Effects of Atrazine exposure on human bone marrow-derived mesenchymal stromal cells assessed by combinatorial assay matrix

Front Immunol. 2023 Jul 31:14:1214098. doi: 10.3389/fimmu.2023.1214098. eCollection 2023.

Abstract

Introduction: Mesenchymal Stromal/Stem cells (MSCs) are an essential component of the regenerative and immunoregulatory stem cell compartment of the human body and thus of major importance in human physiology. The MSCs elicit their beneficial properties through a multitude of complementary mechanisms, which makes it challenging to assess their phenotype and function in environmental toxicity screening. We here employed the novel combinatorial assays matrix approach/technology to profile the MSC response to the herbicide Atrazine, which is a common environmental xenobiotic, that is in widespread agricultural use in the US and other countries, but banned in the EU. Our here presented approach is representative for screening the impact of environmental xenobiotics and toxins on MSCs as an essential representative component of human physiology and well-being.

Methods: We here employed the combinatorial assay matrix approach, including a panel of well standardized assays, such as flow cytometry, multiplex secretome analysis, and metabolic assays, to define the phenotype and functionality of human-donor-derived primary MSCs exposed to the representative xenobiotic Atrazine. This assay matrix approach is now also endorsed for characterization of cell therapies by leading regulatory agencies, such as FDA and EMA.

Results: Our results show that the exposure to Atrazine modulates the metabolic activity, size, and granularity of MSCs in a dose and time dependent manner. Intriguingly, Atrazine exposure leads to a broad modulation of the MSCs secretome (both upregulation and downmodulation of certain factors) with the identification of Interleukin-8 as the topmost upregulated representative secretory molecule. Interestingly, Atrazine attenuates IFNγ-induced upregulation of MHC-class-II, but not MHC-class-I, and early phosphorylation signals on MSCs. Furthermore, Atrazine exposure attenuates IFNγ responsive secretome of MSCs. Mechanistic knockdown analysis identified that the Atrazine-induced effector molecule Interleukin-8 affects only certain but not all the related angiogenic secretome of MSCs.

Discussion: The here described Combinatorial Assay Matrix Technology identified that Atrazine affects both the innate/resting and cytokine-induced/stimulated assay matrix functionality of human MSCs, as identified through the modulation of selective, but not all effector molecules, thus vouching for the great usefulness of this approach to study the impact of xenobiotics on this important human cellular subset involved in the regenerative healing responses in humans.

Keywords: cellular phenotype and function; combinatorial assay matrix technology; environmental herbicide atrazine; immunomodulation and regeneration; mesenchymal stromal/stem cells (MSCs).

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Atrazine* / toxicity
  • Bone Marrow
  • Humans
  • Interleukin-8
  • Mesenchymal Stem Cells*
  • Xenobiotics

Substances

  • Atrazine
  • Interleukin-8
  • Xenobiotics

Grants and funding

Mercer University School of Medicine. The funding body played no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript. This study was partly supported by the WES Leukemia Research Foundation (RC). This research was also supported by Mercer University School of Medicine’s Research funds (RC). HZ and RP were supported by the cancer research scholarship from the Landings Women’s Golf Association (Savannah, GA), CU and TF were supported by the graduate program of Biomedical Sciences at Mercer University. GM’s contributions were made possible by the German Research Foundation/Deutsche Forschungsgemeinschaft (DFG; EX-PAND-PD CA2816/1-1) and the German Federal Ministry of Education and Research (BMBF) funding through the BSRT (GSC203) and BCRT, and in part by funding from the European Union’s Horizon 2020 research and innovation program under grant agreements No. 733006 (PACE) and No. 779293 (HIPGEN).