Immunity
Volume 55, Issue 9, 13 September 2022, Pages 1594-1608.e6
Journal home page for Immunity

Article
Renal NF-κB activation impairs uric acid homeostasis to promote tumor-associated mortality independent of wasting

https://doi.org/10.1016/j.immuni.2022.07.022Get rights and content

Highlights

  • Fly yki3SA-gut tumors cause bacterial overload via suppression of gut PGRP-SC2

  • Gut bacterial clearance increases survival of yki3SA-tumor-bearing flies

  • Renal-specific IMD-NF-κB blockade increases survival of yki3SA-tumor-bearing flies

  • Renal IMD-NF-κB blockade diminishes yki3SA-tumor-associated uric acid accumulation

Summary

Tumor-induced host wasting and mortality are general phenomena across species. Many groups have previously demonstrated endocrinal impacts of malignant tumors on host wasting in rodents and Drosophila. Whether and how environmental factors and host immune response contribute to tumor-associated host wasting and survival, however, are largely unknown. Here, we report that flies bearing malignant yki3SA-gut tumors exhibited the exponential increase of commensal bacteria, which were mostly acquired from the environment, and systemic IMD-NF-κB activation due to suppression of a gut antibacterial amidase PGRP-SC2. Either gut microbial elimination or specific IMD-NF-κB blockade in the renal-like Malpighian tubules potently improved mortality of yki3SA-tumor-bearing flies in a manner independent of host wasting. We further indicate that renal IMD-NF-κB activation caused uric acid (UA) overload to reduce survival of tumor-bearing flies. Therefore, our results uncover a fundamental mechanism whereby gut commensal dysbiosis, renal immune activation, and UA imbalance potentiate tumor-associated host death.

Introduction

Cancer cachexia, or tumor-induced host wasting, is a process characterized by body weight loss, atrophy of skeletal and adipose tissue, hyperglycemia, and mortality (Fearon et al., 2013; Tisdale, 2002). Due to the unclear pathogenic mechanisms, different model organisms from rodents to Drosophila have been used to mimic the wasting development and address the unmet clinical needs. Recent studies using tumor-bearing mice have demonstrated the cachectic roles of circulating secreted proteins, such as activins, myostatin, transforming growth factor β (TGF-β), parathyroid hormone-related protein (PTHrP), interleukin-6 (IL-6), IL-1, tumor necrosis factor α (TNF-α), and interferon γ (IFN-γ), in mediating host organ wasting in response to tumor growth (Ballaro et al., 2016; Baracos et al., 2018). However, most of these mice studies were performed in specific-pathogen-free (SPF) conditions and ignored the potential participation of environmental microorganisms such as bacteria, fungi, and viruses that inevitably interact with the host to modulate host immune response and physiological outputs. Whether and how environmental factors and host immune response contribute to tumor-induced organ wasting or mortality are largely unknown.

Drosophila has emerged as an evolutionarily conserved model organism to investigate the mechanism of cancer cachexia in the past years. We have established that induction of an active transcription factor yki3SA, a homolog of the human oncogene YAP1, in intestinal stem cells (ISCs) leads to overproliferation of gut tumor cells and subsequent wasting phenotypes, including muscle dysfunction, lipid loss, hyperglycemia, and mortality (Kwon et al., 2015). We further characterized the molecular mechanisms through which malignant yki3SA-gut-tumor cells produce cachectic ligands, including ImpL2, Pvf1, and Upd3, to remotely impair anabolism-catabolism balance in host organs via modulation of insulin, MEK, and Jak/Stak signaling pathways, causing wasting phenotypes (Ding et al., 2021; Kwon et al., 2015; Song et al., 2019). Other groups using different tumor models in Drosophila and mice also observe similar cachectic roles of tumor-secreted proteins in the wasting development (Dong et al., 2021; Figueroa-Clarevega and Bilder, 2015; Kandarian et al., 2018; Newton et al., 2020; Nie et al., 2019; Yeom et al., 2021). These findings collectively demonstrate that tumor-induced wasting is a general phenomenon and that Drosophila can be used to dissect the molecular mechanisms involved in tumor-host interaction. Moreover, different from SPF mice, tumor-bearing flies are cultured at a less-restricted condition with direct interaction with natural air and external microorganisms and provide a convenient tool to investigate microbial-host interaction in the context of malignant tumor growth.

Drosophila has also emerged as an excellent model to conduct genetic analysis of microbial recognition, immune defense, and host physiological perturbation (Buchon et al., 2014; Capo et al., 2019; Charroux and Royet, 2012; Lee and Brey, 2013) due to the simpler gut commensal communities and conserved immune regulation. Two innate immune NF-κB pathways, Toll and immune deficiency (IMD), have been identified as the major signaling cascades to regulate the humoral reactions in response to the diverse bacteria and fungi (Lemaitre and Hoffmann, 2007). The Toll-NF-κB signaling is triggered by Gram-positive bacterial Lysine-type peptidoglycans (PGNs) and fungal β-(1,3)-glucan via binding to peptidoglycan recognition proteins (PGRPs) and glucan-binding proteins (GNBPs), respectively, to cleave cytokine Spaetzle (Spz) and activate Toll receptor as well as the Dorsal-Dif transcriptional complex (Ferrandon et al., 2007). On the other hand, Gram-negative bacterial DAP-type PGNs trigger IMD-NF-κB signaling through interaction with PGRP-LC to activate IMD and the downstream transcriptional factor Relish (Rel) (Ferrandon et al., 2007). A subgroup of PGRPs, such as PGRP-LB with the amidase enzymatic activity, have been found to degrade PGNs and suppress the immune responses upon infection (Charroux et al., 2018; Zaidman-Remy et al., 2006). Activated Toll and IMD pathways in multiple tissues like gut and fat body induce expression of anti-microbial peptide (AMP) genes such as Drosomycin (Drs) and Diptericin (Dpt), respectively, and others to restrain the microbial growth in a feedback loop (Lemaitre and Hoffmann, 2007; Royet and Dziarski, 2007). In addition, activation of NF-κB pathways in Drosophila also involves regulation of energy balance and host death (Buchon et al., 2014; Garschall and Flatt, 2018; Molaei et al., 2019), the major features of tumor-induced host wasting or mortality. It raises a hypothesis whereby systemic immune response plays pathogenic roles in the context of tumor growth.

In this study, we observed a robust systemic activation of IMD-NF-κB signaling in yki3SA-tumor bearing flies due to suppressed production of gut PGRP-SC2, a key antibacterial amidase characterized by us, and increased abundance of commensal bacteria. We found that bacteria-induced IMD-NF-κB activation in the renal organ impaired UA metabolism to decrease survival of yki3SA flies without affecting tumor growth or energy wasting. Thus, our results indicate that gut-microbial dysbiosis and renal-IMD-NF-κB activation promote mortality independent of host wasting in yki3SA-tumor-bearing flies.

Section snippets

yki3SA-tumor-bearing flies exhibit systemic activation of IMD-NF-κB signaling and gut microbial overload

In a previous study, our RNA sequencing (RNA-seq) data have shown that AMP-encoding genes are up-regulated in the muscle of yki3SA-tumor-bearing flies compared with control flies (Figure 1A; Ding et al., 2021). qPCR analysis in whole flies also showed that mRNA amounts of AMPs downstream IMD-NF-κB signaling such as DptA, DptB, AttA, AttD, but not Toll-NF-κB signaling like Drs, were increased by >10-fold (Figure 1B), suggesting the activation of systemic IMD-NF-κB signaling in yki3SA flies. As

Discussion

Whether and how environmental factors and host immune response participate in wasting development and mortality regulation in tumor-bearing subjects is a missing piece in the cancer-cachexia field. Using Drosophila as a model organism to study gut bacteria that are mostly acquired from the environment, we establish that yki3SA-gut-bearing tumors suppressed the production of antibacterial-amidase PGRP-SC2 and unleashed bacterial growth to activate IMD-NF-κB signaling specifically in the renal

Key resources table

REAGENT or RESOURCESOURCEIDENTIFIER
Bacterial strains
Escherichia coli (E. coli)VazymeCat#C502
Erwinia carotovora carotovora15 (Ecc15)(Basset et al., 2000)N/A
Enterococcus faecalis (E. faecalis)(Wang et al., 2022)N/A
Salmonella typhimurium (S. typhimurium)(Kroger et al., 2012)N/A
Micrococcus luteus (M. luteus)CGMCCCat#1.9051
Staphylococcus aureus (S. aureus)(Qiu et al., 2022)N/A
Pseudomonas aeruginosa (P. aeruginosa)CCTCCCat#AB2010174
Pseudomonas putida (P. putida)CCTCCCat#AB2014017

Acknowledgments

We thank the TsingHua Fly Center, TRiP at Harvard Medical School, Bloomington Drosophila Stock Center, and NIG in Japan for providing fly stocks; Lei Pan (Institute Pasteur of Shanghai) and Yingying Pu (Wuhan University) for bacteria strains; Heinrich Jasper (BUCK Institute) for UAS-PGRP-SC2 flies; Pankaj Kapahi‬ (BUCK Institute) for c42-GAL4 flies; Zongzhao Zhai (Hunan Normal University) for RelE20 and PGRP-LCE12 flies; Yong Liu and Hong-bing Shu (Wuhan University) for comments and

References (63)

  • I. Kounatidis et al.

    NF-kappaB Immunity in the Brain Determines Fly Lifespan in Healthy Aging and Age-Related Neurodegeneration

    Cell Rep.

    (2017)
  • Y. Kwon et al.

    Systemic organ wasting induced by localized expression of the secreted insulin/IGF antagonist ImpL2

    Dev. Cell

    (2015)
  • X. Li et al.

    Renal Purge of Hemolymphatic Lipids Prevents the Accumulation of ROS-Induced Inflammatory Oxidized Lipids and Protects Drosophila from Tissue Damage

    Immunity

    (2020)
  • M. Martin et al.

    Analysis of Drosophila STING Reveals an Evolutionarily Conserved Antimicrobial Function

    Cell Rep.

    (2018)
  • P. Mellroth et al.

    A scavenger function for a Drosophila peptidoglycan recognition protein

    J. Biol. Chem.

    (2003)
  • M. Molaei et al.

    NF-kappaB Shapes Metabolic Adaptation by Attenuating Foxo-Mediated Lipolysis in Drosophila

    Dev. Cell

    (2019)
  • J.C. Paredes et al.

    Negative regulation by amidase PGRPs shapes the Drosophila antibacterial response and protects the fly from innocuous infection

    Immunity

    (2011)
  • K. Petkau et al.

    Constitutive Immune Activity Promotes Tumorigenesis in Drosophila Intestinal Progenitor Cells

    Cell Rep.

    (2017)
  • H.S. Shin et al.

    Uric acid as a prognostic factor for survival time: a prospective cohort study of terminally ill cancer patients

    J. Pain Symptom Manage.

    (2006)
  • W. Song et al.

    Tumor-Derived Ligands Trigger Tumor Growth and Host Wasting via Differential MEK Activation

    Dev. Cell

    (2019)
  • K. Troha et al.

    Nephrocytes Remove Microbiota-Derived Peptidoglycan from Systemic Circulation to Maintain Immune Homeostasis

    Immunity

    (2019)
  • E. van Dam et al.

    Sugar-Induced Obesity and Insulin Resistance Are Uncoupled from Shortened Survival in Drosophila

    Cell Metab.

    (2020)
  • A. Zaidman-Rémy et al.

    The Drosophila amidase PGRP-LB modulates the immune response to bacterial infection

    Immunity

    (2006)
  • Z. Zhai et al.

    Cell-Specific Imd-NF-kappaB Responses Enable Simultaneous Antibacterial Immunity and Intestinal Epithelial Cell Shedding upon Bacterial Infection

    Immunity

    (2018)
  • X. Zhao et al.

    Glutamate metabolism directs energetic trade-offs to shape host-pathogen susceptibility in Drosophila

    Cell Metab.

    (2021)
  • R. Ballarò et al.

    Animal models for cancer cachexia

    Curr. Opin. Support. Palliat. Care

    (2016)
  • V.E. Baracos et al.

    Cancer-associated cachexia

    Nat. Rev. Dis. Primers

    (2018)
  • A. Basset et al.

    The phytopathogenic bacteria Erwinia carotovora infects Drosophila and activates an immune response

    Proc. Natl. Acad. Sci. USA

    (2000)
  • J. Bischof et al.

    An optimized transgenesis system for Drosophila using germ-line-specific phiC31 integrases

    Proc. Natl. Acad. Sci. USA

    (2007)
  • N. Buchon et al.

    Immunity in Drosophila melanogaster--from microbial recognition to whole-organism physiology

    Nat. Rev. Immunol.

    (2014)
  • F. Capo et al.

    The Intestine of Drosophila melanogaster: An Emerging Versatile Model System to Study Intestinal Epithelial Homeostasis and Host-Microbial Interactions in Humans

    Microorganisms

    (2019)
  • Cited by (10)

    View all citing articles on Scopus
    3

    Lead contact

    View full text