دورية أكاديمية

Early Immunomodulatory Program Triggered by Protolerogenic Bifidobacterium pseudolongum Drives Cardiac Transplant Outcomes.

التفاصيل البيبلوغرافية
العنوان: Early Immunomodulatory Program Triggered by Protolerogenic Bifidobacterium pseudolongum Drives Cardiac Transplant Outcomes.
المؤلفون: Gavzy SJ; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD.; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD., Kensiski A; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Saxena V; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Lakhan R; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Hittle L; University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD., Wu L; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Iyyathurai J; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Dhakal H; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Lee ZL; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Li L; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD.; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD., Lee YS; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD.; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD., Zhang T; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD.; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD., Lwin HW; University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD., Shirkey MW; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Paluskievicz CM; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD.; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD., Piao W; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD., Mongodin EF; University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD., Ma B; University of Maryland School of Medicine, Institute for Genome Sciences, Baltimore, MD.; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD., Bromberg JS; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD.; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD.; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD.
المصدر: Transplantation [Transplantation] 2024 Jul 01; Vol. 108 (7), pp. e91-e105. Date of Electronic Publication: 2024 Apr 08.
نوع المنشور: Journal Article
اللغة: English
بيانات الدورية: Publisher: Lippincott Williams & Wilkins Country of Publication: United States NLM ID: 0132144 Publication Model: Print-Electronic Cited Medium: Internet ISSN: 1534-6080 (Electronic) Linking ISSN: 00411337 NLM ISO Abbreviation: Transplantation Subsets: MEDLINE
أسماء مطبوعة: Publication: Hagerstown, MD : Lippincott Williams & Wilkins
Original Publication: Baltimore, Williams & Wilkins.
مواضيع طبية MeSH: Heart Transplantation*/adverse effects , Gastrointestinal Microbiome*/immunology , Bifidobacterium* , Graft Rejection*/immunology , Graft Rejection*/microbiology , Graft Rejection*/prevention & control, Animals ; Male ; Time Factors ; Graft Survival ; Dysbiosis ; Mice, Inbred C57BL ; Immunity, Innate ; Immunomodulation ; Phenotype ; Probiotics/therapeutic use ; Lymph Nodes/microbiology ; Lymph Nodes/immunology
مستخلص: Background: Despite ongoing improvements to regimens preventing allograft rejection, most cardiac and other organ grafts eventually succumb to chronic vasculopathy, interstitial fibrosis, or endothelial changes, and eventually graft failure. The events leading to chronic rejection are still poorly understood and the gut microbiota is a known driving force in immune dysfunction. We previously showed that gut microbiota dysbiosis profoundly influences the outcome of vascularized cardiac allografts and subsequently identified biomarker species associated with these differential graft outcomes.
Methods: In this study, we further detailed the multifaceted immunomodulatory properties of protolerogenic and proinflammatory bacterial species over time, using our clinically relevant model of allogenic heart transplantation.
Results: In addition to tracing longitudinal changes in the recipient gut microbiome over time, we observed that Bifidobacterium pseudolongum induced an early anti-inflammatory phenotype within 7 d, whereas Desulfovibrio desulfuricans resulted in a proinflammatory phenotype, defined by alterations in leukocyte distribution and lymph node (LN) structure. Indeed, in vitro results showed that B pseudolongum and D desulfuricans acted directly on primary innate immune cells. However, by 40 d after treatment, these 2 bacterial strains were associated with mixed effects in their impact on LN architecture and immune cell composition and loss of colonization within gut microbiota, despite protection of allografts from inflammation with B pseudolongum treatment.
Conclusions: These dynamic effects suggest a critical role for early microbiota-triggered immunologic events such as innate immune cell engagement, T-cell differentiation, and LN architectural changes in the subsequent modulation of protolerant versus proinflammatory immune responses in organ transplant recipients.
Competing Interests: This work was supported by the National Institute of Health (NIH), National Heart, Lung, and Blood Institute award R01HL148672 (J.S.B./B.M.), U01AI170050 (B.M./J.S.B.), and National Institute of Allergy and Infectious Diseases training grant T32AI95190-10 (S.J.G.). An earlier version of this article has been released as a preprint on bioRxiv ( https://doi.org/10.1101/2022.10.13.511915 ). E.F.M. contributed to this work as an employee of the University of Maryland School of Medicine. The views expressed in this article are his own and do not necessarily represent the views of the NIH or the US Government. The other authors declare no conflicts of interest.
(Copyright © 2024 The Author(s). Published by Wolters Kluwer Health, Inc.)
References: Gago M, Cornell LD, Kremers WK, et al. Kidney allograft inflammation and fibrosis, causes and consequences. Am J Transplant. 2012;12:1199–1207.
Israni AK, Leduc R, Jacobson PA, et al.; DeKAF Genomics Investigators. Inflammation in the setting of chronic allograft dysfunction post-kidney transplant: phenotype and genotype. Clin Transplant. 2013;27:348–358.
Moreso F, Seron D, O'Valle F, et al. Immunephenotype of glomerular and interstitial infiltrating cells in protocol renal allograft biopsies and histological diagnosis. Am J Transplant. 2007;7:2739–2747.
Asleh R, Briasoulis A, Pereira NL, et al. Hypercholesterolemia after conversion to sirolimus as primary immunosuppression and cardiac allograft vasculopathy in heart transplant recipients. J Heart Lung Transplant. 2018;37:1372–1380.
Jansen MA, Otten HG, de Weger RA, et al. Immunological and fibrotic mechanisms in cardiac allograft vasculopathy. Transplantation. 2015;99:2467–2475.
Kim M, Bergmark BA, Zelniker TA, et al. Early aspirin use and the development of cardiac allograft vasculopathy. J Heart Lung Transplant. 2017;36:1344–1349.
Mehra MR. The scourge and enigmatic journey of cardiac allograft vasculopathy. J Heart Lung Transplant. 2017;36:1291–1293.
Lamb KE, Lodhi S, Meier-Kriesche HU. Long-term renal allograft survival in the United States: a critical reappraisal. Am J Transplant. 2011;11:450–462.
Sellares J, de Freitas DG, Mengel M, et al. Inflammation lesions in kidney transplant biopsies: association with survival is due to the underlying diseases. Am J Transplant. 2011;11:489–499.
Koren O, Spor A, Felin J, et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4592–4598.
Wang D, Xia M, Yan X, et al. Gut microbiota metabolism of anthocyanin promotes reverse cholesterol transport in mice via repressing miRNA-10b Circ Res. 2012;111:967–981.
Atkinson MA, Chervonsky A. Does the gut microbiota have a role in type 1 diabetes? Early evidence from humans and animal models of the disease. Diabetologia. 2012;55:2868–2877.
Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012;336:1268–1273.
Shankar J, Nguyen MH, Crespo MM, et al. Looking beyond respiratory cultures: microbiome-cytokine signatures of bacterial pneumonia and tracheobronchitis in lung transplant recipients. Am J Transplant. 2016;16:1766–1778.
Kakihana K, Fujioka Y, Suda W, et al. Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut. Blood. 2016;128:2083–2088.
Lei YM, Chen L, Wang Y, et al. The composition of the microbiota modulates allograft rejection. J Clin Invest. 2016;126:2736–2744.
Hossain MS, Jaye DL, Pollack BP, et al. Flagellin, a TLR5 agonist, reduces graft-versus-host disease in allogeneic hematopoietic stem cell transplantation recipients while enhancing antiviral immunity. J Immunol. 2011;187:5130–5140.
Jenq RR, Ubeda C, Taur Y, et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J Exp Med. 2012;209:903–911.
Oh PL, Martínez I, Sun Y, et al. Characterization of the ileal microbiota in rejecting and nonrejecting recipients of small bowel transplants. Am J Transplant. 2012;12:753–762.
Fricke WF, Maddox C, Song Y, et al. Human microbiota characterization in the course of renal transplantation. Am J Transplant. 2014;14:416–427.
Dickson RP, Erb-Downward JR, Freeman CM, et al. Changes in the lung microbiome following lung transplantation include the emergence of two distinct Pseudomonas species with distinct clinical associations. PLoS One. 2014;9:e97214.
Lee JR, Muthukumar T, Dadhania D, et al. Gut microbiota and tacrolimus dosing in kidney transplantation. PLoS One. 2015;10:e0122399.
Lee JR, Muthukumar T, Dadhania D, et al. Gut microbial community structure and complications after kidney transplantation: a pilot study. Transplantation. 2014;98:697–705.
Lemahieu W, Maes B, Verbeke K, et al. Cytochrome P450 3A4 and P-glycoprotein activity and assimilation of tacrolimus in transplant patients with persistent diarrhea. Am J Transplant. 2005;5:1383–1391.
Willner DL, Hugenholtz P, Yerkovich ST, et al. Reestablishment of recipient-associated microbiota in the lung allograft is linked to reduced risk of bronchiolitis obliterans syndrome. Am J Respir Crit Care Med. 2013;187:640–647.
Ma B, Gavzy SJ, France M, et al. Rapid intestinal and systemic metabolic reprogramming in an immunosuppressed environment. BMC Microbiol. 2023;23:394.
Dethlefsen L, Relman DA. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci USA. 2011;108(Suppl 1):4554–4561.
von Rosenvinge EC, Song Y, White JR, et al. Immune status, antibiotic medication and pH are associated with changes in the stomach fluid microbiota. ISME J. 2013;7:1354–1366.
Vrbanac A, Patras KA, Jarmusch AK, et al. Evaluating organism-wide changes in the metabolome and microbiome following a single dose of antibiotic. mSystems 2020;5:e00340–e00320.
Bromberg JS, Hittle L, Xiong Y, et al. Gut microbiota-dependent modulation of innate immunity and lymph node remodeling affects cardiac allograft outcomes. JCI Insight 2018;3:e121045.
Ma B, Gavzy SJ, Saxena V, et al. Strain-specific alterations in gut microbiome and host immune responses elicited by tolerogenic Bifidobacterium pseudolongum . Sci Rep. 2023;13:1023.
Mongodin EF, Hittle LL, Nadendla S, Brinkman CC, Xiong Y, Bromberg JS. Complete genome sequence of a strain of Bifidobacterium pseudolongum isolated from mouse feces and associated with Improved Organ Transplant Outcome. Genome Announc. 2017;5:e01089–17. doi: 10.1128/genomeA.01089-17. (PMID: 10.1128/genomeA.01089-17)
Shannon CE. A mathematical theory of communication. ACM SIGMOBILE Mob Comput Commun Rev 2001;5:3–55.
Ormerod KL, Wood DLA, Lachner N, et al. Genomic characterization of the uncultured Bacteroidales family S24-7 inhabiting the guts of homeothermic animals. Microbiome 2016;4:36.
Everard A, Belzer C, Geurts L, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci USA. 2013;110:9066–9071.
Mongodin EF, Hittle LL, Nadendla S, et al. Complete genome sequence of a strain of Bifidobacterium pseudolongum isolated from mouse feces and associated with improved organ transplant outcome. Genome Announc 2017;5:e01089–e01017.
Kim M, Galan C, Hill AA, et al. Critical role for the microbiota in CX(3)CR1(+) intestinal mononuclear phagocyte regulation of intestinal T cell responses. Immunity. 2018;49:151–163.e5.
Peterson LW, Artis D. Intestinal epithelial cells: regulators of barrier function and immune homeostasis. Nat Rev Immunol. 2014;14:141–153.
Qian S, Li W, Li Y, et al. Systemic administration of cellular interleukin-10 can exacerbate cardiac allograft rejection in mice. Transplantation. 1996;62:1709–1714.
Swarte JC, Li Y, Hu S, et al. Gut microbiome dysbiosis is associated with increased mortality after solid organ transplantation. Sci Transl Med. 2022;14:eabn7566.
Sorbara MT, Pamer EG. Interbacterial mechanisms of colonization resistance and the strategies pathogens use to overcome them. Mucosal Immunol 2019;12:1–9.
Maldonado-Gomez MX, Martínez I, Bottacini F, et al. Stable engraftment of Bifidobacterium longum AH1206 in the human gut depends on individualized features of the resident microbiome. Cell Host Microbe. 2016;20:515–526.
Gabarre P, Loens C, Tamzali Y, et al. Immunosuppressive therapy after solid organ transplantation and the gut microbiota: bidirectional interactions with clinical consequences. Am J Transplant. 2022;22:1014–1030.
Chiu L, Bazin T, Truchetet M-E, et al. Protective microbiota: from localized to long-reaching co-immunity. Front Immunol. 2017;8.
Honda M, Surewaard BGJ, Watanabe M, et al. Perivascular localization of macrophages in the intestinal mucosa is regulated by Nr4a1 and the microbiome. Nat Commun. 2020;11.
Chyou S, Benahmed F, Chen J, et al. Coordinated regulation of lymph node vascular-stromal growth first by CD11c+ cells and then by T and B cells. J Immunol. 2011;187:5558–5567.
Kumar V, Dasoveanu DC, Chyou S, et al. A dendritic-cell-stromal axis maintains immune responses in lymph nodes. Immunity. 2015;42:719–730.
Saito Y, Respatika D, Komori S, et al. SIRPalpha(+) dendritic cells regulate homeostasis of fibroblastic reticular cells via TNF receptor ligands in the adult spleen. Proc Natl Acad Sci USA. 2017;114:E10151–E10160.
Komori S, Saito Y, Respatika D, et al. SIRPalpha(+) dendritic cells promote the development of fibroblastic reticular cells in murine peripheral lymph nodes. Eur J Immunol. 2019;49:1364–1371.
Warren KJ, Iwami D, Harris DG, et al. Laminins affect T cell trafficking and allograft fate. J Clin Invest. 2014;124:2204–2218.
Li L, Shirkey MW, Zhang T, et al. The lymph node stromal laminin alpha5 shapes alloimmunity. J Clin Invest. 2020;130:2602–2619.
Simon T, Li L, Wagner C, et al. Differential regulation of T-cell immunity and tolerance by stromal laminin expressed in the lymph node. Transplantation. 2019;103:2075–2089.
معلومات مُعتمدة: R01 AI114496 United States AI NIAID NIH HHS; R01 HL148672 United States HL NHLBI NIH HHS; T32 AI095190 United States AI NIAID NIH HHS; U01 AI170050 United States AI NIAID NIH HHS
تواريخ الأحداث: Date Created: 20240408 Date Completed: 20240625 Latest Revision: 20240627
رمز التحديث: 20240627
مُعرف محوري في PubMed: PMC11188630
DOI: 10.1097/TP.0000000000004939
PMID: 38587506
قاعدة البيانات: MEDLINE
الوصف
تدمد:1534-6080
DOI:10.1097/TP.0000000000004939