using GRAS bacteria with immunomodulatory surface protein properties.
Collapse
|
34
|
Target-Specific Fluorescence-Mediated Tomography for Non-Invasive and Dynamic Assessment of Early Neutrophil Infiltration in Murine Experimental Colitis. Cells 2019; 8:cells8111328. [PMID: 31661876 PMCID: PMC6912230 DOI: 10.3390/cells8111328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/18/2019] [Accepted: 10/26/2019] [Indexed: 12/13/2022] Open
Abstract
The role of neutrophils in the pathogenesis of inflammatory bowel disease (IBD) is still only incompletely understood. Here, we evaluated target-specific fluorescence-mediated tomography (FMT) for visualization of neutrophil infiltration in murine experimental DSS-induced colitis. Colitis was assessed using clinical, endoscopic, and histopathological parameters. Intestinal neutrophil infiltration was determined at day 0, 4, and 10 by targeted FMT after injection of a neutrophil-specific fluorescence-labelled monoclonal antibody (Gr-1). Complementary, immunofluorescence tissue sections with Gr-1 and ELISA-based assessment of tissue myeloperoxidase (MPO) served as the gold standard for the quantification of neutrophil infiltration. Colitic animals showed decreasing body weight, presence of fecal occult blood, and endoscopic signs of inflammation. FMT revealed a significantly increased level of fluorescence only four days after colitis induction as compared to pre-experimental conditions (pmol tracer 73.2 ± 18.1 versus 738.6 ± 80.7; p < 0.05), while neither body weight nor endoscopic assessment showed significant changes at this early time. Confirmatory, post-mortem immunofluorescence studies and measurements of tissue MPO confirmed the presence of increased neutrophil infiltration in colitic mice compared to controls. Concluding, Gr-1 targeted FMT can detect early colonic infiltration of neutrophils in experimental colitis even before clinical symptoms or endoscopic alterations occur. Therefore, FMT might be an important tool for repetitive and non-invasive monitoring of inflammatory cell infiltrate in intestinal inflammation.
Collapse
|
35
|
Loktionov A. Eosinophils in the gastrointestinal tract and their role in the pathogenesis of major colorectal disorders. World J Gastroenterol 2019; 25:3503-3526. [PMID: 31367153 PMCID: PMC6658389 DOI: 10.3748/wjg.v25.i27.3503] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/22/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023] Open
Abstract
Eosinophils are currently regarded as versatile mobile cells controlling and regulating multiple biological pathways and responses in health and disease. These cells store in their specific granules numerous biologically active substances (cytotoxic cationic proteins, cytokines, growth factors, chemokines, enzymes) ready for rapid release. The human gut is the main destination of eosinophils that are produced and matured in the bone marrow and then transferred to target tissues through the circulation. In health the most important functions of gut-residing eosinophils comprise their participation in the maintenance of the protective mucosal barrier and interactions with other immune cells in providing immunity to microbiota of the gut lumen. Eosinophils are closely involved in the development of inflammatory bowel disease (IBD), when their cytotoxic granule proteins cause damage to host tissues. However, their roles in Crohn’s disease and ulcerative colitis appear to follow different immune response patterns. Eosinophils in IBD are especially important in altering the structure and protective functions of the mucosal barrier and modulating massive neutrophil influx to the lamina propria followed by transepithelial migration to colorectal mucus. IBD-associated inflammatory process involving eosinophils then appears to expand to the mucus overlaying the internal gut surface. The author hypothesises that immune responses within colorectal mucus as well as ETosis exerted by both neutrophils and eosinophils on the both sides of the colonic epithelial barrier act as additional pathogenetic factors in IBD. Literature analysis also shows an association between elevated eosinophil levels and better colorectal cancer (CRC) prognosis, but mechanisms behind this effect remain to be elucidated. In conclusion, the author emphasises the importance of investigating colorectal mucus in IBD and CRC patients as a previously unexplored milieu of disease-related inflammatory responses.
Collapse
|
36
|
Westreich ST, Ardeshir A, Alkan Z, Kable ME, Korf I, Lemay DG. Fecal metatranscriptomics of macaques with idiopathic chronic diarrhea reveals altered mucin degradation and fucose utilization. MICROBIOME 2019; 7:41. [PMID: 30885266 PMCID: PMC6423747 DOI: 10.1186/s40168-019-0664-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/11/2019] [Indexed: 05/20/2023]
Abstract
BACKGROUND Idiopathic chronic diarrhea (ICD) is a common cause of morbidity and mortality among juvenile rhesus macaques. Characterized by chronic inflammation of the colon and repeated bouts of diarrhea, ICD is largely unresponsive to medical interventions, including corticosteroid, antiparasitic, and antibiotic treatments. Although ICD is accompanied by large disruptions in the composition of the commensal gut microbiome, no single pathogen has been concretely identified as responsible for the onset and continuation of the disease. RESULTS Fecal samples were collected from 12 ICD-diagnosed macaques and 12 age- and sex-matched controls. RNA was extracted for metatranscriptomic analysis of organisms and functional annotations associated with the gut microbiome. Bacterial, fungal, archaeal, protozoan, and macaque (host) transcripts were simultaneously assessed. ICD-afflicted animals were characterized by increased expression of host-derived genes involved in inflammation and increased transcripts from bacterial pathogens such as Campylobacter and Helicobacter and the protozoan Trichomonas. Transcripts associated with known mucin-degrading organisms and mucin-degrading enzymes were elevated in the fecal microbiomes of ICD-afflicted animals. Assessment of colon sections using immunohistochemistry and of the host transcriptome suggests differential fucosylation of mucins between control and ICD-afflicted animals. Interrogation of the metatranscriptome for fucose utilization genes reveals possible mechanisms by which opportunists persist in ICD. Bacteroides sp. potentially cross-fed fucose to Haemophilus whereas Campylobacter expressed a mucosa-associated transcriptome with increased expression of adherence genes. CONCLUSIONS The simultaneous profiling of bacterial, fungal, archaeal, protozoan, and macaque transcripts from stool samples reveals that ICD of rhesus macaques is associated with increased gene expression by pathogens, increased mucin degradation, and altered fucose utilization. The data suggest that the ICD-afflicted host produces fucosylated mucins that are leveraged by potentially pathogenic microbes as a carbon source or as adhesion sites.
Collapse
Affiliation(s)
| | - Amir Ardeshir
- California National Primate Research Center, University of California, Davis, California USA
| | - Zeynep Alkan
- USDA ARS Western Human Nutrition Research Center, Davis, California USA
| | - Mary E. Kable
- USDA ARS Western Human Nutrition Research Center, Davis, California USA
- Department of Nutrition, University of California, Davis, California USA
| | - Ian Korf
- Genome Center, University of California, Davis, California USA
| | - Danielle G. Lemay
- Genome Center, University of California, Davis, California USA
- USDA ARS Western Human Nutrition Research Center, Davis, California USA
- Department of Nutrition, University of California, Davis, California USA
| |
Collapse
|
37
|
Chen Y, Zhang H, Liu R, Mats L, Zhu H, Pauls KP, Deng Z, Tsao R. Antioxidant and anti-inflammatory polyphenols and peptides of common bean (Phaseolus vulga L.) milk and yogurt in Caco-2 and HT-29 cell models. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
38
|
Zhang H, Xu CN, Mine Y. Effects of a synthetic di‐phosphoserine peptide (SS‐2) on gene expression profiling against TNF‐α induced inflammation. Int J Food Sci Technol 2019. [DOI: 10.1111/ijfs.14077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Hua Zhang
- Department of Food Science University of Guelph Guelph ON N1G 2W1 Canada
| | - Cai Na Xu
- Department of Food Science University of Guelph Guelph ON N1G 2W1 Canada
| | - Yoshinori Mine
- Department of Food Science University of Guelph Guelph ON N1G 2W1 Canada
| |
Collapse
|
39
|
P. McKernan D. Toll-like receptors and immune cell crosstalk in the intestinal epithelium. AIMS ALLERGY AND IMMUNOLOGY 2019. [DOI: 10.3934/allergy.2019.1.13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
40
|
Governa P, Marchi M, Cocetta V, De Leo B, Saunders PTK, Catanzaro D, Miraldi E, Montopoli M, Biagi M. Effects of Boswellia Serrata Roxb. and Curcuma longa L. in an In Vitro Intestinal Inflammation Model Using Immune Cells and Caco-2. Pharmaceuticals (Basel) 2018; 11:ph11040126. [PMID: 30463367 PMCID: PMC6316569 DOI: 10.3390/ph11040126] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel diseases, which consist of chronic inflammatory conditions of the colon and the small intestine, are considered a global disease of our modern society. Recently, the interest toward the use of herbal therapies for the management of inflammatory bowel diseases has increased because of their effectiveness and favourable safety profile, compared to conventional drugs. Boswellia serrata Roxb. and Curcuma longa L. are amongst the most promising herbal drugs, however, their clinical use in inflammatory bowel diseases is limited and little is known on their mechanism of action. The aim of this work was to investigate the effects of two phytochemically characterized extracts of B. serrata and C. longa in an in vitro model of intestinal inflammation. Their impact on cytokine release and reactive oxygen species production, as well as the maintenance of the intestinal barrier function and on intestinal mucosa immune cells infiltration, has been evaluated. The extracts showed a good protective effect on the intestinal epithelium at 1 µg/mL, with TEER values increasing by approximately 1.5 fold, compared to LPS-stimulated cells. C. longa showed an anti-inflammatory mechanism of action, reducing IL-8, TNF-α and IL-6 production by approximately 30%, 25% and 40%, respectively, compared to the inflammatory stimuli. B. serrata action was linked to its antioxidant effect, with ROS production being reduced by 25%, compared to H2O2-stimulated Caco-2 cells. C. longa and B. serrata resulted to be promising agents for the management of inflammatory bowel diseases by modulating in vitro parameters which have been identified in the clinical conditions.
Collapse
Affiliation(s)
- Paolo Governa
- Department of Physical Sciences, Hearth and Environment, University of Siena, Via Laterina 8, 53100 Siena, Italy.
- Department of Biotechnology, Chemistry and Pharmacy⁻Department of Excellence 2018⁻2022, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| | - Maddalena Marchi
- Department of Physical Sciences, Hearth and Environment, University of Siena, Via Laterina 8, 53100 Siena, Italy.
| | - Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Egidio Meneghetti 2, 35131 Padua, Italy.
| | - Bianca De Leo
- MRC Centres for Inflammation Research and Reproductive Health, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Philippa T K Saunders
- MRC Centre for Inflammation Research, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Daniela Catanzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Egidio Meneghetti 2, 35131 Padua, Italy.
| | - Elisabetta Miraldi
- Department of Physical Sciences, Hearth and Environment, University of Siena, Via Laterina 8, 53100 Siena, Italy.
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo Egidio Meneghetti 2, 35131 Padua, Italy.
- Venetian Institute of Molecular Medicine, Via Orus 2, 35129 Padua, Italy.
| | - Marco Biagi
- Department of Physical Sciences, Hearth and Environment, University of Siena, Via Laterina 8, 53100 Siena, Italy.
| |
Collapse
|
41
|
Sarna VK, Skodje GI, Reims HM, Risnes LF, Dahal-Koirala S, Sollid LM, Lundin KEA. HLA-DQ:gluten tetramer test in blood gives better detection of coeliac patients than biopsy after 14-day gluten challenge. Gut 2018; 67:1606-1613. [PMID: 28779027 DOI: 10.1136/gutjnl-2017-314461] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 06/10/2017] [Accepted: 06/22/2017] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Initiation of a gluten-free diet without proper diagnostic work-up of coeliac disease is a frequent and demanding problem. Recent diagnostic guidelines suggest a gluten challenge of at least 14 days followed by duodenal biopsy in such patients. The rate of false-negative outcome of this approach remains unclear. We studied responses to 14-day gluten challenge in subjects with treated coeliac disease. DESIGN We challenged 20 subjects with biopsy-verified coeliac disease, all in confirmed mucosal remission, for 14 days with 5.7 grams per oral gluten daily. Duodenal biopsies were collected. Blood was analysed by multiplex assay for cytokine detection, and by flow cytometry using HLA-DQ:gluten tetramers. RESULTS Nineteen participants completed the challenge. Villous blunting appeared at end of challenge in 5 of 19 subjects. Villous height to crypt depth ratio reduced with at least 0.4 concomitantly with an increase in intraepithelial lymphocyte count of at least 50% in 9 of 19 subjects. Interleukin-8 plasma concentration increased by more than 100% after 4 hours in 7 of 19 subjects. Frequency of blood CD4+ effector-memory gut-homing HLA-DQ:gluten tetramer-binding T cells increased by more than 100% on day 6 in 12 of 15 evaluated participants. CONCLUSION A 14-day gluten challenge was not enough to establish significant mucosal architectural changes in majority of patients with coeliac disease (sensitivity ≈25%-50%). Increase in CD4+ effector-memory gut-homing HLA-DQ:gluten tetramer-binding T cells in blood 6 days after gluten challenge is a more sensitive and less invasive biomarker that should be validated in a larger study. TRIAL REGISTRATION NUMBER NCT02464150.
Collapse
Affiliation(s)
- Vikas K Sarna
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Norway.,K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Norway
| | - Gry I Skodje
- K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Norway.,Department of Clinical Service, Oslo University Hospital, Norway
| | - Henrik M Reims
- Department of Pathology, Oslo University Hospital, Norway
| | - Louise F Risnes
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Norway.,Centre for Immune Regulation, University of Oslo and Oslo University Hospital, Norway
| | - Shiva Dahal-Koirala
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Norway.,Centre for Immune Regulation, University of Oslo and Oslo University Hospital, Norway
| | - Ludvig M Sollid
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Norway.,K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Norway.,Centre for Immune Regulation, University of Oslo and Oslo University Hospital, Norway
| | - Knut E A Lundin
- K. G. Jebsen Coeliac Disease Research Centre, University of Oslo, Norway.,Centre for Immune Regulation, University of Oslo and Oslo University Hospital, Norway.,Department of Gastroenterology, Oslo University Hospital, Norway
| |
Collapse
|
42
|
Szabady RL, Louissaint C, Lubben A, Xie B, Reeksting S, Tuohy C, Demma Z, Foley SE, Faherty CS, Llanos-Chea A, Olive AJ, Mrsny RJ, McCormick BA. Intestinal P-glycoprotein exports endocannabinoids to prevent inflammation and maintain homeostasis. J Clin Invest 2018; 128:4044-4056. [PMID: 30102254 DOI: 10.1172/jci96817] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 06/19/2018] [Indexed: 01/09/2023] Open
Abstract
Neutrophil influx into the intestinal lumen is a critical response to infectious agents, but is also associated with severe intestinal damage observed in idiopathic inflammatory bowel disease. The chemoattractant hepoxilin A3, an eicosanoid secreted from intestinal epithelial cells by the apically restricted efflux pump multidrug resistance protein 2 (MRP2), mediates this neutrophil influx. Information about a possible counterbalance pathway that could signal the lack of or resolution of an apical inflammatory signal, however, has yet to be described. We now report a system with such hallmarks. Specifically, we identify endocannabinoids as the first known endogenous substrates of the apically restricted multidrug resistance transporter P-glycoprotein (P-gp) and reveal a mechanism, which we believe is novel, for endocannabinoid secretion into the intestinal lumen. Knockdown or inhibition of P-gp reduced luminal secretion levels of N-acyl ethanolamine-type endocannabinoids, which correlated with increased neutrophil transmigration in vitro and in vivo. Additionally, loss of CB2, the peripheral cannabinoid receptor, led to increased pathology and neutrophil influx in models of acute intestinal inflammation. These results define a key role for epithelial cells in balancing the constitutive secretion of antiinflammatory lipids with the stimulated secretion of proinflammatory lipids via surface efflux pumps in order to control neutrophil infiltration into the intestinal lumen and maintain homeostasis in the healthy intestine.
Collapse
Affiliation(s)
- Rose L Szabady
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Christopher Louissaint
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anneke Lubben
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Bailu Xie
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Shaun Reeksting
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Christine Tuohy
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Zachary Demma
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sage E Foley
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Christina S Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Alejandro Llanos-Chea
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrew J Olive
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Randall J Mrsny
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Beth A McCormick
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
43
|
Yang G, Chen S, Deng B, Tan C, Deng J, Zhu G, Yin Y, Ren W. Implication of G Protein-Coupled Receptor 43 in Intestinal Inflammation: A Mini-Review. Front Immunol 2018; 9:1434. [PMID: 29988393 PMCID: PMC6023978 DOI: 10.3389/fimmu.2018.01434] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Accepted: 06/11/2018] [Indexed: 12/27/2022] Open
Abstract
Short chain fatty acids (SCFAs, e.g., acetate, propionate, and butyrate) are a subset of fatty acids that are produced by gut microbiota during the fermentation of dietary fiber. They modulate different processes in the gastrointestinal tract and play various positive roles in mediating the intestinal health. Most beneficial roles of SCFAs in the gastrointestinal tract are mediated by directly activating its receptor, G protein-coupled receptor 43 (GPR43, also known as FFAR2). Various recent studies have demonstrated the role of GPR43 in intestinal inflammatory diseases, such as inflammatory bowel diseases. These SCFAs-mediated regulations of intestinal health are associated with neutrophil chemotaxis, T cell differentiation, activation, and subsequent cytokines production. Therefore, GPR43 could potentially be a drug target for intestinal inflammatory diseases. In this review, we review the current knowledge on the regulatory mechanisms associated with GPR43 in intestinal inflammation. The role of GPR43-mediated regulation of antibody responses is also discussed.
Collapse
Affiliation(s)
- Guan Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Siyuan Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Baichuan Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Chengquan Tan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jinping Deng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Guoqiang Zhu
- Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yulong Yin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, Subtropical Institute of Animal Nutrition and Feed, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
44
|
Rabah H, Ménard O, Gaucher F, do Carmo FLR, Dupont D, Jan G. Cheese matrix protects the immunomodulatory surface protein SlpB of Propionibacterium freudenreichii during in vitro digestion. Food Res Int 2018; 106:712-721. [DOI: 10.1016/j.foodres.2018.01.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 12/19/2022]
|
45
|
Wang B, Yang A, Zhao Z, He C, Liu Y, Colman RW, Dai J, Wu Y. The Plasma Kallikrein-Kininogen Pathway Is Critical in the Pathogenesis of Colitis in Mice. Front Immunol 2018; 9:21. [PMID: 29467753 PMCID: PMC5808240 DOI: 10.3389/fimmu.2018.00021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/04/2018] [Indexed: 12/17/2022] Open
Abstract
The kallikrein-kinin system (KKS) consists of two serine proteases, prekallikrein (pKal) and factor XII (FXII), and a cofactor, high-molecular-weight kininogen (HK). Upon activation of the KKS, HK is cleaved to release bradykinin. Although the KKS is activated in humans and animals with inflammatory bowel disease (IBD), its role in the pathogenesis of IBD has not been characterized. In the present study, we determined the role of the KKS in the pathogenesis of IBD using mice that lack proteins involved in the KKS. In two colitis models, induced by dextran sulfate sodium (DSS) or 2,4,6-trinitrobenzene sulfonic acid (TNBS), mice deficient in HK, pKal, or bradykinin receptors displayed attenuated phenotypes, including body weight loss, disease activity index, colon length shortening, histological scoring, and colonic production of cytokines. Infiltration of neutrophils and inflammatory monocytes in the colonic lamina propria was reduced in HK-deficient mice. Reconstitution of HK-deficient mice through intravenous injection of HK recovered their susceptibility to DSS-induced colitis, increased IL-1β levels in the colon tissue and bradykinin concentrations in plasma. In contrast to the phenotypes of other mice lacking other proteins involved in the KKS, mice lacking FXII had comparable colonic inflammation to that observed in wild-type mice. The concentration of bradykinin was significantly increased in the plasma of wild-type mice after DSS-induced colitis. In vitro analysis revealed that DSS-induced pKal activation, HK cleavage, and bradykinin plasma release were prevented by the absence of pKal or the inhibition of Kal. Unlike DSS, TNBS-induced colitis did not trigger HK cleavage. Collectively, our data strongly suggest that Kal, acting independently of FXII, contributes to experimental colitis by promoting bradykinin release from HK.
Collapse
Affiliation(s)
- Bo Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Aizhen Yang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zhenzhen Zhao
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Chao He
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuanyuan Liu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Robert W. Colman
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, United States
| | - Jihong Dai
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, United States
| | - Yi Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- The Sol Sherry Thrombosis Research Center, Temple University School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
46
|
Tsai MC, Kao LT, Lin HC, Lee CZ, Chung SD. Chronic prostatitis/chronic pelvic pain syndrome is associated with previous colonoscopy. Can Urol Assoc J 2017; 11:E367-E371. [PMID: 29382459 DOI: 10.5489/cuaj.4279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION This study aimed to examine the association between chronic prostatitis/chronic pelvic pain syndrome (CP/CPPS) and receipt of a prior colonoscopic examination using a population-based database. METHODS We used the Taiwan Longitudinal Health Insurance Database 2005 to retrieve the study sample. This study included 3933 patients with CP/CPPS and 3933 age-matched controls. We designated the date of receiving the first diagnosis of CP/CPPS as the index date for cases. We defined the first an ambulatory care visit occurring in the matched year as the index date for the controls. Conditional logistic regressions was used to calculate the odds ratio (OR) and 95% confidence interval (CI) for having previously received colonoscopy between cases and controls. RESULTS We found that 349 (4.44%) of the 7866 sampled patients had previously undergone colonoscopy, including 223 (5.67%) cases and 126 (3.20%) controls (p<0.001). A conditional logistic regression analysis revealed that the adjusted OR of receiving a colonoscopy within three years before the index date was 1.77 (95% CI 1.42-2.23) for cases compared to controls. Furthermore, we found that the youngest group of cases (<40 years) had the greatest adjusted OR for having received colonoscopy within three years before the index date compared to controls (OR 2.81; 95% CI 1.45-5.44); however, in contrast, no significant difference in the adjusted odds of having previously received colonoscopy was observed between cases and controls among the oldest age group (≥60 years). CONCLUSIONS We concluded that there was an association between antecedent colonoscopy and CP/CPPS.
Collapse
Affiliation(s)
- Ming-Chieh Tsai
- Division of Gastroenterology, Department of Internal Medicine, Cathay General Hospital, Hsinchu Branch, Hsinchu; Taiwan.,School of Health Care Administration, Taipei Medical University, Taipei; Taiwan
| | - Li-Ting Kao
- Graduate Institute of Life Science, National Defense Medical Centre, Taipei; Taiwan
| | - Herng-Ching Lin
- School of Health Care Administration, Taipei Medical University, Taipei; Taiwan.,Research Centre of Sleep Medicine, College of Medicine, Taipei Medical University, Taipei; Taiwan
| | - Cha-Ze Lee
- Department of Internal Medicine, National Taiwan University Hospital, Taipei; Taiwan
| | - Shiu-Dong Chung
- Research Centre of Sleep Medicine, College of Medicine, Taipei Medical University, Taipei; Taiwan.,Division of Urology, Department of Surgery, Far Eastern Memorial Hospital, Banciao, Taipei; Taiwan.,Graduate Program in Biomedical Informatics, College of Informatics, Yuan-Ze University, Chungli; Taiwan
| |
Collapse
|
47
|
Zhou GX, Liu ZJ. Potential roles of neutrophils in regulating intestinal mucosal inflammation of inflammatory bowel disease. J Dig Dis 2017; 18:495-503. [PMID: 28857501 DOI: 10.1111/1751-2980.12540] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/15/2017] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel diseases (IBD), comprising of ulcerative colitis and Crohn's disease, are inflammatory disorders of the gastrointestinal tract characterized by chronically relapsing mucosal inflammation. Neutrophils, as the effector cells of acute inflammation, have long been reported to play a role in the maintenance of intestinal homeostasis and pathogenesis of IBD. At the early stage of mucosal inflammation in patients with IBD, neutrophils flood into intestinal mucosa, phagocytose pathogenic microbes, and promote mucosal healing and resolution of inflammation. However, large numbers of neutrophils infiltrating in the inflamed mucosa and accumulating in the epithelia cause damage of mucosal architecture, compromised epithelial barrier and production of inflammatory mediators. In this review we discuss the critical roles of neutrophils in modulating innate and adaptive immune responses in intestinal mucosa, and, importantly, clarify the potential roles of neutrophils related to their production of inflammatory mediators, transenthothelial and transepithelial migration into intestinal mucosa, and the underlying mechanisms in regulating mucosal inflammation of IBD. Moreover, we also describe a new subset of neutrophils (i.e., CD177+ neutrophils) and illustrate its protective role in modulating intestinal mucosal immune responses in IBD.
Collapse
Affiliation(s)
- Guang Xi Zhou
- Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| | - Zhan Ju Liu
- Department of Gastroenterology, The Shanghai Tenth People's Hospital of Tongji University, Shanghai, China
| |
Collapse
|
48
|
Zhang H, Hassan YI, Renaud J, Liu R, Yang C, Sun Y, Tsao R. Bioaccessibility, bioavailability, and anti-inflammatory effects of anthocyanins from purple root vegetables using mono- and co-culture cell models. Mol Nutr Food Res 2017; 61. [PMID: 28691370 DOI: 10.1002/mnfr.201600928] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/05/2017] [Accepted: 05/30/2017] [Indexed: 01/21/2023]
Abstract
SCOPE Immune-inflammatory signaling and metabolic effects are the main pillars for bioactivity of anthocyanins derived from highly pigmented root vegetables. This study aims to assess the bioaccessibility and bioavailability of purple carrot and potato derived anthocyanins and the molecular mechanisms of their ability to ameliorate cellular inflammation in a mono- and co-culture cell models. METHODS AND RESULTS An in vitro gastrointestinal model was used and demonstrated bioaccessibility of 44.62 and 71.8% for anthocyanins of purple carrot and potato, respectively. These accessible anthocyanins significantly inhibited cellular inflammation in Caco-2 cells. Intact cyanidinglycoside or petunidinglycoside (respectively from carrots and potatoes) were transported across a transmembrane cell model and detected by LC-MS/MS. Computational docking and glucose uptake analyses suggested uptake of anthocyanins was mediated by hexose transporters. Subsequent experiment using an inflamed Caco-2 BBe1/THP-1 co-culture cell model showed these transported anthocyanins inhibited IL-8 and TNF-α secretion,and expression of pro-inflammatory cytokines by blocking NF-κB, and MAPK mediated inflammatory cellular signaling cascades, but with varying degrees due to structural features. CONCLUSION Anthocyanins from purple carrots and potatoes possess a promising anti-inflammatory effect in model gut system. They can be absorbed and act differently but are in general beneficial for inflammation-mediated diseases.
Collapse
Affiliation(s)
- Hua Zhang
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| | - Yousef I Hassan
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| | - Justin Renaud
- London Research and Development Centre, Agriculture and Agri-Food Canada, London, ON, Canada
| | - Ronghua Liu
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| | - Cheng Yang
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| | - Yong Sun
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| | - Rong Tsao
- Guelph Food Research Centre, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| |
Collapse
|
49
|
Pazos MA, Lanter BB, Yonker LM, Eaton AD, Pirzai W, Gronert K, Bonventre JV, Hurley BP. Pseudomonas aeruginosa ExoU augments neutrophil transepithelial migration. PLoS Pathog 2017; 13:e1006548. [PMID: 28771621 PMCID: PMC5557605 DOI: 10.1371/journal.ppat.1006548] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 08/15/2017] [Accepted: 07/22/2017] [Indexed: 12/21/2022] Open
Abstract
Excessive neutrophil infiltration of the lungs is a common contributor to immune-related pathology in many pulmonary disease states. In response to pathogenic infection, airway epithelial cells produce hepoxilin A3 (HXA3), initiating neutrophil transepithelial migration. Migrated neutrophils amplify this recruitment by producing a secondary gradient of leukotriene B4 (LTB4). We sought to determine whether this two-step eicosanoid chemoattractant mechanism could be exploited by the pathogen Pseudomonas aeruginosa. ExoU, a P. aeruginosa cytotoxin, exhibits phospholipase A2 (PLA2) activity in eukaryotic hosts, an enzyme critical for generation of certain eicosanoids. Using in vitro and in vivo models of neutrophil transepithelial migration, we evaluated the impact of ExoU expression on eicosanoid generation and function. We conclude that ExoU, by virtue of its PLA2 activity, augments and compensates for endogenous host neutrophil cPLA2α function, leading to enhanced transepithelial migration. This suggests that ExoU expression in P. aeruginosa can circumvent immune regulation at key signaling checkpoints in the neutrophil, resulting in exacerbated neutrophil recruitment. Pseudomonas aeruginosa is an opportunistic pathogen that causes acute pneumonia in immune compromised patients, and infects 70–80% of patients suffering from cystic fibrosis. Infections can result in excessive airway inflammation, which lead to immune-mediated lung damage, in particular through the action of recruited white blood cells known as neutrophils. Certain strains of P. aeruginosa produce the exotoxin ExoU, which has been associated with increased virulence. ExoU causes host cell lysis by hydrolyzing host membrane lipids through its phospholipase activity. However, host phospholipases play a key role in immune signaling by mediating the production of lipids known as eicosanoids. We investigated whether separate from its cytolytic activity, ExoU could modulate host immune responses through its phospholipase activity by hijacking eicosanoid production. Using in vitro and in vivo models of neutrophil recruitment, we find that ExoU producing strains of P. aeruginosa elicit higher levels of the eicosanoid chemoattractant leukotriene B4 from migrated neutrophils. This results in increased neutrophil transepithelial migration. This work reveals a new mechanism for how bacterial pathogens alter our immune function, and highlights a new potential therapeutic strategy for moderating Pseudomonas pathogenesis in patients with cystic fibrosis and acute pneumonia.
Collapse
Affiliation(s)
- Michael A. Pazos
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
- Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Bernard B. Lanter
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
- Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lael M. Yonker
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
- Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Alex D. Eaton
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
| | - Waheed Pirzai
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
| | - Karsten Gronert
- Vision Science Program, School of Optometry, University of California at Berkeley, Berkeley, California, United States of America
| | - Joseph V. Bonventre
- Renal Division and Biomedical Engineering Division, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Bryan P. Hurley
- Mucosal Immunology & Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, United States of America
- Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
50
|
Neutrophil Elastase and Interleukin 17 Expressed in the Pig Colon during Brachyspira hyodysenteriae Infection Synergistically with the Pathogen Induce Increased Mucus Transport Speed and Production via Mitogen-Activated Protein Kinase 3. Infect Immun 2017; 85:IAI.00262-17. [PMID: 28559407 DOI: 10.1128/iai.00262-17] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023] Open
Abstract
Brachyspira hyodysenteriae colonizes the pig colon, resulting in mucoid hemorrhagic diarrhea and mucus layer changes. These changes are characterized by a disorganized mucus structure and massive mucus induction with de novo expression of MUC5AC and increased production of MUC2. To investigate the mechanisms behind this altered mucin environment, we quantified the mRNA levels of mucin pathway genes and factors from the immune system in the colons of infected and control pigs and observed upregulation of neutrophil elastase, SPDEF, FOXA3, MAPK3/ERK1, IL-17A, IL-1β, IL-6, and IL-8 expression. In vitro, colonic mucus-producing mucosal surfaces were treated with these factors along with B. hyodysenteriae infection and analyzed for their effect on mucin production. Neutrophil elastase and infection synergistically induced mucus production and transport speed, and interleukin 17A (IL-17A) also had similar effects, in both the presence and absence of infection. A mitogen-activated protein kinase 3 (MAPK3)/extracellular signal-regulated kinase 1 (ERK1) inhibitor suppressed these effects. Therefore, we suggest that the SPDEF, FOXA3, and MAPK3/ERK1 signaling pathways are behind the transcriptional program regulating mucin biosynthesis in the colon during B. hyodysenteriae infection. In addition to furthering the knowledge on this economically important disease, this mechanism may be useful for the development of therapies aimed at conditions where enhancing mucus production may be beneficial, such as chronic inflammatory disorders of the colon.
Collapse
|