New Senomorphic Strategy for Age-Related Diseases: TNIK Inhibition

3 mins read
A collection of white and blue capsules arranged on a white background, with some blue capsules forming the letters "AI." The image represents the intersection of artificial intelligence and drug discovery, reflecting advancements in AI-driven pharmaceutical research, including the development of senomorphic therapeutics like TNIK inhibitors.

The Promise of Targeting Senescence in Aging and Disease

Aging is the most significant risk factor for numerous chronic diseases, including idiopathic pulmonary fibrosis (IPF), neurodegeneration, and cardiovascular diseases. Cellular senescence—a fundamental hallmark of aging—plays a critical role in driving these pathologies through the accumulation of non-proliferative, metabolically active cells that secrete pro-inflammatory molecules known as the senescence-associated secretory phenotype (SASP). While the complete eradication of senescent cells using senolytic drugs has shown promise, a complementary approach called senomorphics offers an alternative by modulating the harmful effects of SASP without inducing apoptosis.

One of the latest breakthroughs in the field of senotherapeutics is the discovery of Traf2- and Nck-interacting kinase (TNIK) inhibition as a novel senomorphic strategy. Recent studies have identified INS018_055, a small-molecule TNIK inhibitor, as a potent modulator of senescence, fibrosis, and aging-related inflammation. This article explores how AI-driven drug discovery led to the identification of TNIK as a key aging-related target and how TNIK inhibition may shape the future of longevity therapeutics.


Understanding Cellular Senescence and the Role of Senomorphics

Cellular senescence is a complex biological process in which cells enter a state of irreversible growth arrest due to various stressors, including DNA damage, telomere attrition, and oxidative stress. While senescence serves as a protective mechanism against tumorigenesis, it also contributes to aging and chronic diseases by disrupting tissue homeostasis through SASP secretion. Senotherapeutics are broadly classified into two categories: senolytics, which selectively eliminate senescent cells (e.g., dasatinib and quercetin, ABT-263), and senomorphics, which modulate SASP and reduce senescence-related inflammation without inducing cell death (e.g., rapamycin, metformin). TNIK inhibition represents a novel senomorphic strategy that aims to reprogram senescent cells to a less inflammatory state, thereby mitigating tissue dysfunction without the risks associated with senolytic therapies.


AI-Driven Discovery of TNIK as a Senomorphic Target

Recent advancements in artificial intelligence (AI)-driven drug discovery have transformed the way aging-related targets are identified. Using AI-powered platforms, researchers pinpointed TNIK as a critical player in multiple aging-associated pathways. TNIK plays a significant role in TGF-β signaling, a major driver of fibrosis and senescence; Wnt/β-catenin signaling, a key regulator of cell proliferation and extracellular matrix (ECM) remodeling; and pro-inflammatory SASP regulation, contributing to the chronic inflammation characteristic of aging tissues. By integrating multi-omic data, AI models predicted that TNIK inhibition could serve as an effective strategy to counteract SASP-driven tissue degeneration, leading to the development of INS018_055, a selective TNIK inhibitor that demonstrated senomorphic activity in preclinical studies.


INS018_055: A Senomorphic Drug for Aging-Related Diseases

A series of automated high-throughput screenings using AI-driven robotics laboratories evaluated the effects of INS018_055 in various senescence models. In chemotherapy-induced senescence, IMR-90 (human lung fibroblasts) and MRC-5 cells were exposed to doxorubicin to induce senescence. INS018_055 treatment significantly reduced the number of SA-β-gal-positive cells, a hallmark marker of senescence, while also suppressing SASP markers such as IL-6, IL-8, and TGF-β1, indicating a senomorphic effect. In a replicative senescence model, late-passage fibroblasts (IMR-90) exhibited classic senescence markers, and chronic treatment with INS018_055 attenuated SASP without affecting total cell viability. Unlike senolytics, INS018_055 did not deplete senescent cells but rather reprogrammed them to a less inflammatory state.


How TNIK Inhibition Works

INS018_055 exerts its senomorphic effects by targeting key aging pathways. By suppressing TGF-β/SMAD2/3 signaling, TNIK inhibition reduces TGF-β signaling, which limits fibrosis and chronic inflammation. Additionally, Wnt/β-catenin pathway modulation disrupts pro-fibrotic transcriptional programs, reducing ECM deposition. At the same time, SASP attenuation results in decreased secretion of IL-6, IL-8, and IL-1B, mitigating chronic, low-grade inflammation. Western blot analyses confirmed that INS018_055 downregulated phospho-TNIK and phospho-SMAD2/3, directly impacting pro-senescence transcriptional networks.


Clinical Implications and Future Directions

The potential for treating idiopathic pulmonary fibrosis (IPF) is one of the most exciting clinical applications of INS018_055. IPF is characterized by fibroblast senescence and excessive ECM deposition, and INS018_055 has demonstrated both anti-fibrotic and anti-senescence effects in lung fibroblast models. Currently, Phase II clinical trials (NCT05938920, NCT05975983) are assessing its efficacy in human subjects. Beyond IPF, TNIK is highly expressed in the brain and heart, suggesting that TNIK inhibition may have applications in neurodegeneration and cardiovascular aging. Future studies will explore INS018_055’s potential in treating conditions such as Alzheimer’s disease, sarcopenia, and vascular aging.

The success of INS018_055 also validates AI’s ability to identify and optimize novel longevity therapeutics. The integration of AI-powered robotics laboratories is expected to accelerate drug validation and clinical translation, paving the way for a new generation of anti-aging interventions.


A Paradigm Shift in Senomorphic Therapy

The discovery of TNIK inhibition as a senomorphic strategy marks a significant advancement in the field of longevity therapeutics. By selectively modulating SASP and reprogramming senescent cells, INS018_055 offers a promising alternative to traditional senolytics. As clinical trials progress, AI-driven drug discovery is set to redefine how we develop interventions for aging-related diseases. With INS018_055 paving the way for next-generation senotherapeutics, the future of longevity research is poised for groundbreaking innovations in precision geroscience.


Sources

  1. The hallmarks of aging. Cell, 2013.
  2. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. Journal of Clinical Investigation, 2013.
  3. Targeting cellular senescence with senotherapeutics: senolytics and senomorphics. FEBS Journal, 2023.
  4. AI-Driven Drug Discovery Identifies TNIK as a Senescence Modulator. Aging and Disease, 2025.
  5. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nature Medicine, 2016.
  6. TNIK in Wnt Signaling. Molecular Biology Reports, 2019.
  7. Rapamycin inhibits the secretory phenotype of senescent cells by a Nrf2‐independent mechanism. Aging Cell, 2017.
  8. TNIK Suppression Reduces Fibrosis. Frontiers in Aging Neuroscience, 2022.
  9. The Senescence-Associated Secretory Phenotype (SASP) in the Challenging Future of Cancer Therapy and Age-Related Diseases. Biology, 2020.

Leave a Reply

Abstract blue line art illustration of diverse gut microbiota, including rod-shaped, spherical, and spiral bacterial forms, representing the complexity and richness of the human gut microbiome. This visual metaphorically conveys the microbiome's integral role in regulating aging, cognition, immunity, and overall longevity.
Previous Story

Microbiome and Longevity Reviewed! Insights from Long-Lived Humans

Latest from Biotechnology

Don't Miss