1
|
Mayorova TD, Koch TL, Kachar B, Jung JH, Reese TS, Smith CL. Placozoan secretory cell types implicated in feeding, innate immunity and regulation of behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.18.613768. [PMID: 39372748 PMCID: PMC11452194 DOI: 10.1101/2024.09.18.613768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Placozoa are millimeter-sized, flat, irregularly shaped ciliated animals that crawl on surfaces in warm oceans feeding on biofilms, which they digest externally. They stand out from other animals due to their simple body plans. They lack organs, body cavities, muscles and a nervous system and have only seven broadly defined morphological cell types, each with a unique distribution. Analyses of single cell transcriptomes of four species of placozoans revealed greater diversity of secretory cell types than evident from morphological studies, but the locations of many of these new cell types were unknown and it was unclear which morphological cell types they represent. Furthermore, there were contradictions between the conclusions of previous studies and the single cell RNAseq studies. To address these issues, we used mRNA probes for genes encoding secretory products expressed in different metacells in Trichoplax adhaerens to localize cells in whole mounts and in dissociated cell cultures, where their morphological features could be visualized and identified. The nature and functions of their secretory granules were further investigated with electron microscopic techniques and by imaging secretion in live animals during feeding episodes. We found that two cell types participate in disintegrating prey, one resembling a lytic cell type in mammals and another combining features of zymogen gland cells and enterocytes. We identified secretory epithelial cells expressing glycoproteins or short peptides implicated in defense. We located seven peptidergic cell types and two types of mucocytes. Our findings reveal mechanisms that placozoans use to feed and protect themselves from pathogens and clues about neuropeptidergic signaling. We compare placozoan secretory cell types with cell types in other animal phyla to gain insight about general evolutionary trends in cell type diversification, as well as pathways leading to the emergence of synapomorphies.
Collapse
Affiliation(s)
- Tatiana D. Mayorova
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - Thomas Lund Koch
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Bechara Kachar
- Section on Structural Cell Biology, Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States of America
| | - Jae Hoon Jung
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - Thomas S. Reese
- Laboratory of Neurobiology, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| | - Carolyn L. Smith
- Light Imaging Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
2
|
Cidem A, Chang GRL, Yen CC, Chen MS, Yang SH, Chen CM. Lactoferrin targeting INTL1 receptor inhibits hepatocellular carcinoma progression via apoptosis and cell cycle signaling pathways. Sci Rep 2024; 14:31210. [PMID: 39732873 DOI: 10.1038/s41598-024-82514-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
Hepatocellular carcinoma (HCC) constitutes 90% of liver cancer cases and ranks as the third leading cause of cancer-related mortality, necessitating urgent development of alternative therapies. Lactoferrin (LF), a natural iron-binding glycoprotein with reported anticancer effects, is investigated for its potential in liver cancer treatment, an area with limited existing studies. This study focuses on evaluating LF's anti-liver cancer effects on HCC cells and assessing the preventive efficacy of oral LF administration in a murine model. Data showed that LF exerted anti-proliferative effects on HepG2, Hep3B, and SK-Hep1 cells while having no cytotoxicity on healthy liver cells (FL83B). Mechanistically, LF induces mitochondrial-mediated apoptosis and G0/G1 cell cycle arrest in HepG2 cells, associated with increased phosphorylation of p38 MAPK and JNK for apoptosis, and ERK phosphorylation for cell cycle arrest. Intelectin-1 (INTL1) is identified as the receptor facilitating LF endocytosis in HepG2 cells, and downregulation of INTL1 inhibits LF-induced signaling pathways. Notably, oral LF administration prevents HCC development in nude mice with orthotopic HepG2 cell injection. This study unveils the mechanistic basis of LF action in HepG2 cells, showcasing its potential in HCC prevention. Importantly, we report the novel identification of INTL1 as the LF receptor in HepG2 cells, providing valuable insights for future exploration of LF and its derivatives in liver cancer therapy.
Collapse
Affiliation(s)
- Abdulkadir Cidem
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, Kuo Kuang Rd., Taichung, 402, Taiwan
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25250, Turkey
| | - Gary Ro-Lin Chang
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, Kuo Kuang Rd., Taichung, 402, Taiwan
| | - Chih-Ching Yen
- Department of Internal Medicine, China Medical University Hospital, College of Health Care, China Medical University, Taichung, 404, Taiwan
| | - Ming-Shan Chen
- Department of Anesthesiology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chia-Yi, 600, Taiwan
| | - Shang-Hsun Yang
- Department of Physiology, Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, College of Life Sciences, National Chung Hsing University, Kuo Kuang Rd., Taichung, 402, Taiwan.
- The iEGG and Animal Biotechnology Center, and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, 402, Taiwan.
- Center for General Educational, National Quemoy University, Kinmen, 892, Taiwan.
| |
Collapse
|
3
|
Liu L, Davidorf B, Dong P, Peng A, Song Q, He Z. Decoding the mosaic of inflammatory bowel disease: Illuminating insights with single-cell RNA technology. Comput Struct Biotechnol J 2024; 23:2911-2923. [PMID: 39421242 PMCID: PMC11485491 DOI: 10.1016/j.csbj.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 10/19/2024] Open
Abstract
Inflammatory bowel diseases (IBD), comprising ulcerative colitis (UC) and Crohn's disease (CD), are complex chronic inflammatory intestinal conditions with a multifaceted pathology, influenced by immune dysregulation and genetic susceptibility. The challenges in understanding IBD mechanisms and implementing precision medicine include deciphering the contributions of individual immune and non-immune cell populations, pinpointing specific dysregulated genes and pathways, developing predictive models for treatment response, and advancing molecular technologies. Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful tool to address these challenges, offering comprehensive transcriptome profiles of various cell types at the individual cell level in IBD patients, overcoming limitations of bulk RNA sequencing. Additionally, single-cell proteomics analysis, T-cell receptor repertoire analysis, and epigenetic profiling provide a comprehensive view of IBD pathogenesis and personalized therapy. This review summarizes significant advancements in single-cell sequencing technologies for enhancing our understanding of IBD, covering pathogenesis, diagnosis, treatment, and prognosis. Furthermore, we discuss the challenges that persist in the context of IBD research, including the need for longitudinal studies, integration of multiple single-cell and spatial transcriptomics technologies, and the potential of microbial single-cell RNA-seq to shed light on the role of the gut microbiome in IBD.
Collapse
Affiliation(s)
- Liang Liu
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin Davidorf
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peixian Dong
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Alice Peng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Qianqian Song
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Zhiheng He
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
4
|
Rosa L, Cutone A, Ianiro G, Valenti P, Paesano R. Lactoferrin in the treatment of interstitial cystitis: a retrospective pilot study. Biochem Cell Biol 2024; 102:506-514. [PMID: 39088844 DOI: 10.1139/bcb-2024-0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2024] Open
Abstract
Interstitial cystitis (IC), defined as a painful bladder syndrome (PBS), is a chronic condition that manifests itself as a suprapubic pain associated with an enhancing of frequency/urgency of urination, and for which there is no cure. Here, we present a retrospective pilot study on women affected from IC/PBS and treated with bovine lactoferrin (bLf). A total of 31 women, affected (20) or unaffected (11) from hereditary thrombophilia (HT), presented the median of 6 episodes of IC/PBS during the 6 months before the study. Treatment consisted of 17 weeks of orally administered Valpalf® capsules, containing bLf plus sodium bicarbonate and citrate. Out of 31 patients, only 3 women had one episode of IC/PBS during the follow-up period, while no episode was observed in 28 women. In the HT group, a significant decrease in both serum IL-6 and D-dimers was found after Valpalf® treatment. Moreover, in Valpalf®-treated women, cystoscopy revealed a global improvement in the appearance of the bladder, especially in term of inflammation/irritation and presence of Hunner ulcers. Even if our results must be corroborated by randomized double-blinded controlled trials on a larger number of patients, our observations indicate that bLf treatment is efficient in relieving IC/PBS symptoms, without side effects.
Collapse
Affiliation(s)
- Luigi Rosa
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, Pesche, Italy
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
5
|
Manzoni P, Messina A, Germano C, Picone S, Masturzo B, Sainaghi PP, Sola D, Rizzi M. Lactoferrin Supplementation in Preventing and Protecting from SARS-CoV-2 Infection: Is There Any Role in General and Special Populations? An Updated Review of Literature. Int J Mol Sci 2024; 25:10248. [PMID: 39408576 PMCID: PMC11476995 DOI: 10.3390/ijms251910248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/07/2024] [Accepted: 09/21/2024] [Indexed: 10/19/2024] Open
Abstract
At the beginning of the pandemic, SARS-CoV-2 infection represented a great medical burden worldwide, as targeted and effective therapeutic options were lacking. This resulted in the revival of existing molecules and the increasing popularity of over-the-counter nutritional supplements. Among the latter, lactoferrin has been investigated as an adjuvant in COVID-19 therapy with conflicting results, mainly depending on different study designs. Considering that lactoferrin is one of the main components of human breast milk with anti-microbial and anti-inflammatory activity, it is conceivable that such bioactive molecule could be effective in supporting anti-SARS-CoV-2 infection therapy, especially in infants and pregnant women, two subpopulations that have been poorly evaluated in the existing clinical trials. This narrative review is intended to offer insight into the existing literature on lactoferrin's biological functions and protective effects against COVID-19, with a special focus on pregnant women and their infants.
Collapse
Affiliation(s)
- Paolo Manzoni
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Alessandro Messina
- School of Medicine, University of Turin, 10124 Turin, Italy;
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Chiara Germano
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Simonetta Picone
- Neonatology and Neonatal Intensive Care Unit, Policlinico Casilino, 00169 Rome, Italy
| | - Bianca Masturzo
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Pier Paolo Sainaghi
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- IRCAD (Interdisciplinary Research Center of Autoimmune Diseases), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| | - Daniele Sola
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Laboratory of Metabolic Research, IRCCS Istituto Auxologico Italiano, S. Giuseppe Hospital, 28824 Piancavallo, Italy
| | - Manuela Rizzi
- IRCAD (Interdisciplinary Research Center of Autoimmune Diseases), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Department of Health Sciences (DiSS), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| |
Collapse
|
6
|
Kim B, Jang S, Jang H, Kim JS, Jeon TI, Park JG, Shin IS, Cho KO, Moon C. Lectin histochemistry in the small intestines of piglets naturally infected with porcine epidemic diarrhea virus. J Vet Sci 2024; 25:e66. [PMID: 39363654 PMCID: PMC11450395 DOI: 10.4142/jvs.24179] [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: 06/18/2024] [Revised: 07/21/2024] [Accepted: 08/04/2024] [Indexed: 10/05/2024] Open
Abstract
IMPORTANCE Porcine epidemic diarrhea virus (PEDV) binds to particular cell surface receptors to penetrate cells. The virus specifically identifies certain carbohydrate structures present on the surface of the cell to facilitate the binding process. Nevertheless, the influence of viral infections on specific alterations of glycoconjugates in the small intestines remains unexplored. OBJECTIVE This work aimed to examine the alterations in glycoconjugates in the small intestines of piglets naturally infected with PEDV using lectin histochemistry. METHODS Six piglets including three PEDV-infected and three non-infected piglets were evaluated. Small intestinal samples were histopathologically examined, and lectin histochemistry was performed. RESULTS Piglets infected with PEDV had significant histological abnormalities in their small intestines, such as pronounced villous atrophy, varying degrees of villous fusion, and diverse mucosal alterations. Specific regions of the duodenum, jejunum, and ileum showed discernible variations in glycoconjugate distribution, as determined by lectin histochemistry. Compared with the controls, the PEDV-infected piglets showed significant changes in N-acetylglucosamine- and galactose-binding lectins (particularly wheat germ agglutinin and Arachis hypogaea (peanut) agglutinin) in multiple intestinal regions. CONCLUSIONS AND RELEVANCE These findings can enhance understanding of how viruses such as PEDV impact the glycoconjugate composition of the small intestines and emphasize the potential connection between the pathogenesis of PEDV and glycoconjugate.
Collapse
Affiliation(s)
- Bohye Kim
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Sungwoong Jang
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Hyewon Jang
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Joong-Sun Kim
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Tae-Il Jeon
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Jun-Gyu Park
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - In-Sik Shin
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Kyoung-Oh Cho
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea
| | - Changjong Moon
- College of Veterinary Medicine and Host-Directed Antiviral Research Center, Chonnam National University, Gwangju 61186, Korea.
| |
Collapse
|
7
|
Gori A, Brindisi G, Daglia M, del Giudice MM, Dinardo G, Di Minno A, Drago L, Indolfi C, Naso M, Trincianti C, Tondina E, Brunese FP, Ullah H, Varricchio A, Ciprandi G, Zicari AM. Exploring the Role of Lactoferrin in Managing Allergic Airway Diseases among Children: Unrevealing a Potential Breakthrough. Nutrients 2024; 16:1906. [PMID: 38931261 PMCID: PMC11206375 DOI: 10.3390/nu16121906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of allergic diseases has dramatically increased among children in recent decades. These conditions significantly impact the quality of life of allergic children and their families. Lactoferrin, a multifunctional glycoprotein found in various biological fluids, is emerging as a promising immunomodulatory agent that can potentially alleviate allergic diseases in children. Lactoferrin's multifaceted properties make it a compelling candidate for managing these conditions. Firstly, lactoferrin exhibits potent anti-inflammatory and antioxidant activities, which can mitigate the chronic inflammation characteristic of allergic diseases. Secondly, its iron-binding capabilities may help regulate the iron balance in allergic children, potentially influencing the severity of their symptoms. Lactoferrin also demonstrates antimicrobial properties, making it beneficial in preventing secondary infections often associated with respiratory allergies. Furthermore, its ability to modulate the immune response and regulate inflammatory pathways suggests its potential as an immune-balancing agent. This review of the current literature emphasises the need for further research to elucidate the precise roles of lactoferrin in allergic diseases. Harnessing the immunomodulatory potential of lactoferrin could provide a novel add-on approach to managing allergic diseases in children, offering hope for improved outcomes and an enhanced quality of life for paediatric patients and their families. As lactoferrin continues to capture the attention of researchers, its properties and diverse applications make it an intriguing subject of study with a rich history and a promising future.
Collapse
Affiliation(s)
- Alessandra Gori
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.B.)
| | - Giulia Brindisi
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.B.)
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (M.D.); (A.D.M.); (H.U.)
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Michele Miraglia del Giudice
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.M.d.G.); (G.D.); (C.I.)
| | - Giulio Dinardo
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.M.d.G.); (G.D.); (C.I.)
| | - Alessandro Di Minno
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (M.D.); (A.D.M.); (H.U.)
- CEINGE-Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Naples, Italy
| | - Lorenzo Drago
- Laboratory of Clinical Microbiology & Microbiome, Department of Biomedical Sciences for Health, University of Milan, 20122 Milan, Italy;
- UOC Laboratory of Clinical Medicine, MultiLab Department, IRCCS Multimedica, 20138 Milan, Italy
| | - Cristiana Indolfi
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (M.M.d.G.); (G.D.); (C.I.)
| | - Matteo Naso
- Allergy Center, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.N.); (C.T.)
| | - Chiara Trincianti
- Allergy Center, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (M.N.); (C.T.)
| | - Enrico Tondina
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | | | - Hammad Ullah
- Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Naples, Italy; (M.D.); (A.D.M.); (H.U.)
| | - Attilio Varricchio
- Department of Otolaryngology, University of Molise, 86100 Campobasso, Italy;
| | - Giorgio Ciprandi
- Allergy Clinic, Casa di Cura Villa Montallegro, 16145 Genoa, Italy;
| | - Anna Maria Zicari
- Department of Mother-Child, Urological Science, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.B.)
| |
Collapse
|
8
|
Kim K, Park S, Lee Y, Baek J, Kim Y, Hwang SW, Lee JL, Park SH, Yang SK, Han B, Song K, Yoon YS, Lee HS, Ye BD. Transcriptomic Profiling and Cellular Composition of Creeping Fat in Crohn's disease. J Crohns Colitis 2024; 18:223-232. [PMID: 37594364 DOI: 10.1093/ecco-jcc/jjad141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND AND AIMS Creeping fat [CF] is a poorly understood feature of Crohn's disease [CD], characterized by the wrapping of mesenteric adipose tissue [MAT] around the inflamed intestine. The aim of this study was to investigate the transcriptional profile and compositional features of CF. METHODS We collected 59 MAT samples: 23 paired samples from patients with CD (CF [CD-CF] and MAT around the uninflamed intestine [CD-MAT]) and 13 MAT samples from non-CD patients [Con-MAT]. Differentially expressed gene [DEG], functional pathway, cell deconvolution, and gene co-expression network analyses were performed. RESULTS By comparing three different MAT samples, we identified a total of 529 DEGs [|log2FoldChange| > 1.5; false discovery rate < 0.05]. Of these, 323 genes showed an incremental pattern from Con-MAT to CD-MAT, and to CD-CF, while 105 genes displayed a decremental pattern. Genes with an incremental pattern were related to immune cell responses, including B- and T-cell activation, while genes with a decremental pattern were involved in cell trafficking and migration. Cell deconvolution analysis revealed significant changes in cellular composition between the CD-CF and Con-MAT groups, with increased proportions of B-cells/plasma cells [p = 1.16 × 10-4], T-cells [p = 3.66 × 10-3], and mononuclear phagocytes [p = 3.53 × 10-2] in the CD-CF group. In contrast, only the B-cell/plasma cell component showed a significant increase [p = 1.62 × 10-2] in the CD-MAT group compared to Con-MAT. CONCLUSION The distinct transcriptional profiles and altered cellular components of each MAT found in our study provide insight into the mechanisms behind CF and highlight its possible role in the pathogenesis of CD.
Collapse
Affiliation(s)
- Kyuwon Kim
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sojung Park
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yoonho Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jiwon Baek
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yongjae Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sung Wook Hwang
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jong Lyul Lee
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Division of Colon and Rectal Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang Hyoung Park
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Buhm Han
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea
| | - Kyuyoung Song
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yong Sik Yoon
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Division of Colon and Rectal Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ho-Su Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Byong Duk Ye
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
9
|
Ianiro G, Niro A, Rosa L, Valenti P, Musci G, Cutone A. To Boost or to Reset: The Role of Lactoferrin in Energy Metabolism. Int J Mol Sci 2023; 24:15925. [PMID: 37958908 PMCID: PMC10650157 DOI: 10.3390/ijms242115925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Many pathological conditions, including obesity, diabetes, hypertension, heart disease, and cancer, are associated with abnormal metabolic states. The progressive loss of metabolic control is commonly characterized by insulin resistance, atherogenic dyslipidemia, inflammation, central obesity, and hypertension, a cluster of metabolic dysregulations usually referred to as the "metabolic syndrome". Recently, nutraceuticals have gained attention for the generalized perception that natural substances may be synonymous with health and balance, thus becoming favorable candidates for the adjuvant treatment of metabolic dysregulations. Among nutraceutical proteins, lactoferrin (Lf), an iron-binding glycoprotein of the innate immune system, has been widely recognized for its multifaceted activities and high tolerance. As this review shows, Lf can exert a dual role in human metabolism, either boosting or resetting it under physiological and pathological conditions, respectively. Lf consumption is safe and is associated with several benefits for human health, including the promotion of oral and gastrointestinal homeostasis, control of glucose and lipid metabolism, reduction of systemic inflammation, and regulation of iron absorption and balance. Overall, Lf can be recommended as a promising natural, completely non-toxic adjuvant for application as a long-term prophylaxis in the therapy for metabolic disorders, such as insulin resistance/type II diabetes and the metabolic syndrome.
Collapse
Affiliation(s)
- Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Antonella Niro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (L.R.); (P.V.)
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (L.R.); (P.V.)
| | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| | - Antimo Cutone
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (G.I.); (A.N.); (G.M.)
| |
Collapse
|
10
|
Conesa C, Bellés A, Grasa L, Sánchez L. The Role of Lactoferrin in Intestinal Health. Pharmaceutics 2023; 15:1569. [PMID: 37376017 DOI: 10.3390/pharmaceutics15061569] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
The intestine represents one of the first barriers where microorganisms and environmental antigens come into tight contact with the host immune system. A healthy intestine is essential for the well-being of humans and animals. The period after birth is a very important phase of development, as the infant moves from a protected environment in the uterus to one with many of unknown antigens and pathogens. In that period, mother's milk plays an important role, as it contains an abundance of biologically active components. Among these components, the iron-binding glycoprotein, lactoferrin (LF), has demonstrated a variety of important benefits in infants and adults, including the promotion of intestinal health. This review article aims to provide a compilation of all the information related to LF and intestinal health, in infants and adults.
Collapse
Affiliation(s)
- Celia Conesa
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Andrea Bellés
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lourdes Sánchez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| |
Collapse
|
11
|
Matute JD, Duan J, Flak MB, Griebel P, Tascon-Arcila JA, Doms S, Hanley T, Antanaviciute A, Gundrum J, Mark Welch JL, Sit B, Abtahi S, Fuhler GM, Grootjans J, Tran F, Stengel ST, White JR, Krupka N, Haller D, Clare S, Lawley TD, Kaser A, Simmons A, Glickman JN, Bry L, Rosenstiel P, Borisy G, Waldor MK, Baines JF, Turner JR, Blumberg RS. Intelectin-1 binds and alters the localization of the mucus barrier-modifying bacterium Akkermansia muciniphila. J Exp Med 2023; 220:e20211938. [PMID: 36413219 PMCID: PMC9683900 DOI: 10.1084/jem.20211938] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 09/06/2022] [Accepted: 10/13/2022] [Indexed: 01/25/2023] Open
Abstract
Intelectin-1 (ITLN1) is a lectin secreted by intestinal epithelial cells (IECs) and upregulated in human ulcerative colitis (UC). We investigated how ITLN1 production is regulated in IECs and the biological effects of ITLN1 at the host-microbiota interface using mouse models. Our data show that ITLN1 upregulation in IECs from UC patients is a consequence of activating the unfolded protein response. Analysis of microbes coated by ITLN1 in vivo revealed a restricted subset of microorganisms, including the mucolytic bacterium Akkermansia muciniphila. Mice overexpressing intestinal ITLN1 exhibited decreased inner colonic mucus layer thickness and closer apposition of A. muciniphila to the epithelial cell surface, similar to alterations reported in UC. The changes in the inner mucus layer were microbiota and A. muciniphila dependent and associated with enhanced sensitivity to chemically induced and T cell-mediated colitis. We conclude that by determining the localization of a select group of bacteria to the mucus layer, ITLN1 modifies this critical barrier. Together, these findings may explain the impact of ITLN1 dysregulation on UC pathogenesis.
Collapse
Affiliation(s)
- Juan D. Matute
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Division of Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Jinzhi Duan
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Magdalena B. Flak
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Paul Griebel
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jose A. Tascon-Arcila
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Shauni Doms
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Thomas Hanley
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Agne Antanaviciute
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | | | | | - Brandon Sit
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA
- Department of Microbiology, Harvard Medical School, Boston, MA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
| | - Shabnam Abtahi
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Gwenny M. Fuhler
- Department of Gastroenterology & Hepatology, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Joep Grootjans
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology and Metabolism & Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Florian Tran
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Stephanie T. Stengel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Niklas Krupka
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Dirk Haller
- Nutrition and Immunology, Technische Universität München, Freising, Germany
| | - Simon Clare
- Wellcome Trust Sanger Institute, Hinxton, UK
| | | | - Arthur Kaser
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, and Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Alison Simmons
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jonathan N. Glickman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Lynn Bry
- Massachusetts Host-Microbiome Center, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Matthew K. Waldor
- Division of Infectious Diseases, Brigham and Women’s Hospital, Boston, MA
- Department of Microbiology, Harvard Medical School, Boston, MA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
| | - John F. Baines
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Jerrold R. Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Richard S. Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
12
|
Radhakrishnan A, Chellapandian H, Ramasamy P, Jeyachandran S. Back2Basics: animal lectins: an insight into a highly versatile recognition protein. JOURNAL OF PROTEINS AND PROTEOMICS 2023; 14:43-59. [PMID: 36597476 PMCID: PMC9799708 DOI: 10.1007/s42485-022-00102-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/15/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022]
Abstract
The rapid advancement of molecular research has contributed to the discovery of 'Lectin', a carbohydrate-binding protein which specifically interacts with receptors on surface glycan moieties that regulate various critical cellular activities. The first animal lectin reported was 'the asialoglycoprotein receptor' in mammalian cells which helped analyze how animal lectins differ in glycoconjugate binding. Animal lectins are classified into several families, depending on their diverse cellular localization, and the binding specificities of their Carbohydrate-Recognition Domain (CRD) modules. Earlier characterization of animal lectins classified them into two structural families, the C-type (Ca2+-dependent binding) and S-type galectins (sulfhydryl-dependent binding) lectins. The C-type lectin includes the most significant animal lectins, such as endocytic receptors, mannose receptors, selectins, and collectins. The recent developments in research based on the complexity of the carbohydrate ligands, the metabolic processes they perform, their expression levels, and their reliance on divalent cations have identified more than 100 animal lectins and classified them into around 13 different families, such as Calnexin, F-lectin, Intelectin, Chitinase-like lectin, F-box lectin, etc. Understanding their structure and expression patterns have aided in defining their significant functions including cell adhesion, antimicrobial activity, innate immunity, disease diagnostic biomarkers, and drug delivery through specific carbohydrate-protein interactions. Such extensive potential roles of animal lectins made it equally important to plant lectins among researchers. Hence, the review focuses on providing an overview of animal lectins, their taxonomy, structural characteristics, and functions in diverse aspects interconnected to their specific carbohydrate and glycoconjugate binding. Graphical abstract
Collapse
Affiliation(s)
- Akshaya Radhakrishnan
- PG & Research Department of Biotechnology & Microbiology, National College, Tiruchirappalli, Tamil Nadu 620001 India
| | - Hethesh Chellapandian
- PG & Research Department of Biotechnology & Microbiology, National College, Tiruchirappalli, Tamil Nadu 620001 India
| | - Pasiyappazham Ramasamy
- Department of Physiology, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 600077 India
| | - Sivakamavalli Jeyachandran
- PG & Research Department of Biotechnology & Microbiology, National College, Tiruchirappalli, Tamil Nadu 620001 India
- Centre for Biotechnology & Biosignal Transduction, Department of Orthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 600077 India
| |
Collapse
|
13
|
Nonnecke EB, Castillo PA, Akahoshi DT, Goley SM, Bevins CL, Lönnerdal B. Characterization of an intelectin-1 ( Itln1) knockout mouse model. Front Immunol 2022; 13:894649. [PMID: 36072603 PMCID: PMC9441953 DOI: 10.3389/fimmu.2022.894649] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/28/2022] [Indexed: 01/26/2023] Open
Abstract
Intelectins are carbohydrate-binding proteins implicated in innate immunity and highly conserved across chordate evolution, including both ascidians and humans. Human intelectin-1 (ITLN1) is highly abundant within the intestinal mucosa and binds microbial but not host glycans. Genome-wide association studies identified SNPs in ITLN1 that are linked to susceptibility for Crohn's disease. Moreover, ITLN1 has been implicated in the pathophysiology of obesity and associated metabolic disease. To gain insight on biological activities of human ITLN1 in vivo, we developed a C57BL/6 mouse model genetically targeting the gene encoding the functional mouse ortholog. In wild-type C57BL/6 mice, both mRNA and protein analysis showed high expression of Itln1 in the small intestine, but manifold lower levels in colon and other extraintestinal tissues. Whereas intestinal expression of human ITLN1 localizes to goblet cells, our data confirm that mouse Itln1 is expressed in Paneth cells. Compared to wild-type littermate controls, mice homozygous for the Itln1 hypomorphic trapping allele had reduced expression levels of Itln1 expression (~10,000-fold). The knockout mice exhibited increased susceptibility in an acute model of experimentally induced colitis with 2% w/v dextran sulfate sodium (DSS). In a model of chronic colitis using a lower dose of DSS (1.5% w/v), which enabled a detailed view of disease activity across a protracted period, no differences were observed in body weight, fecal texture, hemoccult scores, food/water intake, or colon length at necropsy, but there was a statistically significant genotype over time effect for the combined fecal scores of disease activity. In model of diet-induced obesity, using two western-style diets, which varied in amounts of sugar (as sucrose) and saturated fat (as lard), mice with Itln1 expression ablated showed no increased susceptibility, in terms of weight gain, food intake, plasma markers of obesity compared to wildtype littermates. While the mouse genetic knockout model for Itln1 holds promise for elucidating physiological function(s) for mammalian intelectins, results reported here suggest that Itln1, a Paneth cell product in C57BL/6 mice, likely plays a minor role in the pathophysiology of chemically induced colitis or diet-induced obesity.
Collapse
Affiliation(s)
- Eric B. Nonnecke
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States,*Correspondence: Eric B. Nonnecke, ; Charles L. Bevins,
| | - Patricia A. Castillo
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Douglas T. Akahoshi
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Stephanie M. Goley
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Charles L. Bevins
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, CA, United States,*Correspondence: Eric B. Nonnecke, ; Charles L. Bevins,
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| |
Collapse
|
14
|
Andresen S, Fantone K, Chapla D, Rada B, Moremen KW, Pierce M, Szymanski CM. Human Intelectin-1 Promotes Cellular Attachment and Neutrophil Killing of Streptococcus pneumoniae in a Serotype-Dependent Manner. Infect Immun 2022; 90:e0068221. [PMID: 35499339 PMCID: PMC9119095 DOI: 10.1128/iai.00682-21] [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: 12/25/2021] [Accepted: 04/10/2022] [Indexed: 11/20/2022] Open
Abstract
Human intelectin-1 (hIntL-1) is a secreted glycoprotein capable of binding exocyclic 1,2-diols within surface glycans of human pathogens such as Streptococcus pneumoniae, Vibrio cholerae, and Helicobacter pylori. For the latter, lectin binding was shown to cause bacterial agglutination and increased phagocytosis, suggesting a role for hIntL-1 in pathogen surveillance. In this study, we investigated the interactions between hIntL-1 and S. pneumoniae, the leading cause of bacterial pneumonia. We show that hIntL-1 also agglutinates S. pneumoniae serotype 43, which displays an exocyclic 1,2-diol moiety in its capsular polysaccharide but is unable to kill in a complement-dependent manner or to promote bacterial killing by peripheral blood mononuclear cells. In contrast, hIntL-1 not only significantly increases serotype-specific S. pneumoniae killing by neutrophils but also enhances the attachment of these bacteria to A549 lung epithelial cells. Taken together, our results suggest that hIntL-1 participates in host surveillance through microbe sequestration and enhanced targeting to neutrophils.
Collapse
Affiliation(s)
- Silke Andresen
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Kayla Fantone
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Digantkumar Chapla
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Balázs Rada
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Kelley W. Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Michael Pierce
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Christine M. Szymanski
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
15
|
Nonnecke EB, Castillo PA, Johansson MEV, Hollox EJ, Shen B, Lönnerdal B, Bevins CL. Human intelectin-2 (ITLN2) is selectively expressed by secretory Paneth cells. FASEB J 2022; 36:e22200. [PMID: 35182405 PMCID: PMC9262044 DOI: 10.1096/fj.202101870r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 01/04/2023]
Abstract
Intelectins (intestinal lectins) are highly conserved across chordate evolution and have been implicated in various human diseases, including Crohn's disease (CD). The human genome encodes two intelectin genes, intelectin-1 (ITLN1) and intelectin-2 (ITLN2). Other than its high sequence similarity with ITLN1, little is known about ITLN2. To address this void in knowledge, we report that ITLN2 exhibits discrete, yet notable differences from ITLN1 in primary structure, including a unique amino terminus, as well as changes in amino acid residues associated with the glycan-binding activity of ITLN1. We identified that ITLN2 is a highly abundant Paneth cell-specific product, which localizes to secretory granules, and is expressed as a multimeric protein in the small intestine. In surgical specimens of ileal CD, ITLN2 mRNA levels were reduced approximately five-fold compared to control specimens. The ileal expression of ITLN2 was unaffected by previously reported disease-associated variants in ITLN2 and CD-associated variants in neighboring ITLN1 as well as NOD2 and ATG16L1. ITLN2 mRNA expression was undetectable in control colon tissue; however, in both ulcerative colitis (UC) and colonic CD, metaplastic Paneth cells were found to express ITLN2. Together, the data reported establish the groundwork for understanding ITLN2 function(s) in the intestine, including its possible role in CD.
Collapse
Affiliation(s)
- Eric B Nonnecke
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Patricia A Castillo
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| | - Malin E V Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Bo Shen
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, California, USA
| | - Charles L Bevins
- Department of Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
16
|
Donaldson DS, Shih BB, Mabbott NA. Aging-Related Impairments to M Cells in Peyer's Patches Coincide With Disturbances to Paneth Cells. Front Immunol 2021; 12:761949. [PMID: 34938288 PMCID: PMC8687451 DOI: 10.3389/fimmu.2021.761949] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/17/2021] [Indexed: 11/26/2022] Open
Abstract
The decline in mucosal immunity during aging increases susceptibility, morbidity and mortality to infections acquired via the gastrointestinal and respiratory tracts in the elderly. We previously showed that this immunosenescence includes a reduction in the functional maturation of M cells in the follicle-associated epithelia (FAE) covering the Peyer’s patches, diminishing the ability to sample of antigens and pathogens from the gut lumen. Here, co-expression analysis of mRNA-seq data sets revealed a general down-regulation of most FAE- and M cell-related genes in Peyer’s patches from aged mice, including key transcription factors known to be essential for M cell differentiation. Conversely, expression of ACE2, the cellular receptor for SARS-Cov-2 virus, was increased in the aged FAE. This raises the possibility that the susceptibility of aged Peyer’s patches to infection with the SARS-Cov-2 virus is increased. Expression of key Paneth cell-related genes was also reduced in the ileum of aged mice, consistent with the adverse effects of aging on their function. However, the increased expression of these genes in the villous epithelium of aged mice suggested a disturbed distribution of Paneth cells in the aged intestine. Aging effects on Paneth cells negatively impact on the regenerative ability of the gut epithelium and could indirectly impede M cell differentiation. Thus, restoring Paneth cell function may represent a novel means to improve M cell differentiation in the aging intestine and increase mucosal vaccination efficacy in the elderly.
Collapse
Affiliation(s)
- David S Donaldson
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, United Kingdom
| | - Barbara B Shih
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, United Kingdom
| | - Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, United Kingdom
| |
Collapse
|
17
|
Extensive variation in the intelectin gene family in laboratory and wild mouse strains. Sci Rep 2021; 11:15548. [PMID: 34330944 PMCID: PMC8324875 DOI: 10.1038/s41598-021-94679-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/13/2021] [Indexed: 12/30/2022] Open
Abstract
Intelectins are a family of multimeric secreted proteins that bind microbe-specific glycans. Both genetic and functional studies have suggested that intelectins have an important role in innate immunity and are involved in the etiology of various human diseases, including inflammatory bowel disease. Experiments investigating the role of intelectins in human disease using mouse models are limited by the fact that there is not a clear one-to-one relationship between intelectin genes in humans and mice, and that the number of intelectin genes varies between different mouse strains. In this study we show by gene sequence and gene expression analysis that human intelectin-1 (ITLN1) has multiple orthologues in mice, including a functional homologue Itln1; however, human intelectin-2 has no such orthologue or homologue. We confirm that all sub-strains of the C57 mouse strain have a large deletion resulting in retention of only one intelectin gene, Itln1. The majority of laboratory strains have a full complement of six intelectin genes, except CAST, SPRET, SKIVE, MOLF and PANCEVO strains, which are derived from different mouse species/subspecies and encode different complements of intelectin genes. In wild mice, intelectin deletions are polymorphic in Mus musculus castaneus and Mus musculus domesticus. Further sequence analysis shows that Itln3 and Itln5 are polymorphic pseudogenes due to premature truncating mutations, and that mouse Itln1 has undergone recent adaptive evolution. Taken together, our study shows extensive diversity in intelectin genes in both laboratory and wild-mice, suggesting a pattern of birth-and-death evolution. In addition, our data provide a foundation for further experimental investigation of the role of intelectins in disease.
Collapse
|
18
|
Blanco Vázquez C, Balseiro A, Alonso-Hearn M, Juste RA, Iglesias N, Canive M, Casais R. Bovine Intelectin 2 Expression as a Biomarker of Paratuberculosis Disease Progression. Animals (Basel) 2021; 11:ani11051370. [PMID: 34065919 PMCID: PMC8151335 DOI: 10.3390/ani11051370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/02/2021] [Accepted: 05/06/2021] [Indexed: 01/09/2023] Open
Abstract
Simple Summary The potential of the bovine intelectin 2 as a biomarker of Mycobacterium avium subsp. paratuberculosis infection was investigated using quantitative immunohistochemical analysis of ileocecal valve samples of animals with increasing degrees of lesion severity (focal, multifocal and diffuse histological lesions) and control animals without detected lesions. Significant differences were observed in the mean number of intelectin 2 immunolabelled cells between the three histopathological types and the control. Specifically, the mean number of intelectin 2 labelled cells was indicative of disease progression as the focal group had the highest number of intelectin 2 secreting cells followed by the multifocal, diffuse and control groups indicating that intelectin 2 is a good biomarker for the different stages of Mycobacterium avium subsp. paratuberculosis infection. Quantification of bovine intelectin 2 secreting cells could constitute a good post-mortem tool, complementary to histopathology, to improve detection of Mycobacterium avium subsp. Paratuberculosis infections, especially latent forms of infection. Abstract Paratuberculosis (PTB), a chronic granulomatous enteritis caused by Mycobacterium avium subsp. paratuberculosis (MAP), is responsible for important economic losses in the dairy industry. Our previous RNA-sequencing (RNA-Seq) analysis showed that bovine intelectin 2 (ITLN2) precursor gene was overexpressed in ileocecal valve (ICV) samples of animals with focal (log2 fold-change = 10.6) and diffuse (log2 fold-change = 6.8) PTB-associated lesions compared to animals without lesions. This study analyzes the potential use of ITLN2, a protein that has been described as fundamental in the innate immune response to infections, as a biomarker of MAP infection. The presence of ITLN2 was investigated by quantitative immunohistochemical analysis of ICV samples of 20 Holstein Friesian cows showing focal (n = 5), multifocal (n = 5), diffuse (n = 5) and no histological lesions (n = 5). Significant differences were observed in the mean number of ITLN2 immunostained goblet and Paneth cells between the three histopathological types and the control. The number of immunolabelled cells was higher in the focal histopathological type (116.9 ± 113.9) followed by the multifocal (108.7 ± 140.5), diffuse (76.5 ± 97.8) and control types (41.0 ± 81.3). These results validate ITLN2 as a post-mortem biomarker of disease progression.
Collapse
Affiliation(s)
- Cristina Blanco Vázquez
- Center for Animal Biotechnology, Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), 33394 Deva, Spain; (C.B.V.); (N.I.)
| | - Ana Balseiro
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad de León, 24071 León, Spain;
- Departamento de Sanidad Animal, Instituto de Ganadería de Montaña (CSIC-Universidad de León), Finca Marzanas, Grulleros, 24346 León, Spain
| | - Marta Alonso-Hearn
- Department of Animal Health, NEIKER-Basque Institute for Agricultural Research and Development, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Bizkaia, P812, E-48160 Derio, Spain; (M.A.-H.); (R.A.J.); (M.C.)
| | - Ramón A. Juste
- Department of Animal Health, NEIKER-Basque Institute for Agricultural Research and Development, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Bizkaia, P812, E-48160 Derio, Spain; (M.A.-H.); (R.A.J.); (M.C.)
| | - Natalia Iglesias
- Center for Animal Biotechnology, Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), 33394 Deva, Spain; (C.B.V.); (N.I.)
| | - Maria Canive
- Department of Animal Health, NEIKER-Basque Institute for Agricultural Research and Development, Basque Research and Technology Alliance (BRTA), Parque Científico y Tecnológico de Bizkaia, P812, E-48160 Derio, Spain; (M.A.-H.); (R.A.J.); (M.C.)
| | - Rosa Casais
- Center for Animal Biotechnology, Servicio Regional de Investigación y Desarrollo Agroalimentario (SERIDA), 33394 Deva, Spain; (C.B.V.); (N.I.)
- Correspondence:
| |
Collapse
|
19
|
Ivashenka A, Wunder C, Chambon V, Sandhoff R, Jennemann R, Dransart E, Podsypanina K, Lombard B, Loew D, Lamaze C, Poirier F, Gröne HJ, Johannes L, Shafaq-Zadah M. Glycolipid-dependent and lectin-driven transcytosis in mouse enterocytes. Commun Biol 2021; 4:173. [PMID: 33564097 PMCID: PMC7873212 DOI: 10.1038/s42003-021-01693-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/13/2021] [Indexed: 01/05/2023] Open
Abstract
Glycoproteins and glycolipids at the plasma membrane contribute to a range of functions from growth factor signaling to cell adhesion and migration. Glycoconjugates undergo endocytic trafficking. According to the glycolipid-lectin (GL-Lect) hypothesis, the construction of tubular endocytic pits is driven in a glycosphingolipid-dependent manner by sugar-binding proteins of the galectin family. Here, we provide evidence for a function of the GL-Lect mechanism in transcytosis across enterocytes in the mouse intestine. We show that galectin-3 (Gal3) and its newly identified binding partner lactotransferrin are transported in a glycosphingolipid-dependent manner from the apical to the basolateral membrane. Transcytosis of lactotransferrin is perturbed in Gal3 knockout mice and can be rescued by exogenous Gal3. Inside enterocytes, Gal3 is localized to hallmark structures of the GL-Lect mechanism, termed clathrin-independent carriers. These data pioneer the existence of GL-Lect endocytosis in vivo and strongly suggest that polarized trafficking across the intestinal barrier relies on this mechanism.
Collapse
Affiliation(s)
- Alena Ivashenka
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, Endocytic Trafficking and Intracellular Delivery Team, Paris, France
| | - Christian Wunder
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, Endocytic Trafficking and Intracellular Delivery Team, Paris, France
| | - Valerie Chambon
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, Endocytic Trafficking and Intracellular Delivery Team, Paris, France
| | - Roger Sandhoff
- Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Richard Jennemann
- Lipid Pathobiochemistry Group, German Cancer Research Center, Heidelberg, Germany
| | - Estelle Dransart
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, Endocytic Trafficking and Intracellular Delivery Team, Paris, France
| | - Katrina Podsypanina
- Institut Curie, Université PSL, UMR144 CNRS, Cell Biology and Cancer, Paris, France
| | - Bérangère Lombard
- Institut Curie, Université PSL, Mass Spectrometry and Proteomics Facility, Paris, France
| | - Damarys Loew
- Institut Curie, Université PSL, Mass Spectrometry and Proteomics Facility, Paris, France
| | - Christophe Lamaze
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, Membrane Dynamics and Mechanics of Intracellular Signaling Team, Paris, France
| | - Francoise Poirier
- Institut Jacques Monod, UMR 7592 CNRS - Université Paris Diderot, 15 rue Hélène Brion, Paris, France
| | | | - Ludger Johannes
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, Endocytic Trafficking and Intracellular Delivery Team, Paris, France.
| | - Massiullah Shafaq-Zadah
- Institut Curie, Université PSL, U1143 INSERM, UMR3666 CNRS, Cellular and Chemical Biology Unit, Endocytic Trafficking and Intracellular Delivery Team, Paris, France.
| |
Collapse
|
20
|
Dierick M, Vanrompay D, Devriendt B, Cox E. Lactoferrin, a versatile natural antimicrobial glycoprotein that modulates the host's innate immunity. Biochem Cell Biol 2020; 99:61-65. [PMID: 32585120 DOI: 10.1139/bcb-2020-0080] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lactoferrin is a multifunctional protein found in the secretions of mammals. The antimicrobial activity of lactoferrin was the first to be discovered and was assumed to be solely dependent on its iron-chelating ability. However, lactoferrin has been reported to display proteolytic activity towards bacterial virulence factors and to modulate the host defence by stimulating the immune system and balancing pathogen-induced inflammation. Here, we review the current understandings of the antimicrobial effect, interaction with host cells, and innate immune modulation of lactoferrin, and put forward this moonlighting protein as a possible alternative for antibiotics.
Collapse
Affiliation(s)
- Matthias Dierick
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Daisy Vanrompay
- Laboratory for Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
21
|
Dawson HD, Sang Y, Lunney JK. Porcine cytokines, chemokines and growth factors: 2019 update. Res Vet Sci 2020; 131:266-300. [PMID: 32442727 DOI: 10.1016/j.rvsc.2020.04.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022]
Abstract
Pigs are a major food source worldwide as well as major biomedical models for human physiology and therapeutics. A thorough understanding of porcine immunity is essential to prevent and treat infectious diseases, and develop effective vaccines and therapeutics. The use of pigs as biomedical models is dependent on the growing molecular and immune toolbox. This paper summarizes current knowledge of swine cytokines, chemokines and growth factors, identifying 289 pig proteins, characterizing knowledge of their gene structures and families. It identifies areas in the current swine genome build that need to be clarified. A broad-based literature and vendor search was conducted to identify defined sets of monoclonal and polyclonal antibodies reacting with porcine cytokines, chemokines, growth factors along with availability of cloned recombinant proteins and assays for their quantitation. This process identified numerous reagents that are reportedly reactive with 170 pig cytokines, chemokines, growth factors: 118 have at least one commercial antibody reagent, 66 a cloned recombinant peptide, and 97 with quantitative assays. This affirms the great need to develop and characterize additional reagents. There are panels of reagents for numerous high priority targets that have been essential reagents for characterizing porcine immunity, disease and vaccine responses, and factors regulating development of innate immune responses, polarized macrophages and lymphoid cells including T regulatory cells. Yet there are many areas requiring investment of efforts to more effectively explore the pig immune system. The development of more reagents to understand the complex of cytokines, chemokines, and growth factors will clearly advance these initiatives.
Collapse
Affiliation(s)
- Harry D Dawson
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet, Genomics & Immunology Laboratory, Beltsville, MD 20705-2350, USA
| | - Yongming Sang
- Department of Agricultural and Environmental Sciences, College of Agriculture, Tennessee State University, Nashville, TN 37209-1561, USA
| | - Joan K Lunney
- USDA, ARS, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, MD 20705-2350, USA.
| |
Collapse
|
22
|
Smillie CS, Biton M, Ordovas-Montanes J, Sullivan KM, Burgin G, Graham DB, Herbst RH, Rogel N, Slyper M, Waldman J, Sud M, Andrews E, Velonias G, Haber AL, Jagadeesh K, Vickovic S, Yao J, Stevens C, Dionne D, Nguyen LT, Villani AC, Hofree M, Creasey EA, Huang H, Rozenblatt-Rosen O, Garber JJ, Khalili H, Desch AN, Daly MJ, Ananthakrishnan AN, Shalek AK, Xavier RJ, Regev A. Intra- and Inter-cellular Rewiring of the Human Colon during Ulcerative Colitis. Cell 2020; 178:714-730.e22. [PMID: 31348891 DOI: 10.1016/j.cell.2019.06.029] [Citation(s) in RCA: 763] [Impact Index Per Article: 152.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 03/25/2019] [Accepted: 06/18/2019] [Indexed: 11/29/2022]
Abstract
Genome-wide association studies (GWAS) have revealed risk alleles for ulcerative colitis (UC). To understand their cell type specificities and pathways of action, we generate an atlas of 366,650 cells from the colon mucosa of 18 UC patients and 12 healthy individuals, revealing 51 epithelial, stromal, and immune cell subsets, including BEST4+ enterocytes, microfold-like cells, and IL13RA2+IL11+ inflammatory fibroblasts, which we associate with resistance to anti-TNF treatment. Inflammatory fibroblasts, inflammatory monocytes, microfold-like cells, and T cells that co-express CD8 and IL-17 expand with disease, forming intercellular interaction hubs. Many UC risk genes are cell type specific and co-regulated within relatively few gene modules, suggesting convergence onto limited sets of cell types and pathways. Using this observation, we nominate and infer functions for specific risk genes across GWAS loci. Our work provides a framework for interrogating complex human diseases and mapping risk variants to cell types and pathways.
Collapse
Affiliation(s)
| | - Moshe Biton
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA; Department of Molecular Biology, MGH, Boston, MA, USA
| | - Jose Ordovas-Montanes
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA, USA; Department of Chemistry, MIT, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Division of Infectious Diseases and Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
| | - Keri M Sullivan
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, MGH, Boston, MA, USA
| | - Grace Burgin
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Daniel B Graham
- Department of Molecular Biology, MGH, Boston, MA, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, MGH, Boston, MA, USA; Broad Institute, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA; Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA, USA
| | - Rebecca H Herbst
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Noga Rogel
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Michal Slyper
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Julia Waldman
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Malika Sud
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Elizabeth Andrews
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, MGH, Boston, MA, USA
| | - Gabriella Velonias
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, MGH, Boston, MA, USA
| | - Adam L Haber
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | | | - Sanja Vickovic
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Junmei Yao
- Center for Computational and Integrative Biology, MGH, Boston, MA, USA
| | | | - Danielle Dionne
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Lan T Nguyen
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Alexandra-Chloé Villani
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA; Center for Immunology and Inflammatory Diseases, Department of Medicine, MGH, Boston, MA, USA
| | - Matan Hofree
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | | | - Hailiang Huang
- Medical and Population Genetics, Broad Institute, Cambridge, MA, USA; Analytical and Translational Genetics Unit, MGH, Boston, MA, USA
| | | | - John J Garber
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, MGH, Boston, MA, USA
| | - Hamed Khalili
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, MGH, Boston, MA, USA
| | - A Nicole Desch
- Broad Institute, Cambridge, MA, USA; Center for Computational and Integrative Biology, MGH, Boston, MA, USA
| | - Mark J Daly
- Medical and Population Genetics, Broad Institute, Cambridge, MA, USA; Analytical and Translational Genetics Unit, MGH, Boston, MA, USA; Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Ashwin N Ananthakrishnan
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, MGH, Boston, MA, USA.
| | - Alex K Shalek
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA; Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA, USA; Department of Chemistry, MIT, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| | - Ramnik J Xavier
- Department of Molecular Biology, MGH, Boston, MA, USA; Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, MGH, Boston, MA, USA; Broad Institute, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA; Center for Microbiome Informatics and Therapeutics, MIT, Cambridge, MA, USA; Center for Computational and Integrative Biology, MGH, Boston, MA, USA.
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA; Howard Hughes Medical Institute and Koch Institute for Integrative Cancer Research, Department of Biology, MIT, Cambridge, MA, USA.
| |
Collapse
|
23
|
Viral Hepatitis and Iron Dysregulation: Molecular Pathways and the Role of Lactoferrin. Molecules 2020; 25:molecules25081997. [PMID: 32344579 PMCID: PMC7221917 DOI: 10.3390/molecules25081997] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/16/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
The liver is a frontline immune site specifically designed to check and detect potential pathogens from the bloodstream to maintain a general state of immune hyporesponsiveness. One of the main functions of the liver is the regulation of iron homeostasis. The liver detects changes in systemic iron requirements and can regulate its concentration. Pathological states lead to the dysregulation of iron homeostasis which, in turn, can promote infectious and inflammatory processes. In this context, hepatic viruses deviate hepatocytes' iron metabolism in order to better replicate. Indeed, some viruses are able to alter the expression of iron-related proteins or exploit host receptors to enter inside host cells. Lactoferrin (Lf), a multifunctional iron-binding glycoprotein belonging to the innate immunity, is endowed with potent antiviral activity, mainly related to its ability to block viral entry into host cells by interacting with viral and/or cell surface receptors. Moreover, Lf can act as an iron scavenger by both direct iron-chelation or the modulation of the main iron-related proteins. In this review, the complex interplay between viral hepatitis, iron homeostasis, and inflammation as well as the role of Lf are outlined.
Collapse
|
24
|
Zhang Y, Zhao X, Chen M. Autocrine action of adipokine omentin-1 in the SW480 colon cancer cell line. Oncol Lett 2019; 19:892-898. [PMID: 31897204 PMCID: PMC6924134 DOI: 10.3892/ol.2019.11131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 10/10/2019] [Indexed: 01/22/2023] Open
Abstract
Omentin-1, a 34-kDa protein, has been demonstrated to be associated with colorectal cancer (CRC). Epidemiological and clinical studies have indicated that the levels of circulating omentin-1 are significantly increased in patients with CRC, but the cause of the high omentin-1 levels and whether CRC cells express this adipokine have not been determined. In the present study, human colorectal carcinoma and para-carcinoma tissues were collected from 24 patients with CRC. In addition, SW480 and HCT116 colon cancer cells were cultured in vitro. The expression and localization of omentin-1 protein in human CRC and para-carcinoma tissues were determined by immunohistochemistry. The mRNA and protein expression levels of omentin-1 in human CRC and para-carcinoma tissues were detected by reverse transcription-quantitative PCR (RT-qPCR) and western blotting, respectively. In addition, omentin-1 mRNA expression levels in SW480 and HCT116 cell lines were detected by RT-qPCR. Since SW480 cells exhibited higher omentin-1 mRNA levels compared with those of HCT116 cells, SW480 cells were selected for further experiments. The expression of omentin-1 protein in the supernatant and lysate of SW480 cells obtained at 6, 12, 24 and 48 h was determined by ELISA. The immunohistochemistry results demonstrated that the positive expression of omentin-1 protein was mainly located in the cytoplasm of cancer cells in human CRC tissues. The mRNA and protein expression levels of omentin-1 in the CRC tissues were higher compared with those in para-carcinoma tissues. The expression levels of omentin-1 were detected in the cell lysate and supernatant of SW480 cells; the expression level of omentin-1 protein in the cell lysate was higher compared with that in the supernatant. These results indicated that SW480 cells secret and express the adipokine omentin-1 endogenously. Omentin-1 may serve its potential carcinogenetic role in CRC through endocrine, autocrine and paracrine pathways.
Collapse
Affiliation(s)
- Yaqin Zhang
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xiaotong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Mingwei Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China.,Institute of Traditional Chinese Medicine Diabetes Prevention, Anhui Academy of Traditional Chinese Medicine, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
25
|
Lepanto MS, Rosa L, Paesano R, Valenti P, Cutone A. Lactoferrin in Aseptic and Septic Inflammation. Molecules 2019; 24:molecules24071323. [PMID: 30987256 PMCID: PMC6480387 DOI: 10.3390/molecules24071323] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
Lactoferrin (Lf), a cationic glycoprotein able to chelate two ferric irons per molecule, is synthesized by exocrine glands and neutrophils. Since the first anti-microbial function attributed to Lf, several activities have been discovered, including the relevant anti-inflammatory one, especially associated to the down-regulation of pro-inflammatory cytokines, as IL-6. As high levels of IL-6 are involved in iron homeostasis disorders, Lf is emerging as a potent regulator of iron and inflammatory homeostasis. Here, the role of Lf against aseptic and septic inflammation has been reviewed. In particular, in the context of aseptic inflammation, as anemia of inflammation, preterm delivery, Alzheimer’s disease and type 2 diabetes, Lf administration reduces local and/or systemic inflammation. Moreover, Lf oral administration, by decreasing serum IL-6, reverts iron homeostasis disorders. Regarding septic inflammation occurring in Chlamydia trachomatis infection, cystic fibrosis and inflammatory bowel disease, Lf, besides the anti-inflammatory activity, exerts a significant activity against bacterial adhesion, invasion and colonization. Lastly, a critical analysis of literature in vitro data reporting contradictory results on the Lf role in inflammatory processes, ranging from pro- to anti-inflammatory activity, highlighted that they depend on cell models, cell metabolic status, stimulatory or infecting agents as well as on Lf iron saturation degree, integrity and purity.
Collapse
Affiliation(s)
- Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | | | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
| | - Antimo Cutone
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy.
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy.
| |
Collapse
|
26
|
Mancinelli R, Olivero F, Carpino G, Overi D, Rosa L, Lepanto MS, Cutone A, Franchitto A, Alpini G, Onori P, Valenti P, Gaudio E. Role of lactoferrin and its receptors on biliary epithelium. Biometals 2018; 31:369-379. [PMID: 29550924 DOI: 10.1007/s10534-018-0094-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
Human lactoferrin is an iron-binding glycoprotein present at high concentrations in breast milk and colostrum. It is produced by many exocrine glands and widely distributed in a variety of body fluids. This protein has antimicrobial, immunomodulatory, antioxidant, and anticancer properties. Two important hLf receptors have been identified: LDL receptor related protein (LRP1), a low specificity receptor, and intelectin-1 (ITLN1), a high specificity receptor. No data are present on the role of hLf on the biliary epithelium. Our aims have been to evaluate the expression of Lf and its receptors in human and murine cholangiocytes and its effect on proliferation. Immunohistochemistry and immunofluorescence (IF) were conducted on human healthy and primary biliary cholangitis (PBC) liver samples as well as on liver samples obtained from normal and bile duct ligated (BDL) mice to evaluate the expression of Lf, LRP1 and ITLN1. Cell proliferation in vitro studies were performed on human cholangiocyte cell lines via 3-(4,5-dimetiltiazol-2-il)-2,5-diphenyltetrazolium assay as well as IF to evaluate proliferating cell nuclear antigen (PCNA) expression. Our results show that mouse and human cholangiocytes express Lf, LRP1 and ITLN1, at higher extent in cholangiocytes from BDL and PBC samples. Furthermore, the in vitro addition of bovine Lf (bLf) has a proliferative effect on human cholangiocyte cell line. The results support a proliferative role of hLf on the biliary epithelium; this pro-proliferative effect of hLf and bLf on cholangiocytes could be particularly relevant in human cholangiopathies such as PBC, characterized by cholangiocyte death and ductopenia.
Collapse
Affiliation(s)
- Romina Mancinelli
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy.
| | - Francesca Olivero
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Guido Carpino
- Department of Movement, Human and Health Sciences, Sapienza University of Rome, Rome, Italy
| | - Diletta Overi
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Antimo Cutone
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Antonio Franchitto
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Baylor Scott & White Digestive Disease Research Center, Baylor Scott & White, Department of Medical Physiology, Texas A&M University College of Medicine, Temple, TX, 76504, USA
| | - Paolo Onori
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
27
|
Jensen SR, Schoof EM, Wheeler SE, Hvid H, Ahnfelt-Rønne J, Hansen BF, Nishimura E, Olsen GS, Kislinger T, Brubaker PL. Quantitative Proteomics of Intestinal Mucosa From Male Mice Lacking Intestinal Epithelial Insulin Receptors. Endocrinology 2017; 158:2470-2485. [PMID: 28591806 DOI: 10.1210/en.2017-00194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/31/2017] [Indexed: 12/16/2022]
Abstract
The goal of the present study was to determine whether loss of the insulin receptor alters the molecular landscape of the intestinal mucosa, using intestinal-epithelial insulin receptor knockout (IE-irKO) mice and both genetic (IRfl/fl and Villin-cre) controls. Quantitative proteomic analysis by liquid chromatography mass spectrometry was applied to jejunal and colonic mucosa from mice fed a normal chow diet and mice fed a Western diet (WD). Jejunal mucosa from IE-irKO mice demonstrated alterations in all intestinal cell lineages: Paneth, goblet, absorptive, and enteroendocrine cells. Only goblet and absorptive cells were affected in the colon. Also, a marked effect of WD consumption was found on the gut proteome. A substantial reduction was detected in Paneth cell proteins with antimicrobial activity, including lysozyme C-1, angiogenin-4, cryptdin-related sequence 1C-3 and -2, α-defensin 17, and intelectin-1a. The key protein expressed by goblet cells, mucin-2, was also reduced in the IE-irKO mice. Proteins involved in lipid metabolism, including aldose reductase-related protein 1, 15-hydroxyprostaglandin dehydrogenase, apolipoprotein A-II, and pyruvate dehydrogenase kinase isozyme 4, were increased in the mucosa of WD-fed IE-irKO mice compared with controls. In contrast, expression of the nutrient-responsive gut hormones, glucose-dependent insulinotropic polypeptide and neurotensin, was reduced in the jejunal mucosa of IE-irKO mice, and the expression of proteins of the P-type adenosine triphosphatases and the solute carrier-transporter family was reduced in the colon of WD-fed IE-irKO mice. In conclusion, IE-irKO mice display a distinct molecular phenotype, suggesting a biological role of insulin and its receptor in determining differentiated cell specificity in the intestinal epithelium.
Collapse
Affiliation(s)
- Stina Rikke Jensen
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Metabolic Disease Research, Novo Nordisk A/S, Måløv DK-2760, Denmark
| | - Erwin M Schoof
- Princess Margaret Hospital Cancer Centre, University Health Network, Ontario M5G 2M9, Canada
| | - Sarah E Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Henning Hvid
- Metabolic Disease Research, Novo Nordisk A/S, Måløv DK-2760, Denmark
| | | | - Bo Falck Hansen
- Metabolic Disease Research, Novo Nordisk A/S, Måløv DK-2760, Denmark
| | - Erica Nishimura
- Metabolic Disease Research, Novo Nordisk A/S, Måløv DK-2760, Denmark
| | | | - Thomas Kislinger
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
28
|
Migration ofFasciola hepaticanewly excysted juveniles is inhibited by high-mannose and oligomannose-typeN-glycan-binding lectins. Parasitology 2017; 144:1708-1717. [DOI: 10.1017/s003118201700124x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SUMMARYFasciola hepaticahas both zoonotic importance and high economic impact in livestock worldwide. After ingestion by the definitive host, the Newly Excysted Juveniles (NEJ) penetrate the intestine before reaching the peritoneal cavity. The role of some NEJ-derived proteins in invasion has been documented, but the role of NEJ glycans or lectin-binding receptors during initial infection in the gut is still unknown. To address these questions, the migration of NEJ through rat intestine was recorded at 30 min intervals up to 150 min by twoex vivomethods. Firstly, jejunal sheets were challenged with NEJ incubated with biotinylated lectins. Secondly, untreated NEJ were incubated with distal jejunum pre-treated with lectins. BothConcanavalin A(ConA) andGalanthus nivalis(GNL), which recognize mannose-typeN-glycans, significantly inhibited NEJ migration across the jejunum. Most of the lectins bound to the tegument and oral sucker of the NEJ, but only ConA and GNL maintained this interaction over 150 min. None of the lectins examined significantly reduced NEJ migration when pre-incubated with jejunal sheets, suggesting that host glycans might not be essential for initial binding/recognition of the gut by NEJ. Agents capable of blocking mannose-typeN-glycans on the NEJ tegument may have potential for disrupting infection.
Collapse
|
29
|
Watanabe T, Watanabe-Kominato K, Takahashi Y, Kojima M, Watanabe R. Adipose Tissue-Derived Omentin-1 Function and Regulation. Compr Physiol 2017. [PMID: 28640441 DOI: 10.1002/cphy.c160043] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Omentin-1, also known as intelectin-1, is a recently identified novel adipocytokine of 313 amino acids, which is expressed in visceral (omental and epicardial) fat as well as mesothelial cells, vascular cells, airway goblet cells, small intestine, colon, ovary, and plasma. The level of omentin-1 expression in (pre)adipocytes is decreased by glucose/insulin and stimulated by fibroblast growth factor-21 and dexamethasone. Several lines of experimental evidence have shown that omentin-1 plays crucial roles in the maintenance of body metabolism and insulin sensitivity, and has anti-inflammatory, anti-atherosclerotic, and cardiovascular protective effects via AMP-activated protein kinase/Akt/nuclear factor-κB/mitogen-activated protein kinase (ERK, JNK, and p38) signaling. Clinical studies have indicated the usage of circulating omentin-1 as a biomarker of obesity, metabolic disorders including insulin resistance, diabetes, and metabolic syndrome, and atherosclerotic cardiovascular diseases. It is also possible to use circulating omentin-1 as a biomarker of bone metabolism, inflammatory diseases, cancers, sleep apnea syndrome, preeclampsia, and polycystic ovary syndrome. Decreased omentin-1 levels are generally associated with these diseases. However, omentin-1 increases to counteract the acute phase after onset of these diseases. These findings indicate that omentin-1 may be a negative risk factor for these diseases, and also act as an acute-phase reactant by its anti-inflammatory and atheroprotective effects. Therapeutic strategies to restore omentin-1 levels may be valuable for the prevention or treatment of these diseases. Weight loss, olive oil-rich diet, aerobic training, and treatment with atorvastatin and antidiabetic drugs (metformin, pioglitazone, and exenatide) are effective means of increasing circulating omentin-1 levels. This review provides insights into the potential use of omentin-1 as a biomarker and therapeutic target for these diseases. © 2017 American Physiological Society. Compr Physiol 7:765-781, 2017.
Collapse
Affiliation(s)
- Takuya Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Hachioji-City, Tokyo, Japan
| | - Kaho Watanabe-Kominato
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Hachioji-City, Tokyo, Japan
| | - Yui Takahashi
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Hachioji-City, Tokyo, Japan
| | - Miho Kojima
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Hachioji-City, Tokyo, Japan
| | - Rena Watanabe
- Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, Hachioji-City, Tokyo, Japan
| |
Collapse
|
30
|
Mayers MD, Moon C, Stupp GS, Su AI, Wolan DW. Quantitative Metaproteomics and Activity-Based Probe Enrichment Reveals Significant Alterations in Protein Expression from a Mouse Model of Inflammatory Bowel Disease. J Proteome Res 2017; 16:1014-1026. [PMID: 28052195 DOI: 10.1021/acs.jproteome.6b00938] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tandem mass spectrometry based shotgun proteomics of distal gut microbiomes is exceedingly difficult due to the inherent complexity and taxonomic diversity of the samples. We introduce two new methodologies to improve metaproteomic studies of microbiome samples. These methods include the stable isotope labeling in mammals to permit protein quantitation across two mouse cohorts as well as the application of activity-based probes to enrich and analyze both host and microbial proteins with specific functionalities. We used these technologies to study the microbiota from the adoptive T cell transfer mouse model of inflammatory bowel disease (IBD) and compare these samples to an isogenic control, thereby limiting genetic and environmental variables that influence microbiome composition. The data generated highlight quantitative alterations in both host and microbial proteins due to intestinal inflammation and corroborates the observed phylogenetic changes in bacteria that accompany IBD in humans and mouse models. The combination of isotope labeling with shotgun proteomics resulted in the total identification of 4434 protein clusters expressed in the microbial proteomic environment, 276 of which demonstrated differential abundance between control and IBD mice. Notably, application of a novel cysteine-reactive probe uncovered several microbial proteases and hydrolases overrepresented in the IBD mice. Implementation of these methods demonstrated that substantial insights into the identity and dysregulation of host and microbial proteins altered in IBD can be accomplished and can be used in the interrogation of other microbiome-related diseases.
Collapse
Affiliation(s)
- Michael D Mayers
- Department of Molecular and Experimental Medicine, ‡Department of Integrative Structural and Computational Biology, and §Department of Chemical Physiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Clara Moon
- Department of Molecular and Experimental Medicine, ‡Department of Integrative Structural and Computational Biology, and §Department of Chemical Physiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Gregory S Stupp
- Department of Molecular and Experimental Medicine, ‡Department of Integrative Structural and Computational Biology, and §Department of Chemical Physiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Andrew I Su
- Department of Molecular and Experimental Medicine, ‡Department of Integrative Structural and Computational Biology, and §Department of Chemical Physiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Dennis W Wolan
- Department of Molecular and Experimental Medicine, ‡Department of Integrative Structural and Computational Biology, and §Department of Chemical Physiology, The Scripps Research Institute , 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
31
|
Oshima Y, Seki K, Shibuya M, Naka Y, Yokoyama T, Sato A. Soluble Human Intestinal Lactoferrin Receptor: Ca(2+)-Dependent Binding to Sepharose-Based Matrices. Biol Pharm Bull 2016; 39:435-9. [PMID: 26934934 DOI: 10.1248/bpb.b15-00643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A soluble form of human intestinal lactoferrin receptor (shLFR) is identical to human intelectin-1 (hITLN-1), a galactofuranose-binding protein that acts as a host defense against invading pathogenic microorganisms. We found that recombinant shLFR, expressed in mammalian cells (CHO DG44, COS-1, and RK13), binds tightly to Sepharose 4 Fast Flow (FF)-based matrices in a Ca(2+)-dependent manner. This binding of shLFR to Sepharose 4 FF-based matrices was inhibited by excess D-galactose, but not by D-glucose, suggesting that shLFR recognizes repeating units of α-1,6-linked D-galactose in Sepharose 4 FF. Furthermore, shLFR could bind to both Sepharose 4B- and Sepharose 6B-based matrices that were not crosslinked in a similar manner as to Sepharose 4 FF-based matrices. Therefore, shLFR (hITLN-1) binds to Sepharose-based matrices in a Ca(2+)-dependent manner. This binding property is most likely related to the ability, as host defense lectins, to recognize sepharose (agarobiose)-like structures present on the surface of invading pathogenic microorganisms.
Collapse
Affiliation(s)
- Yuta Oshima
- School of Bioscience and Biotechnology, Tokyo University of Technology
| | | | | | | | | | | |
Collapse
|
32
|
Lundquist P, Artursson P. Oral absorption of peptides and nanoparticles across the human intestine: Opportunities, limitations and studies in human tissues. Adv Drug Deliv Rev 2016; 106:256-276. [PMID: 27496705 DOI: 10.1016/j.addr.2016.07.007] [Citation(s) in RCA: 321] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 07/02/2016] [Accepted: 07/08/2016] [Indexed: 12/23/2022]
Abstract
In this contribution, we review the molecular and physiological barriers to oral delivery of peptides and nanoparticles. We discuss the opportunities and predictivity of various in vitro systems with special emphasis on human intestine in Ussing chambers. First, the molecular constraints to peptide absorption are discussed. Then the physiological barriers to peptide delivery are examined. These include the gastric and intestinal environment, the mucus barrier, tight junctions between epithelial cells, the enterocytes of the intestinal epithelium, and the subepithelial tissue. Recent data from human proteome studies are used to provide information about the protein expression profiles of the different physiological barriers to peptide and nanoparticle absorption. Strategies that have been employed to increase peptide absorption across each of the barriers are discussed. Special consideration is given to attempts at utilizing endogenous transcytotic pathways. To reliably translate in vitro data on peptide or nanoparticle permeability to the in vivo situation in a human subject, the in vitro experimental system needs to realistically capture the central aspects of the mentioned barriers. Therefore, characteristics of common in vitro cell culture systems are discussed and compared to those of human intestinal tissues. Attempts to use the cell and tissue models for in vitro-in vivo extrapolation are reviewed.
Collapse
Affiliation(s)
- P Lundquist
- Department of Pharmacy, Uppsala University, Box 580, SE-752 37 Uppsala, Sweden.
| | - P Artursson
- Department of Pharmacy, Uppsala University, Box 580, SE-752 37 Uppsala, Sweden.
| |
Collapse
|
33
|
Nagata S. Identification and characterization of a novel intelectin in the digestive tract of Xenopus laevis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 59:229-239. [PMID: 26855011 DOI: 10.1016/j.dci.2016.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 02/03/2016] [Accepted: 02/03/2016] [Indexed: 06/05/2023]
Abstract
The intelectin (Intl) family is a group of secretory lectins in chordates that serve multiple functions, including innate immunity, through Ca(2+)-dependent recognition of carbohydrate chains. Although six Intl family lectins have so far been reported in Xenopus laevis, none have been identified in the intestine. Using a monoclonal antibody to the Xenopus embryonic epidermal lectin (XEEL or Intl-1), I identified cross-reactive proteins in the intestines. The proteins were purified by affinity chromatography on a galactose-Sepharose column and found to be oligomers consisting of N-glycosylated 39 kDa and 40.5 kDa subunit peptides. N-terminal amino acid sequencing of these peptides, followed by cDNA cloning, identified two novel Intls (designated Intl-3 and Intl-4) that showed 59-79% amino acid identities with known Xenopus Intl family proteins. From the amino acid sequence, immunoreactivity, and properties of the recombinant protein, Intl-3 was considered the intestinal lectin identified by the anti-XEEL antibody. The purified Intl-3 protein could potentially bind to Escherichia coli and its lipopolysaccharides (LPS), and to Staphylococcus aureus and its peptidoglycans, depending on Ca(2+). In addition, the Intl-3 protein agglutinated E. coli cells in the presence of Ca(2+). Intraperitoneal injection of LPS increased the intestinal and rectal contents of Intl-3 and XCL-1 (or 35K serum lectin) proteins within three days; however, unlike XCL-1, Intl-3 was detectable in neither the sera nor the other tissues regardless of LPS stimulation. Immunohistochemical analyses revealed accumulation of the Intl-3 protein in mucus secretory granules of intestinal goblet cells. The results of this study suggest that Xenopus Intl-3 is involved in the innate immune protection of the digestive tract against bacterial infections.
Collapse
Affiliation(s)
- Saburo Nagata
- Department of Chemical and Biological Sciences, Faculty of Science, Japan Women's University, Mejirodai 2-8-1, Bunkyoku, Tokyo 112-8681, Japan.
| |
Collapse
|
34
|
Mayeur S, Spahis S, Pouliot Y, Levy E. Lactoferrin, a Pleiotropic Protein in Health and Disease. Antioxid Redox Signal 2016; 24:813-36. [PMID: 26981846 DOI: 10.1089/ars.2015.6458] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
SIGNIFICANCE Lactoferrin (Lf) is a nonheme iron-binding glycoprotein strongly expressed in human and bovine milk and it plays many functions during infancy such as iron homeostasis and defense against microorganisms. In humans, Lf is mainly expressed in mucosal epithelial and immune cells. Growing evidence suggests multiple physiological roles for Lf after weaning. RECENT ADVANCES The aim of this review is to highlight the recent advances concerning multifunctional Lf activities. CRITICAL ISSUES First, we will provide an overview of the mechanisms related to Lf intrinsic synthesis or intestinal absorption as well as its interaction with a wide spectrum of mammalian receptors and distribution in organs and cell types. Second, we will discuss the large variety of its physiological functions such as iron homeostasis, transportation, immune regulation, oxidative stress, inflammation, and apoptosis while specifying the mechanisms of action. Third, we will focus on its recent physiopathology implication in metabolic disorders, including obesity, type 2 diabetes, and cardiovascular diseases. Additional efforts are necessary before suggesting the potential use of Lf as a diagnostic marker or as a therapeutic tool. FUTURE DIRECTIONS The main sources of Lf in human cardiometabolic disorders should be clarified to identify new perspectives for future research and develop new strategies using Lf in therapeutics. Antioxid. Redox Signal. 24, 813-836.
Collapse
Affiliation(s)
- Sylvain Mayeur
- 1 Research Centre, CHU Ste-Justine, Université de Montréal , Montreal, Canada .,2 Institute of Nutraceuticals and Functional Foods (INAF) , Université Laval, Quebec, Canada
| | - Schohraya Spahis
- 1 Research Centre, CHU Ste-Justine, Université de Montréal , Montreal, Canada .,2 Institute of Nutraceuticals and Functional Foods (INAF) , Université Laval, Quebec, Canada .,3 Department of Nutrition, Université de Montréal , Montreal, Canada
| | - Yves Pouliot
- 3 Department of Nutrition, Université de Montréal , Montreal, Canada
| | - Emile Levy
- 1 Research Centre, CHU Ste-Justine, Université de Montréal , Montreal, Canada .,2 Institute of Nutraceuticals and Functional Foods (INAF) , Université Laval, Quebec, Canada .,3 Department of Nutrition, Université de Montréal , Montreal, Canada
| |
Collapse
|
35
|
Hatzios SK, Abel S, Martell J, Hubbard T, Sasabe J, Munera D, Clark L, Bachovchin DA, Qadri F, Ryan ET, Davis BM, Weerapana E, Waldor MK. Chemoproteomic profiling of host and pathogen enzymes active in cholera. Nat Chem Biol 2016; 12:268-274. [PMID: 26900865 PMCID: PMC4765928 DOI: 10.1038/nchembio.2025] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 12/31/2015] [Indexed: 12/15/2022]
Abstract
Activity-based protein profiling (ABPP) is a chemoproteomic tool for detecting active enzymes in complex biological systems. We used ABPP to identify secreted bacterial and host serine hydrolases that are active in animals infected with the cholera pathogen Vibrio cholerae. Four V. cholerae proteases were consistently active in infected rabbits, and one, VC0157 (renamed IvaP), was also active in human choleric stool. Inactivation of IvaP influenced the activity of other secreted V. cholerae and rabbit enzymes in vivo, and genetic disruption of all four proteases increased the abundance of intelectin, an intestinal lectin, and its binding to V. cholerae in infected rabbits. Intelectin also bound to other enteric bacterial pathogens, suggesting that it may constitute a previously unrecognized mechanism of bacterial surveillance in the intestine that is inhibited by pathogen-secreted proteases. Our work demonstrates the power of activity-based proteomics to reveal host-pathogen enzymatic dialog in an animal model of infection.
Collapse
Affiliation(s)
- Stavroula K. Hatzios
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Sören Abel
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Department of Pharmacy, University of Tromsø (UiT), The Arctic University of Norway, Tromsø, Norway
| | | | - Troy Hubbard
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Jumpei Sasabe
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Diana Munera
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Lars Clark
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | | | - Firdausi Qadri
- International Centre for Diarrhoeal Disease Research, Bangladesh (ICDDR,B), Dhaka, Bangladesh
| | - Edward T. Ryan
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Brigid M. Davis
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | | | - Matthew K. Waldor
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
36
|
Maeda K, Saigo C, Kito Y, Sakuratani T, Yoshida K, Takeuchi T. Expression of TMEM207 in Colorectal Cancer: Relation between TMEM207 and Intelectin-1. J Cancer 2016; 7:207-13. [PMID: 26819645 PMCID: PMC4716854 DOI: 10.7150/jca.13732] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/01/2015] [Indexed: 02/06/2023] Open
Abstract
Recent research advances highlighted an intestinal goblet cell-produced lectin, intelectin-1 (also known as omentin-1), as a tumor suppressor. One study indicated that downregulation of intelectin-1 may be related to the unfavorable prognosis among patients with colorectal carcinoma at an advanced stage. The present study was aimed at analyzing the expression of a hitherto uncharacterized transmembrane protein TMEM207 in colorectal carcinoma, and we found that the TMEM207 function is linked to intelectin-1 processing. With specific antibodies, TMEM207 immunoreactivity was detected in 38 of 216 colorectal cancer tissue samples. TMEM207 immunoreactivity correlated inversely with lymph node metastatic status (p < 0.01). TMEM207 expression significantly correlated with the mucinous phenotype of colorectal carcinoma. A coimmunoprecipitation assay revealed an interaction between intelectin-1 and TMEM207 in colorectal cancer cells. A proximal ligation assay indicated that intelectin-1 and TMEM207 were colocalized to the cytoplasm of the colorectal cancer cells. A small-interfering-RNA-mediated knockdown of TMEM207 increased polyubiquitination and proteasome degradation of intelectin-1 in cultured colorectal cancer cells and decreased intelectin-1 secretion. These findings indicate that a loss of TMEM207 expression leads to insufficient intelectin-1 production thus promoting colorectal carcinogenesis.
Collapse
Affiliation(s)
- Kenichi Maeda
- 1. Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Chiemi Saigo
- 2. Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yusuke Kito
- 2. Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takuji Sakuratani
- 1. Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kazuhiro Yoshida
- 1. Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tamotsu Takeuchi
- 2. Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
37
|
Lee SJ, Jung YH, Ryu JM, Jang KK, Choi SH, Han HJ. VvpE mediates the intestinal colonization of Vibrio vulnificus by the disruption of tight junctions. Int J Med Microbiol 2016; 306:10-9. [DOI: 10.1016/j.ijmm.2015.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/17/2015] [Accepted: 10/26/2015] [Indexed: 01/01/2023] Open
|
38
|
Vibrio vulnificus VvpE inhibits mucin 2 expression by hypermethylation via lipid raft-mediated ROS signaling in intestinal epithelial cells. Cell Death Dis 2015; 6:e1787. [PMID: 26086960 PMCID: PMC4669833 DOI: 10.1038/cddis.2015.152] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/08/2015] [Accepted: 05/08/2015] [Indexed: 02/07/2023]
Abstract
Mucin is an important physical barrier against enteric pathogens. VvpE is an elastase encoded by Gram-negative bacterium Vibrio vulnificus; however, the functional role of VvpE in intestinal mucin (Muc) production is yet to be elucidated. The recombinant protein (r) VvpE significantly reduced the level of Muc2 in human mucus-secreting HT29-MTX cells. The repression of Muc2 induced by rVvpE was highly susceptible to the knockdown of intelectin-1b (ITLN) and sequestration of cholesterol by methyl-β-cyclodextrin. We found that rVvpE induces the recruitment of NADPH oxidase 2 and neutrophil cytosolic factor 1 into the membrane lipid rafts coupled with ITLN to facilitate the production of reactive oxygen species (ROS). The bacterial signaling of rVvpE through ROS production is uniquely mediated by the phosphorylation of ERK, which was downregulated by the silencing of the PKCδ. Moreover, rVvpE induced region-specific methylation in the Muc2 promoter to promote the transcriptional repression of Muc2. In two mouse models of V. vulnificus infection, the mutation of the vvpE gene from V. vulnificus exhibited an increased survival rate and maintained the level of Muc2 expression in intestine. These results demonstrate that VvpE inhibits Muc2 expression by hypermethylation via lipid raft-mediated ROS signaling in the intestinal epithelial cells.
Collapse
|
39
|
Pathogenesis of human diffusely adhering Escherichia coli expressing Afa/Dr adhesins (Afa/Dr DAEC): current insights and future challenges. Clin Microbiol Rev 2015; 27:823-69. [PMID: 25278576 DOI: 10.1128/cmr.00036-14] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pathogenicity and clinical pertinence of diffusely adhering Escherichia coli expressing the Afa/Dr adhesins (Afa/Dr DAEC) in urinary tract infections (UTIs) and pregnancy complications are well established. In contrast, the implication of intestinal Afa/Dr DAEC in diarrhea is still under debate. These strains are age dependently involved in diarrhea in children, are apparently not involved in diarrhea in adults, and can also be asymptomatic intestinal microbiota strains in children and adult. This comprehensive review analyzes the epidemiology and diagnosis and highlights recent progress which has improved the understanding of Afa/Dr DAEC pathogenesis. Here, I summarize the roles of Afa/Dr DAEC virulence factors, including Afa/Dr adhesins, flagella, Sat toxin, and pks island products, in the development of specific mechanisms of pathogenicity. In intestinal epithelial polarized cells, the Afa/Dr adhesins trigger cell membrane receptor clustering and activation of the linked cell signaling pathways, promote structural and functional cell lesions and injuries in intestinal barrier, induce proinflammatory responses, create angiogenesis, instigate epithelial-mesenchymal transition-like events, and lead to pks-dependent DNA damage. UTI-associated Afa/Dr DAEC strains, following adhesin-membrane receptor cell interactions and activation of associated lipid raft-dependent cell signaling pathways, internalize in a microtubule-dependent manner within urinary tract epithelial cells, develop a particular intracellular lifestyle, and trigger a toxin-dependent cell detachment. In response to Afa/Dr DAEC infection, the host epithelial cells generate antibacterial defense responses. Finally, I discuss a hypothetical role of intestinal Afa/Dr DAEC strains that can act as "silent pathogens" with the capacity to emerge as "pathobionts" for the development of inflammatory bowel disease and intestinal carcinogenesis.
Collapse
|
40
|
The human gastrointestinal tract-specific transcriptome and proteome as defined by RNA sequencing and antibody-based profiling. J Gastroenterol 2015; 50:46-57. [PMID: 24789573 DOI: 10.1007/s00535-014-0958-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/07/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND The gastrointestinal tract (GIT) is subdivided into different anatomical organs with many shared functions and characteristics, but also distinct differences. We have combined a genome-wide transcriptomics analysis with immunohistochemistry-based protein profiling to describe the gene and protein expression patterns that define the human GIT. METHODS RNA sequencing data derived from stomach, duodenum, jejunum/ileum and colon specimens were compared to gene expression levels in 23 other normal human tissues analysed with the same method. Protein profiling based on immunohistochemistry and tissue microarrays was used to sub-localize the corresponding proteins with GIT-specific expression into sub-cellular compartments and cell types. RESULTS Approximately 75% of all human protein-coding genes were expressed in at least one of the GIT tissues. Only 51 genes showed enriched expression in either one of the GIT tissues and an additional 83 genes were enriched in two or more GIT tissues. The list of GIT-enriched genes with validated protein expression patterns included various well-known but also previously uncharacterised or poorly studied genes. For instance, the colon-enriched expression of NXPE family member 1 (NXPE1) was established, while NLR family, pyrin domain-containing 6 (NLRP6) expression was primarily found in the human small intestine. CONCLUSIONS We have applied a genome-wide analysis based on transcriptomics and antibody-based protein profiling to identify genes that are expressed in a specific manner within the human GIT. These genes and proteins constitute important starting points for an improved understanding of the normal function and the different states of disease associated with the GIT.
Collapse
|
41
|
Antisecretory factor peptide AF-16 inhibits the secreted autotransporter toxin-stimulated transcellular and paracellular passages of fluid in cultured human enterocyte-like cells. Infect Immun 2014; 83:907-22. [PMID: 25534938 DOI: 10.1128/iai.02759-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Both the endogenous antisecretory factor (AF) protein and peptide AF-16, which has a sequence that matches that of the active N-terminal region of AF, inhibit the increase in the epithelial transport of fluid and electrolytes induced by bacterial toxins in animal and ex vivo models. We conducted a study to investigate the inhibitory effect of peptide AF-16 against the increase of transcellular passage and paracellular permeability promoted by the secreted autotransporter toxin (Sat) in a cultured cellular model of the human intestinal epithelial barrier. Peptide AF-16 produced a concentration-dependent inhibition of the Sat-induced increase in the formation of fluid domes, in the mucosal-to-serosal passage of D-[1-(14)C]mannitol, and in the rearrangements in the distribution and protein expression of the tight junction (TJ)-associated proteins ZO-1 and occludin in cultured human enterocyte-like Caco-2/TC7 cell monolayers. In addition, we show that peptide AF-16 also inhibits the cholera toxin-induced increase of transcellular passage and the Clostridium difficile toxin-induced effects on paracellular permeability and TJ protein organization in Caco-2/TC7 cell monolayers. Treatment of cell monolayers by the lipid raft disorganizer methyl-β-cyclodextrin abolished the inhibitory activity of peptide AF-16 at the transcellular passage level and did not modify the effect of the peptide at the paracellular level.
Collapse
|
42
|
Su P, Zheng Z, Pang Y, Xue Z, Gou M, Han Y, Liu G, Zan Q, Li Q. Preparation, identfication, and activity assay of lamprey (lampetra japonica) natural intelectins. J Immunoassay Immunochem 2014; 36:368-78. [PMID: 25275839 DOI: 10.1080/15321819.2014.955583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Intelectins play an important role in innate immune response. In a previous study, lamprey inteletins purified by galactose-Sepharose were inactive and insoluble. Herein, we provided a simple and effective method to purify natural intelectins from the serum of lamprey (Lethenteron japonicum) using proteinG agarose. SDS-PAGE, two-dimensional polyacrylamide gel electrophoresis (2D-PAGE), and mass spectrometry (MS) were used to analyze the purified proteins. The purified proteins were identified to be lamprey serum lectin and intelectinB. The activity analysis results indicated that the proteins had certain extent agglutination activity. The effective method will be useful to study their immune functions and molecular mechanisms.
Collapse
Affiliation(s)
- Peng Su
- a College of Life Science , Liaoning Normal University , Dalian , China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Technological advances in the large scale analysis of human genetics have generated profound insights into possible genetic contributions to chronic diseases including the inflammatory bowel diseases (IBDs), Crohn's disease and ulcerative colitis. To date, 163 distinct genetic risk loci have been associated with either Crohn's disease or ulcerative colitis, with a substantial degree of genetic overlap between these 2 conditions. Although many risk variants show a reproducible correlation with disease, individual gene associations only affect a subset of patients, and the functional contribution(s) of these risk variants to the onset of IBD is largely undetermined. Although studies in twins have demonstrated that the development of IBD is not mediated solely by genetic risk, it is nevertheless important to elucidate the functional consequences of risk variants for gene function in relevant cell types known to regulate key physiological processes that are compromised in IBD. This article will discuss IBD candidate genes that are known to be, or are suspected of being, involved in regulating the intestinal epithelial barrier and several of the physiological processes presided over by this dynamic and versatile layer of cells. This will include assembly and regulation of tight junctions, cell adhesion and polarity, mucus and glycoprotein regulation, bacterial sensing, membrane transport, epithelial differentiation, and restitution.
Collapse
|
44
|
Reznikov EA, Comstock SS, Yi C, Contractor N, Donovan SM. Dietary bovine lactoferrin increases intestinal cell proliferation in neonatal piglets. J Nutr 2014; 144:1401-8. [PMID: 25056692 DOI: 10.3945/jn.114.196568] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Lactoferrin is a bioactive milk protein that stimulates cell proliferation in vitro; however, limited in vivo evidence exists to allow lactoferrin to be incorporated into infant formula. Herein, the effect of dietary bovine lactoferrin (bLF) on neonatal intestinal growth and maturation was investigated guided by the hypothesis that bLF would increase cellular proliferation leading to functional differences in neonatal piglets. Colostrum-deprived piglets were fed formula containing 0.4 [control (Ctrl)], 1.0 (LF1), or 3.6 (LF3) g bLF/L for the first 7 or 14 d of life. To provide passive immunity, sow serum was provided orally during the first 36 h of life. Intestinal cell proliferation, histomorphology, mucosal DNA concentration, enzyme activity, gene expression, and fecal bLF content were measured. Intestinal enzyme activity, DNA concentration, and villus length were unaffected by bLF. However, crypt proliferation was 60% greater in LF1- and LF3-fed piglets than in Ctrl piglets, and crypt depth and area were 20% greater in LF3-fed piglets than in Ctrl piglets. Crypt cells from LF3-fed piglets had 3-fold higher β-catenin mRNA expression than did crypt cells from Ctrl piglets. Last, feces of piglets fed bLF contained intact bLF, suggesting that some bLF was resistant to digestion and could potentially affect intestinal proliferation through direct interaction with intestinal epithelial cells. This study is the first to our knowledge to show that dietary bLF stimulates crypt cell proliferation in vivo. The increased β-catenin expression indicates that Wnt signaling may in part mediate the stimulatory effect of bLF on intestinal cell proliferation.
Collapse
Affiliation(s)
| | - Sarah S Comstock
- Food Science and Human Nutrition, University of Illinois, Urbana, IL; and
| | - Cuiyi Yi
- Wyeth Nutrition, King of Prussia, PA
| | | | - Sharon M Donovan
- Division of Nutritional Sciences Food Science and Human Nutrition, University of Illinois, Urbana, IL; and
| |
Collapse
|
45
|
Zhang X, Wu W. Ligand-mediated active targeting for enhanced oral absorption. Drug Discov Today 2014; 19:898-904. [DOI: 10.1016/j.drudis.2014.03.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/21/2014] [Accepted: 03/03/2014] [Indexed: 01/08/2023]
|
46
|
Ren M, Liu C, Zeng X, Yue L, Mao X, Qiao S, Wang J. Amino acids modulates the intestinal proteome associated with immune and stress response in weaning pig. Mol Biol Rep 2014; 41:3611-20. [PMID: 24510411 DOI: 10.1007/s11033-014-3225-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 01/29/2014] [Indexed: 10/25/2022]
Abstract
The objective of this study was to investigate the effects of free amino acids supplementation to protein restricted diet on the intestinal morphology and proteome composition in weaning pigs. Weanling piglets were randomly fed one of the three diets including a corn-soybean based control diet and two lower protein diets with or without free amino acids supplementation for 2 weeks. The jejunum samples of piglets were collected for morphology and proteome analysis. Compared with the control diet, the protein restricted diet had a significant lower average daily gain and higher feed conversion rate. Free amino acids supplementation to the protein restricted diet significantly improved average daily gain and higher feed conversion rate, compared with the protein restricted diet. The villous height in pigs fed the protein restricted diet was lower than that of the control and free amino acids diet. Using two-dimensional gel electrophoresis and mass spectrometry, we identified 16 differentially expressed protein spots in the jejunum of the weaning piglet. These proteins were related to stress and immune response, the metabolism of carbohydrates and lipids, and tissue structure. Based on the proteome and ELISA analysis, free amino acids diet significantly down-regulated the jejunal expression of stress protein heat shock 60 kDa protein. Our results indicated that amino acids supplementation to the protein restricted diet could enhance weight gain and feed efficiency in weanling pigs through improving intestinal nutrient absorption and transportation, gut health, and mucosal immunity.
Collapse
Affiliation(s)
- Man Ren
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2. Yuanmingyuan West Road, Beijing, 100193, China
| | | | | | | | | | | | | |
Collapse
|
47
|
Comstock SS, Reznikov EA, Contractor N, Donovan SM. Dietary bovine lactoferrin alters mucosal and systemic immune cell responses in neonatal piglets. J Nutr 2014; 144:525-32. [PMID: 24553692 DOI: 10.3945/jn.113.190264] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Lactoferrin (LF) is a multifunctional immune protein found at high concentrations in human milk. Herein, the effect of dietary bovine LF (bLF) on mucosal and systemic immune development was investigated. Colostrum-deprived piglets were fed formula containing 130 [control (Ctrl)], 367 (LF1), or 1300 (LF3) mg of bLF/(kg body weight · d). To provide passive immunity, sow serum was provided orally during the first 36 h of life. Blood, spleen, mesenteric lymph node (MLN), and ascending colon (Asc) contents were collected on day 7 (n = 10-14/group) and day 14 (n = 10-12/group). Immune cell populations were quantified by flow cytometry and immunoglobulins (Igs) were measured by ELISA. Additionally, immune cells were isolated from spleen and MLNs (n = 7/group) on day 7 and stimulated ex vivo with phytohemagglutinin or lipopolysaccharide (LPS) ± LF for 72 h. Secreted cytokine concentrations were quantified by multiplex assay. Lymphocyte populations [cluster determinant (CD)4, CD8, and natural killer cells] developed normally and were unaffected by dietary bLF. LF3 piglets tended to have 1.4 to 2 times more serum IgG than Ctrl piglets (P = 0.07) or LF1 piglets (P = 0.03), but IgA in Asc contents was unaffected by bLF. Asc IgA was 4 times higher on day 14 than day 7. Spleen cells from LF3 piglets produced 2 times more interleukin (IL)-10 and tumor necrosis factor (TNF)-α ex vivo than those from Ctrl or LF1 piglets. MLN cells from LF1 and LF3 piglets produced 40% more IL-10 and tended to produce 40% more IL-6 (P = 0.05) than those from Ctrl piglets. However, ex vivo bLF did not affect the cytokine response of spleen or MLN cells to LPS. In summary, dietary bLF alters the capacity of MLN and spleen immune cells to respond to stimulation, supporting a role for LF in the initiation of protective immune responses in these immunologically challenged neonates.
Collapse
|
48
|
Xue Z, Pang Y, Liu X, Zheng Z, Xiao R, Jin M, Han Y, Su P, Lv L, Wang J, Li Q. First evidence of protein G-binding protein in the most primitive vertebrate: serum lectin from lamprey (Lampetra japonica). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:618-630. [PMID: 23806362 DOI: 10.1016/j.dci.2013.06.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 06/16/2013] [Accepted: 06/17/2013] [Indexed: 06/02/2023]
Abstract
The intelectins, a recently identified subgroup of extracellular animal lectins, are glycan-binding receptors that recognize glycan epitopes on foreign pathogens in host systems. Here, we have described NPGBP (novel protein G-binding protein), a novel serum lectin found in the lamprey, Lampetra japonica. RT-PCR yielded a 1005 bp cDNA sequence from the lamprey liver encoding a 334 amino acid secretory protein with homology to mammalian and aquatic organism intelectins. Gene expression analyses showed that the NPGBP gene was expressed in the blood, intestines, kidney, heart, gill, liver, adipose tissue and gonads. NPGBP was isolated by protein G-conjugated agarose immunoprecipitation, and SDS-PAGE analyses showed that NPGBP migrated as a specific band (∼35 and ∼124 kDa under reducing and non-reducing conditions, respectively). These results suggested that NPGBP forms monomers and tetramers. NPGBP gene expression was induced by in vivo bacterial stimulation, and NPGBP showed different agglutination activities against pathogenic Gram-positive bacteria, Gram-negative bacteria and fungi. The induction of NPGBP suggested that it plays an important role in defense against microorganisms in the internal circulation system of the lamprey. When incubated with an unrelated antibody, the specific binding between NPGBP and protein G was competitively inhibited, indicating that NPGBP and the Fc region of Ig bind to the same site on protein G. We thus assume that the tertiary structure of NPGBP is similar to that of the Fc region of Ig. Additionally, NPGBP can effectively promote endothelial cell mitosis. These findings suggest that NPGBP plays a role in the immune defense against microorganisms, and this study represents one of the few examples of the characterization and functional analysis of an aquatic organism intelectin.
Collapse
Affiliation(s)
- Zhuang Xue
- Institute of Marine Genomics and Proteomics, Liaoning Normal University, Dalian 116029, China; School of Life Science, Liaoning Normal University, Dalian 116029, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yan J, Xu L, Zhang Y, Zhang C, Zhang C, Zhao F, Feng L. Comparative genomic and phylogenetic analyses of the intelectin gene family: implications for their origin and evolution. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 41:189-199. [PMID: 23643964 DOI: 10.1016/j.dci.2013.04.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 04/08/2013] [Accepted: 04/23/2013] [Indexed: 06/02/2023]
Abstract
Intelectin is a newly characterized gene family involved in early embryogenesis, host-pathogen interactions and iron metabolism. In this study, we searched the genomes of metazoans by extensive BLAST survey and found no intelectin homologs in invertebrate metazoans but 12 in amphioxus Branchiostoma floridae and 21 in ascidians Ciona intestinalis. Some ascidians oocyte cortical granule lectins (CGLs) have unknown insertion sequences between fibrinogen-related domain (FReD) and Intelectin Domain, the boundaries of which are equivalent to exon structures. In addition to ascidians intelectins/CGLs located in the base, phylogenetic tree comprises four main clades representing mammal, frog, fish, and amphioxus, indicating that intelectin genes undergo extensive lineage-specific duplication or gene conversion. However, genomic neighborhood surrounding analysis shows that clear proto-orthologies are difficult to be established among these counterparts. In addition, sequence comparison and phylogenetic analysis of FReDs from intelectins and other fibrinogen-like proteins from choanoflagellate, anemone, frog and human indicate FReDs of intelectins are unique. Likewise, these choanoflagellate and anemone genes may be close to intelectin gene.
Collapse
Affiliation(s)
- Jie Yan
- Marine Biotechnology Research Center, Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, Institute of Developmental Biology, School of Life Sciences, Shandong University, Jinan, Shandong 250100, PR China.
| | | | | | | | | | | | | |
Collapse
|
50
|
Akiyama Y, Oshima K, Kuhara T, Shin K, Abe F, Iwatsuki K, Nadano D, Matsuda T. A lactoferrin-receptor, intelectin 1, affects uptake, sub-cellular localization and release of immunochemically detectable lactoferrin by intestinal epithelial Caco-2 cells. J Biochem 2013; 154:437-48. [PMID: 23921499 DOI: 10.1093/jb/mvt073] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Intelectin 1 (IntL) is known as a lectin expressed in intestinal epithelia and also as a receptor for an iron-binding protein, lactoferrin (LF). Uptake of LF with bound iron by enterocytes via receptor-mediated endocytosis has been well investigated, whereas subsequent fate of endocytized LF and LF/IntL complexes remains largely unknown. In the present study, we examined contribution of IntL to the uptake, sub-cellular localization and subsequent release of LF by intestinal Caco-2 IntL-transfectants using two-site ELISA and fluorescence confocal microscopy. LF taken up by IntL-transfectants was immunochemically detected mostly as intact protein in the cell lysates, and it was a little larger in amount than that of the mock-transfectants. In the IntL-transfectants cultured on porous membrane, LF taken up from the apical side was detected immunochemically as punctate signals in the apical-side cytoplasmic region near nucleus. The LF signals were co-localized with IntL and, in a time-dependent manner, partially with early endosome antigen 1 (EEA1), but not with alkaline phosphatase. LF taken up, retained and subsequently released by the IntL-transfectants was larger in amount than that of mock-transfectants. Moreover, uptake of LF altered sub-cellular localization of IntL and markedly enhanced the IntL signals within the cells.
Collapse
Affiliation(s)
- Yuka Akiyama
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Chikusa-ku, Nagoya, Aichi 464-8601; and Food Science & Technology Institute, Morinaga Milk Industry Co., Ltd., 5-1-83 Higashihara, Zama, Kanagawa 228-8583, Japan
| | | | | | | | | | | | | | | |
Collapse
|