Full length article
Conditioned medium of adipose derived Mesenchymal Stem Cells reverse insulin resistance through downregulation of stress induced serine kinases

https://doi.org/10.1016/j.ejphar.2020.173215Get rights and content

Abstract:

Insulin resistance (IR) is a constituent part of Type 2 Diabetes Mellitus (T2DM). Conditioned medium from Adipose derived Mesenchymal Stem Cells (ADMSCs–CM) has been shown to reverse IR. However, its effect on cellular stress is not well established. The objective of this study was to explore the effect of ADMSCs-CM on reactive oxygen species, mitochondrial membrane potential (ΔΨm), endoplasmic reticulum (ER) stress and expression of oxidative and inflammatory stress induced serine kinases (SISK) which are pathophysiologically linked to IR. In insulin resistant, 3T3-L1 adipocytes and C2C12 myoblast cell culture models, glucose uptake was assayed by 2-NBDG uptake. Immunomodulatory cytokines, intracellular reactive oxygen species generation, ΔΨm and protein expression of JNK1, IKKβ and phospho-IRS1 (307) were analyzed using FACS. mRNA expression of ER stress markers (CHOP1 and IRE1) and SISK (JNK1, IKKβ, ERK1 and S6K1) were analyzed using RT-PCR. ADMSCs-CM effectively improve glucose uptake as evidenced by 2-NBDG uptake assay. FACS analysis showed that ADMSCs-CM possessed significantly higher levels of IL-6 and IL-10. ADMSCs-CM decreased intracellular generation of reactive oxygen species where it restored ΔΨm in C2C12 cells. ADMSCs-CM mediated reduction in ER stress was confirmed by down-regulation in CHOP1 and IRE1 mRNA expression. ADMSCs-CM treatment showed significant down-regulation of SISK mRNA expression including IKKβ, JNK, ERK and S6K1. Our results unequivocally demonstrate for the first time the mechanism of action of ADMSCs-CM in amelioration IR by reducing oxidative and inflammatory cellular stress. This study identifies SISK as potential therapeutic targets for T2DM therapy.

Introduction

The global epidemic of Type 2 Diabetes Mellitus (T2DM) has progressed exponentially, unhindered due to lack of effective therapies(World Health Organization, 2016). Kidney failure, neuropathy, retinopathy and vascular complications that lead to ischemic heart disease are among the many co-morbidities associated with T2DM(Jeanie et al., n.d). Impaired insulin signaling and dysfunctional β cells are hallmarks of T2DM which accounts for 90–95% of all Diabetes Mellitus cases(Raz et al., 2013). Pharmacological intervention become necessary as life style modifications proved to be difficult in the management of T2DM in the general population(Deborah M Muoio and Newgard, 2008a, Muoio and Newgard, 2008b). A combination of improvement in insulin sensitivity with simultaneous enhancement in β-cell function is believed to be ideal strategy to tackle T2DM pathogenesis. Currently available therapeutics temporarily ameliorate IR, however in a long run they can neither improve insulin sensitivity nor β-cell dysfunction warranting new therapeutic approach(Inzucchi, 2002).

Mesenchymal Stem Cells (MSCs) based therapies have emerged as potential candidate for the treatment of metabolic and immune disorders(Zang et al., 2017). MSCs are considered advantageous as they are easy to isolate and culture, lack immunogenicity and ethical issues, possess multi-lineage differentiation potential(Lalu et al., 2012). Also, MSCs possess unique secretome containing potent immunomodulatory and anti-inflammatory bioactive proteins and metabolites(Ranganath et al., 2016).

In the context of T2DM, the ability of MSCs to differentiate into Insulin Producing Cells (IPCs) was believed to be primary mechanisms to ameliorate hyperglycemia in T2DM. However, recent evidences suggest that paracrine effect of MSCs can improve insulin sensitivity by activating the insulin receptor substrate 1 (IRS1) signaling pathwaySi et al., 2012. Our group has demonstrated that conditioned medium from adipose derived MSCs (ADMSCs-CM) ameliorate IR through improved insulin signaling, Akt phosphorylation and GLUT4 expression in adipose and skeletal muscle cell models(Shree and Bhonde, 2017). Umbilical cord MSCs (UC-MSCs) have been reported to reprogram classically activated macrophage (M1, pro-inflammatory) into alternately activated phenotype (M2, anti-inflammatory) alleviating IR in T2DM rats(Xie et al., 2016). UCMSCs are also reported to alleviate IR through regulation of NLRP3 inflammasome in T2DM rats(Sun et al., 2017). However, knowledge on cellular and molecular mechanisms through which MSCs improves insulin sensitivity remains obscure.

IR arises from multiple etiological factors, of them excessive nutrient overload, accumulation of fatty acids derivatives, pro-inflammatory milieu, endoplasmic reticulum (ER) stress, mitochondrial dysfunction and oxidative stress are major drivers of insulin resistance(Hotamisligil, 2006). At molecular level, insulin signaling pathway negatively regulated by serine/threonine kinases activated by inflammatory cytokines, fatty acids derivatives, such as ceramides and diacylglycerol, reactive oxygen species(Czech, 2017; Deborah M Muoio and Newgard, 2008a; Tangvarasittichai, 2015). Kinases, including inhibitory-kB kinase β (IKKβ), c-Jun N-terminal kinases (JNK), extracellular signal regulated kinases (ERK) and p70S6 kinase (S6K1) when activated by metabolic and inflammatory stresses promote inhibitory serine phosphorylation on Insulin Receptor Substrate 1 (IRS1) promoting IR(Tanti and Jager, 2009a). High fat diet induced pre-diabetes exhibit decreased ΔΨm and ATP production in skeletal muscle suggest key role of mitochondrial dysfunction in IR(Xu et al., 2018).

In this study, we have examined effect of ADMSCs-CM on reactive oxygen species generation, mitochondrial membrane potential, ER stress markers, expression of stress induced serine kinases and its downstream effect on levels of inhibitory phosphorylation at ser-307 at IRS1 in differentiated and insulin resistant 3T3-L1 adipocytes and C2C12 myoblast cell culture models.

Section snippets

Adipose derived Mesenchymal Stem Cells culture

ADMSCs were obtained from Manipal School of Regenerative Medicine, Bengaluru (India). ADMSCs were cultured in α-MEM medium (Gibco) with 10% FBS and 1% Antibiotic-antimycotic solution (Gibco). Further, passage 4 cells were characterized for expression of CD90, CD73, CD105 (PE Tagged), CD34, CD45 and HLADR (FITC Tagged) (BD Biosciences) as previously described by us(Shree and Bhonde, 2017). Cells were further characterized for its chondrogenic (Stem-pro, A1007101), adipogenic (Stempro, A1007001)

Characterization of ADMSCs

Passage 4 cells subjected to CD antigen expression analysis using FACS were found to be positive for CD90, CD73, CD105 (>95%) and negative for CD34, CD45 and HLADR (MHC class II) (<2%). Further, cells were able to differentiate into adipogenic, osteogenic and chondrogenic lineage confirming its tri-lineage potential as depicted in Fig. 1.

Cytokines analysis of ADMSCs-CM

MSCs exert its immunomodulatory action through secretion of cytokines and bioactive proteins(Zang et al., 2017, Zhang et al., 2017). We performed cytokine

Discussion

IR is the major driver in the pathogenesis of T2DM which arises from metabolic overload, physical inactivity, hypoxia, psychological stress and environmental pollutants. These multi-etiological factors induce a network of cellular stress, stress responses and stress response dysregulations that inhibits downstream insulin signaling in insulin target cells(Onyango, 2018). Control of T2DM with conventional pharmaceutical agents is proving insufficient to tackle this multi-factorial etiology and

Conclusion

In conclusion, our data for the first time reveals that ADMSCs-CM rescues oxidative, inflammatory and ER stress and downregulates expression of stress induced serine kinases asserting its use as a prospective therapy for T2DM. The results obtained are attributed to the cumulative Secretome of ADMSCs-CM.

CRediT authorship contribution statement

Avinash Sanap: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Data curation, Writing - original draft. Ramesh Bhonde: Conceptualization, Methodology, Writing - review & editing, Visualization, Resources. Kalpana Joshi: Formal analysis, Writing - review & editing, Supervision, Validation.

Declaration of competing interest

All authors declare no conflict of interest.

Acknowledgements

We would like to thank Council of Scientific and Industrial Research (CSIR), Government of India for providing research fellowship to the first author. We are thankful to Sinhgad College of Engineering, Pune (India) and Dr. D. Y. Patil Vidyapeeth, Pune (India) for the extended support provided to carry out the work. Thanks are also due to Dr. Nitya Shree and Mr. Avinash Kharat for their guidance and assistance in manuscript editing.

References (47)

  • M. Zhang et al.

    The condition medium of mesenchymal stem cells promotes proliferation, adhesion and neuronal differentiation of retinal progenitor cells

    Neurosci. Lett.

    (2017)
  • M.C. Arkan et al.

    IKK-β links inflammation to obesity-induced insulin resistance

    Nat. Med.

    (2005)
  • A. Besse-Patin et al.

    An Intimate relationship between ROS and insulin signalling: implications for antioxidant treatment of fatty liver disease

    Int. J. Cell Biol.

    (2014)
  • S.P. Blaber et al.

    Analysis of in vitro secretion profiles from adipose-derived cell populations

    J. Transl. Med.

    (2012)
  • F. Bost et al.

    The Extracellular Signal – Regulated Kinase Isoform ERK1 Is Specifically Required for in Vitro and in Vivo Adipogenesis 54

    (2005)
  • M.P. Czech

    Perspective Insulin Action and Resistance in Obesity and Type 2 Diabetes 23

    (2017)
  • E.J. Estrada et al.
    (2008)
  • Z. Gao et al.
    (2014)
  • C.Y. Han

    Roles of reactive oxygen species on insulin resistance in adipose tissue

    Diabetes Metab. J.

    (2016)
  • J. Hirosumi et al.

    A central role for JNK in obesity and insulin resistance

    Nature

    (2002)
  • G.S. Hotamisligil
    (2006)
  • S.E. Inzucchi

    Oral antihyperglycemic therapy for type 2 diabetes scientific review

    J. Am. Med. Assoc.

    (2002)
  • Jeanie B. Tryggestad, MD and Steven M. Willi, M., n.d. Complications and comorbidities of T2DM in adolescents: findings...
  • Cited by (10)

    • Mesenchymal stem cell conditioned medium ameliorates diabetic serum-induced insulin resistance in 3T3-L1 cells

      2021, Chronic Diseases and Translational Medicine
      Citation Excerpt :

      DS contains a combination of pro-inflammatory cytokines and fatty acid derivatives such as ceramides and diacylglycerol which can trigger metabolic and inflammatory stress. Stress-induced serine kinases (IKK-β, c-Jun N-terminal kinases, extracellular signal regulated kinases, and p70S6 kinase) can negate normal insulin signaling by promoting inhibitory serine phosphorylation at IRS1-307 with concomitant increase in IR.28 DS can be a better alternative as compared to the pharmacological factors for inducing IR in vitro.

    • Treat liver to beat diabetes

      2020, Medical Hypotheses
      Citation Excerpt :

      Autologous MSCs transplantation also improve liver cirrhosis [36,37]. Our work has also evidenced that MSCs conditioned medium may also function as prospective insulin sensitizer which along with improving liver function can ameliorate peripheral insulin resistance [38–39] Therefore, application of MSCs to improve liver function is a much feasible option for T2DM therapy through improving liver function. The hypothesis revolves around interdependence of liver and pancreas due to their common developmental similarity and intra-organ communication.

    View all citing articles on Scopus
    View full text