1
|
Liang S, Chang Z, Lu M, Guo Z, Luo D, Xing G, Xie M, Huang W, Hou S. Host lipid metabolism influences the variation in resistance of Pekin ducks to duck hepatitis A virus genotype 3. Int J Biol Macromol 2024:139168. [PMID: 39733889 DOI: 10.1016/j.ijbiomac.2024.139168] [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: 11/12/2023] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/31/2024]
Abstract
Duck viral hepatitis (DVH) is a common and serious acute infectious disease that has a significantly impact on the duck farming industry. Duck hepatitis A virus type 3 (DHAV-3) is the major causative agent of DVH in East Asia. Host factor indicators of resistance to DHAV-3 in Pekin ducks were investigated using resistant (Z7R) and susceptible (Z7S) duck lines. Before DHAV-3 infection, Z7R had significantly higher HDL-C and LDL-C levels than Z7S. The results of population verification showed that Pekin ducks with HDL-C and/or LDL-C concentrations within their maximum 5 % confidence interval were highly resistant to DHAV-3. RNA-seq identified fifteen differentially expressed genes, primarily involved in lipid metabolism. Additionally, lipidomics identified one hundred distinct metabolites involved in glycerophospholipid metabolism. The ACSL6 gene was found to be significantly associated with OAHFA, PC, and PE. ACSL6, PE, PC, HDLC, and LDL-C co-regulated hepatic lipid metabolism. In conclusion, our results reveal that HDL-C and LDL-C may serve as markers of anti-DHAV-3 infection and lipid metabolism may be related to a potential mechanism of antiviral activity in Pekin ducks, providing a theoretical basis for future studies on the interaction between lipid metabolism and DHAV-3.
Collapse
Affiliation(s)
- Suyun Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhuo Chang
- Beijing General Station of Animal Husbandry, Beijing 100107, China
| | - Meixi Lu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Zhanbao Guo
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Dawei Luo
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Guangnan Xing
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Ming Xie
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Wei Huang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Shuisheng Hou
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
2
|
Nowacki JS, Jones GS, D'Orazio SEF. Listeria monocytogenes use multiple mechanisms to disseminate from the intestinal lamina propria to the mesenteric lymph nodes. Microbiol Spectr 2024:e0259524. [PMID: 39714174 DOI: 10.1128/spectrum.02595-24] [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: 10/14/2024] [Accepted: 11/23/2024] [Indexed: 12/24/2024] Open
Abstract
Listeria monocytogenes are facultative intracellular bacterial pathogens that cause foodborne disease in humans. The bacteria can use the surface protein InlA to invade intestinal epithelial cells or transcytose across M cells in the gut, but it is not well understood how the bacteria traffic from the underlying lamina propria to the draining mesenteric lymph nodes (MLN). Previous studies indicated that L. monocytogenes associated with both monocytes and dendritic cells in the intestinal lamina propria. We show here that CCR2-/- mice had a significant reduction in Ly6Chi monocytes in the MLN but no change in bacterial burden following foodborne infection; thus, dissemination of L. monocytogenes associated with monocytes is not required for colonization of the MLN. To block CCR7-mediated trafficking of dendritic cells from the lamina propria, we treated mice with anti-VEGFR3 antibody (clone AFL4) prior to and during infection but did not see a change in dendritic numbers in the MLN as had been previously reported with other anti-VEGFR3-specific antibodies. However, increasing the number of circulating dendritic cells by treating mice with rFlt3L resulted in a significant increase in L. monocytogenes in the lymph nodes that drain the small intestine and the spleen. Whole-mount fluorescent microscopy of lymphatic vessels following ligated loop infection revealed both free-floating L. monocytogenes and cell-associated bacteria within lymphatic vessels. Together, these results suggest that L. monocytogenes can use multiple, redundant mechanisms to disseminate from the gut tissue to the MLN. IMPORTANCE Consumption of the foodborne bacterial pathogen Listeria monocytogenes results in a wide spectrum of human disease from mild self-limiting gastroenteritis to life-threatening infections of the bloodstream, brain, and placenta. It is not well understood how the bacteria migrate from the intestines to the draining mesenteric lymph nodes, which are thought to serve as the last barrier to prevent systemic infections. Results presented here reveal multiple redundant mechanisms L. monocytogenes can use to disseminate from the ileum or colon to the mesenteric lymph nodes.
Collapse
Affiliation(s)
- Joshua S Nowacki
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Grant S Jones
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| | - Sarah E F D'Orazio
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
3
|
Sinton MC, Kajimura S. From fat storage to immune hubs: the emerging role of adipocytes in coordinating the immune response to infection. FEBS J 2024. [PMID: 39428707 DOI: 10.1111/febs.17302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/21/2024] [Accepted: 10/04/2024] [Indexed: 10/22/2024]
Abstract
Adipose tissue is a rich source of diverse cell populations, including immune cells, adipocytes and stromal cells. Interactions between these different cell types are now appreciated to be critical for maintaining tissue structure and function, by governing processes such as adipogenesis, lipolysis and differentiation of white to beige adipocytes. Interactions between these cells also drive inflammation in obesity, leading to an expansion of adipose tissue immune cells, and the secretion of proinflammatory cytokines from immune cells and from adipocytes themselves. However, in evolutionary terms, obesity is a recent phenomenon, raising the question of why adipocytes evolved to express factors that influence the immune response. Studies of various pathogens indicate that adipocytes are highly responsive to infection, altering their metabolic profiles in a way that can be used to release nutrients and fuel the immune response. In the case of infection with the extracellular parasite Trypanosoma brucei, attenuating the ability of adipocytes to sense the cytokine IL-17 results in a loss of control of the local immune response and an increased pathogen load. Intriguingly, comparisons of the adipocyte response to infection suggest that the immune responses of these cells occur in a pathogen-dependent manner, further confirming their complexity. Here, with a focus on murine adipose tissue, we discuss the emerging concept that, in addition to their canonical function, adipocytes are immune signalling hubs that integrate and disseminate signals from the immune system to generate a local environment conducive to pathogen clearance.
Collapse
Affiliation(s)
- Matthew C Sinton
- Division of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, UK
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, UK
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
4
|
Zhao D, Ravikumar V, Leach TJ, Kraushaar D, Lauder E, Li L, Sun Y, Oravecz-Wilson K, Keller ET, Chen F, Maneix L, Jenq RR, Britton R, King KY, Santibanez AE, Creighton CJ, Rao A, Reddy P. Inflammation-induced epigenetic imprinting regulates intestinal stem cells. Cell Stem Cell 2024; 31:1447-1464.e6. [PMID: 39232559 DOI: 10.1016/j.stem.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/25/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Abstract
It remains unknown whether and how intestinal stem cells (ISCs) adapt to inflammatory exposure and whether the adaptation leaves scars that will affect their subsequent regeneration. We investigated the consequences of inflammation on Lgr5+ ISCs in well-defined clinically relevant models of acute gastrointestinal graft-versus-host disease (GI GVHD). Utilizing single-cell transcriptomics, as well as organoid, metabolic, epigenomic, and in vivo models, we found that Lgr5+ ISCs undergo metabolic changes that lead to the accumulation of succinate, which reprograms their epigenome. These changes reduced the ability of ISCs to differentiate and regenerate ex vivo in serial organoid cultures and also in vivo following serial transplantation. Furthermore, ISCs demonstrated a reduced capacity for in vivo regeneration despite resolution of the initial inflammatory exposure, demonstrating the persistence of the maladaptive impact induced by the inflammatory encounter. Thus, inflammation imprints the epigenome of ISCs in a manner that persists and affects their sensitivity to adapt to future stress or challenges.
Collapse
Affiliation(s)
- Dongchang Zhao
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Visweswaran Ravikumar
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tyler J Leach
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Daniel Kraushaar
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Emma Lauder
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA; Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, USA
| | - Lu Li
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Yaping Sun
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Katherine Oravecz-Wilson
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, USA
| | - Evan T Keller
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Fengju Chen
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Laure Maneix
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Robert R Jenq
- Department of Genomic Medicine and Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Robert Britton
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Katherine Y King
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Ana E Santibanez
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Chad J Creighton
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA
| | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pavan Reddy
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Department of Internal Medicine, Houston, TX 77030, USA; Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
5
|
Naseri S, Asgarpanah J, Ziai SA. Immunomodulatory and antioxidant effect of liposomal auraptene against cyclophosphamide-induced immunosuppression in BALB/c mice. Exp Gerontol 2024; 195:112552. [PMID: 39173782 DOI: 10.1016/j.exger.2024.112552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION Cyclophosphamide (CP), which is a commonly used chemotherapy drug, can lead to a range of side effects such as immunosuppression, bone marrow suppression, leukopenia, and oxidative stress. This study aims to explore the effects of Auraptene (AUR), which has immunomodulatory and antioxidant properties, on immune function in mice that are experiencing suppression induced by CP. MATERIALS AND METHODS The experiment involved 60 male BALB/c mice that underwent a 10-day treatment. On days 1, 3, and 9, CP was given at 80 mg/kg IP doses to induce immunosuppression. The mice were divided into five groups: Control group, CP group, CP + liposomal AUR 0.2 mg/kg (AUR 0.2), CP + liposomal AUR 0.25 mg/kg (AUR 0.25), and liposomal vehicle group. Various parameters were measured, including mouse weight, immune organ weight index (spleen and thymus), spleen and thymus histopathology, levels of inflammatory cytokines (IL2, IL10, IL4, IFN-γ), TH1/TH2 balance ratio, IgG and IgM immunoglobulin levels, white blood cell count, platelets, neutrophils, lymphocytes, and oxidative activity measured by MDA, SOD, and Total Antioxidant. RESULTS In the group treated with CP, the mice showed a significant decrease in weight compared to the control group. In contrast, the group treated with AUR maintained their weight and did not show a significant difference from the control group. AUR 0.25 reduced the damage to the spleen and thymus caused by CP. Additionally, AUR 0.25 demonstrated a significant decrease in IL4 and IL10 levels compared to the CP group (p = 0.04), approaching the levels of the control group. Furthermore, IL2 and IFN-γ levels in the AUR 0.25 group significantly increased (p = 0.04) compared to the CP group, reaching levels similar to the control group. AUR also increased serum IgM and IgG levels two to three times compared to the CP group, approaching the levels of the control group. MDA levels in the AUR 0.25 group decreased to normal and control levels. AUR 0.25 also showed increased SOD and Total Antioxidant levels. Additionally, white blood cells, platelets, neutrophils, and lymphocytes in the AUR group significantly increased compared to the CP group, reaching normal levels similar to the control group. The TH1/TH2 ratio in the AUR group exhibited a significant increase of two and a half times (p = 0.002) compared to the CP group. CONCLUSION These results show that AUR protects against the side effects of CP by increasing the function of the humoral and cellular immune system through the balance of TH1/TH2 and increasing the level of immunoglobulins, as well as increasing the antioxidant activity and the protective role of cytotoxicity.
Collapse
Affiliation(s)
- Saeed Naseri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Jinous Asgarpanah
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Seyed Ali Ziai
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Infectious Diseases and Tropical Medicine Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Alberti A, Araujo Coelho DR, Vieira WF, Moehlecke Iser B, Lampert RMF, Traebert E, Silva BBD, Oliveira BHD, Leão GM, Souza GD, Dallacosta FM, Kades G, Madeira K, Chupel MU, Grossl FS, Souza R, Hur Soares B, Endrigo Ruppel da Rocha R, da Silva Sipriano E, Fernandes Martins D, Agostinetto L. Factors Associated with the Development of Depression and the Influence of Obesity on Depressive Disorders: A Narrative Review. Biomedicines 2024; 12:1994. [PMID: 39335507 PMCID: PMC11429137 DOI: 10.3390/biomedicines12091994] [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: 07/13/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/30/2024] Open
Abstract
Depression affects several aspects of life, including socioeconomic status, relationships, behavior, emotions, and overall health. The etiology of depression is complex and influenced by various factors, with obesity emerging as a significant contributor. This narrative review aims to investigate the factors associated with the development of depression, with a particular focus on the role of obesity. The literature search was conducted on PubMed, Embase, and PsycINFO from May to July 2024. The review highlights the impact of environmental and socioeconomic conditions; lifestyle choices, including physical activity and dietary habits; stress; traumatic experiences; neurotransmitter imbalances; medical and psychological conditions; hormone fluctuations; and epigenetic factors on depression. A key emphasis is placed on the inflammatory processes linked to obesity, which may drive the bidirectional relationship between obesity and depression. The findings suggest that obesity is associated with an increased risk of depression, potentially due to chronic inflammation, neurochemical dysregulation, and the emotional and social challenges related to weight stigma and obesity management. Understanding these interconnected factors is important for developing targeted interventions to address both obesity and depression, leading to improved quality of life for those affected.
Collapse
Affiliation(s)
- Adriano Alberti
- Department of Biological and Health Sciences Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça 88132-260, Brazil
- Graduate Program in Environment and Health, University of Planalto Catarinense-UNIPLAC, Lages 88509-900, Brazil
| | | | - Willians Fernando Vieira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo 5508-000, Brazil
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-864, Brazil
- Laboratory of Neuroimmune Interface of Pain Research, Faculdade São Leopoldo Mandic, Instituto São Leopoldo Mandic, Campinas 13045-755, Brazil
| | - Betine Moehlecke Iser
- Department of Biological and Health Sciences Posgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Tubarão 88704-900, Brazil
| | - Rose Meiry Fernandez Lampert
- Department of Biological and Health Sciences Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça 88132-260, Brazil
| | - Eliane Traebert
- Department of Biological and Health Sciences Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça 88132-260, Brazil
| | - Bruna Becker da Silva
- Department of Biological and Health Sciences Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça 88132-260, Brazil
| | - Bruna Hoffmann de Oliveira
- Department of Biological and Health Sciences Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça 88132-260, Brazil
| | - Graziela Marques Leão
- Department of Biological and Health Sciences Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça 88132-260, Brazil
| | - Gabriela de Souza
- Department of Biological and Health Sciences Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça 88132-260, Brazil
| | | | - Gabriela Kades
- Department of Biosciences and Health, University of West Santa Catarina, Joaçaba 89600-000, Brazil
| | - Kristian Madeira
- Department of Mathematics and Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma 88806-000, Brazil
| | - Matheus Uba Chupel
- Hurvitz Brain Sciences, Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Fernando Schorr Grossl
- Department of Biosciences and Health, University of West Santa Catarina, Joaçaba 89600-000, Brazil
| | - Renan Souza
- Department of Biosciences and Health, University of West Santa Catarina, Joaçaba 89600-000, Brazil
| | - Ben Hur Soares
- Department of Physical Education and Physiotherapy, University of Passo Fundo, Passo Fundo 99052-900, Brazil
| | - Ricelli Endrigo Ruppel da Rocha
- Department of the Graduate Program in Development and Society-PPGEDS (UNIARP), University of Alto Vale do Rio do Peixe, Caçador 89500-199, Brazil
| | - Erica da Silva Sipriano
- Department of Mathematics and Health Sciences, University of the Extreme South of Santa Catarina (UNESC), Criciúma 88806-000, Brazil
| | - Daniel Fernandes Martins
- Department of Biological and Health Sciences Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça 88132-260, Brazil
| | - Lenita Agostinetto
- Graduate Program in Environment and Health, University of Planalto Catarinense-UNIPLAC, Lages 88509-900, Brazil
| |
Collapse
|
7
|
Bolden M, Davis XD, Sherwood ER, Bohannon JK, Caslin HL. Weight loss-induced adipose macrophage memory improves local Staphylococcus aureus clearance in male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.03.606494. [PMID: 39211192 PMCID: PMC11361095 DOI: 10.1101/2024.08.03.606494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Different stimuli can induce innate immune memory to improve pathogen defense or worsen cardiometabolic disease. However, it is less clear if the same stimuli can induce both the protective and detrimental effects of innate immune memory. We previously showed that weight loss induces innate immune memory in adipose macrophages that correlates with worsened diabetes risk after weight regain. In this study, we investigated the effect of weight loss on macrophage cytokine production and overall survival in a mouse model of infection. Male C57Bl/6J mice were put on high-fat or low-fat diets over 18 weeks to induce weight gain or weight loss. Lean mice served as controls. All mice were then infected IV with 2.5×10^6 CFU Staphylococcus aureus . Tissues were collected from 10 mice/group at day 3 and the remaining animals were followed for survival. Weight gain mice had the highest blood neutrophils and the highest bacterial burden in the kidney. However, there was no significant difference in survival. The weight loss group had the highest plasma TNF-α and a significant reduction in bacterial burden in the adipose tissue that correlated with increased adipose macrophage cytokine production. Thus, weight loss-induced adipose macrophage memory may both improve local S.aureus clearance and worsen diabetes risk upon weight regain. Collectively, these findings support the notion that innate immune memory is an evolutionarily protective mechanism that also contributes to the development of cardiometabolic diseases.
Collapse
|
8
|
Dahlquist KJV, Huggins MA, Yousefzadeh MJ, Soto-Palma C, Cholensky SH, Pierson M, Smith DM, Hamilton SE, Camell CD. PD1 blockade improves survival and CD8 + cytotoxic capacity, without increasing inflammation, during normal microbial experience in old mice. NATURE AGING 2024; 4:915-925. [PMID: 38689133 DOI: 10.1038/s43587-024-00620-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/29/2024] [Indexed: 05/02/2024]
Abstract
By 2030, individuals 65 years of age or older will make up approximately 20% of the world's population1. Older individuals are at the highest risk for mortality from infections, largely due to the pro-inflammatory, dysfunctional immune response, which is collectively known as immunosenescence2. During aging, CD8+ T cells acquire an exhausted phenotype, including increased expression of inhibitory receptors, such as programmed cell death 1 (PD1), a decline in effector function and elevated expression of inflammatory factors3-7. PD1 reduces T cell receptor activity via SHP2-dependent dephosphorylation of multiple pathways; accordingly, inhibiting PD1 activity through monoclonal antibodies increases CD8+ T cell effector response in young mice8-11. Attempts to improve CD8+ T cell responses by blocking inhibitory receptors are attractive; however, they can lead to adverse immune events due to overamplification of T cell receptor signaling and T cell activation12,13. Here we investigated the effect of monoclonal anti-PD1 immunotherapy during normal microbial experience, otherwise known as exposure to dirty mice, to determine whether it either improves exhausted CD8+ T cell responses in old mice or leads to a heightened inflammatory response and increased mortality.
Collapse
Affiliation(s)
- Korbyn J V Dahlquist
- Biochemistry, Molecular Biology and Biophysics Graduate Program, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Matthew A Huggins
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Matthew J Yousefzadeh
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, Columbia Center for Translational Immunology, Columbia Center for Healthy Longevity, Columbia University, New York, NY, USA
| | - Carolina Soto-Palma
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Stephanie H Cholensky
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Mark Pierson
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Declan M Smith
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
| | - Sara E Hamilton
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Christina D Camell
- Biochemistry, Molecular Biology and Biophysics Graduate Program, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA.
- Department of Biochemistry, Molecular Biology and Biophysics, Institute on the Biology of Aging and Metabolism, University of Minnesota, Minneapolis, MN, USA.
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
9
|
Chen C, Han P, Qing Y. Metabolic heterogeneity in tumor microenvironment - A novel landmark for immunotherapy. Autoimmun Rev 2024; 23:103579. [PMID: 39004158 DOI: 10.1016/j.autrev.2024.103579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/10/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The surrounding non-cancer cells and tumor cells that make up the tumor microenvironment (TME) have various metabolic rhythms. TME metabolic heterogeneity is influenced by the intricate network of metabolic control within and between cells. DNA, protein, transport, and microbial levels are important regulators of TME metabolic homeostasis. The effectiveness of immunotherapy is also closely correlated with alterations in TME metabolism. The response of a tumor patient to immunotherapy is influenced by a variety of variables, including intracellular metabolic reprogramming, metabolic interaction between cells, ecological changes within and between tumors, and general dietary preferences. Although immunotherapy and targeted therapy have made great strides, their use in the accurate identification and treatment of tumors still has several limitations. The function of TME metabolic heterogeneity in tumor immunotherapy is summarized in this article. It focuses on how metabolic heterogeneity develops and is regulated as a tumor progresses, the precise molecular mechanisms and potential clinical significance of imbalances in intracellular metabolic homeostasis and intercellular metabolic coupling and interaction, as well as the benefits and drawbacks of targeted metabolism used in conjunction with immunotherapy. This offers insightful knowledge and important implications for individualized tumor patient diagnosis and treatment plans in the future.
Collapse
Affiliation(s)
- Chen Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China
| | - Peng Han
- Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, China.
| | - Yanping Qing
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
10
|
Song M, Lv K, Xu Z, Li J, Sun J, Shi J, Xu Y. N6 methyladenosine eraser FTO suppresses Staphylococcus aureus-induced ferroptosis of bone marrow mesenchymal stem cells to ameliorate osteomyelitis through regulating the MDM2/TLR4/SLC7A11 signaling pathway. Cell Biol Int 2024; 48:450-460. [PMID: 38165230 DOI: 10.1002/cbin.12115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/22/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Osteomyelitis is a bone destructive inflammatory disease caused by infection. Ferroptosis is closely related to multiple inflammatory diseases, but the role of ferroptosis in Staphylococcus aureus (SA)-induced osteomyelitis remains unknown. In the present study, we found that SA treatment promoted the accumulation of iron, Fe2+ , lipid peroxide, and malondialdehyde, increased TFRC and reduced FTH1 and GPX4 to trigger ferroptosis in rat bone marrow mesenchymal stem cells (BMSCs). Interestingly, increased level of N6 methyl adenosine (m6A) modification along with decreased expression level of m6A eraser FTO were observed in SA-induced BMSCs, while upregulating FTO alleviated SA-triggered ferroptosis and protected cell viability in BMSCs. Mechanistically, MDM2 was identified as a target of FTO-mediated m6A demethylation, and FTO upregulation promoted MDM2 instability to downregulated TLR4 signal and elevate the expression of SLC7A11 and GPX4 in SA-induced BMSCs. Functional recovery experiments verified that overexpressing MDM2 or TLR4 reversed the inhibiting effect of FTO upregulation on ferroptosis in SA-treated BMSCs. Additionally, FTO upregulation restrained ferroptosis and pathological damage to bone tissue in SA-induced osteomyelitis model rats. Altogether, m6A eraser FTO alleviates SA-induced ferroptosis in osteomyelitis models partly through inhibiting the MDM2-TLR4 axis.
Collapse
Affiliation(s)
- Muguo Song
- Graduate School of Kunming Medical University, Kunming, China
- Orthopaedics Department, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Kehan Lv
- Graduate School of Kunming Medical University, Kunming, China
- Orthopaedics Department, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Zhi Xu
- Graduate School of Kunming Medical University, Kunming, China
- Orthopaedics Department, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Junyi Li
- Graduate School of Kunming Medical University, Kunming, China
- Orthopaedics Department, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Jian Sun
- Graduate School of Kunming Medical University, Kunming, China
- Orthopaedics Department, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Jian Shi
- Orthopaedics Department, 920th Hospital of Joint Logistics Support Force, Kunming, China
| | - Yongqing Xu
- Orthopaedics Department, 920th Hospital of Joint Logistics Support Force, Kunming, China
| |
Collapse
|
11
|
Zheng X, Liu Y, Liu Y, Zang J, Wang K, Yang Z, Chen N, Sun J, Huang L, Li Y, Xue L, Zhi H, Zhang X, Yu M, Chen S, Dong H, Li Y. Arginine-assembly as NO nano-donor prevents the negative feedback of macrophage repolarization by mitochondrial dysfunction for cancer immunotherapy. Biomaterials 2024; 306:122474. [PMID: 38271788 DOI: 10.1016/j.biomaterials.2024.122474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/03/2023] [Accepted: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Repolarizing the tumor-associated macrophages (TAMs) towards the antitumoral M1-like phenotype has been a promising approach for cancer immunotherapy. However, the anti-cancer immune response is severely limited mainly by the repolarized M1-like macrophages belatedly returning to the M2-like phenotype (i.e., negative feedback). Inspired by nitric oxide (NO) effectively preventing repolarization of inflammatory macrophages in inflammatory diseases, herein, we develop an arginine assembly, as NO nano-donor for NO generation to prevent the negative feedback of the macrophage repolarization. The strategy is to first apply reversible tagging of hydrophobic terephthalaldehyde to create an arginine nano-assembly, and then load a toll-like receptor 7/8 agonist resiquimod (R848) (R848@Arg). Through this strategy, a high loading efficiency of 40 % for the arginine and repolarization characteristics for TAMs can be achieved. Upon the macrophage repolarization by R848, NO can be intracellularly generated from the released arginine by the upregulated inducible nitric oxide synthase. Mechanistically, NO effectively prevented the negative feedback of the repolarized macrophage by mitochondrial dysfunction via blocking oxidative phosphorylation. Notably, R848@Arg significantly increased the tumor inhibition ratio by 3.13-fold as compared to the free R848 by maintaining the M1-like phenotype infiltrating into tumor. The Arg-assembly as NO nano-donor provides a promising method for effective repolarization of macrophages.
Collapse
Affiliation(s)
- Xiao Zheng
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Ying Liu
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Yiqiong Liu
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Jie Zang
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Kun Wang
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Zichen Yang
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Nana Chen
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Jiuyuan Sun
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Li Huang
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Yan Li
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Liangyi Xue
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Hui Zhi
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Xiaoyou Zhang
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Min Yu
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Shiyu Chen
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Haiqing Dong
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China
| | - Yongyong Li
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092,China.
| |
Collapse
|
12
|
Yang J, Liu J, Kuang W, Lin Y, Zhong S, Kraithong S, Zhang X, Wong IN, Huang R. Structural characterization and ferroptosis-related immunomodulatory of a novel exopolysaccharide isolated from marine fungus Aspergillus medius. Int J Biol Macromol 2024; 265:130703. [PMID: 38458279 DOI: 10.1016/j.ijbiomac.2024.130703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Marine fungal exopolysaccharides play a crucial role in immunoregulation. In this investigation, a novel polysaccharide was extracted from the culture medium of the marine fungus Aspergillus medius SCAU-236. Compositional analysis revealed a structure composed of glucose units with (1,4)-α-D-Glcp, (1,3,4)-β-D-Glcp, and (1,4,6)-α-D-Glcp, along with side chains of 1-α-D-Glcp linked to carbon 6 of (1,4,6)-α-D-Glcp and carbon 3 of (1,3,4)-β-D-Glcp. Functional evaluations on RAW264.7 macrophage cells demonstrated Aspergillus medius polysaccharide (ASMP)'s effects on cell proliferation, nitric oxide levels, and the secretion of TNF-α, IL-6, and IL-1β cytokines. Additionally, metabolomics indicated ASMP's potential to modulate macrophage immune function by impacting key regulatory molecules, including COX-2, iNOS, Nrf2, SLC7A11, GPX4, and ACSL4. The Nrf2/SLC7A11/GPX4 axis and ACSL4 were suggested to be involved in ASMP-induced ferroptosis, leading to increased reactive oxygen species (ROS) levels and lipid peroxidation. These findings propose a unique mechanism by which ASMP exerts immunomodulatory effects through ferroptosis induction, contributing to the understanding of marine-derived compounds in immunomodulation research.
Collapse
Affiliation(s)
- Jiajia Yang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jun Liu
- Laboratory of Pathogenic Biology, Guangdong Medical University, Zhanjiang 524023, China
| | - Weiyang Kuang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yuqi Lin
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Saiyi Zhong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Supaluck Kraithong
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoyong Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Io Nam Wong
- Faculty of Medicine, Macau University of Science and Technology, 999078, Macau.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
13
|
Kim K, Park S, Lee Y, Baek J, Kim Y, Hwang SW, Lee JL, Park SH, Yang SK, Han B, Song K, Yoon YS, Lee HS, Ye BD. Transcriptomic Profiling and Cellular Composition of Creeping Fat in Crohn's disease. J Crohns Colitis 2024; 18:223-232. [PMID: 37594364 DOI: 10.1093/ecco-jcc/jjad141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND AND AIMS Creeping fat [CF] is a poorly understood feature of Crohn's disease [CD], characterized by the wrapping of mesenteric adipose tissue [MAT] around the inflamed intestine. The aim of this study was to investigate the transcriptional profile and compositional features of CF. METHODS We collected 59 MAT samples: 23 paired samples from patients with CD (CF [CD-CF] and MAT around the uninflamed intestine [CD-MAT]) and 13 MAT samples from non-CD patients [Con-MAT]. Differentially expressed gene [DEG], functional pathway, cell deconvolution, and gene co-expression network analyses were performed. RESULTS By comparing three different MAT samples, we identified a total of 529 DEGs [|log2FoldChange| > 1.5; false discovery rate < 0.05]. Of these, 323 genes showed an incremental pattern from Con-MAT to CD-MAT, and to CD-CF, while 105 genes displayed a decremental pattern. Genes with an incremental pattern were related to immune cell responses, including B- and T-cell activation, while genes with a decremental pattern were involved in cell trafficking and migration. Cell deconvolution analysis revealed significant changes in cellular composition between the CD-CF and Con-MAT groups, with increased proportions of B-cells/plasma cells [p = 1.16 × 10-4], T-cells [p = 3.66 × 10-3], and mononuclear phagocytes [p = 3.53 × 10-2] in the CD-CF group. In contrast, only the B-cell/plasma cell component showed a significant increase [p = 1.62 × 10-2] in the CD-MAT group compared to Con-MAT. CONCLUSION The distinct transcriptional profiles and altered cellular components of each MAT found in our study provide insight into the mechanisms behind CF and highlight its possible role in the pathogenesis of CD.
Collapse
Affiliation(s)
- Kyuwon Kim
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sojung Park
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yoonho Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jiwon Baek
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yongjae Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sung Wook Hwang
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jong Lyul Lee
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Division of Colon and Rectal Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang Hyoung Park
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Buhm Han
- Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea
| | - Kyuyoung Song
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Yong Sik Yoon
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Division of Colon and Rectal Surgery, Department of Surgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ho-Su Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Byong Duk Ye
- Department of Gastroenterology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
- Inflammatory Bowel Disease Center, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| |
Collapse
|
14
|
Bradford BJ, Contreras GA. Adipose Tissue Inflammation: Linking Physiological Stressors to Disease Susceptibility. Annu Rev Anim Biosci 2024; 12:261-281. [PMID: 38064480 DOI: 10.1146/annurev-animal-021122-113212] [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: 02/16/2024]
Abstract
The study of adipose tissue (AT) is enjoying a renaissance. White, brown, and beige adipocytes are being investigated in adult animals, and the critical roles of small depots like perivascular AT are becoming clear. But the most profound revision of the AT dogma has been its cellular composition and regulation. Single-cell transcriptomic studies revealed that adipocytes comprise well under 50% of the cells in white AT, and a substantial portion of the rest are immune cells. Altering the function of AT resident leukocytes can induce or correct metabolic syndrome and, more surprisingly, alter adaptive immune responses to infection. Although the field is dominated by obesity research, conditions such as rapid lipolysis, infection, and heat stress impact AT immune dynamics as well. Recent findings in rodents lead to critical questions that should be explored in domestic livestock as potential avenues for improved animal resilience to stressors, particularly as animals age.
Collapse
Affiliation(s)
- Barry J Bradford
- Department of Animal Science, College of Agriculture and Natural Resources, Michigan State University, East Lansing, Michigan, USA;
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA;
| |
Collapse
|
15
|
Zhou YW, Ren Y, Lu MM, Xu LL, Cheng WX, Zhang MM, Ding LP, Chen D, Gao JG, Du J, Jin CL, Chen CX, Li YF, Cheng T, Jiang PL, Yang YD, Qian PX, Xu PF, Jin X. Crohn's disease as the intestinal manifestation of pan-lymphatic dysfunction: An exploratory proposal based on basic and clinical data. World J Gastroenterol 2024; 30:34-49. [PMID: 38293325 PMCID: PMC10823898 DOI: 10.3748/wjg.v30.i1.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/08/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024] Open
Abstract
Crohn's disease (CD) is caused by immune, environmental, and genetic factors. It can involve the entire gastrointestinal tract, and although its prevalence is rapidly increasing its etiology remains unclear. Emerging biological and small-molecule drugs have advanced the treatment of CD; however, a considerable proportion of patients are non-responsive to all known drugs. To achieve a breakthrough in this field, innovations that could guide the further development of effective therapies are of utmost urgency. In this review, we first propose the innovative concept of pan-lymphatic dysfunction for the general distribution of lymphatic dysfunction in various diseases, and suggest that CD is the intestinal manifestation of pan-lymphatic dysfunction based on basic and clinical preliminary data. The supporting evidence is fully summarized, including the existence of lymphatic system dysfunction, recognition of the inside-out model, disorders of immune cells, changes in cell plasticity, partial overlap of the underlying mechanisms, and common gut-derived fatty and bile acid metabolism. Another benefit of this novel concept is that it proposes adopting the zebrafish model for studying intestinal diseases, especially CD, as this model is good at presenting and mimicking lymphatic dysfunction. More importantly, the ensuing focus on improving lymphatic function may lead to novel and promising therapeutic strategies for CD.
Collapse
Affiliation(s)
- Yu-Wei Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yue Ren
- Department of Gastroenterology, The Second Hospital of Jiaxing, Jiaxing 314000, Zhejiang Province, China
| | - Miao-Miao Lu
- Endoscopy Center, Children’s Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ling-Ling Xu
- Department of Gastroenterology, The Second People’s Hospital of Yuhang District, Hangzhou 310000, Zhejiang Province, China
| | - Wei-Xin Cheng
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Meng-Meng Zhang
- Department of Gastroenterology, Hangzhou Shangcheng District People’s Hospital, Hangzhou 310003, Zhejiang Province, China
| | - Lin-Ping Ding
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Jian-Guo Gao
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Juan Du
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Ci-Liang Jin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Chun-Xiao Chen
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yun-Fei Li
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Tao Cheng
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Peng-Lei Jiang
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yi-Da Yang
- Department of Infectious Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Peng-Xu Qian
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Peng-Fei Xu
- Women’s Hospital and Institute of Genetics, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Xi Jin
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
16
|
Nie R, Zhang QY, Tan J, Feng ZY, Huang K, Sheng N, Jiang YL, Song YT, Zou CY, Zhao LM, Li HX, Wang R, Zhou XL, Hu JJ, Wu CY, Li-Ling J, Xie HQ. EGCG modified small intestine submucosa promotes wound healing through immunomodulation. COMPOSITES PART B: ENGINEERING 2023; 267:111005. [DOI: 10.1016/j.compositesb.2023.111005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
|
17
|
Fu X, Wang Y, Zhao F, Cui R, Xie W, Liu Q, Yang W. Shared biological mechanisms of depression and obesity: focus on adipokines and lipokines. Aging (Albany NY) 2023; 15:5917-5950. [PMID: 37387537 PMCID: PMC10333059 DOI: 10.18632/aging.204847] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
Depression and obesity are both common disorders currently affecting public health, frequently occurring simultaneously within individuals, and the relationship between these disorders is bidirectional. The association between obesity and depression is highly co-morbid and tends to significantly exacerbate metabolic and related depressive symptoms. However, the neural mechanism under the mutual control of obesity and depression is largely inscrutable. This review focuses particularly on alterations in systems that may mechanistically explain the in vivo homeostatic regulation of the obesity and depression link, such as immune-inflammatory activation, gut microbiota, neuroplasticity, HPA axis dysregulation as well as neuroendocrine regulators of energy metabolism including adipocytokines and lipokines. In addition, the review summarizes potential and future treatments for obesity and depression and raises several questions that need to be answered in future research. This review will provide a comprehensive description and localization of the biological connection between obesity and depression to better understand the co-morbidity of obesity and depression.
Collapse
Affiliation(s)
- Xiying Fu
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun 130041, P.R. China
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Yicun Wang
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Fangyi Zhao
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Ranji Cui
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wei Xie
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Qianqian Liu
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| | - Wei Yang
- Jilin Provincial Key Laboratory for Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, P.R. China
- Department of Neurology, The Second Hospital of Jilin University, Changchun 130041, P.R. China
| |
Collapse
|
18
|
Schrom E, Kinzig A, Forrest S, Graham AL, Levin SA, Bergstrom CT, Castillo-Chavez C, Collins JP, de Boer RJ, Doupé A, Ensafi R, Feldman S, Grenfell BT, Halderman JA, Huijben S, Maley C, Moses M, Perelson AS, Perrings C, Plotkin J, Rexford J, Tiwari M. Challenges in cybersecurity: Lessons from biological defense systems. Math Biosci 2023:109024. [PMID: 37270102 DOI: 10.1016/j.mbs.2023.109024] [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: 01/24/2023] [Revised: 04/27/2023] [Accepted: 05/20/2023] [Indexed: 06/05/2023]
Abstract
Defending against novel, repeated, or unpredictable attacks, while avoiding attacks on the 'self', are the central problems of both mammalian immune systems and computer systems. Both systems have been studied in great detail, but with little exchange of information across the different disciplines. Here, we present a conceptual framework for structured comparisons across the fields of biological immunity and cybersecurity, by framing the context of defense, considering different (combinations of) defensive strategies, and evaluating defensive performance. Throughout this paper, we pose open questions for further exploration. We hope to spark the interdisciplinary discovery of general principles of optimal defense, which can be understood and applied in biological immunity, cybersecurity, and other defensive realms.
Collapse
Affiliation(s)
- Edward Schrom
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, United States of America
| | - Ann Kinzig
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, United States of America
| | - Stephanie Forrest
- Biodesign Center for Biocomputation, Security and Society, Arizona State University, Tempe, AZ 85287, United States of America; School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ 85287, United States of America; Santa Fe Institute, Santa Fe, NM 87501, United States of America
| | - Andrea L Graham
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, United States of America; Santa Fe Institute, Santa Fe, NM 87501, United States of America
| | - Simon A Levin
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, United States of America.
| | - Carl T Bergstrom
- Department of Biology, University of Washington, Seattle, WA 98195, United States of America
| | - Carlos Castillo-Chavez
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ 85287, United States of America
| | - James P Collins
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, United States of America
| | - Rob J de Boer
- Theoretical Biology and Bioinformatics, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Adam Doupé
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ 85287, United States of America; Center for Cybersecurity and Trusted Foundations, Global Security Initiative, Arizona State University, Tempe, AZ 85287, United States of America
| | - Roya Ensafi
- Department of Electrical Engineering and Computer Science, Computer Science and Engineering Division, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Stuart Feldman
- Schmidt Futures, New York, NY 10011, United States of America
| | - Bryan T Grenfell
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, United States of America; Princeton School of Public and International Affairs, Princeton University, Princeton, NJ 08544, United States of America
| | - J Alex Halderman
- Department of Electrical Engineering and Computer Science, Computer Science and Engineering Division, University of Michigan, Ann Arbor, MI 48109, United States of America; Center for Computer Security and Society, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Silvie Huijben
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, United States of America
| | - Carlo Maley
- Arizona Cancer Evolution Center, Arizona State University, Tempe, AZ 85287, United States of America; Biodesign Center for Biocomputation, Security and Society, Arizona State University, Tempe, AZ 85287, United States of America
| | - Melanie Moses
- Department of Computer Science, University of New Mexico, Albuquerque, NM 87131, United States of America; Department of Biology, University of New Mexico, Albuquerque, NM 87131, United States of America; Santa Fe Institute, Santa Fe, NM 87501, United States of America
| | - Alan S Perelson
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM 87545, United States of America; Santa Fe Institute, Santa Fe, NM 87501, United States of America
| | - Charles Perrings
- School of Life Sciences, Arizona State University, Tempe, AZ 85287, United States of America
| | - Joshua Plotkin
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, United States of America
| | - Jennifer Rexford
- Department of Computer Science, Princeton University, Princeton, NJ 08540, United States of America
| | - Mohit Tiwari
- Department of Electrical and Computer Engineering, University of Texas, Austin, TX 78712, United States of America
| |
Collapse
|
19
|
Barthelemy J, Bogard G, Wolowczuk I. Beyond energy balance regulation: The underestimated role of adipose tissues in host defense against pathogens. Front Immunol 2023; 14:1083191. [PMID: 36936928 PMCID: PMC10019896 DOI: 10.3389/fimmu.2023.1083191] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 03/06/2023] Open
Abstract
Although the adipose tissue (AT) is a central metabolic organ in the regulation of whole-body energy homeostasis, it is also an important endocrine and immunological organ. As an endocrine organ, AT secretes a variety of bioactive peptides known as adipokines - some of which have inflammatory and immunoregulatory properties. As an immunological organ, AT contains a broad spectrum of innate and adaptive immune cells that have mostly been studied in the context of obesity. However, overwhelming evidence supports the notion that AT is a genuine immunological effector site, which contains all cell subsets required to induce and generate specific and effective immune responses against pathogens. Indeed, AT was reported to be an immune reservoir in the host's response to infection, and a site of parasitic, bacterial and viral infections. In addition, besides AT's immune cells, preadipocytes and adipocytes were shown to express innate immune receptors, and adipocytes were reported as antigen-presenting cells to regulate T-cell-mediated adaptive immunity. Here we review the current knowledge on the role of AT and AT's immune system in host defense against pathogens. First, we will summarize the main characteristics of AT: type, distribution, function, and extraordinary plasticity. Second, we will describe the intimate contact AT has with lymph nodes and vessels, and AT immune cell composition. Finally, we will present a comprehensive and up-to-date overview of the current research on the contribution of AT to host defense against pathogens, including the respiratory viruses influenza and SARS-CoV-2.
Collapse
Affiliation(s)
| | | | - Isabelle Wolowczuk
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (Inserm), Centre Hospitalier Universitaire de Lille (CHU Lille), Institut Pasteur de Lille, U1019 - UMR 9017 - Center for Infection and Immunity of Lille (CIIL), Lille, France
| |
Collapse
|
20
|
Yuan S, Xie G, Yang X, Chen Y, Zhang H. Portable paper-based electrochemiluminescence test incorporating lateral-flow immunosensors for detection of interferon-γ levels. Front Bioeng Biotechnol 2023; 11:1131840. [PMID: 36824352 PMCID: PMC9941175 DOI: 10.3389/fbioe.2023.1131840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Tuberculosis (TB) poses a serious threat to human health and social development. Accurate diagnosis of mycobacterium tuberculosis infection plays a critical role in the prevention and treatment of tuberculosis. Interferon-γ (INF-γ) release assay (IGRA) is currently the only quantitative tuberculosis infection diagnosis method. An accurate, fast, and easily handled INF-γ detection method is the key to obtaining accurate results. Herein, we report a novel paper-based electrochemiluminescence (ECL) method based on lateral flow immunosensors that combines the easy handling characteristics of immunochromatography and the high sensitivity of electrochemiluminescence to detect IFN-γ. To our knowledge this is the first INF-γ detection method that combines immunochromatography with electrochemiluminescence. The paper-based ECL-LFI test consists of a sample pad, conjugation pad (with binding antibody IFN-γ-Ab1 conjugated with ruthenium tripyridine), detection pad (with capture antibody IFN-γ-Ab2 immobilized on nanospheres), absorbent pad, and electrode for signal activation. The ECL signal is obtained by cyclic voltammetry scanning at a speed of 0.1 V/s in the detection area of the paper-based ECL-LFI test. In our experiments, the paper-based ECL-LFI test exhibited a minimum detection limit of 2.57 pg/mL within 12 min, and a broad detection range of 2.57-5,000 pg/mL, with repeatability of 8.10% and stability of 4.97%. With the advantage of high accuracy and sensitivity, easy handling, and low user training requirements, this ECL-LFI test might be used as point-of-care testing (POCT) in the IGRA for tuberculosis diagnosis.
Collapse
Affiliation(s)
- Shichao Yuan
- Department of Basic Medical Research, General Hospital of Southern Theater Command of PLA, Guangzhou, China
| | - Guihua Xie
- Guangzhou Leide Biotechnology Co, Ltd, Guangzhou, China
| | - Xiang Yang
- Guangzhou Leide Biotechnology Co, Ltd, Guangzhou, China
| | - Yu Chen
- Department of Basic Medical Research, General Hospital of Southern Theater Command of PLA, Guangzhou, China
| | - Hongbin Zhang
- Department of Basic Medical Research, General Hospital of Southern Theater Command of PLA, Guangzhou, China,*Correspondence: Hongbin Zhang,
| |
Collapse
|
21
|
Dumesic PA, Wilensky SE, Bose S, Van Vranken JG, Gygi SP, Spiegelman BM. RBM43 links adipose inflammation and energy expenditure through translational regulation of PGC1α. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.06.522985. [PMID: 36712038 PMCID: PMC9881917 DOI: 10.1101/2023.01.06.522985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Adipose thermogenesis involves specialized mitochondrial function that counteracts metabolic disease through dissipation of chemical energy as heat. However, inflammation present in obese adipose tissue can impair oxidative metabolism. Here, we show that PGC1α, a key governor of mitochondrial biogenesis and thermogenesis, is negatively regulated at the level of mRNA translation by the little-known RNA-binding protein RBM43. Rbm43 is expressed selectively in white adipose depots that have low thermogenic potential, and is induced by inflammatory cytokines. RBM43 suppresses mitochondrial and thermogenic gene expression in a PGC1α-dependent manner and its loss protects cells from cytokine-induced mitochondrial impairment. In mice, adipocyte-selective Rbm43 disruption increases PGC1α translation, resulting in mitochondrial biogenesis and adipose thermogenesis. These changes are accompanied by improvements in glucose homeostasis during diet-induced obesity that are independent of body weight. The action of RBM43 suggests a translational mechanism by which inflammatory signals associated with metabolic disease dampen mitochondrial function and thermogenesis.
Collapse
|
22
|
Three-in-one customized bioink for islet organoid: GelMA/ECM/PRP orchestrate pro-angiogenic and immunoregulatory function. Colloids Surf B Biointerfaces 2022; 221:113017. [DOI: 10.1016/j.colsurfb.2022.113017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/30/2022] [Accepted: 11/10/2022] [Indexed: 11/15/2022]
|
23
|
Kabat AM, Hackl A, Sanin DE, Zeis P, Grzes KM, Baixauli F, Kyle R, Caputa G, Edwards-Hicks J, Villa M, Rana N, Curtis JD, Castoldi A, Cupovic J, Dreesen L, Sibilia M, Pospisilik JA, Urban JF, Grün D, Pearce EL, Pearce EJ. Resident T H2 cells orchestrate adipose tissue remodeling at a site adjacent to infection. Sci Immunol 2022; 7:eadd3263. [PMID: 36240286 DOI: 10.1126/sciimmunol.add3263] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Type 2 immunity is associated with adipose tissue (AT) homeostasis and infection with parasitic helminths, but whether AT participates in immunity to these parasites is unknown. We found that the fat content of mesenteric AT (mAT) declined in mice during infection with a gut-restricted helminth. This was associated with the accumulation of metabolically activated, interleukin-33 (IL-33), thymic stromal lymphopoietin (TSLP), and extracellular matrix (ECM)-producing stromal cells. These cells shared transcriptional features, including the expression of Dpp4 and Pi16, with multipotent progenitor cells (MPC) that have been identified in numerous tissues and are reported to be capable of differentiating into fibroblasts and adipocytes. Concomitantly, mAT became infiltrated with resident T helper 2 (TH2) cells that responded to TSLP and IL-33 by producing stromal cell-stimulating cytokines, including transforming growth factor β1 (TGFβ1) and amphiregulin. These TH2 cells expressed genes previously associated with type 2 innate lymphoid cells (ILC2), including Nmur1, Calca, Klrg1, and Arg1, and persisted in mAT for at least 11 months after anthelmintic drug-mediated clearance of infection. We found that MPC and TH2 cells localized to ECM-rich interstitial spaces that appeared shared between mesenteric lymph node, mAT, and intestine. Stromal cell expression of epidermal growth factor receptor (EGFR), the receptor for amphiregulin, was required for immunity to infection. Our findings point to the importance of MPC and TH2 cell interactions within the interstitium in orchestrating AT remodeling and immunity to an intestinal infection.
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.,Bloomberg Kimmel Institute and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alexandra Hackl
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - David E Sanin
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.,Bloomberg Kimmel Institute and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Patrice Zeis
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.,International Max Planck Research School for Molecular and Cellular Biology (IMPRS-MCB), Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany
| | - Katarzyna M Grzes
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.,Bloomberg Kimmel Institute and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Francesc Baixauli
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Ryan Kyle
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - George Caputa
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Joy Edwards-Hicks
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Matteo Villa
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Nisha Rana
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Jonathan D Curtis
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.,Bloomberg Kimmel Institute and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Angela Castoldi
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Jovana Cupovic
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Leentje Dreesen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maria Sibilia
- Institute of Cancer Research, Medical University of Vienna, Comprehensive Cancer Center, Borschkegasse 8a, Vienna A-1090, Austria
| | - J Andrew Pospisilik
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Joseph F Urban
- USDA, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, and Belstville Agricultural Research Service, Animal Parasitic Disease Laboratory, Beltsville, MD 20705, USA
| | - Dominic Grün
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.,Centre for Integrative Biological Signaling Studies (CIBSS), University of Freiburg, Freiburg 79104, Germany.,Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität, Würzburg 97078, Germany.,Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg 97080, Germany
| | - Erika L Pearce
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.,Bloomberg Kimmel Institute and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Edward J Pearce
- Max Planck Institute for Immunobiology and Epigenetics, Freiburg 79108, Germany.,Bloomberg Kimmel Institute and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Faculty of Biology, University of Freiburg, Freiburg 79104, Germany.,Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| |
Collapse
|
24
|
Mathur N, Severinsen MCK, Jensen ME, Naver L, Schrölkamp M, Laye MJ, Watt MJ, Nielsen S, Krogh-Madsen R, Pedersen BK, Scheele C. Human visceral and subcutaneous adipose stem and progenitor cells retain depot-specific adipogenic properties during obesity. Front Cell Dev Biol 2022; 10:983899. [PMID: 36340033 PMCID: PMC9629396 DOI: 10.3389/fcell.2022.983899] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/03/2022] [Indexed: 11/29/2022] Open
Abstract
Abdominal obesity associates with cardiometabolic disease and an accumulation of lipids in the visceral adipose depot, whereas lipid accumulation in the subcutaneous depot is more benign. We aimed to further investigate whether the adipogenic properties where cell-intrinsic, or dependent on a depot-specific or obesity-produced microenvironment. We obtained visceral and subcutaneous biopsies from non-obese women (n = 14) or women living with morbid obesity (n = 14) and isolated adipose stem and progenitor cells (ASPCs) from the stromal vascular fraction of non-obese (n = 13) and obese (n = 13). Following in vitro differentiation into mature adipocytes, we observed a contrasting pattern with a lower gene expression of adipogenic markers and a higher gene expression of immunogenic markers in the visceral compared to the subcutaneous adipocytes. We identified the immunogenic factor BST2 as a marker for visceral ASPCs. The effect of obesity and insulin resistance on adipogenic and immunogenic markers in the in vitro differentiated cells was minor. In contrast, differentiation with exogenous Tumor necrosis factor resulted in increased immunogenic signatures, including increased expression of BST2, and decreased adipogenic signatures in cells from both depots. Our data, from 26 women, underscore the intrinsic differences between human visceral and subcutaneous adipose stem and progenitor cells, suggest that dysregulation of adipocytes in obesity mainly occurs at a post-progenitor stage, and highlight an inflammatory microenvironment as a major constraint of human adipogenesis.
Collapse
Affiliation(s)
- Neha Mathur
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Neha Mathur, ; Mai C. K. Severinsen, ; Camilla Scheele,
| | - Mai C. K. Severinsen
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Neha Mathur, ; Mai C. K. Severinsen, ; Camilla Scheele,
| | - Mette E. Jensen
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Naver
- Department of Gastroenterology, Hvidovre Hospital, Hvidovre, Denmark
| | - Maren Schrölkamp
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matthew J. Laye
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Matthew J. Watt
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Søren Nielsen
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Krogh-Madsen
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Klarlund Pedersen
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Scheele
- The Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Neha Mathur, ; Mai C. K. Severinsen, ; Camilla Scheele,
| |
Collapse
|
25
|
Zhang N, Peng Y, Zhao L, He P, Zhu J, Liu Y, Liu X, Liu X, Deng G, Zhang Z, Feng M. Integrated Analysis of Gut Microbiome and Lipid Metabolism in Mice Infected with Carbapenem-Resistant Enterobacteriaceae. Metabolites 2022; 12:metabo12100892. [PMID: 36295794 PMCID: PMC9609999 DOI: 10.3390/metabo12100892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/22/2022] Open
Abstract
The disturbance in gut microbiota composition and metabolism has been implicated in the process of pathogenic bacteria infection. However, the characteristics of the microbiota and the metabolic interaction of commensals−host during pathogen invasion remain more than vague. In this study, the potential associations of gut microbes with disturbed lipid metabolism in mice upon carbapenem-resistant Escherichia coli (CRE) infection were explored by the biochemical and multi-omics approaches including metagenomics, metabolomics and lipidomics, and then the key metabolites−reaction−enzyme−gene interaction network was constructed. Results showed that intestinal Erysipelotrichaceae family was strongly associated with the hepatic total cholesterol and HDL-cholesterol, as well as a few sera and fecal metabolites involved in lipid metabolism such as 24, 25-dihydrolanosterol. A high-coverage lipidomic analysis further demonstrated that a total of 529 lipid molecules was significantly enriched and 520 were depleted in the liver of mice infected with CRE. Among them, 35 lipid species showed high correlations (|r| > 0.8 and p < 0.05) with the Erysipelotrichaceae family, including phosphatidylglycerol (42:2), phosphatidylglycerol (42:3), phosphatidylglycerol (38:5), phosphatidylcholine (42:4), ceramide (d17:1/16:0), ceramide (d18:1/16:0) and diacylglycerol (20:2), with correlation coefficients higher than 0.9. In conclusion, the systematic multi-omics study improved the understanding of the complicated connection between the microbiota and the host during pathogen invasion, which thereby is expected to lead to the future discovery and establishment of novel control strategies for CRE infection.
Collapse
Affiliation(s)
- Ning Zhang
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Yuanyuan Peng
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Linjing Zhao
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Shanghai University of Engineering Science, Shanghai 201620, China
- Correspondence: ; Tel.: +86-21-6779-1214
| | - Peng He
- Minhang Hospital & School of Pharmacy, Fudan University, Shanghai 200433, China
- Shanghai Engineering Research Center of Immunotherapeutic, Shanghai 201203, China
| | - Jiamin Zhu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Yumin Liu
- Instrumental Analysis Centre, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xijian Liu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Xiaohui Liu
- School of Chemistry and Chemical Engineering, Shanghai Engineering Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Guoying Deng
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Zhong Zhang
- Nursing Department, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Meiqing Feng
- Minhang Hospital & School of Pharmacy, Fudan University, Shanghai 200433, China
- Shanghai Engineering Research Center of Immunotherapeutic, Shanghai 201203, China
| |
Collapse
|
26
|
Zhang H, Lu L, Zhao C, Liu Q, Zhou Q, Zhang Y, Pu Y, Wang S, Liu R, Yin L. Lipid metabolism disorders contribute to hepatotoxicity of ICR mice induced by nitrosamines exposure. ENVIRONMENT INTERNATIONAL 2022; 167:107423. [PMID: 35908391 DOI: 10.1016/j.envint.2022.107423] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/15/2022] [Accepted: 07/16/2022] [Indexed: 06/15/2023]
Abstract
Health risks caused by crucial environmental carcinogens N-nitrosamines triggered ubiquitous attention. As the liver exerted vital function through metabolic process, lipid metabolism disorders have been confirmed as potential drivers for toxicological effects, and the mechanisms of lipid regulation related to hepatotoxicity induced by N-nitrosamines remained largely unclear. In this study, we comprehensively explored the disturbance of hepatic lipid homeostasis in mice induced by nitrosamines. The results implied that nitrosamines exposure induced hepatotoxicity accompanied by liver injury, inflammatory infiltration, and hepatic edema. Lipidomics profiling analysis indicated the decreased levels of phosphatidic acids (PA), phosphatidylcholines (PC), phosphatidylethanolamines (PE), lyso-phosphatidylcholines (LPC), lyso-phosphatidylethanolamines (LPE), diacylglycerols (DAG) and triacylglycerols (TAG), the elevation of ceramides (Cer) and decomposition of free fatty acids (FFA) in high-dose nitrosamines exposure group. Importantly, nitrosamines exposure promoted fatty acid oxidation (FAO) by facilitating fatty acid uptake and decomposition, together with the upregulation of genes associated with FAO accompanied by the activation of inflammatory cytokines TNF-α, IL-1β and NLRP3. Furthermore, fatty acid translocase CD36-mediated fatty acid oxidation was correlated with the enhancement of oxidative stress in the liver caused by nitrosamines exposure. Overall, our results contributed to the new strategies to interpret the early toxic effects of nitrosamines exposure.
Collapse
Affiliation(s)
- Hu Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Lu Lu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Chao Zhao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Qiwei Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Qian Zhou
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Ying Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Shizhi Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
27
|
Kobayashi T, Brenner D. A FAsT contribution: Adipocytes rewire their metabolism to acquire immune functions. Cell Metab 2022; 34:656-657. [PMID: 35508107 DOI: 10.1016/j.cmet.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Adipose tissue has been linked to inflammation and various physiological processes. In this issue of Cell Metabolism, Caputa et al. describe that perinodal adipocytes adapt their metabolism to actively participate in an immune response against intracellular Listeria monocytogenes.
Collapse
Affiliation(s)
- Takumi Kobayashi
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology and Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Immunology and Genetics, Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 Avenue des Hauts Fourneaux, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis (ORCA), Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|