1
|
Wu HHL, Bhagavath V, Nguyen LT, Chinnadurai R, Goldys EM, Pollock CA, Saad S. Association Between Glycemic Control and Complications With Concentration of Urinary Exfoliated Proximal Tubule Kidney Cells in People With Diabetes Mellitus. J Diabetes Res 2025; 2025:1273073. [PMID: 39850513 PMCID: PMC11756946 DOI: 10.1155/jdr/1273073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/20/2024] [Accepted: 01/06/2025] [Indexed: 01/25/2025] Open
Abstract
Background: Emerging evidence suggests cell exfoliation could be operating under the control of cell metabolism. It is unclear if there are associations between the concentration of exfoliated kidney proximal tubule cells (PTCs) in urine with glycemic control and complications. Our study is aimed at exploring this. Methods: Urine samples were collected from 122 adult study participants and stored at -80°C. Exfoliated PTCs were extracted from thawed urine using a validated specific immunomagnetic separation method based on anti-CD13 and anti-SGLT-2 antibodies. The number of PTCs was assessed using brightfield microscopy. Study participants were grouped into those with no diabetes mellitus (DM) and those with DM. Individuals with DM were further subgrouped into those with and without retinopathy. Adjusted Poisson regression analysis was conducted for the DM cohort, investigating associations between demographic, clinical, and biochemical parameters with mean urinary exfoliated PTCs. Results: The adjusted Poisson regression analysis noted sex to have a significant association with mean number of urinary exfoliated PTCs, with a lower incidence rate in males compared to females (incidence rate ratio (IRR) 0.56, 95% CI 0.35-0.89, p = 0.014). Each 1% increase in glycated haemoglobin (HbA1c) was associated with an increase of 1.03 times in mean exfoliated PTCs (IRR 1.03, 95% CI 1.01-1.04, p = 0.007), and DM patients with retinopathy had an increase of 1.68 times in mean exfoliated PTCs compared to those without retinopathy (IRR 1.68, 95% CI 1.07-2.62, p = 0.024). No significant associations were observed with albuminuria or estimated glomerular filtration rate (eGFR). Conclusions: Our results indicate increased shedding of PTCs into the urinary tract in patients with poorer glycemic control, particularly those with diabetic retinopathy and in females.
Collapse
Affiliation(s)
- Henry H. L. Wu
- Renal Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital & The University of Sydney, Sydney, Australia
- ARC Centre of Excellence for Nanoscale Biophotonics, School of Biomedical Engineering, The University of New South Wales, Sydney, Australia
| | - Venkatesha Bhagavath
- Biostatistics Support and Consultation Services, Northern Sydney Local Health District, Sydney, Australia
| | - Long The Nguyen
- Renal Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital & The University of Sydney, Sydney, Australia
| | - Rajkumar Chinnadurai
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Ewa M. Goldys
- ARC Centre of Excellence for Nanoscale Biophotonics, School of Biomedical Engineering, The University of New South Wales, Sydney, Australia
| | - Carol A. Pollock
- Renal Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital & The University of Sydney, Sydney, Australia
- Department of Renal Medicine, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, Australia
| | - Sonia Saad
- Renal Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital & The University of Sydney, Sydney, Australia
| |
Collapse
|
2
|
Cordeiro S, Oliveira BB, Valente R, Ferreira D, Luz A, Baptista PV, Fernandes AR. Breaking the mold: 3D cell cultures reshaping the future of cancer research. Front Cell Dev Biol 2024; 12:1507388. [PMID: 39659521 PMCID: PMC11628512 DOI: 10.3389/fcell.2024.1507388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Despite extensive efforts to unravel tumor behavior and develop anticancer therapies, most treatments fail when advanced to clinical trials. The main challenge in cancer research has been the absence of predictive cancer models, accurately mimicking the tumoral processes and response to treatments. The tumor microenvironment (TME) shows several human-specific physical and chemical properties, which cannot be fully recapitulated by the conventional 2D cell cultures or the in vivo animal models. These limitations have driven the development of novel in vitro cancer models, that get one step closer to the typical features of in vivo systems while showing better species relevance. This review introduces the main considerations required for developing and exploiting tumor spheroids and organoids as cancer models. We also detailed their applications in drug screening and personalized medicine. Further, we show the transition of these models into novel microfluidic platforms, for improved control over physiological parameters and high-throughput screening. 3D culture models have provided key insights into tumor biology, more closely resembling the in vivo TME and tumor characteristics, while enabling the development of more reliable and precise anticancer therapies.
Collapse
Affiliation(s)
- Sandra Cordeiro
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Beatriz B. Oliveira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ruben Valente
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Daniela Ferreira
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - André Luz
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V. Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alexandra R. Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- i4HB, Associate Laboratory – Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
3
|
II T, CHAMBERS JK, NAKASHIMA K, GOTO-KOSHINO Y, UCHIDA K. Intraepithelial lymphocytes are associated with epithelial injury in feline intestinal T-cell lymphoma. J Vet Med Sci 2024; 86:101-110. [PMID: 38072403 PMCID: PMC10849855 DOI: 10.1292/jvms.23-0339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/07/2023] [Accepted: 11/25/2023] [Indexed: 01/30/2024] Open
Abstract
Our previous study indicated that cytotoxicity of intraepithelial lymphocytes is a poor prognostic factor in feline intestinal T-cell lymphoma (FITL), but the effect of cytotoxic lymphocytes on mucosal epithelium is still unknown. Thus, we investigated the association between cytotoxic lymphocytes and mucosal epithelium in 71 cases of feline intestinal T-cell lymphoma (FITL): epithelial injury, basement membrane injury, cleaved-caspase-3 positivity of epithelial cells, and the number and Ki67 positivity of intraepithelial lymphocytes in granzyme B (GRB)+ and GRB- FITLs were evaluated. Epithelial injury score and the number of intraepithelial lymphocytes in granzyme B (GRB)+ FITL were significantly higher than those of GRB- FITL (P<0.05, P<0.05), but no significant differences were found in the basement membrane injury score, the percentage of cleaved-caspase-3+ epithelial cells, and the percentage of Ki67+ intraepithelial lymphocytes. There was a significant correlation between the epithelial injury score and the number of intraepithelial lymphocytes (P<0.05), but no significant correlation was observed between the epithelial injury score and Ki67+ percentage of intraepithelial lymphocytes. Because epithelial cell cleaved-caspase-3 positivity was observed in FITL, regardless of GRB expression in lymphocytes, GRB-mediated apoptosis may not contribute to epithelial injury in FITL. The association between increased number of intraepithelial lymphocytes and epithelial injury suggests that intraepithelial lymphocytes infiltration may contribute to epithelial injury in FITL.
Collapse
Affiliation(s)
- Tatsuhito II
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - James K CHAMBERS
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ko NAKASHIMA
- Japan Small Animal Medical Center (JSAMC), Tokorozawa, Saitama, Japan
| | - Yuko GOTO-KOSHINO
- Veterinary Medical Center, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki UCHIDA
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
McConnel CS, Slanzon GS, Parrish LM, Trombetta SC, Shaw LF, Moore DA, Sischo WM. Transcriptional changes detected in fecal RNA from neonatal dairy calves of different breeds following gastrointestinal disease of varying severity. PLoS One 2022; 17:e0278664. [PMID: 36454999 PMCID: PMC9714867 DOI: 10.1371/journal.pone.0278664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Gastrointestinal (GI) disease is a major health concern in preweaned dairy calves. The objective of this fixed cohort study was to use RNA isolated from preweaned Holstein and Jersey heifer calf feces to study the molecular adaptations to variable clinical GI disease. The study was conducted on a commercial calf ranch in the western U.S. Enrolled calves were assessed twice daily for variations in demeanor, milk intake, and hydration. Fecal consistency scores were recorded at enrollment (day 1), and on the day (day 10) that a fecal sample was collected for differential gene expression (DGE). Calves with diarrhea on either day were classified as having either uncomplicated, localized GI disease (scours), or systemic GI disease (systemic enteritis). Eighty-four calves' fecal RNA was evaluated for DGE, of which 33 calves (n = 20 Holstein; n = 13 Jersey) were consistently healthy. The remaining 51 calves (n = 23 Holstein; n = 28 Jersey) experienced varying severity of GI disease during the sampling window. Genes of interest were related to the inflammatory response (i.e., IFNG, NFKB1, NOD2, TLR2, and TLR4) and cell membrane or cytoplasmic transport (i.e., AQP3, FABP2, KRT8 and SLC5A1). Breed-specific findings indicated that AQP3, IFNG, and TLR4 were upregulated in Holsteins with systemic enteritis, whereas KRT8 was downregulated in systemically affected Jerseys. Holsteins did not appear affected by scours aside from a tendency for DGE of toll-like receptors (TLRs) on the day of diarrhea. However, Jersey calves consistently demonstrated a tendency to upregulate IFNG, NFKB1, and TLR4 when affected with either scours or systemic enteritis. These findings were more pronounced in systemically affected Jersey calves and were observed as a delayed response to both scours and systemic enteritis. These findings support previous observations suggesting that Holstein calves may be better equipped than Jersey calves to rapidly fight pathogen invasion.
Collapse
Affiliation(s)
- C. S. McConnel
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
- * E-mail:
| | - G. S. Slanzon
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - L. M. Parrish
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - S. C. Trombetta
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - L. F. Shaw
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - D. A. Moore
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| | - W. M. Sischo
- Department of Veterinary Clinical Sciences, Field Disease Investigation Unit, Washington State University, Pullman, Washington, United States of America
| |
Collapse
|
5
|
Wu HHL, Goldys EM, Pollock CA, Saad S. Exfoliated Kidney Cells from Urine for Early Diagnosis and Prognostication of CKD: The Way of the Future? Int J Mol Sci 2022; 23:7610. [PMID: 35886957 PMCID: PMC9324667 DOI: 10.3390/ijms23147610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/30/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic kidney disease (CKD) is a global health issue, affecting more than 10% of the worldwide population. The current approach for formal diagnosis and prognostication of CKD typically relies on non-invasive serum and urine biomarkers such as serum creatinine and albuminuria. However, histological evidence of tubulointerstitial fibrosis is the 'gold standard' marker of the likelihood of disease progression. The development of novel biomedical technologies to evaluate exfoliated kidney cells from urine for non-invasive diagnosis and prognostication of CKD presents opportunities to avoid kidney biopsy for the purpose of prognostication. Efforts to apply these technologies more widely in clinical practice are encouraged, given their potential as a cost-effective approach, and no risk of post-biopsy complications such as bleeding, pain and hospitalization. The identification of biomarkers in exfoliated kidney cells from urine via western blotting, enzyme-linked immunosorbent assay (ELISA), immunofluorescence techniques, measurement of cell and protein-specific messenger ribonucleic acid (mRNA)/micro-RNA and other techniques have been reported. Recent innovations such as multispectral autofluorescence imaging and single-cell RNA sequencing (scRNA-seq) have brought additional dimensions to the clinical application of exfoliated kidney cells from urine. In this review, we discuss the current evidence regarding the utility of exfoliated proximal tubule cells (PTC), podocytes, mesangial cells, extracellular vesicles and stem/progenitor cells as surrogate markers for the early diagnosis and prognostication of CKD. Future directions for development within this research area are also identified.
Collapse
Affiliation(s)
- Henry H. L. Wu
- Renal Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW 2065, Australia; (H.H.L.W.); (C.A.P.)
- School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia;
| | - Ewa M. Goldys
- School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia;
| | - Carol A. Pollock
- Renal Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW 2065, Australia; (H.H.L.W.); (C.A.P.)
| | - Sonia Saad
- Renal Research Laboratory, Kolling Institute of Medical Research, The University of Sydney, Sydney, NSW 2065, Australia; (H.H.L.W.); (C.A.P.)
| |
Collapse
|
6
|
Brogna C, Brogna B, Bisaccia DR, Lauritano F, Marino G, Montano L, Cristoni S, Prisco M, Piscopo M. Could SARS-CoV-2 Have Bacteriophage Behavior or Induce the Activity of Other Bacteriophages? Vaccines (Basel) 2022; 10:vaccines10050708. [PMID: 35632464 PMCID: PMC9143435 DOI: 10.3390/vaccines10050708] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/09/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 02/04/2023] Open
Abstract
SARS-CoV-2 has become one of the most studied viruses of the last century. It was assumed that the only possible host for these types of viruses was mammalian eukaryotic cells. Our recent studies show that microorganisms in the human gastrointestinal tract affect the severity of COVID-19 and for the first time provide indications that the virus might replicate in gut bacteria. In order to further support these findings, in the present work, cultures of bacteria from the human microbiome and SARS-CoV-2 were analyzed by electron and fluorescence microscopy. The images presented in this article, in association with the nitrogen (15N) isotope-labeled culture medium experiment, suggest that SARS-CoV-2 could also infect bacteria in the gut microbiota, indicating that SARS-CoV-2 could act as a bacteriophage. Our results add new knowledge to the understanding of the mechanisms of SARS-CoV-2 infection and fill gaps in the study of the interactions between SARS-CoV-2 and non-mammalian cells. These findings could be useful in suggesting specific new pharmacological solutions to support the vaccination campaign.
Collapse
Affiliation(s)
- Carlo Brogna
- Department of Research, Craniomed Group Facility Srl., 20091 Bresso, Italy; (D.R.B.); (F.L.)
- Correspondence: (C.B.); (M.P.)
| | - Barbara Brogna
- Department of Radiology, Moscati Hospital, Contrada Amoretta, 83100 Avellino, Italy;
| | - Domenico Rocco Bisaccia
- Department of Research, Craniomed Group Facility Srl., 20091 Bresso, Italy; (D.R.B.); (F.L.)
| | - Francesco Lauritano
- Department of Research, Craniomed Group Facility Srl., 20091 Bresso, Italy; (D.R.B.); (F.L.)
| | - Giuliano Marino
- Marsan Consulting Srl., Public Health Company, Via dei Fiorentini, 80133 Naples, Italy;
| | - Luigi Montano
- Andrology Unit and Service of Life Style Medicine in Uro-Andrology, Local Health Authority (ASL), 84124 Salerno, Italy;
| | | | - Marina Prisco
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy;
| | - Marina Piscopo
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy;
- Correspondence: (C.B.); (M.P.)
| |
Collapse
|
7
|
Yoon G, Davidson LA, Goldsby JS, Mullens DA, Ivanov I, Donovan SM, Chapkin RS. Exfoliated epithelial cell transcriptome reflects both small and large intestinal cell signatures in piglets. Am J Physiol Gastrointest Liver Physiol 2021; 321:G41-G51. [PMID: 33949197 PMCID: PMC8321797 DOI: 10.1152/ajpgi.00017.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/08/2023]
Abstract
Assessing intestinal development and host-microbe interactions in healthy human infants requires noninvasive approaches. We have shown that the transcriptome of exfoliated epithelial cells in feces can differentiate breast-fed and formula-fed infants and term and preterm infants. However, it is not fully understood which regions of the intestine that the exfoliated cells represent. Herein, the transcriptional profiles of exfoliated cells with that of the ileal and colonic mucosa were compared. We hypothesized that exfoliated cells in the distal colon would reflect mucosal signatures of more proximal regions of the gut. Two-day-old piglets (n = 8) were fed formulas for 20 days. Luminal contents and mucosa were collected from ileum (IL), ascending colon (AC), and descending (DC) colon, and mRNA was extracted and sequenced. On average, ∼13,000 genes were mapped in mucosal tissues and ∼10,000 in luminal contents. The intersection of detected genes between three mucosa regions and DC exfoliome indicated an approximately 99% overlap. On average, 49% of the genes in IL, AC, and DC mucosa were present in the AC and DC exfoliome. Genes expressed predominantly in specific anatomic sites (stomach, pancreas, small intestine, colon) were detectable in exfoliated cells. In addition, gene markers for all intestinal epithelial cell types were expressed in the exfoliome representing a diverse array of cell types arising from both the small and large intestine. Genes were mapped to nutrient absorption and transport and immune function. Thus, the exfoliome represents a robust reservoir of information in which to assess intestinal development and responses to dietary interventions.NEW & NOTEWORTHY The transcriptome of exfoliated epithelial cells in stool contain gene signatures from both small and large intestinal mucosa affording a noninvasive approach to assess gut health and function.
Collapse
Affiliation(s)
- Grace Yoon
- 1Department of Statistics, Texas A&M University, College Station, Texas
| | - Laurie A. Davidson
- 2Department of Nutrition, Texas A&M University, College Station Texas,3Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, Texas
| | - Jennifer S. Goldsby
- 2Department of Nutrition, Texas A&M University, College Station Texas,3Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, Texas
| | - Destiny A. Mullens
- 3Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, Texas,4Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas
| | - Ivan Ivanov
- 4Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas
| | - Sharon M. Donovan
- 5Department of Food Science and Human Nutrition, University of Illinois, Urbana-Champaign, Illinois
| | - Robert S. Chapkin
- 2Department of Nutrition, Texas A&M University, College Station Texas,3Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, Texas
| |
Collapse
|
8
|
Phosphatidic acid-mediated binding and mammalian cell internalization of the Vibrio cholerae cytotoxin MakA. PLoS Pathog 2021; 17:e1009414. [PMID: 33735319 PMCID: PMC8009392 DOI: 10.1371/journal.ppat.1009414] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/15/2020] [Revised: 03/30/2021] [Accepted: 02/22/2021] [Indexed: 01/08/2023] Open
Abstract
Vibrio cholerae is a noninvasive intestinal pathogen extensively studied as the causative agent of the human disease cholera. Our recent work identified MakA as a potent virulence factor of V. cholerae in both Caenorhabditis elegans and zebrafish, prompting us to investigate the potential contribution of MakA to pathogenesis also in mammalian hosts. In this study, we demonstrate that the MakA protein could induce autophagy and cytotoxicity of target cells. In addition, we observed that phosphatidic acid (PA)-mediated MakA-binding to the host cell plasma membranes promoted macropinocytosis resulting in the formation of an endomembrane-rich aggregate and vacuolation in intoxicated cells that lead to induction of autophagy and dysfunction of intracellular organelles. Moreover, we functionally characterized the molecular basis of the MakA interaction with PA and identified that the N-terminal domain of MakA is required for its binding to PA and thereby for cell toxicity. Furthermore, we observed that the ΔmakA mutant outcompeted the wild-type V. cholerae strain A1552 in the adult mouse infection model. Based on the findings revealing mechanistic insights into the dynamic process of MakA-induced autophagy and cytotoxicity we discuss the potential role played by the MakA protein during late stages of cholera infection as an anti-colonization factor. Vibrio cholerae is the cause of cholera, an infectious disease causing watery diarrhea that can lead to fatal dehydration. The bacteria can readily adapt to different environments, such as from its natural aquatic habitats to the human digestive system. Recently, we reported a novel V. cholerae cytotoxin, MakA that functions as a potent virulence factor in C. elegans and zebrafish. Here we identified phosphatidic acid as a lipid target for MakA interaction with mammalian cells. This interaction promoted macropinocytosis resulting in the formation of an endomembrane-rich aggregate in intoxicated cells that ultimately lead to activation of autophagy. Importantly, data from bacterial colonization in a mouse infection model suggested that MakA might act as an anti-colonization factor of V. cholerae, presumably expressed during later stage(s) of infection. MakA might be explored as a new target for diagnostics and therapeutic developments against V. cholerae infections. Our findings will contribute to further understanding of the virulence, colonization and post-infection spread of V. cholerae.
Collapse
|
9
|
Primary differentiated respiratory epithelial cells respond to apical measles virus infection by shedding multinucleated giant cells. Proc Natl Acad Sci U S A 2021; 118:2013264118. [PMID: 33836570 DOI: 10.1073/pnas.2013264118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022] Open
Abstract
Measles virus (MeV) is highly infectious by the respiratory route and remains an important cause of childhood mortality. However, the process by which MeV infection is efficiently established in the respiratory tract is controversial with suggestions that respiratory epithelial cells are not susceptible to infection from the apical mucosal surface. Therefore, it has been hypothesized that infection is initiated in lung macrophages or dendritic cells and that epithelial infection is subsequently established through the basolateral surface by infected lymphocytes. To better understand the process of respiratory tract initiation of MeV infection, primary differentiated respiratory epithelial cell cultures were established from rhesus macaque tracheal and nasal tissues. Infection of these cultures with MeV from the apical surface was more efficient than from the basolateral surface with shedding of viable MeV-producing multinucleated giant cell (MGC) syncytia from the surface. Despite presence of MGCs and infectious virus in supernatant fluids after apical infection, infected cells were not detected in the adherent epithelial sheet and transepithelial electrical resistance was maintained. After infection from the basolateral surface, epithelial damage and large clusters of MeV-positive cells were observed. Treatment with fusion inhibitory peptides showed that MeV production after apical infection was not dependent on infection of the basolateral surface. These results are consistent with the hypothesis that MeV infection is initiated by apical infection of respiratory epithelial cells with subsequent infection of lymphoid tissue and systemic spread.
Collapse
|
10
|
Assessing the Multivariate Relationship between the Human Infant Intestinal Exfoliated Cell Transcriptome (Exfoliome) and Microbiome in Response to Diet. Microorganisms 2020; 8:microorganisms8122032. [PMID: 33353204 PMCID: PMC7766018 DOI: 10.3390/microorganisms8122032] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/08/2020] [Revised: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota and the host exist in a mutualistic relationship, with the functional composition of the microbiota strongly influencing the health and well-being of the host. In addition to the standard differential expression analysis of host genes to assess the complex cross-talk between environment (diet), microbiome, and host intestinal physiology, data-driven integrative approaches are needed to identify potential biomarkers of both host genes and microbial communities that characterize these interactions. Our findings demonstrate that the complementary application of univariate differential gene expression analysis and multivariate approaches such as sparse Canonical Correlation Analysis (sCCA) and sparse Principal Components Analysis (sPCA) can be used to integrate data from both the healthy infant gut microbial community and host transcriptome (exfoliome) using stool derived exfoliated cells shed from the gut. These approaches reveal host genes and microbial functional categories related to the feeding phenotype of the infants. Our findings also confirm that combinatorial noninvasive -omic approaches provide an integrative genomics-based perspective of neonatal host-gut microbiome interactions.
Collapse
|
11
|
Pradhan S, Karve SS, Weiss AA, Hawkins J, Poling HM, Helmrath MA, Wells JM, McCauley HA. Tissue Responses to Shiga Toxin in Human Intestinal Organoids. Cell Mol Gastroenterol Hepatol 2020; 10:171-190. [PMID: 32145469 PMCID: PMC7240222 DOI: 10.1016/j.jcmgh.2020.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/10/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Shiga toxin (Stx)-producing Escherichia coli (eg, O157:H7) infection produces bloody diarrhea, while Stx inhibits protein synthesis and causes the life-threatening systemic complication of hemolytic uremic syndrome. The murine intestinal tract is resistant to O157:H7 and Stx, and human cells in culture fail to model the complex tissue responses to intestinal injury. We used genetically identical, human stem cell-derived intestinal tissues of varying complexity to study Stx toxicity in vitro and in vivo. METHODS In vitro susceptibility to apical or basolateral exposure to Stx was assessed using human intestinal organoids (HIOs) derived from embryonic stem cells, or enteroids derived from multipotent intestinal stem cells. HIOs contain a lumen, with a single layer of differentiated epithelium surrounded by mesenchymal cells. Enteroids only contain epithelium. In vivo susceptibility was assessed using HIOs, with or without an enteric nervous system, transplanted into mice. RESULTS Stx induced necrosis and apoptotic death in both epithelial and mesenchymal cells. Responses that require protein synthesis (cellular proliferation and wound repair) also were observed. Epithelial barrier function was maintained even after epithelial cell death was seen, and apical to basolateral translocation of Stx was seen. Tissue cross-talk, in which mesenchymal cell damage caused epithelial cell damage, was observed. Stx induced mesenchymal expression of the epithelial marker E-cadherin, the initial step in mesenchymal-epithelial transition. In vivo responses of HIO transplants injected with Stx mirrored those seen in vitro. CONCLUSIONS Intestinal tissue responses to protein synthesis inhibition by Stx are complex. Organoid models allow for an unprecedented examination of human tissue responses to a deadly toxin.
Collapse
Affiliation(s)
- Suman Pradhan
- Department of Molecular Genetics, University of Cincinnati, Cincinnati, Ohio
| | - Sayali S Karve
- Department of Molecular Genetics, University of Cincinnati, Cincinnati, Ohio
| | - Alison A Weiss
- Department of Molecular Genetics, University of Cincinnati, Cincinnati, Ohio.
| | | | | | | | - James M Wells
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Heather A McCauley
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
12
|
Sharma AK, Pafčo B, Vlčková K, Červená B, Kreisinger J, Davison S, Beeri K, Fuh T, Leigh SR, Burns MB, Blekhman R, Petrželková KJ, Gomez A. Mapping gastrointestinal gene expression patterns in wild primates and humans via fecal RNA-seq. BMC Genomics 2019; 20:493. [PMID: 31200636 PMCID: PMC6567582 DOI: 10.1186/s12864-019-5813-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/03/2018] [Accepted: 05/20/2019] [Indexed: 12/30/2022] Open
Abstract
Background Limited accessibility to intestinal epithelial tissue in wild animals and humans makes it challenging to study patterns of intestinal gene regulation, and hence to monitor physiological status and health in field conditions. To explore solutions to this limitation, we have used a noninvasive approach via fecal RNA-seq, for the quantification of gene expression markers in gastrointestinal cells of free-range primates and a forager human population. Thus, a combination of poly(A) mRNA enrichment and rRNA depletion methods was used in tandem with RNA-seq to quantify and compare gastrointestinal gene expression patterns in fecal samples of wild Gorilla gorilla gorilla (n = 9) and BaAka hunter-gatherers (n = 10) from The Dzanga Sangha Protected Areas, Central African Republic. Results Although only a small fraction (< 4.9%) of intestinal mRNA signals was recovered, the data was sufficient to detect significant functional differences between gorillas and humans, at the gene and pathway levels. These intestinal gene expression differences were specifically associated with metabolic and immune functions. Additionally, non-host RNA-seq reads were used to gain preliminary insights on the subjects’ dietary habits, intestinal microbiomes, and infection prevalence, via identification of fungi, nematode, arthropod and plant RNA. Conclusions Overall, the results suggest that fecal RNA-seq, targeting gastrointestinal epithelial cells can be used to evaluate primate intestinal physiology and gut gene regulation, in samples obtained in challenging conditions in situ. The approach used herein may be useful to obtain information on primate intestinal health, while revealing preliminary insights into foraging ecology, microbiome, and diet. Electronic supplementary material The online version of this article (10.1186/s12864-019-5813-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Barbora Pafčo
- The Czech Academy of Sciences, Institute of Vertebrate Biology, Květná 8, 603 65, Brno, Czech Republic.,Department of Pathology and Parasitology, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1946/1, 612 42, Brno, Czech Republic
| | - Klára Vlčková
- The Czech Academy of Sciences, Institute of Vertebrate Biology, Květná 8, 603 65, Brno, Czech Republic.,Department of Pathology and Parasitology, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1946/1, 612 42, Brno, Czech Republic
| | - Barbora Červená
- The Czech Academy of Sciences, Institute of Vertebrate Biology, Květná 8, 603 65, Brno, Czech Republic.,Department of Pathology and Parasitology, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences Brno, Palackého tř. 1946/1, 612 42, Brno, Czech Republic
| | - Jakub Kreisinger
- The Czech Academy of Sciences, Institute of Vertebrate Biology, Květná 8, 603 65, Brno, Czech Republic.,Department of Zoology, Faculty of Science, Charles University, Viničná 7, 128 44, Praha, Czech Republic
| | - Samuel Davison
- Department of Animal Science, University of Minnesota, Twin Cities, USA
| | - Karen Beeri
- Vanderbilt University medical center Technologies for Advanced Genomics, Vanderbilt University medical center, Nashville, TN, USA
| | - Terence Fuh
- WWF Central African Republic, Bangui, Central African Republic
| | - Steven R Leigh
- Department of Anthropology, University of Colorado, Boulder, CO, USA
| | - Michael B Burns
- Loyola University Chicago, Quinlan Life Sciences Building, Chicago, IL, USA
| | - Ran Blekhman
- Department of Genetics, Cell Biology, and Development, University of Minnesota, Twin Cities, MN, USA.,Department of Ecology, Evolution and Behavior, University of Minnesota, Twin Cities, MN, USA
| | - Klára J Petrželková
- The Czech Academy of Sciences, Institute of Vertebrate Biology, Květná 8, 603 65, Brno, Czech Republic. .,The Czech Academy of Sciences, Biology Centre, Institute of Parasitology, Branišovská 31, 370 05, České Budějovice, Czech Republic. .,Liberec Zoo, Lidové sady 425/1, 460 01, Liberec, Czech Republic.
| | - Andres Gomez
- Department of Animal Science, University of Minnesota, Twin Cities, USA.
| |
Collapse
|
13
|
Freedman JC, Navarro MA, Morrell E, Beingesser J, Shrestha A, McClane BA, Uzal FA. Evidence that Clostridium perfringens Enterotoxin-Induced Intestinal Damage and Enterotoxemic Death in Mice Can Occur Independently of Intestinal Caspase-3 Activation. Infect Immun 2018; 86:e00931-17. [PMID: 29685988 PMCID: PMC6013662 DOI: 10.1128/iai.00931-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/18/2017] [Accepted: 04/15/2018] [Indexed: 01/13/2023] Open
Abstract
Clostridium perfringens enterotoxin (CPE) is responsible for the gastrointestinal symptoms of C. perfringens type A food poisoning and some cases of nonfoodborne gastrointestinal diseases, such as antibiotic-associated diarrhea. In the presence of certain predisposing medical conditions, this toxin can also be absorbed from the intestines to cause enterotoxemic death. CPE action in vivo involves intestinal damage, which begins at the villus tips. The cause of this CPE-induced intestinal damage is unknown, but CPE can induce caspase-3-mediated apoptosis in cultured enterocyte-like Caco-2 cells. Therefore, the current study evaluated whether CPE activates caspase-3 in the intestines and, if so, whether this effect is required for the development of intestinal tissue damage or enterotoxemic lethality. Using a mouse ligated small intestinal loop model, CPE was shown to cause intestinal caspase-3 activation in a dose- and time-dependent manner. Most of this caspase-3 activation occurred in epithelial cells shed from villus tips. However, CPE-induced caspase-3 activation occurred after the onset of tissue damage. Furthermore, inhibition of intestinal caspase-3 activity did not affect the onset of intestinal tissue damage. Similarly, inhibition of intestinal caspase-3 activity did not reduce CPE-induced enterotoxemic lethality in these mice. Collectively, these results demonstrate that caspase-3 activation occurs in the CPE-treated intestine but that this effect is not necessary for the development of CPE-induced intestinal tissue damage or enterotoxemic lethality.
Collapse
Affiliation(s)
- John C Freedman
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mauricio A Navarro
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, California, USA
| | - Eleonora Morrell
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, California, USA
| | - Juliann Beingesser
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, California, USA
| | - Archana Shrestha
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bruce A McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Francisco A Uzal
- California Animal Health and Food Safety Laboratory, San Bernardino Branch, School of Veterinary Medicine, University of California-Davis, San Bernardino, California, USA
| |
Collapse
|
14
|
Rosa F, Busato S, Avaroma FC, Linville K, Trevisi E, Osorio JS, Bionaz M. Transcriptional changes detected in fecal RNA of neonatal dairy calves undergoing a mild diarrhea are associated with inflammatory biomarkers. PLoS One 2018; 13:e0191599. [PMID: 29373601 PMCID: PMC5786293 DOI: 10.1371/journal.pone.0191599] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/21/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022] Open
Abstract
After birth, a newborn calf has to adapt to an extrauterine life characterized by several physiological changes. In particular, maturation of the gastrointestinal tract in a new environment loaded with potential pathogens, which can predispose neonatal calves to develop diarrhea, and is a major cause of morbidity and mortality during the first 4 wks of life. We aimed to investigate the inflammatory adaptations at a transcriptomic level in the gastrointestinal (GI) tract to a mild diarrhea in neonatal dairy calves using RNA isolated from fresh fecal samples. Eight newborn Jersey male calves were used from birth to 5 wks of age and housed in individual pens. After birth, calves received 1.9 L of colostrum from their respective dams. Calves had ad-libitum access to water and starter grain (22% CP) and were fed twice daily a total of 5.6 L pasteurized whole milk. Starter intake, body weight (BW), fecal score, withers height (WH), and rectal temperature (RT) were recorded throughout the experiment. Blood samples were collected weekly for metabolic and inflammatory profiling from wk 0 to wk 5. Fresh fecal samples were collected weekly and immediately flash frozen until RNA was extracted using a Trizol-based method, and subsequently, an RT-qPCR analysis was performed. Orthogonal contrasts were used to evaluate linear or quadratic effects over time. Starter intake, BW, and WH increased over time. Fecal score was greatest (2.6 ± 0.3) during wk 2. The concentrations of IL-6, ceruloplasmin, and haptoglobin had a positive quadratic effect with maximal concentrations during wk 2, which corresponded to the maximal fecal score observed during the same time. The concentration of serum amyloid A decreased over time. The mRNA expression of the proinflammatory related genes TLR4, TNFA, IL8, and IL1B had a positive quadratic effect of time. A time effect was observed for the cell membrane sodium-dependent glucose transporter SLC5A1, for the major carbohydrate facilitated transporter SLC2A2, and water transport function AQP3, where SLC5A1 and AQP3 had a negative quadratic effect over time. Our data support the use of the fecal RNA as a noninvasive tool to investigate intestinal transcriptomic profiling of dairy calves experiencing diarrhea, which would be advantageous for future research including nutritional effects and health conditions.
Collapse
Affiliation(s)
- Fernanda Rosa
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Sebastiano Busato
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Fatima C. Avaroma
- Escuela Agrícola Panamericana El Zamorano, El Zamorano, Francisco Morazán, Honduras
| | - Kali Linville
- Department of Dairy and Food Sciences, South Dakota State University, Brookings, South Dakota, United States of America
| | - Erminio Trevisi
- Istituto di Zootecnica, Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | - Johan S. Osorio
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, Oregon, United States of America
| |
Collapse
|
15
|
Li YI, Libby EF, Lewis MJ, Liu J, Shacka JJ, Hurst DR. Increased autophagic response in a population of metastatic breast cancer cells. Oncol Lett 2016; 12:523-529. [PMID: 27347175 PMCID: PMC4906619 DOI: 10.3892/ol.2016.4613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/17/2015] [Accepted: 04/29/2016] [Indexed: 02/05/2023] Open
Abstract
Breast cancer cells are heterogeneous in their ability to invade and fully metastasize, and thus also in their capacity to survive the numerous stresses encountered throughout the multiple steps of the metastatic cascade. Considering the role of autophagy as a survival response to stress, the present study hypothesized that distinct populations of breast cancer cells may possess an altered autophagic capacity that influences their metastatic potential. It was observed that a metastatic breast cancer cell line, MDA-MB-231, that was sensitive to autophagic induction additionally possessed the ability to proliferate following nutrient deprivation. Furthermore, a selected subpopulation of these cells that survived multiple exposures to starvation conditions demonstrated a heightened response to autophagic induction compared to their parent cells. Although this subpopulation maintained a more grape-like pattern in three-dimensional culture compared to the extended spikes of the parent population, autophagic induction in this subpopulation elicited an invasive phenotype with extended spikes. Taken together, these results suggest that autophagic induction may contribute to the ability of distinct breast cancer cell populations to survive and invade.
Collapse
Affiliation(s)
- Y I Li
- State Key Laboratory of Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Emily Falk Libby
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Monica J Lewis
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Jianzhong Liu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - John J Shacka
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | - Douglas R Hurst
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| |
Collapse
|
16
|
Apoptosis, Necrosis, and Necroptosis in the Gut and Intestinal Homeostasis. Mediators Inflamm 2015; 2015:250762. [PMID: 26483605 PMCID: PMC4592906 DOI: 10.1155/2015/250762] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/04/2015] [Accepted: 09/08/2015] [Indexed: 12/13/2022] Open
Abstract
Intestinal epithelial cells (IECs) form a physiochemical barrier that separates the intestinal lumen from the host's internal milieu and is critical for electrolyte passage, nutrient absorption, and interaction with commensal microbiota. Moreover, IECs are strongly involved in the intestinal mucosal inflammatory response as well as in mucosal innate and adaptive immune responses. Cell death in the intestinal barrier is finely controlled, since alterations may lead to severe disorders, including inflammatory diseases. The emerging picture indicates that intestinal epithelial cell death is strictly related to the maintenance of tissue homeostasis. This review is focused on previous reports on different forms of cell death in intestinal epithelium.
Collapse
|
17
|
Donovan SM, Wang M, Monaco MH, Martin CR, Davidson LA, Ivanov I, Chapkin RS. Noninvasive molecular fingerprinting of host-microbiome interactions in neonates. FEBS Lett 2014; 588:4112-9. [PMID: 25042036 DOI: 10.1016/j.febslet.2014.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/24/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 01/12/2023]
Abstract
The early postnatal period is a critical window for intestinal and immune maturation. Intestinal development and microbiome diversity and composition differ between breast- (BF) and formula-fed (FF) infants. Mechanistic examination into host-microbe relationships in healthy infants has been hindered by ethical constraints surrounding tissue biopsies. Thus, a statistically rigorous analytical framework to simultaneously examine both host and microbial responses to dietary/environmental factors using exfoliated intestinal epithelial cells was developed. Differential expression of ∼1200 genes, including genes regulating intestinal proliferation, differentiation and barrier function, was observed between BF and FF term infants. Canonical correlation analysis uncovered a relationship between microbiome virulence genes and host immunity and defense genes. Lastly, exfoliated cells from preterm and term infants were compared. Pathways associated with immune cell function and inflammation were up-regulated in preterm, whereas cell growth-related genes were up-regulated in the term infants. Thus, coordinate measurement of the transcriptomes of exfoliated epithelial cells and microbiome allows inquiry into mutualistic host-microbe interactions in the infant, which can be used to prospectively study gut development or, retrospectively, to identify potential triggers of disease in banked samples.
Collapse
Affiliation(s)
- Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA.
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA
| | - Marcia H Monaco
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA
| | - Camilia R Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Laurie A Davidson
- Department of Nutrition & Food Science and Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX 77843-2253, USA; Center for Translational Environmental Health Research, Texas A&M University, College Station, TX 77843-2253, USA
| | - Ivan Ivanov
- Department of Nutrition & Food Science and Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX 77843-2253, USA; Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843-2253, USA; Center for Translational Environmental Health Research, Texas A&M University, College Station, TX 77843-2253, USA
| | - Robert S Chapkin
- Department of Nutrition & Food Science and Program in Integrative Nutrition and Complex Diseases, Texas A&M University, College Station, TX 77843-2253, USA; Center for Translational Environmental Health Research, Texas A&M University, College Station, TX 77843-2253, USA
| |
Collapse
|
18
|
Beauséjour M, Thibodeau S, Demers MJ, Bouchard V, Gauthier R, Beaulieu JF, Vachon PH. Suppression of anoikis in human intestinal epithelial cells: differentiation state-selective roles of α2β1, α3β1, α5β1, and α6β4 integrins. BMC Cell Biol 2013; 14:53. [PMID: 24289209 PMCID: PMC4219346 DOI: 10.1186/1471-2121-14-53] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2013] [Accepted: 11/26/2013] [Indexed: 12/26/2022] Open
Abstract
Background Regulation of anoikis in human intestinal epithelial cells (IECs) implicates differentiation state-specific mechanisms. Human IECs express distinct repertoires of integrins according to their state of differentiation. Therefore, we investigated whether α2β1, α3β1, α5β1, and α6β4 integrins perform differentiation state-specific roles in the suppression of IEC anoikis. Results Human (HIEC, Caco-2/15) IECs were exposed to specific antibodies that block the binding activity of integrin subunits (α2, α3, α5, α6, β1 or β4) to verify whether or not their inhibition induced anoikis. The knockdown of α6 was also performed by shRNA. Additionally, apoptosis/anoikis was induced by pharmacological inhibition of Fak (PF573228) or Src (PP2). Anoikis/apoptosis was assayed by DNA laddering, ISEL, and/or caspase activity (CASP-8, -9, or -3). Activation levels of Fak and Src, as well as functional Fak-Src interactions, were also assessed. We report herein that differentiated IECs exhibit a greater sensitivity to anoikis than undifferentiated ones. This involves an earlier onset of anoikis when kept in suspension, as well as significantly greater contributions from β1 and β4 integrins in the suppression of anoikis in differentiated cells, and functional distinctions between β1 and β4 integrins in engaging both Fak and Src, or Src only, respectively. Likewise, Fak performs significantly greater contributions in the suppression of anoikis in differentiated cells. Additionally, we show that α2β1 and α5β1 suppress anoikis in undifferentiated cells, whereas α3β1 does so in differentiated ones. Furthermore, we provide evidence that α6β4 contributes to the suppression of anoikis in a primarily α6 subunit-dependent manner in undifferentiated cells, whereas this same integrin in differentiated cells performs significantly greater contributions in anoikis suppression than its undifferentiated state-counterpart, in addition to doing so through a dependence on both of its subunits. Conclusions Our findings indicate that the suppression of human IEC anoikis implicates differentiation state-selective repertoires of integrins, which in turn results into distinctions in anoikis regulation, and sensitivity, between undifferentiated and differentiated IECs. These data further the functional understanding of the concept that the suppression of anoikis is subjected to cell differentiation state-selective mechanisms.
Collapse
Affiliation(s)
- Marco Beauséjour
- Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, J1H5N4 Sherbrooke, Québec, Canada.
| | | | | | | | | | | | | |
Collapse
|
19
|
Chen SF, Lin YS, Jao SW, Chang YC, Liu CL, Lin YJ, Nieh S. Pulmonary Adenocarcinoma in Malignant Pleural Effusion Enriches Cancer Stem Cell Properties during Metastatic Cascade. PLoS One 2013; 8:e54659. [PMID: 23658677 PMCID: PMC3641054 DOI: 10.1371/journal.pone.0054659] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/12/2012] [Accepted: 12/17/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Metastasis occurs in a series of discrete steps involving invasion, angiogenesis, lymphovascular space permeation, and establishment of secondary tumors. Malignant pleural effusion (MPE), a type of tumor metastasis, is usually a poor prognostic sign for patients with pulmonary adenocarcinoma, although its underlying mechanism has received less attention than other types of metastases have. The objective of the current study was to confirm whether cancer stem cells (CSCs) in MPE contribute to the "metastatic cascade" through the epithelial - mesenchymal transition (EMT), anoikis, and adaptation in the microenvironment. METHODS Pulmonary tissue and corresponding cell blocks of MPE samples from 20 patients with primary adenocarcinoma were analyzed by immunohistochemical staining with CSC-representative markers (CD133, Nanog, and OCT-4) and EMT-associated markers (E-cadherin and vimentin). Correlations between these variables and clinico-pathological parameters were analyzed. Primary cultures from eight cases of MPE were investigated to characterize the CSC properties, including marker expression, sphere formation, and differentiation. RESULTS Expressions of CSC-representative markers for 20 cases of MPE cell blocks were quite diverse and variable ranging from 15% to 90%. Stronger expression of CSC-representative markers and alteration of EMT-associated markers were found at the invasive fronts and in MPEs compared with the expression in primary pulmonary tumor tissues. The expression of OCT-4 in MPEs significantly related to distant metastasis and stage, as well as inversely correlated with patient survival. Primary cultures confirmed the CSC properties in MPE. Five of eight cases of MPE yielded adequate cell clusters, which also showed variable expressions of CSC markers in addition to sphere formation and the ability for differentiation and metastasis. CONCLUSION This pilot study offers a better understanding of the metastatic cascade. Establishing a model of MPE will provide further insight into the role of CSCs in metastasis and may explain the high therapeutic failure rates for patients with MPE.
Collapse
Affiliation(s)
- Su-Feng Chen
- Department of Dental Hygiene, China Medical University, Taichung, Taiwan
- Department of Pathology, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Yaoh-Shiang Lin
- Department of Otolaryngology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Shu-Wen Jao
- Division of Colon and Rectal Surgery, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Yun-Ching Chang
- Graduate Institute of Life Sciences, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
- Department of Pathology, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Chia-Lin Liu
- Graduate Institute of Life Sciences, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Yu-Ju Lin
- Department of Pathology, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
| | - Shin Nieh
- Department of Pathology, National Defense Medical Center and Tri-Service General Hospital, Taipei, Taiwan
- * E-mail:
| |
Collapse
|