Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Sep 27;22(1):868.
doi: 10.1186/s12967-024-05627-4.

Survival advantage of native and engineered T cells is acquired by mitochondrial transfer from mesenchymal stem cells

Affiliations

Survival advantage of native and engineered T cells is acquired by mitochondrial transfer from mesenchymal stem cells

Angela C Court et al. J Transl Med. .

Abstract

Background: Apoptosis, a form of programmed cell death, is critical for the development and homeostasis of the immune system. Chimeric antigen receptor T (CAR-T) cell therapy, approved for hematologic cancers, retains several limitations and challenges associated with ex vivo manipulation, including CAR T-cell susceptibility to apoptosis. Therefore, strategies to improve T-cell survival and persistence are required. Mesenchymal stem/stromal cells (MSCs) exhibit immunoregulatory and tissue-restoring potential. We have previously shown that the transfer of umbilical cord MSC (UC-MSC)-derived mitochondrial (MitoT) prompts the genetic reprogramming of CD3+ T cells towards a Treg cell lineage. The potency of T cells plays an important role in effective immunotherapy, underscoring the need for improving their metabolic fitness. In the present work, we evaluate the effect of MitoT on apoptotis of native T lymphocytes and engineered CAR-T cells.

Methods: We used a cell-free approach using artificial MitoT (Mitoception) of UC-MSC derived MT to peripheral blood mononuclear cells (PBMCs) followed by RNA-seq analysis of CD3+ MitoTpos and MitoTneg sorted cells. Target cell apoptosis was induced with Staurosporine (STS), and cell viability was evaluated with Annexin V/7AAD and TUNEL assays. Changes in apoptotic regulators were assessed by flow cytometry, western blot, and qRT-PCR. The effect of MitoT on 19BBz CAR T-cell apoptosis in response to electroporation with a non-viral transposon-based vector was assessed with Annexin V/7AAD.

Results: Gene expression related to apoptosis, cell death and/or responses to different stimuli was modified in CD3+ T cells after Mitoception. CD3+MitoTpos cells were resistant to STS-induced apoptosis compared to MitoTneg cells, showing a decreased percentage in apoptotic T cells as well as in TUNEL+ cells. Additionally, MitoT prevented the STS-induced collapse of the mitochondrial membrane potential (MMP) levels, decreased caspase-3 cleavage, increased BCL2 transcript levels and BCL-2-related BARD1 expression in FACS-sorted CD3+ T cells. Furthermore, UC-MSC-derived MitoT reduced both early and late apoptosis in CAR-T cells following electroporation, and exhibited an increasing trend in cytotoxic activity levels.

Conclusions: Artificial MitoT prevents STS-induced apoptosis of human CD3+ T cells by interfering with the caspase pathway. Furthermore, we observed that MitoT confers protection to apoptosis induced by electroporation in MitoTpos CAR T-engineered cells, potentially improving their metabolic fitness and resistance to environmental stress. These results widen the physiological perspective of organelle-based therapies in immune conditions while offering potential avenues to enhance CAR-T treatment outcomes where their viability is compromised.

Keywords: Chimeric antigen receptor T (CAR-T) cells; Induced-apoptosis; Mesenchymal stromal/stem cells; Mitochondria transfer.

PubMed Disclaimer

Conflict of interest statement

Maroun Khoury is the CEO and CSO of Cells for Cells and Regenero. Angela Court received stipend from Regenero. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Figures

Fig. 1
Fig. 1
Experimental workflow of MitoT to native and CAR engineered T cells. This schematic diagram illustrates the experimental methodology used to isolate mitochondria from umbilical cord MSCs after their transfer into T cells and subsequently challenge them by staurorsporine or through DNA electroporation for CAR-T generation. Apoptosis was then assessed using various markers and membrane potential measurements
Fig. 2
Fig. 2
Transfer of UC-MSC-derived MT modifies T lymphocyte response to stimulus and apoptosis gene expression. A Volcano plot analysis derived from FPKM values showing DE genes (p < 0.05, Student’s t test). Red dots: underexpressed genes (FC MitoTpos/MitoTneg < − 1.5). Blue dots: overexpressed genes (FC MitoTpos/MitoTneg > 1.5). B Pathway enrichment analysis (PEA) for DE genes between CD3+ MitoTpos (n = 4) and CD3+ MitoTneg (n = 4) cells. C Significantly enriched GO terms related to apoptosis, cell death, and/or responses to different stimuli of DE genes (p < 0.05, n = 4) between MitoTpos and MitoTneg cells. D Unsupervised hierarchical clustering heatmap of differentially expressed genes associated with GO terms from MitoTpos (n = 4) and MitoTneg (n = 4) samples, from whole transcriptome RNA sequencing analysis. Red transcripts represent genes directly involved in the apoptotic process regulation
Fig. 3
Fig. 3
MitoT from MSC-derived MT grants apoptosis resistance to T cells. A Experimental design of artificial MitoT (mitoception) used for apoptosis assays. B Representative FACS plots of Annexin V-apoptosis assay on CD3+ MitoTpos and MitoTneg populations after 4 h of staurosporine (STS) treatment. C Percentage of apoptotic CD3+ MitoTpos cells compared to MitoTneg cells after STS treatment. The left bars represented no STS-treatment control (n = 3). D Representative TUNEL assay (left panel) and percentage of TUNEL+ cells (right panel), by FACS, of CD3+ MitoTpos cells compared to MitoTneg cells after STS treatment (n = 3). E Representative western blot of caspase-3 cleavage on MitoTpos and MitoTneg CD3+ T lymphocytes after 4 h of STS treatment. β-Actin was used as loading control. F Quantification of proteins showed in E, represented as caspase 3/pro-caspase-3 ratio in CD3+ MitoTpos cells compared to MitoTneg cells (n = 3). G Representative FACS plots of the MMP by flow cytometry using JC-1 labeling of T lymphocytes after STS treatment. H Mitochondrial depolarization, as the ratio of red to green fluorescence, in CD3+ MitoTpos cells compared to MitoTneg cells after STS treatment by FACS analysis (n = 3). For figures C, D, F and H: graphs show mean ± SEM and statistical analysis by unpaired t-test. All replicates are biological
Fig. 4
Fig. 4
MitoT increases the expression of anti-apoptotic Bcl-2/BARD1 pathways. A Fold change of differentially expressed (DE) genes contained within the GO terms related to apoptosis, cell death, and/or responses to different stimuli. B qRT-PCR analysis of BARD1 mRNA expression levels in FACS-sorted CD3+ T cells after 24-h post-mitoception with MSC derived-MT (n = 4). C Representative western blots of BARD1 in FACS-sorted CD3+ MitoTpos cells and MitoTneg cells after 24 or 48 h post-mitoception. D Fold change quantification of proteins showed in C (n = 3). E Ingenuity Pathway analysis for Bcl-2 gene network for the DE genes denoted in A. F qRT-PCR of Bcl-2 family gene expression in FACS-sorted CD3+ MitoTpos cells and MitoTneg cells after 24 h post-mitoception (n = 4). For figures B and D: graphs show mean ± SEM and statistical analysis by unpaired t-test. For figure F: graph shows mean ± SEM and statistical analysis by unpaired Mann–Whitney test (*p < 0.05, relative to MitoTneg control). All replicates are biological
Fig. 5
Fig. 5
Anti-apoptotic effect on cMyc-tagged CAR-T cells. A Experimental design of artificial MitoT used for apoptosis assay on CAR-T cells. B Representative plots (left panel) and percentage of CARpos cells (right panel), by FACS, of CD3+ MitoTpos cells compared to MitoTneg cells after cell-electroporation (n = 4). C Representative FACS plots of Annexin V-7AAD apoptosis assay on CD3+ MitoTpos and MitoTneg cells in CARneg and CARpos populations after cell-electroporation. D Percentage of live, apoptotic, and necrotic CD3+ MitoTpos and MitoTneg cells in CARneg and CARpos populations after cell-electroporation (n = 4). E, F Mean of cytotoxicity activity of lymphocytes mitocepted (MitoTpos) or not (MitoTneg) and electroporated with 19BBz or Mock control, against Nalm-6-GFP B-cell line, after 8 days of expansion. The effector cells were incubated in a 1:2 (E) or 1:1 (F) ratio with the target cells (E:T) for 48 or 96 h. Right bar graph shows the average of remaining target cells (counts) (n = 4). G Representative plots (left panel) and percentage of CARpos IFNγ+ cells (right panel), by FACS, of CD3+ lymphocytes mitocepted (MitoTpos) or not (MitoTneg) after 8 days of expansion (n = 4). H Cytokines quantification, by ELISA, on supernatant from lymphocytes expanded and co-cultured for 24 h with target cells (1:1 ratio) (n = 4). Graphs show mean ± SEM and statistical analysis by unpaired t-test for figures B and D, paired t-test for figures E, F and G, and One-way ANOVA for figure H. All replicates are biological

Similar articles

Cited by

References

    1. Taylor RC, Cullen SP, Martin SJ. Apoptosis: controlled demolition at the cellular level. Nat Rev Mol Cell Biol. 2008;9:231–41. - PubMed
    1. Singh R, Letai A, Sarosiek K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat Rev Mol Cell Biol. 2019;20:175–93. - PMC - PubMed
    1. Opferman JT. Apoptosis in the development of the immune system. Cell Death Differ. 2008;15:234–42. - PubMed
    1. Rathmell JC, Thompson CB. Pathways of apoptosis in lymphocyte development, homeostasis, and disease. Cell. 2002;109(Suppl):S97–107. - PubMed
    1. Simula L, Corrado M, Accordi B, Di Rita A, Nazio F, Antonucci Y, et al. JNK1 and ERK1/2 modulate lymphocyte homeostasis via BIM and DRP1 upon AICD induction. Cell Death Differ. 2020;27:2749–67. - PMC - PubMed

Substances

LinkOut - more resources