ザクロ由来化合物が免疫老化を逆転―新たな免疫強化法(Pomegranate-Derived Compound Strengthens Immune Defense)

ad

2025-11-03 ゲーテ大学

ゲーテ大学フランクフルトとゲオルク・シュパイヤー研究所のフロリアン・グレーテン教授、ドミニク・デンク医師らは、ザクロ由来成分「ウロリチンA」が加齢による免疫老化を逆転させることをヒト臨床試験で実証した。『Nature Aging』に掲載された第Ⅰ相試験では、50名の中年成人に4週間ウロリチンAまたはプラセボを投与。ミトコンドリアの再生を促す「マイトファジー」を活性化することで、免疫の“若返り細胞”であるナイーブT細胞が顕著に増加し、免疫応答が強化された。被験者では細菌排除能や免疫記憶形成も向上し、安全性も確認された。研究チームは以前から、ウロリチンAが抗腫瘍性T細胞を活性化し大腸がん治療を改善することを報告しており、今回の成果はその臨床応用の第一歩となる。現在、免疫療法中のがん患者を対象にした後続試験が進行中であり、老化・感染症・がん治療の新戦略として期待されている。

<関連情報>

マイトファジー誘導剤ウロリチンAの加齢性免疫低下に対する効果:ランダム化プラセボ対照試験 Effect of the mitophagy inducer urolithin A on age-related immune decline: a randomized, placebo-controlled trial

Dominic Denk,Anurag Singh,Herbert G. Kasler,Davide D’Amico,Julia Rey,Lucía Alcober-Boquet,Johanna M. Gorol,Christoph Steup,Ritesh Tiwari,Ryan Kwok,Rafael J. Argüello,Julie Faitg,Kathrin Sprinzl,Stefan Zeuzem,Valentina Nekljudova,Sibylle Loibl,Eric Verdin,Chris Rinsch & Florian R. Greten
Nature Aging  Published:31 October 2025
DOI:https://doi.org/10.1038/s43587-025-00996-x

Fig. 2

Abstract

Mitochondrial dysfunction and stem cell exhaustion contribute to age-related immune decline, yet clinical interventions targeting immune aging are lacking. Recently, we demonstrated that urolithin A (UA), a mitophagy inducer, expands T memory stem cells (TSCM) and naive T cells in mice. In this randomized, double-blind, placebo-controlled trial, 50 healthy middle-aged adults received oral UA (1,000 mg day-1) or placebo for 4 weeks; time points of analysis were baseline and day 28. Primary outcomes were phenotypical changes in peripheral CD3+ T cell subsets and immune metabolic remodeling. UA expanded peripheral naive-like, less terminally exhausted CD8+ cells (treatment difference 0.50 percentage points; 95% CI = 0.16 to 0.83; P = 0.0437) while also increasing CD8+ fatty acid oxidation capacity (treatment difference = 14.72 percentage points; 95% confidence interval (CI) = 6.46 to 22.99; P = 0.0061). Secondary outcomes included changes in plasma cytokine levels (IL-6, TNF, IL-1β, IL-10), immune populations assessed via flow cytometry, immune cell function, and mitochondrial content. Analysis revealed augmented mitochondrial biogenesis in CD8+ cells, increased peripheral CD56dimCD16bright NK cells, and nonclassical CD14loCD16hi monocytes in UA-treated participants, as well as improved activation-elicited TNF secretion in T cells and bacterial uptake by monocytes. Exploratory single-cell RNA sequencing demonstrated UA-driven transcriptional shifts across immune populations, modulating pathways linked to inflammation and metabolism. These findings indicate that short-term UA supplementation modulates human immune cell composition and function, supporting its potential to counteract age-related immune decline and inflammaging. ClinicalTrials.gov registration number: NCT05735886.

 

ウロリチンA誘導マイトファジーによる優れた抗腫瘍免疫力を持つT記憶幹細胞の増殖 Expansion of T memory stem cells with superior anti-tumor immunity by Urolithin A-induced mitophagy

Dominic Denk ∙ Valentina Petrocelli ∙ Claire Conche ∙ … ∙ Pénélope A. Andreux ∙ Chris Rinsch ∙ Florian R. Greten
Immunity  Published:October 18, 2022
DOI:https://doi.org/10.1016/j.immuni.2022.09.014

Highlights

  • UA induces Pink1-dependent mitophagy in CD8+ cells causing PGAM5 release
  • Cytosolic PGAM5 augments Wnt signaling to drive TSCM formation
  • TSCM induction augments anti-tumor immunity in vivo
  • Urolithin can be used for ex vivo expansion of CAR-expressing TSCM

Graphical abstract undfig1

Summary

T memory stem cells (TSCM) display increased self-renewal and prolonged survival capabilities, thus preventing T cell exhaustion and promoting effective anti-tumor T cell responses. TSCM cells can be expanded by Urolithin A (UA), which is produced by the commensal gut microbiome from foods rich in ellagitannins and is known to improve mitochondrial health. Oral UA administration to tumor-bearing mice conferred strong anti-tumor CD8+ T cell immunity, whereas ex vivo UA pre-treated T cells displayed improved anti-tumor function upon adoptive cell transfer. UA-induced TSCM formation depended on Pink1-mediated mitophagy triggering cytosolic release of the mitochondrial phosphatase Pgam5. Cytosolic Pgam5 dephosphorylated β-catenin, which drove Wnt signaling and compensatory mitochondrial biogenesis. Collectively, we unravel a critical signaling pathway linking mitophagy to TSCM formation and suggest that the well-tolerated metabolic compound UA represents an attractive option to improve immune therapy.

医療・健康
ad
ad
Follow
ad
タイトルとURLをコピーしました