1
|
Silva CAC, Fidelle M, Almonte AA, Derosa L, Zitvogel L. Gut Microbiota-Related Biomarkers in Immuno-Oncology. Annu Rev Pharmacol Toxicol 2025; 65:333-354. [PMID: 39259979 DOI: 10.1146/annurev-pharmtox-061124-102218] [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: 09/13/2024]
Abstract
Carcinogenesis is associated with the emergence of protracted intestinal dysbiosis and metabolic changes. Increasing evidence shows that gut microbiota-related biomarkers and microbiota-centered interventions are promising strategies to overcome resistance to immunotherapy. However, current standard methods for evaluating gut microbiota composition are cost- and time-consuming. The development of routine diagnostic tools for intestinal barrier alterations and dysbiosis constitutes a critical unmet medical need that can guide routine treatment and microbiota-centered intervention decisions in patients with cancer. In this review, we explore the influence of gut microbiota on cancer immunotherapy and highlight gut-associated biomarkers that have the potential to be transformed into simple diagnostic tools, thus guiding standard treatment decisions in the field of immuno-oncology. Mechanistic insights toward leveraging the complex relationship between cancer immunosurveillance, gut microbiota, and metabolism open exciting opportunities for developing novel biomarkers in immuno-oncology.
Collapse
Affiliation(s)
- Carolina Alves Costa Silva
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Marine Fidelle
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Andrew A Almonte
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Lisa Derosa
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| | - Laurence Zitvogel
- Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicêtre, France
- Clinicobiome, Gustave Roussy Cancer Campus (GRCC), and INSERM U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; ,
| |
Collapse
|
2
|
Tindle C, Fonseca AG, Taheri S, Katkar GD, Lee J, Maity P, Sayed IM, Ibeawuchi SR, Vidales E, Pranadinata RF, Fuller M, Stec DL, Anandachar MS, Perry K, Le HN, Ear J, Boland BS, Sandborn WJ, Sahoo D, Das S, Ghosh P. A living organoid biobank of patients with Crohn's disease reveals molecular subtypes for personalized therapeutics. Cell Rep Med 2024; 5:101748. [PMID: 39332415 PMCID: PMC11513829 DOI: 10.1016/j.xcrm.2024.101748] [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: 01/04/2024] [Revised: 07/15/2024] [Accepted: 08/31/2024] [Indexed: 09/29/2024]
Abstract
Crohn's disease (CD) is a complex and heterogeneous condition with no perfect preclinical model or cure. To address this, we explore adult stem cell-derived organoids that retain their tissue identity and disease-driving traits. We prospectively create a biobank of CD patient-derived organoid cultures (PDOs) from colonic biopsies of 53 subjects across all clinical subtypes and healthy subjects. Gene expression analyses enabled benchmarking of PDOs as tools for modeling the colonic epithelium in active disease and identified two major molecular subtypes: immune-deficient infectious CD (IDICD) and stress and senescence-induced fibrostenotic CD (S2FCD). Each subtype shows internal consistency in the transcriptome, genome, and phenome. The spectrum of morphometric, phenotypic, and functional changes within the "living biobank" reveals distinct differences between the molecular subtypes. Drug screens reverse subtype-specific phenotypes, suggesting phenotyped-genotyped CD PDOs can bridge basic biology and patient trials by enabling preclinical phase "0" human trials for personalized therapeutics.
Collapse
Affiliation(s)
- Courtney Tindle
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA
| | - Ayden G Fonseca
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA
| | - Sahar Taheri
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Gajanan D Katkar
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jasper Lee
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Priti Maity
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA
| | - Ibrahim M Sayed
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stella-Rita Ibeawuchi
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Eleadah Vidales
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA
| | - Rama F Pranadinata
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA
| | - Mackenzie Fuller
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA
| | - Dominik L Stec
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Kevin Perry
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA
| | - Helen N Le
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jason Ear
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Brigid S Boland
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - William J Sandborn
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Debashis Sahoo
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Soumita Das
- HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; HUMANOID™ Center of Research Excellence (CoRE), University of California, San Diego, La Jolla, CA 92093, USA; Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
3
|
Tocci S, Das S, Sayed IM. An Update on Blastocystis: Possible Mechanisms of Blastocystis-Mediated Colorectal Cancer. Microorganisms 2024; 12:1924. [PMID: 39338600 PMCID: PMC11433781 DOI: 10.3390/microorganisms12091924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Blastocystis is an anaerobic parasite that colonizes the intestinal tract of humans and animals. When it was first discovered, Blastocystis was considered to be a normal flora with beneficial effects on human health, such as maintaining gut hemostasis and improving intestinal barrier integrity. Later, with increasing research on Blastocystis, reports showed that Blastocystis sp. is associated with gastrointestinal disorders, colorectal cancer (CRC), and neurological disorders. The association between Blastocystis sp. and CRC has been confirmed in several countries. Blastocystis sp. can mediate CRC via similar mechanisms to CRC-associated bacteria, including infection-mediated inflammation, increased oxidative stress, induced gut dysbiosis, and damage to intestinal integrity, leading to a leaky gut. IL-8 is the main inflammatory cytokine released from epithelial cells and can promote CRC development. The causal association of Blastocystis sp. with other diseases needs further investigation. In this review, we have provided an update on Blastocystis sp. and summarized the debate about the beneficial and harmful effects of this parasite. We have also highlighted the possible mechanisms of Blastocystis-mediated CRC.
Collapse
Affiliation(s)
- Stefania Tocci
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Soumita Das
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Ibrahim M Sayed
- Department of Biomedical & Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| |
Collapse
|
4
|
Broering MF, Tocci S, Sout NT, Reutelingsperger C, Farsky SHP, Das S, Sayed IM. Development of an Inflamed High Throughput Stem-cell-based Gut Epithelium Model to Assess the Impact of Annexin A1. Stem Cell Rev Rep 2024; 20:1299-1310. [PMID: 38498294 DOI: 10.1007/s12015-024-10708-4] [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] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE AND DESIGN Annexin A1 (ANXA1) plays a role in maintaining intestinal hemostasis, especially following mucosal inflammation. The published data about ANXA1 was derived from experimental animal models where there is an overlapping between epithelial and immune cells. There is no in vitro gut epithelial model that can assess the direct effect of ANXA1 on the gut epithelium. METHODS We developed high-throughput stem-cell-based murine epithelial cells and bacterial lipopolysaccharides (LPS) were used to induce inflammation. The impact of ANXA1 and its functional part (Ac2-26) was evaluated in the inflamed model. Intestinal integrity was assessed by the transepithelial electrical resistance (TEER), and FITC-Dextran permeability. Epithelial junction proteins were assessed using confocal microscopy and RT-qPCR. Inflammatory cytokines were evaluated by RT-qPCR and ELISA. RESULTS LPS challenge mediated a damage in the epithelial cells as shown by a drop in the TEER and an increase in FITC-dextran permeability; reduced the expression of epithelial junctional proteins (Occludin, ZO-1, and Cadherin) and increased the expression of the gut leaky protein, Claudin - 2. ANXA1 and Ac2-26 treatment reduced the previous damaging effects. In addition, ANXA1 and Ac2-26 inhibited the inflammatory responses mediated by the LPS and increased the transcription of the anti-inflammatory cytokine, IL-10. CONCLUSION ANXA1 and Ac2-26 directly protect the epithelial integrity by affecting the expression of epithelial junction and inflammatory markers. The inflamed gut model is a reliable tool to study intestinal inflammatory diseases, and to evaluate the efficacy of potential anti-inflammatory drugs and the screening of new drugs that could be candidates for inflammatory bowel disease.
Collapse
Affiliation(s)
- Milena Fronza Broering
- Department of Biomedical and Nutritional Sciences, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, São Paulo, 05508-000, Brazil
| | - Stefania Tocci
- Department of Biomedical and Nutritional Sciences, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| | - Noah T Sout
- Department of Biomedical and Nutritional Sciences, University of Massachusetts-Lowell, Lowell, MA, 01854, USA
| | - Chris Reutelingsperger
- Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht University, Maastricht, 6211 LK, The Netherlands
| | - Sandra H P Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo, São Paulo, 05508-000, Brazil
| | - Soumita Das
- Department of Biomedical and Nutritional Sciences, University of Massachusetts-Lowell, Lowell, MA, 01854, USA.
| | - Ibrahim M Sayed
- Department of Biomedical and Nutritional Sciences, University of Massachusetts-Lowell, Lowell, MA, 01854, USA.
| |
Collapse
|
5
|
Sinha S, Alcantara J, Perry K, Castillo V, Espinoza CR, Taheri S, Vidales E, Tindle C, Adel A, Amirfakhri S, Sawires JR, Yang J, Bouvet M, Sahoo D, Ghosh P. Machine-Learning Identifies a Strategy for Differentiation Therapy in Solid Tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.13.557628. [PMID: 37745574 PMCID: PMC10515918 DOI: 10.1101/2023.09.13.557628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
BACKGROUND Although differentiation therapy can cure some hematologic malignancies, its curative potential remains unrealized in solid tumors. This is because conventional computational approaches succumb to the thunderous noise of inter-/intratumoral heterogeneity. Using colorectal cancers (CRCs) as an example, here we outline a machine learning(ML)-based approach to track, differentiate, and selectively target cancer stem cells (CSCs). METHODS A transcriptomic network was built and validated using healthy colon and CRC tissues in diverse gene expression datasets (~5,000 human and >300 mouse samples). Therapeutic targets and perturbation strategies were prioritized using ML, with the goal of reinstating the expression of a transcriptional identifier of the differentiated colonocyte, CDX2, whose loss in poorly differentiated (CSC-enriched) CRCs doubles the risk of relapse/death. The top candidate target was then engaged with a clinical-grade drug and tested on 3 models: CRC lines in vitro, xenografts in mice, and in a prospective cohort of healthy (n = 3) and CRC (n = 23) patient-derived organoids (PDOs). RESULTS The drug shifts the network predictably, induces CDX2 and crypt differentiation, and shows cytotoxicity in all 3 models, with a high degree of selectivity towards all CDX2-negative cell lines, xenotransplants, and PDOs. The potential for effective pairing of therapeutic efficacy (IC50) and biomarker (CDX2-low state) is confirmed in PDOs using multivariate analyses. A 50-gene signature of therapeutic response is derived and tested on 9 independent cohorts (~1700 CRCs), revealing the impact of CDX2-reinstatement therapy could translate into a ~50% reduction in the risk of mortality/recurrence. CONCLUSIONS Findings not only validate the precision of the ML approach in targeting CSCs, and objectively assess its impact on clinical outcome, but also exemplify the use of ML in yielding clinical directive information for enhancing personalized medicine.
Collapse
|
6
|
Sharma A, Achi SC, Ibeawuchi SR, Anandachar MS, Gementera H, Chaudhury U, Usmani F, Vega K, Sayed IM, Das S. The crosstalk between microbial sensors ELMO1 and NOD2 shape intestinal immune responses. Virulence 2023; 14:2171690. [PMID: 36694274 PMCID: PMC9980453 DOI: 10.1080/21505594.2023.2171690] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/26/2023] Open
Abstract
Microbial sensors play an essential role in maintaining cellular homoeostasis. Our knowledge is limited on how microbial sensing helps in differential immune response and its link to inflammatory diseases. Recently we have confirmed that ELMO1 (Engulfment and Cell Motility Protein-1) present in cytosol is involved in pathogen sensing, engulfment, and intestinal inflammation. Here, we show that ELMO1 interacts with another sensor, NOD2 (Nucleotide-binding oligomerization domain-containing protein 2), that recognizes bacterial cell wall component muramyl dipeptide (MDP). The polymorphism of NOD2 is linked to Crohn's disease (CD) pathogenesis. Interestingly, we found that overexpression of ELMO1 and mutant NOD2 (L1007fs) were not able to clear the CD-associated adherent invasive E. coli (AIEC-LF82). The functional implications of ELMO1-NOD2 interaction in epithelial cells were evaluated by using enteroid-derived monolayers (EDMs) from ELMO1 and NOD2 KO mice. Subsequently we also assessed the immune response in J774 macrophages depleted of either ELMO1 or NOD2 or both. The infection of murine EDMs with AIEC-LF82 showed higher bacterial load in ELMO1-KO, NOD2 KO EDMs, and ELMO1 KO EDMs treated with NOD2 inhibitors. The murine macrophage cells showed that the downregulation of ELMO1 and NOD2 is associated with impaired bacterial clearance that is linked to reduce pro-inflammatory cytokines and reactive oxygen species. Our results indicated that the crosstalk between microbial sensors in enteric infection and inflammatory diseases impacts the fate of the bacterial load and disease pathogenesis.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Pathology, University of California San Diego; San Diego, California, USA
| | | | - Stella-Rita Ibeawuchi
- Department of Pathology, University of California San Diego; San Diego, California, USA
| | | | - Hobie Gementera
- Department of Pathology, University of California San Diego; San Diego, California, USA
| | - Uddeep Chaudhury
- Department of Pathology, University of California San Diego; San Diego, California, USA
| | - Fatima Usmani
- Department of Pathology, University of California San Diego; San Diego, California, USA
| | - Kevin Vega
- Department of Pathology, University of California San Diego; San Diego, California, USA
| | - Ibrahim M Sayed
- Department of Pathology, University of California San Diego; San Diego, California, USA
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Soumita Das
- Department of Pathology, University of California San Diego; San Diego, California, USA
- Department of Biomedical and Nutritional Science, University of Massachusetts-Lowell, Lowell, USA
| |
Collapse
|
7
|
Shemtov SJ, Emani R, Bielska O, Covarrubias AJ, Verdin E, Andersen JK, Winer DA. The intestinal immune system and gut barrier function in obesity and ageing. FEBS J 2023; 290:4163-4186. [PMID: 35727858 PMCID: PMC9768107 DOI: 10.1111/febs.16558] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/29/2022] [Accepted: 06/20/2022] [Indexed: 08/13/2023]
Abstract
Obesity and ageing predispose to numerous, yet overlapping chronic diseases. For example, metabolic abnormalities, including insulin resistance (IR) and type 2 diabetes (T2D) are important causes of morbidity and mortality. Low-grade chronic inflammation of tissues, such as the liver, visceral adipose tissue and neurological tissues, is considered a significant contributor to these chronic diseases. Thus, it is becoming increasingly important to understand what drives this inflammation in affected tissues. Recent evidence, especially in the context of obesity, suggests that the intestine plays an important role as the gatekeeper of inflammatory stimuli that ultimately fuels low-grade chronic tissue inflammation. In addition to metabolic diseases, abnormalities in the intestinal mucosal barrier have been linked to a range of other chronic inflammatory conditions, such as neurodegeneration and ageing. The flow of inflammatory stimuli from the gut is in part controlled by local immunological inputs impacting the intestinal barrier. Here, we will review the impact of obesity and ageing on the intestinal immune system and its downstream consequences on gut barrier function, which is strongly implicated in the pathogenesis of obesity and age-related diseases. In particular, we will discuss the effects of age-related intestinal dysfunction on neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarah J. Shemtov
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Rohini Emani
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Olga Bielska
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Anthony J. Covarrubias
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095 USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095 USA
| | - Eric Verdin
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Julie K. Andersen
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
| | - Daniel A. Winer
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California, USA
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA
- Division of Cellular & Molecular Biology, Diabetes Research Group, Toronto General Research Institute (TGRI), University Health Network, 101 College Street, Toronto, ON, M5G 1L7, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King’s College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Immunology, University of Toronto, 1 King’s College Circle, Toronto, ON, M5S 1A8, Canada
| |
Collapse
|
8
|
Jain S, Marotta F, Haghshenas L, Yadav H. Treating Leaky Syndrome in the Over 65s: Progress and Challenges. Clin Interv Aging 2023; 18:1447-1451. [PMID: 37671072 PMCID: PMC10476862 DOI: 10.2147/cia.s409801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 08/03/2023] [Indexed: 09/07/2023] Open
Abstract
As we age, our organ functions gradually decline. Circulating factors in the blood and the integrity of organ barriers can become dysfunctional, resulting in a condition known as leaky syndrome. This condition involves the unregulated exchange or leakage of components between organs. However, the triggers of leaky syndrome, as well as its role in aging-related disorders and illnesses, remain largely unknown. In this editorial, we discuss potential mechanisms that originate from the gut and resident microbes (microbiome) to contribute in leaky syndrome. Furthermore, we explore how the food we consume can impact the development of leaky syndrome, potentially influencing the biology of aging and challenges to diagnose the leaky gut condition accurately and clinically.
Collapse
Affiliation(s)
- Shalini Jain
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
| | | | - Leila Haghshenas
- Department of Clinical Bioinformatics, Harvard Medical School, Boston, MA, USA
| | - Hariom Yadav
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, USA
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida, Tampa, FL, USA
- Department of Internal Medicine- Digestive Diseases and Nutrition, University of South Florida, Tampa, FL, USA
| |
Collapse
|
9
|
Tindle C, Katkar GD, Fonseca AG, Taheri S, Lee J, Maity P, Sayed IM, Ibeawuchi SR, Vidales E, Pranadinata RF, Fuller M, Stec DL, Anandachar MS, Perry K, Le HN, Ear J, Boland BS, Sandborn WJ, Sahoo D, Das S, Ghosh P. A Living Organoid Biobank of Crohn's Disease Patients Reveals Molecular Subtypes for Personalized Therapeutics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.11.532245. [PMID: 36993763 PMCID: PMC10054961 DOI: 10.1101/2023.03.11.532245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Crohn's disease (CD) is a complex, clinically heterogeneous disease of multifactorial origin; there is no perfect pre-clinical model, little insight into the basis for such heterogeneity, and still no cure. To address these unmet needs, we sought to explore the translational potential of adult stem cell-derived organoids that not only retain their tissue identity, but also their genetic and epigenetic disease-driving traits. We prospectively created a biobank of CD patient-derived organoid cultures (PDOs) using biopsied tissues from colons of 34 consecutive subjects representing all clinical subtypes (Montreal Classification B1-B3 and perianal disease). PDOs were generated also from healthy subjects. Comparative gene expression analyses enabled benchmarking of PDOs as tools for modeling the colonic epithelium in active disease and revealed that despite the clinical heterogeneity there are two major molecular subtypes: immune-deficient infectious-CD [IDICD] and stress and senescence-induced fibrostenotic-CD [S2FCD]. The transcriptome, genome and phenome show a surprising degree of internal consistency within each molecular subtype. The spectrum of morphometric, phenotypic, and functional changes within the "living biobank" reveals distinct differences between the molecular subtypes. These insights enabled drug screens that reversed subtype-specific phenotypes, e.g., impaired microbial clearance in IDICD was reversed using agonists for nuclear receptors, and senescence in S2FCD was rectified using senotherapeutics, but not vice versa . Phenotyped-genotyped CD-PDOs may fill the gap between basic biology and patient trials by enabling pre-clinical Phase '0' human trials for personalized therapeutics. GRAPHIC ABSTRACT In Brief This work creates a prospectively biobanked phenotyped-genotyped Crohn's disease patient-derived organoids (CD-PDOs) as platforms for molecular subtyping of disease and for ushering personalized therapeutics. HIGHLIGHTS Prospectively biobanked CD-organoids recapitulate the disease epithelium in patientsThe phenome-transcriptome-genome of CD-organoids converge on two molecular subtypesOne subtype shows impaired microbial clearance, another increased cellular senescencePhenotyped-genotyped PDOs are then used for integrative and personalized therapeutics.
Collapse
|
10
|
Noble A, Pring ET, Durant L, Man R, Dilke SM, Hoyles L, James SA, Carding SR, Jenkins JT, Knight SC. Altered immunity to microbiota, B cell activation and depleted γδ/resident memory T cells in colorectal cancer. Cancer Immunol Immunother 2022. [PMID: 35316367 DOI: 10.1007/s00262-021-03135-8/figures/5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
The role of microbiota:immune system dysregulation in the etiology of colorectal cancer (CRC) is poorly understood. CRC develops in gut epithelium, accompanied by low level inflammatory signaling, intestinal microbial dysbiosis and immune dysfunction. We examined populations of intraepithelial lymphocytes in non-affected colonic mucosa of CRC and healthy donors and circulating immune memory to commensal bacterial species and yeasts. γδ T cells and resident memory T cells, populations with a regulatory CD39-expressing phenotype, were found at lower frequencies in the colonic tissue of CRC donors compared to healthy controls. Patterns of T cell proliferative responses to a panel of commensal bacteria were distinct in CRC, while B cell memory responses to several bacteria/yeast were significantly increased, accompanied by increased proportions of effector memory B cells, transitional B cells and plasmablasts in blood. IgA responses to mucosal microbes were unchanged. Our data describe a novel immune signature with similarities to and differences from that of inflammatory bowel disease. They implicate B cell dysregulation as a potential contributor to parainflammation and identify pathways of weakened barrier function and tumor surveillance in CRC-susceptible individuals.
Collapse
Affiliation(s)
- Alistair Noble
- Gut Microbes and Health Program, Quadram Institute Bioscience, Norwich, UK
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| | - Edward T Pring
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK
| | - Lydia Durant
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| | - Ripple Man
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK
| | - Stella M Dilke
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK
| | - Lesley Hoyles
- Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Steve A James
- Gut Microbes and Health Program, Quadram Institute Bioscience, Norwich, UK
| | - Simon R Carding
- Gut Microbes and Health Program, Quadram Institute Bioscience, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - John T Jenkins
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK
| | - Stella C Knight
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK.
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK.
| |
Collapse
|
11
|
Noble A, Pring ET, Durant L, Man R, Dilke SM, Hoyles L, James SA, Carding SR, Jenkins JT, Knight SC. Altered immunity to microbiota, B cell activation and depleted γδ/resident memory T cells in colorectal cancer. Cancer Immunol Immunother 2022; 71:2619-2629. [PMID: 35316367 PMCID: PMC9519644 DOI: 10.1007/s00262-021-03135-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022]
Abstract
The role of microbiota:immune system dysregulation in the etiology of colorectal cancer (CRC) is poorly understood. CRC develops in gut epithelium, accompanied by low level inflammatory signaling, intestinal microbial dysbiosis and immune dysfunction. We examined populations of intraepithelial lymphocytes in non-affected colonic mucosa of CRC and healthy donors and circulating immune memory to commensal bacterial species and yeasts. γδ T cells and resident memory T cells, populations with a regulatory CD39-expressing phenotype, were found at lower frequencies in the colonic tissue of CRC donors compared to healthy controls. Patterns of T cell proliferative responses to a panel of commensal bacteria were distinct in CRC, while B cell memory responses to several bacteria/yeast were significantly increased, accompanied by increased proportions of effector memory B cells, transitional B cells and plasmablasts in blood. IgA responses to mucosal microbes were unchanged. Our data describe a novel immune signature with similarities to and differences from that of inflammatory bowel disease. They implicate B cell dysregulation as a potential contributor to parainflammation and identify pathways of weakened barrier function and tumor surveillance in CRC-susceptible individuals.
Collapse
Affiliation(s)
- Alistair Noble
- Gut Microbes and Health Program, Quadram Institute Bioscience, Norwich, UK
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| | - Edward T Pring
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK
| | - Lydia Durant
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
| | - Ripple Man
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK
| | - Stella M Dilke
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK
| | - Lesley Hoyles
- Department of Biosciences, Nottingham Trent University, Nottingham, UK
| | - Steve A James
- Gut Microbes and Health Program, Quadram Institute Bioscience, Norwich, UK
| | - Simon R Carding
- Gut Microbes and Health Program, Quadram Institute Bioscience, Norwich, UK
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - John T Jenkins
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK
| | - Stella C Knight
- Antigen Presentation Research Group, Imperial College London, Northwick Park and St. Mark's Campus, Harrow, UK.
- St. Mark's Hospital, London North West University Healthcare NHS Trust, Harrow, UK.
| |
Collapse
|
12
|
Sharma A, Achi SC, Ibeawuchi S, Anandachar MS, Gementera H, Chaudhury U, Usmani F, Vega K, Sayed IM, Das S. The crosstalk between microbial sensors ELMO1 and NOD2 shape intestinal immune responses.. [DOI: 10.1101/2022.07.09.499433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2023]
Abstract
ABSTRACTMicrobial sensors play an essential role in maintaining cellular homeostasis. Our knowledge is limited on how microbial sensing helps in differential immune response and its link to inflammatory diseases. Recently, we have shown that cytosolic sensor ELMO1 (Engulfment and Cell Motility Protein-1) binds to effectors from pathogenic bacteria and controls intestinal inflammation. Here, we show that ELMO1 interacts with another sensor, NOD2 (Nucleotide-binding oligomerization domain-containing protein 2), that recognizes bacterial cell wall component muramyl dipeptide (MDP). The polymorphism of NOD2 is linked to Crohn’s disease (CD) pathogenesis. Interestingly, we found that overexpression of ELMO1 and mutant NOD2 (L1007fs) were not able to clear the CD-associated adherent invasive E. coli (AIEC-LF82). To understand the interplay of microbial sensing of ELMO1-NOD2 in epithelial cells and macrophages, we used enteroid-derived monolayers (EDMs) from ELMO1 and NOD2 KO mice and ELMO1 and NOD2-depleted murine macrophage cell lines. The infection of murine EDMs with AIEC-LF82 showed higher bacterial load in ELMO1-KO, NOD2 KO EDMs, and ELMO1 KO EDMs treated with NOD2 inhibitors. The murine macrophage cells showed that the downregulation of ELMO1 and NOD2 is associated with impaired bacterial clearance that is linked to reduced pro-inflammatory cytokines and reactive oxygen species. Our results indicated that the crosstalk between microbial sensors in enteric infection and inflammatory diseases impacts the fate of the bacterial load and disease pathogenesis.
Collapse
|
13
|
Olivier S, Diounou H, Foretz M, Guilmeau S, Daniel N, Marette A, Rolli-Derkinderen M, Viollet B. [AMPK activity is a gatekeeper of the intestinal epithelial barrier]. Med Sci (Paris) 2022; 38:136-138. [PMID: 35179465 DOI: 10.1051/medsci/2021251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Séverine Olivier
- Université de Paris, Institut Cochin, Inserm, CNRS, 24 rue du faubourg Saint-Jacques, 75014 Paris, France
| | - Hanna Diounou
- Université de Paris, Institut Cochin, Inserm, CNRS, 24 rue du faubourg Saint-Jacques, 75014 Paris, France
| | - Marc Foretz
- Université de Paris, Institut Cochin, Inserm, CNRS, 24 rue du faubourg Saint-Jacques, 75014 Paris, France
| | - Sandra Guilmeau
- Université de Paris, Institut Cochin, Inserm, CNRS, 24 rue du faubourg Saint-Jacques, 75014 Paris, France
| | - Noëmie Daniel
- Université de Paris, Institut Cochin, Inserm, CNRS, 24 rue du faubourg Saint-Jacques, 75014 Paris, France
| | - André Marette
- Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ) et Institut sur la nutrition et les aliments fonctionnels (INAF)
| | - Malvyne Rolli-Derkinderen
- Université de Nantes, Unité de recherche TENS (Le système nerveux entérique dans les maladies de l'intestin et du cerveau), Inserm, 44093 Nantes, France
| | - Benoit Viollet
- Université de Paris, Institut Cochin, Inserm, CNRS, 24 rue du faubourg Saint-Jacques, 75014 Paris, France
| |
Collapse
|
14
|
Ren B, Wang L, Mulati A, Liu Y, Liu Z, Liu X. Methionine Restriction Improves Gut Barrier Function by Reshaping Diurnal Rhythms of Inflammation-Related Microbes in Aged Mice. Front Nutr 2022; 8:746592. [PMID: 35004799 PMCID: PMC8733897 DOI: 10.3389/fnut.2021.746592] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 12/03/2021] [Indexed: 12/25/2022] Open
Abstract
Age-related gut barrier dysfunction and dysbiosis of the gut microbiome play crucial roles in human aging. Dietary methionine restriction (MR) has been reported to extend lifespan and reduce the inflammatory response; however, its protective effects on age-related gut barrier dysfunction remain unclear. Accordingly, we focus on the effects of MR on inflammation and gut function. We found a 3-month methionine-restriction reduced inflammatory factors in the serum of aged mice. Moreover, MR reduced gut permeability in aged mice and increased the levels of the tight junction proteins mRNAs, including those of occludin, claudin-1, and zona occludens-1. MR significantly reduced bacterial endotoxin lipopolysaccharide concentration in aged mice serum. By using 16s rRNA sequencing to analyze microbiome diurnal rhythmicity during 24 h, we found MR moderately recovered the cyclical fluctuations of the gut microbiome which was disrupted in aged mice, leading to time-specific enhancement of the abundance of short-chain fatty acid-producing and lifespan-promoting microbes. Moreover, MR dampened the oscillation of inflammation-related TM7-3 and Staphylococcaceae. In conclusion, the effects of MR on the gut barrier were likely related to alleviation of the oscillations of inflammation-related microbes. MR can enable nutritional intervention against age-related gut barrier dysfunction.
Collapse
Affiliation(s)
- Bo Ren
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China.,School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Luanfeng Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Aiziguli Mulati
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yan Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| |
Collapse
|
15
|
Mohammed HO, Ahmed Alaa El-Din E, Farag AI. Impact of e-cigarettes on colonic mucosa and the role of recovery: involvement of oxidative and inflammatory pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:64561-64571. [PMID: 34312757 PMCID: PMC8313116 DOI: 10.1007/s11356-021-15575-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/18/2021] [Indexed: 04/16/2023]
Abstract
Electronic cigarettes (e-cigarettes) (EC) are often advertised as a safer alternative to conventional cigarettes. Its widespread use has led to increased interest in its adverse health effects, thanks to few restrictions and a lack of regulatory guidelines. The study aimed to evaluate the influence of exposure to e-cigarette aerosol inhalation in rat colon model and conduct a follow-up after cessation of exposure. The experiment included 30 male adult Albino rats. The animals were divided into three groups: group I (control), non-exposed animals; group II (exposed), was exposed to electronic cigarette liquid vapor for four consecutive weeks; and group III (recovery), was followed up for another 4 weeks after exposure to an e-cigarette as exposed group and for the same duration. In the exposed group, malondialdehyde (MDA) and total nitric oxide (NO) increased significantly in colonic tissue, while superoxide dismutase (SOD) decreased. On histological examination, colonic mucosa showed distortion and loss of its epithelial lining with heavy inflammatory cell infiltration. Also, there was a significant decrease in periodic acid-Schiff-positive goblet cells and area percent of proliferating cell nuclear antigen expression. Tumor necrosis factor-alpha (TNFα) expression significantly increased in colonic mucosa. After 4 weeks of EC cessation, the colonic mucosal histological structure showed recovery with downregulated TNFα immunoexpression and restored oxidant/antioxidant balance. In conclusion, the usage of electronic cigarettes resulted in marked pathological alterations in the colonic mucosa, which could be attributed to oxidative and inflammatory stresses. In contrast, the cessation of exposure led to recovery.
Collapse
Affiliation(s)
- Heba O. Mohammed
- Department of Human Anatomy & Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Eman Ahmed Alaa El-Din
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Zagazig University, Zagazig, 44519 Egypt
| | - Azza I. Farag
- Department of Human Anatomy & Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
16
|
Vo DT, Fuller MR, Tindle C, Anandachar MS, Das S, Sahoo D, Ghosh P. SPT6 loss permits the transdifferentiation of keratinocytes into an intestinal fate that resembles Barrett's metaplasia. iScience 2021; 24:103121. [PMID: 34622168 PMCID: PMC8481972 DOI: 10.1016/j.isci.2021.103121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/13/2021] [Accepted: 09/09/2021] [Indexed: 12/15/2022] Open
Abstract
Transient depletion of the transcription elongation factor SPT6 in the keratinocyte has been recently shown to inhibit epidermal differentiation and stratification; instead, they transdifferentiate into a gut-like lineage. We show here that this phenomenon of transdifferentiation recapitulates Barrett's metaplasia, the only human pathophysiologic condition in which a stratified squamous epithelium that is injured due to chronic acid reflux is trans-committed into an intestinal fate. The evidence we present here not only lend support to the notion that the keratinocytes are potentially the cell of origin of Barrett's metaplasia but also provide mechanistic insights linking transient acid exposure, downregulation of SPT6, stalled transcription of the master regulator of epidermal fate TP63, loss of epidermal fate, and metaplastic progression. Because Barrett's metaplasia in the esophagus is a pre-neoplastic condition with no preclinical human models, these findings have a profound impact on the modeling Barrett's metaplasia-in-a-dish.
Collapse
Affiliation(s)
- Daniella T. Vo
- Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, MC 0703, Leichtag Building 132, La Jolla, CA 92093-0703, USA
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, La Jolla, USA
| | - MacKenzie R. Fuller
- Departments of Medicine and Cell and Molecular Medicine, University of California San Diego, 9500 Gilman Drive (MC 0651), George E. Palade Bldg, Rm 232, La Jolla, CA 92093, USA
- HUMANOID Center of Research Excellence (CoRE), University of California San Diego, La Jolla, USA
| | - Courtney Tindle
- Departments of Medicine and Cell and Molecular Medicine, University of California San Diego, 9500 Gilman Drive (MC 0651), George E. Palade Bldg, Rm 232, La Jolla, CA 92093, USA
- HUMANOID Center of Research Excellence (CoRE), University of California San Diego, La Jolla, USA
| | - Mahitha Shree Anandachar
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, George E. Palade Bldg, Rm 256, La Jolla, CA 92093, USA
| | - Soumita Das
- HUMANOID Center of Research Excellence (CoRE), University of California San Diego, La Jolla, USA
- Department of Pathology, University of California San Diego, 9500 Gilman Drive, George E. Palade Bldg, Rm 256, La Jolla, CA 92093, USA
- Moore Comprehensive Cancer Center, University of California San Diego, La Jolla, USA
| | - Debashis Sahoo
- Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, MC 0703, Leichtag Building 132, La Jolla, CA 92093-0703, USA
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, La Jolla, USA
- Moore Comprehensive Cancer Center, University of California San Diego, La Jolla, USA
| | - Pradipta Ghosh
- Departments of Medicine and Cell and Molecular Medicine, University of California San Diego, 9500 Gilman Drive (MC 0651), George E. Palade Bldg, Rm 232, La Jolla, CA 92093, USA
- HUMANOID Center of Research Excellence (CoRE), University of California San Diego, La Jolla, USA
- Moore Comprehensive Cancer Center, University of California San Diego, La Jolla, USA
- Department of Medicine, University of California San Diego, La Jolla, USA
| |
Collapse
|
17
|
Sayed IM, Tindle C, Fonseca AG, Ghosh P, Das S. Functional assays with human patient-derived enteroid monolayers to assess the human gut barrier. STAR Protoc 2021; 2:100680. [PMID: 34337445 PMCID: PMC8313751 DOI: 10.1016/j.xpro.2021.100680] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Here, we describe the use of polarized patient enteroid-derived monolayers (EDMs) to assess the impact of e-cigarettes on the human gut barrier. These EDMs can be adapted to culture in a 96-well plate for high-throughput screening. We model the effect of e-cigarettes by combining pathogens, enteroids, and e-cigarette vapor-infused media and assess gut barrier integrity, bacterial internalization, and inflammatory response of the gut epithelium. This protocol can be used to assess the effects of e-cigarette components on gut functions. For complete details on the use and execution of this protocol, please refer to Sharma et al. (2021).
Collapse
Affiliation(s)
- Ibrahim M. Sayed
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | - Courtney Tindle
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
- HUMANOID CoRE, University of California, San Diego, CA 92093, USA
| | - Ayden G. Fonseca
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
- HUMANOID CoRE, University of California, San Diego, CA 92093, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
- HUMANOID CoRE, University of California, San Diego, CA 92093, USA
- Department of Medicine, University of California, San Diego, CA 92093, USA
- Moore’s Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
- Veterans Affairs Medical Center, VA San Diego Healthcare System, La Jolla, San Diego, CA 92093, USA
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA 92093, USA
- HUMANOID CoRE, University of California, San Diego, CA 92093, USA
- Moore’s Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
- Veterans Affairs Medical Center, VA San Diego Healthcare System, La Jolla, San Diego, CA 92093, USA
| |
Collapse
|
18
|
Tindle C, Fuller M, Fonseca A, Taheri S, Ibeawuchi SR, Beutler N, Katkar GD, Claire A, Castillo V, Hernandez M, Russo H, Duran J, Crotty Alexander LE, Tipps A, Lin G, Thistlethwaite PA, Chattopadhyay R, Rogers TF, Sahoo D, Ghosh P, Das S. Adult stem cell-derived complete lung organoid models emulate lung disease in COVID-19. eLife 2021; 10:e66417. [PMID: 34463615 PMCID: PMC8463074 DOI: 10.7554/elife.66417] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
Background SARS-CoV-2, the virus responsible for COVID-19, causes widespread damage in the lungs in the setting of an overzealous immune response whose origin remains unclear. Methods We present a scalable, propagable, personalized, cost-effective adult stem cell-derived human lung organoid model that is complete with both proximal and distal airway epithelia. Monolayers derived from adult lung organoids (ALOs), primary airway cells, or hiPSC-derived alveolar type II (AT2) pneumocytes were infected with SARS-CoV-2 to create in vitro lung models of COVID-19. Results Infected ALO monolayers best recapitulated the transcriptomic signatures in diverse cohorts of COVID-19 patient-derived respiratory samples. The airway (proximal) cells were critical for sustained viral infection, whereas distal alveolar differentiation (AT2→AT1) was critical for mounting the overzealous host immune response in fatal disease; ALO monolayers with well-mixed proximodistal airway components recapitulated both. Conclusions Findings validate a human lung model of COVID-19, which can be immediately utilized to investigate COVID-19 pathogenesis and vet new therapies and vaccines. Funding This work was supported by the National Institutes for Health (NIH) grants 1R01DK107585-01A1, 3R01DK107585-05S1 (to SD); R01-AI141630, CA100768 and CA160911 (to PG) and R01-AI 155696 (to PG, DS and SD); R00-CA151673 and R01-GM138385 (to DS), R01- HL32225 (to PT), UCOP-R00RG2642 (to SD and PG), UCOP-R01RG3780 (to P.G. and D.S) and a pilot award from the Sanford Stem Cell Clinical Center at UC San Diego Health (P.G, S.D, D.S). GDK was supported through The American Association of Immunologists Intersect Fellowship Program for Computational Scientists and Immunologists. L.C.A's salary was supported in part by the VA San Diego Healthcare System. This manuscript includes data generated at the UC San Diego Institute of Genomic Medicine (IGC) using an Illumina NovaSeq 6000 that was purchased with funding from a National Institutes of Health SIG grant (#S10 OD026929).
Collapse
Affiliation(s)
- Courtney Tindle
- Department of Cellular and Molecular Medicine, University of California San DiegoSan DiegoUnited States
- HUMANOID CoRE, University of California San DiegoSan DiegoUnited States
| | - MacKenzie Fuller
- Department of Cellular and Molecular Medicine, University of California San DiegoSan DiegoUnited States
- HUMANOID CoRE, University of California San DiegoSan DiegoUnited States
| | - Ayden Fonseca
- Department of Cellular and Molecular Medicine, University of California San DiegoSan DiegoUnited States
- HUMANOID CoRE, University of California San DiegoSan DiegoUnited States
| | - Sahar Taheri
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San DiegoSan DiegoUnited States
| | | | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research InstituteLa JollaUnited States
| | - Gajanan Dattatray Katkar
- Department of Cellular and Molecular Medicine, University of California San DiegoSan DiegoUnited States
| | - Amanraj Claire
- Department of Cellular and Molecular Medicine, University of California San DiegoSan DiegoUnited States
- HUMANOID CoRE, University of California San DiegoSan DiegoUnited States
| | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, University of California San DiegoSan DiegoUnited States
| | - Moises Hernandez
- Division of Cardiothoracic Surgery, University of California San DiegoSan DiegoUnited States
| | - Hana Russo
- Department of Pathology, University of California San DiegoSan DiegoUnited States
| | - Jason Duran
- Division of Cardiology, Department of Internal Medicine, UC San Diego Medical CenterSan DiegoUnited States
| | - Laura E Crotty Alexander
- Pulmonary Critical Care Section, Veterans Affairs (VA) San Diego Healthcare SystemLa JollaUnited States
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San DiegoLa Jolla, CAUnited States
| | - Ann Tipps
- Department of Pathology, University of California San DiegoSan DiegoUnited States
| | - Grace Lin
- Department of Pathology, University of California San DiegoSan DiegoUnited States
| | | | - Ranajoy Chattopadhyay
- Department of Cellular and Molecular Medicine, University of California San DiegoSan DiegoUnited States
- HUMANOID CoRE, University of California San DiegoSan DiegoUnited States
- Cell Applications Inc.La Jolla, CAUnited States
| | - Thomas F Rogers
- Department of Immunology and Microbiology, The Scripps Research InstituteLa JollaUnited States
- Division of Infectious Diseases, Department of Medicine, University of California, San DiegoLa JollaUnited States
- Department of Immunology and Microbiology, The Scripps Research InstituteLa JollaUnited States
| | - Debashis Sahoo
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San DiegoSan DiegoUnited States
- Department of Pediatrics, University of California, San DiegoLa Jolla, CAUnited States
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San DiegoSan DiegoUnited States
- HUMANOID CoRE, University of California San DiegoSan DiegoUnited States
- Department of Medicine, University of California, San DiegoLa Jolla, CAUnited States
| | - Soumita Das
- HUMANOID CoRE, University of California San DiegoSan DiegoUnited States
- Department of Pathology, University of California San DiegoSan DiegoUnited States
| |
Collapse
|
19
|
Artificial intelligence guided discovery of a barrier-protective therapy in inflammatory bowel disease. Nat Commun 2021; 12:4246. [PMID: 34253728 PMCID: PMC8275683 DOI: 10.1038/s41467-021-24470-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
Modeling human diseases as networks simplify complex multi-cellular processes, helps understand patterns in noisy data that humans cannot find, and thereby improves precision in prediction. Using Inflammatory Bowel Disease (IBD) as an example, here we outline an unbiased AI-assisted approach for target identification and validation. A network was built in which clusters of genes are connected by directed edges that highlight asymmetric Boolean relationships. Using machine-learning, a path of continuum states was pinpointed, which most effectively predicted disease outcome. This path was enriched in gene-clusters that maintain the integrity of the gut epithelial barrier. We exploit this insight to prioritize one target, choose appropriate pre-clinical murine models for target validation and design patient-derived organoid models. Potential for treatment efficacy is confirmed in patient-derived organoids using multivariate analyses. This AI-assisted approach identifies a first-in-class gut barrier-protective agent in IBD and predicted Phase-III success of candidate agents. Traditional drug discovery process use differential, Bayesian and other network based approaches. We developed a Boolean approach for building disease maps and prioritizing pre-clinical models to discover a first-in-class therapy to restore and protect the leaky gut barrier in inflammatory bowel disease.
Collapse
|
20
|
Tindle C, Fuller M, Fonseca A, Taheri S, Ibeawuchi SR, Beutler N, Katkar G, Claire A, Castillo V, Hernandez M, Russo H, Duran J, Crotty Alexander LE, Tipps A, Lin G, Thistlethwaite PA, Chattopadhyay R, Rogers TF, Sahoo D, Ghosh P, Das S. Adult Stem Cell-derived Complete Lung Organoid Models Emulate Lung Disease in COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2020.10.17.344002. [PMID: 33106807 PMCID: PMC7587781 DOI: 10.1101/2020.10.17.344002] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
SARS-CoV-2, the virus responsible for COVID-19, causes widespread damage in the lungs in the setting of an overzealous immune response whose origin remains unclear. We present a scalable, propagable, personalized, cost-effective adult stem cell-derived human lung organoid model that is complete with both proximal and distal airway epithelia. Monolayers derived from adult lung organoids (ALOs), primary airway cells, or hiPSC-derived alveolar type-II (AT2) pneumocytes were infected with SARS-CoV-2 to create in vitro lung models of COVID-19. Infected ALO-monolayers best recapitulated the transcriptomic signatures in diverse cohorts of COVID-19 patient-derived respiratory samples. The airway (proximal) cells were critical for sustained viral infection whereas distal alveolar differentiation (AT2→AT1) was critical for mounting the overzealous host immune response in fatal disease; ALO monolayers with well-mixed proximodistal airway components recapitulated both. Findings validate a human lung model of COVID-19 which can be immediately utilized to investigate COVID-19 pathogenesis, and vet new therapies and vaccines.
Collapse
|
21
|
Sayed IM, El-Hafeez AAA, Maity PP, Das S, Ghosh P. Modeling colorectal cancers using multidimensional organoids. Adv Cancer Res 2021; 151:345-383. [PMID: 34148617 PMCID: PMC8221168 DOI: 10.1016/bs.acr.2021.02.005] [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] [Indexed: 10/21/2022]
Abstract
Organoids have revolutionized cancer research as highly adaptable models that enable an array of experimental techniques to interrogate tissue morphology and function. Because they preserve the genetic, phenotypic, and behavioral traits of their source tissue, organoids have gained traction as the most relevant models for drug discovery, tracking therapeutic response and for personalized medicine. As organoids are indisputably becoming a mainstay of cancer research, this review specifically addresses how colon-derived organoids can be perfected as multidimensional, scalable, reproducible models of healthy, pre-neoplastic and neoplastic conditions of the colon and for use in high-throughput "Phase-0" human clinical trials-in-a-dish.
Collapse
Affiliation(s)
- Ibrahim M Sayed
- Department of Pathology, University of California, San Diego, CA, United States
| | - Amer Ali Abd El-Hafeez
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States
| | - Priti P Maity
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA, United States; Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA, United States; HUMANOID Center of Research Excellence (CoRE), University of California, San Diego, CA, United States.
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA, United States; Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA, United States; Department of Medicine, University of California, San Diego, CA, United States; Veterans Affairs Medical Center, San Diego, CA, United States; HUMANOID Center of Research Excellence (CoRE), University of California, San Diego, CA, United States.
| |
Collapse
|
22
|
Sharma A, Lee J, Fonseca AG, Moshensky A, Kothari T, Sayed IM, Ibeawuchi SR, Pranadinata RF, Ear J, Sahoo D, Crotty-Alexander LE, Ghosh P, Das S. E-cigarettes compromise the gut barrier and trigger inflammation. iScience 2021. [PMID: 33537654 DOI: 10.1101/2020.07.29.227348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
E-cigarette usage continues to rise, yet the safety of e-cigarette aerosols is questioned. Using murine models of acute and chronic e-cigarette aerosol inhalation, murine colon transcriptomics, and murine and human gut-derived organoids in co-culture models, we assessed the effects of e-cigarette use on the gut barrier. Histologic and transcriptome analyses revealed that chronic, but not acute, nicotine-free e-cigarette use increased inflammation and reduced expression of tight junction (TJ) markers. Exposure of murine and human enteroid-derived monolayers (EDMs) to nicotine-free e-cigarette aerosols alone or in co-culture with bacteria also causes barrier disruption, downregulation of TJ protein, and enhanced inflammation in response to infection. These data highlight the harmful effects of "non-nicotine" component of e-cigarettes on the gut barrier. Considering the importance of an intact gut barrier for host fitness and the impact of gut mucosal inflammation on a multitude of chronic diseases, these findings are broadly relevant to both medicine and public health.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | - Jasper Lee
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | - Ayden G Fonseca
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
| | - Alex Moshensky
- Department of Medicine, University of California, San Diego, CA 92093, USA
| | - Taha Kothari
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | - Ibrahim M Sayed
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | | | - Rama F Pranadinata
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
| | - Jason Ear
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
- Department of Medicine, University of California, San Diego, CA 92093, USA
| | - Debashis Sahoo
- Department of Pediatrics, University of California, San Diego, CA 92093, USA
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California, San Diego, CA 92093, USA
- Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
| | - Laura E Crotty-Alexander
- Department of Medicine, University of California, San Diego, CA 92093, USA
- Veterans Affairs Medical Center, VA San Diego Healthcare System, La Jolla, San Diego, CA 92093, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
- Department of Medicine, University of California, San Diego, CA 92093, USA
- Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
- Veterans Affairs Medical Center, VA San Diego Healthcare System, La Jolla, San Diego, CA 92093, USA
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA 92093, USA
- Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
| |
Collapse
|
23
|
Sharma A, Lee J, Fonseca AG, Moshensky A, Kothari T, Sayed IM, Ibeawuchi SR, Pranadinata RF, Ear J, Sahoo D, Crotty-Alexander LE, Ghosh P, Das S. E-cigarettes compromise the gut barrier and trigger inflammation. iScience 2021; 24:102035. [PMID: 33537654 PMCID: PMC7841355 DOI: 10.1016/j.isci.2021.102035] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/15/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
E-cigarette usage continues to rise, yet the safety of e-cigarette aerosols is questioned. Using murine models of acute and chronic e-cigarette aerosol inhalation, murine colon transcriptomics, and murine and human gut-derived organoids in co-culture models, we assessed the effects of e-cigarette use on the gut barrier. Histologic and transcriptome analyses revealed that chronic, but not acute, nicotine-free e-cigarette use increased inflammation and reduced expression of tight junction (TJ) markers. Exposure of murine and human enteroid-derived monolayers (EDMs) to nicotine-free e-cigarette aerosols alone or in co-culture with bacteria also causes barrier disruption, downregulation of TJ protein, and enhanced inflammation in response to infection. These data highlight the harmful effects of "non-nicotine" component of e-cigarettes on the gut barrier. Considering the importance of an intact gut barrier for host fitness and the impact of gut mucosal inflammation on a multitude of chronic diseases, these findings are broadly relevant to both medicine and public health.
Collapse
Affiliation(s)
- Aditi Sharma
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | - Jasper Lee
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | - Ayden G. Fonseca
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
| | - Alex Moshensky
- Department of Medicine, University of California, San Diego, CA 92093, USA
| | - Taha Kothari
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | - Ibrahim M. Sayed
- Department of Pathology, University of California, San Diego, CA 92093, USA
| | | | - Rama F. Pranadinata
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
| | - Jason Ear
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
- Department of Medicine, University of California, San Diego, CA 92093, USA
| | - Debashis Sahoo
- Department of Pediatrics, University of California, San Diego, CA 92093, USA
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California, San Diego, CA 92093, USA
- Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
| | - Laura E. Crotty-Alexander
- Department of Medicine, University of California, San Diego, CA 92093, USA
- Veterans Affairs Medical Center, VA San Diego Healthcare System, La Jolla, San Diego, CA 92093, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA
- Department of Medicine, University of California, San Diego, CA 92093, USA
- Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
- Veterans Affairs Medical Center, VA San Diego Healthcare System, La Jolla, San Diego, CA 92093, USA
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA 92093, USA
- Rebecca and John Moore Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
| |
Collapse
|
24
|
Olivier S, Pochard C, Diounou H, Castillo V, Divoux J, Alcantara J, Leclerc J, Guilmeau S, Huet C, Charifi W, Varin TV, Daniel N, Foretz M, Neunlist M, Salomon BL, Ghosh P, Marette A, Rolli-Derkinderen M, Viollet B. Deletion of intestinal epithelial AMP-activated protein kinase alters distal colon permeability but not glucose homeostasis. Mol Metab 2021; 47:101183. [PMID: 33548500 PMCID: PMC7921883 DOI: 10.1016/j.molmet.2021.101183] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/21/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Objective The intestinal epithelial barrier (IEB) restricts the passage of microbes and potentially harmful substances from the lumen through the paracellular space, and rupture of its integrity is associated with a variety of gastrointestinal disorders and extra-digestive diseases. Increased IEB permeability has been linked to disruption of metabolic homeostasis leading to obesity and type 2 diabetes. Interestingly, recent studies have uncovered compelling evidence that the AMP-activated protein kinase (AMPK) signaling pathway plays an important role in maintaining epithelial cell barrier function. However, our understanding of the function of intestinal AMPK in regulating IEB and glucose homeostasis remains sparse. Methods We generated mice lacking the two α1 and α2 AMPK catalytic subunits specifically in intestinal epithelial cells (IEC AMPK KO) and determined the physiological consequences of intestinal-specific deletion of AMPK in response to high-fat diet (HFD)-induced obesity. We combined histological, functional, and integrative analyses to ascertain the effects of gut AMPK loss on intestinal permeability in vivo and ex vivo and on the development of obesity and metabolic dysfunction. We also determined the impact of intestinal AMPK deletion in an inducible mouse model (i-IEC AMPK KO) by measuring IEB function, glucose homeostasis, and the composition of gut microbiota via fecal 16S rRNA sequencing. Results While there were no differences in in vivo intestinal permeability in WT and IEC AMPK KO mice, ex vivo transcellular and paracellular permeability measured in Ussing chambers was significantly increased in the distal colon of IEC AMPK KO mice. This was associated with a reduction in pSer425 GIV phosphorylation, a marker of leaky gut barrier. However, the expression of tight junction proteins in intestinal epithelial cells and pro-inflammatory cytokines in the lamina propria were not different between genotypes. Although the HFD-fed AMPK KO mice displayed suppression of the stress polarity signaling pathway and a concomitant increase in colon permeability, loss of intestinal AMPK did not exacerbate body weight gain or adiposity. Deletion of AMPK was also not sufficient to alter glucose homeostasis or the acute glucose-lowering action of metformin in control diet (CD)- or HFD-fed mice. CD-fed i-IEC AMPK KO mice also presented higher permeability in the distal colon under homeostatic conditions but, surprisingly, this was not detected upon HFD feeding. Alteration in epithelial barrier function in the i-IEC AMPK KO mice was associated with a shift in the gut microbiota composition with higher levels of Clostridiales and Desulfovibrionales. Conclusions Altogether, our results revealed a significant role of intestinal AMPK in maintaining IEB integrity in the distal colon but not in regulating glucose homeostasis. Our data also highlight the complex interaction between gut microbiota and host AMPK. Deletion of intestinal AMPKα1 and α2 suppresses the stress-polarity signaling (SPS) pathway. Loss of the SPS pathway is associated with increased paracellular permeability in the distal colon. Intestinal AMPK is dispensable for the acute glucose-lowering action of metformin. Loss of intestinal AMPK alters the gut microbiota composition.
Collapse
Affiliation(s)
- Séverine Olivier
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Camille Pochard
- University of Nantes, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Hanna Diounou
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Vanessa Castillo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jordane Divoux
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Joshua Alcantara
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Jocelyne Leclerc
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Sandra Guilmeau
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Camille Huet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Wafa Charifi
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Thibault V Varin
- Québec Heart and Lung Research Institute (IUCPQ) & Institute for Nutrition and Functional Foods (INAF), Laval University Québec, Québec, Canada
| | - Noëmie Daniel
- Québec Heart and Lung Research Institute (IUCPQ) & Institute for Nutrition and Functional Foods (INAF), Laval University Québec, Québec, Canada
| | - Marc Foretz
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Michel Neunlist
- University of Nantes, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | - Benoit L Salomon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - André Marette
- Québec Heart and Lung Research Institute (IUCPQ) & Institute for Nutrition and Functional Foods (INAF), Laval University Québec, Québec, Canada
| | - Malvyne Rolli-Derkinderen
- University of Nantes, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France.
| | - Benoit Viollet
- Université de Paris, Institut Cochin, INSERM, CNRS, F-75014 Paris, France.
| |
Collapse
|
25
|
Li J, Xu X, Tiwari M, Chen Y, Fuller M, Bansal V, Tamayo P, Das S, Ghosh P, Sen GL. SPT6 promotes epidermal differentiation and blockade of an intestinal-like phenotype through control of transcriptional elongation. Nat Commun 2021; 12:784. [PMID: 33542242 PMCID: PMC7862286 DOI: 10.1038/s41467-021-21067-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
In adult tissue, stem and progenitor cells must tightly regulate the balance between proliferation and differentiation to sustain homeostasis. How this exquisite balance is achieved is an area of active investigation. Here, we show that epidermal genes, including ~30% of induced differentiation genes already contain stalled Pol II at the promoters in epidermal stem and progenitor cells which is then released into productive transcription elongation upon differentiation. Central to this process are SPT6 and PAF1 which are necessary for the elongation of these differentiation genes. Upon SPT6 or PAF1 depletion there is a loss of human skin differentiation and stratification. Unexpectedly, loss of SPT6 also causes the spontaneous transdifferentiation of epidermal cells into an intestinal-like phenotype due to the stalled transcription of the master regulator of epidermal fate P63. Our findings suggest that control of transcription elongation through SPT6 plays a prominent role in adult somatic tissue differentiation and the inhibition of alternative cell fate choices.
Collapse
Affiliation(s)
- Jingting Li
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA
| | - Xiaojun Xu
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Manisha Tiwari
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA
| | - Yifang Chen
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA
| | - Mackenzie Fuller
- Departments of Medicine and Cellular and Molecular Medicine, HUMANOID Center of Research Excellence, University of California, San Diego, La Jolla, CA, USA
- Department of Pathology, HUMANOID Center of Research Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Varun Bansal
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA
| | - Pablo Tamayo
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
- Division of Medical Genetics, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Soumita Das
- Department of Pathology, HUMANOID Center of Research Excellence, University of California, San Diego, La Jolla, CA, USA
| | - Pradipta Ghosh
- Departments of Medicine and Cellular and Molecular Medicine, HUMANOID Center of Research Excellence, University of California, San Diego, La Jolla, CA, USA
| | - George L Sen
- Department of Dermatology, Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
26
|
Lan Y, Dong M, Li Y, Diao Y, Chen Z, Li Y. SIRT1-induced deacetylation of Akt expedites platelet phagocytosis and delays HEMEC aging. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 23:1323-1333. [PMID: 33717652 PMCID: PMC7920857 DOI: 10.1016/j.omtn.2021.01.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 01/20/2021] [Indexed: 12/25/2022]
Abstract
Maintaining the health of the endothelium is of critical importance to prevention against cell aging. The current study was performed to clarify the role of sirtuin1 (SIRT1) in platelet phagocytosis in cell aging and identified its downstream molecular mechanism. Platelet phagocytosis by human endometrial microvascular endothelial cells (HEMECs) was characterized by transmission electron and fluorescence microscopy. Functional experiments were conducted to examine platelet phagocytosis and cell aging using the overexpression or knockdown plasmids of SIRT1 and G alpha-interacting, vesicle-associated protein (GIRDIN) as well as Akt inhibitor and activator. It was found that SIRT1 facilitated platelet phagocytosis by HEMECs, contributing to inhibition of cell aging. Akt activation facilitated platelet phagocytosis and repressed cell aging. GIRDIN overexpression accelerated platelet phagocytosis by HEMECs, leading to a delay in cell aging. GIRDIN phosphorylation at Ser1417 was induced by Akt activation, while activation of Akt was induced by SIRT1-mediated deacetylation, consequently augmenting platelet phagocytosis and delaying cell aging. Taken together, SIRT1 delayed aging of HEMECs by deacetylating Akt, phosphorylating GIRDIN, and inducing platelet phagocytosis. The study highlights a possible target for the prevention of HEMEC aging.
Collapse
Affiliation(s)
- Yong Lan
- Department of Vascular Surgery, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Min Dong
- Department of Cardiology, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Yongjun Li
- Department of Vascular Surgery, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Yongpeng Diao
- Department of Vascular Surgery, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Zuoguang Chen
- Department of Vascular Surgery, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital, Beijing 100730, P.R. China
| | - Yangfang Li
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| |
Collapse
|
27
|
Sayed IM, Chakraborty A, Abd El-Hafeez AA, Sharma A, Sahan AZ, Huang WJM, Sahoo D, Ghosh P, Hazra TK, Das S. The DNA Glycosylase NEIL2 Suppresses Fusobacterium-Infection-Induced Inflammation and DNA Damage in Colonic Epithelial Cells. Cells 2020; 9:E1980. [PMID: 32872214 PMCID: PMC7565382 DOI: 10.3390/cells9091980] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer, while the majority (80-85%) of CRCs are sporadic and are microsatellite stable (MSS), and approximately 15-20% of them display microsatellite instability (MSI). Infection and chronic inflammation are known to induce DNA damage in host tissues and can lead to oncogenic transformation of cells, but the role of DNA repair proteins in microbe-associated CRCs remains unknown. Using CRC-associated microbes such as Fusobacterium nucleatum (Fn) in a coculture with murine and human enteroid-derived monolayers (EDMs), here, we show that, among all the key DNA repair proteins, NEIL2, an oxidized base-specific DNA glycosylase, is significantly downregulated after Fn infection. Fn infection of NEIL2-null mouse-derived EDMs showed a significantly higher level of DNA damage, including double-strand breaks and inflammatory cytokines. Several CRC-associated microbes, but not the commensal bacteria, induced the accumulation of DNA damage in EDMs derived from a murine CRC model, and Fn had the most pronounced effect. An analysis of publicly available transcriptomic datasets showed that the downregulation of NEIL2 is often encountered in MSS compared to MSI CRCs. We conclude that the CRC-associated microbe Fn induced the downregulation of NEIL2 and consequent accumulation of DNA damage and played critical roles in the progression of CRCs.
Collapse
Affiliation(s)
- Ibrahim M. Sayed
- Department of Pathology, University of California, San Diego, CA 92093, USA; (I.M.S.); (A.S.); (A.Z.S.)
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Anirban Chakraborty
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX-77555, USA; (A.C.); (T.K.H.)
| | - Amer Ali Abd El-Hafeez
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA.; (A.A.A.E.-H.); (W.J.M.H.); (P.G.)
| | - Aditi Sharma
- Department of Pathology, University of California, San Diego, CA 92093, USA; (I.M.S.); (A.S.); (A.Z.S.)
| | - Ayse Z. Sahan
- Department of Pathology, University of California, San Diego, CA 92093, USA; (I.M.S.); (A.S.); (A.Z.S.)
| | - Wendy Jia Men Huang
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA.; (A.A.A.E.-H.); (W.J.M.H.); (P.G.)
| | - Debashis Sahoo
- Department of Pediatrics, University of California, San Diego, CA 92093, USA;
- Department of Computer Science and Engineering, Jacob’s School of Engineering, La Jolla, CA 92093, USA
| | - Pradipta Ghosh
- Department of Cellular and Molecular Medicine, University of California, San Diego, CA 92093, USA.; (A.A.A.E.-H.); (W.J.M.H.); (P.G.)
- Department of Medicine, University of California, San Diego, CA 92093, USA
- Moores Cancer Center, University of California, San Diego, CA 92093, USA
| | - Tapas K. Hazra
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX-77555, USA; (A.C.); (T.K.H.)
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA 92093, USA; (I.M.S.); (A.S.); (A.Z.S.)
| |
Collapse
|