Next Article in Journal
Tobacco Hornworm (Manduca sexta) Oral Secretion Elicits Reactive Oxygen Species in Isolated Tomato Protoplasts
Next Article in Special Issue
Stiffness and Aging in Cardiovascular Diseases: The Dangerous Relationship between Force and Senescence
Previous Article in Journal
Aβ Beyond the AD Pathology: Exploring the Structural Response of Membranes Exposed to Nascent Aβ Peptide
Previous Article in Special Issue
New Insights for BPIFB4 in Cardiovascular Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

miR-34a: A Promising Target for Inflammaging and Age-Related Diseases

by
Angela Raucci
1,* and
Maria Cristina Vinci
2,*
1
Unit of Experimental Cardio-Oncology and Cardiovascular Aging, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
2
Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Via Carlo Parea 4, 20138 Milan, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(21), 8293; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218293
Submission received: 28 October 2020 / Accepted: 3 November 2020 / Published: 5 November 2020
The term “inflammaging” describes the chronic, low-grade systemic inflammation that occurs during physiological aging in the absence of an overt infection [1,2]. Inflammaging is a hallmark of all age-related diseases, including cardiovascular diseases (CVDs) and associated risk factors (e.g., diabetes), that affects morbidity and mortality in the elderly [1,2]. Along with specialized immune cells, senescent cells are recognized as the largest contributors to inflammaging thanks to the acquisition of a senescence-associated secretory phenotype (SASP) that enable them to secrete a variety of soluble molecules including proinflammatory cytokines and chemokines, growth factors and matrix degrading proteins [2,3]. Furthermore, senescence may promote cell trans-differentiation toward a pathological phenotype [4]. Vascular calcification (VC) is an age-related complication of atherosclerosis, type 2 diabetes mellitus (T2DM) and chronic kidney disease characterized by the transition of vascular smooth muscle cells (VSMCs) to an osteo-chondrogenic phenotype with consequent hydroxyapatite crystals deposition and mineralization of the arterial wall [5,6]. Senescent VSMCs have greater propensity to experience the osteoblastic switch since express bone-related genes, such as Runt-related transcription factor 2 (Runx2), alkaline phosphatase and osteocalcin and secrete pro-calcification SASP factors, like interleukin 6 (IL-6), bone morphogenetic protein 2 and osteoprotegerin responsible for the spreading of senescence and mineralization of neighboring VSMCs [7,8].
MicroRNA-34a (miRNA-34a) is a senescent-associated miRNA whose expression has been shown to increase in different tissues and organs with age [9,10]. miR-34a is a promoter of senescence-induced VC. Mir34a deletion in mice reduces the expression of the VC markers such as SRY (sex-determining region Y)-box 9 (Sox9) and Runx2 and senescence proteins p16 and p21 and, consequently soft tissue and aorta medial calcification [4]. In vitro, replicative senescent human aortic smooth muscle cells (HASMCs) show higher levels of miR-34a and its ectopic overexpression in proliferative cells induces growth arrest and senescence through direct downregulation of AXL receptor tyrosine kinase (Axl) and sirtuin 1 favoring HASMCs mineralization in hyperphosphatemia conditions [4,9].
Our recent work, published in the Special Issue “Mechanisms of Inflammation in Degenerative Cardiovascular Conditions 2.0” of this journal, demonstrates that miR-34a enhances the secretion of several SASP factors in HASMCs, such as pro-inflammatory molecules (IL6, IL12, IL13 and Growth-Regulated Oncogene-alfa (GRO-α)), the metalloprotease inhibitor TIMP2 and the Insulin-like Growth Factor Binding Protein 3 (IGFBP3) [11]. Preconditioning with miR-34a-induced “secretome” enhances HASMCs senescence and mineralization indicating that this miRNA is able to endorse the activation of the VSMCs SASP to fuel the inflammatory conditions responsible for the spreading of vascular cells senescence and calcification [11]. Accordingly, Mir34a genetic ablation prevents the induction of IL6 expression occurring during aortas medial calcification onset. Importantly, we also found a positive correlation between circulating miR-34a and IL6 in a population of healthy subjects spanning from 20–90 years [11]. Altogether, our findings pinpoint miR-34a as a driver of vascular and systemic low-grade inflammaging and, hence, a causal promoter of age-associated diseases.
T2DM shares a number of important features with aging, including inflammaging and VC [12,13]. Indeed, high glucose triggers numerous inflammation and endoplasmic reticulum (ER) pathways that contribute to VSMCs senescence and calcification [13,14]. To date, there is strong evidence that diabetic milieu may epigenetically skew CD34+ stem cell differentiation, a cell population endowed of both regenerative and hematopoietic properties, towards more inflammatory cell populations [15]. To this regard, clinical and preclinical studies described abnormal elevation of monocyte subsets with higher inflammatory phenotype, alteration in macrophage polarization, as well as in the levels of circulating cytokines [16,17,18]. Interestingly, a very recent study demonstrated that non-classical monocytes exhibit the hallmarks of senescence, suggesting that their pro-inflammatory nature could be the manifestation of SASP [19]. In the Special Issue “Bone Marrow and Stem Cell Alterations in Diabetes: Causes, Consequences and Therapeutics” of this journal we recently reviewed the pathological contribution of bone marrow (BM) stem cells to diabetic cardiovascular complications [20]. In particular, we described the ability of diabetic milieu to redirect stem cell differentiation into cell populations with calcifying phenotype (osteoprogenitor cells) [21]. These cells, hypothetical “side products” of differentiation drift, witness the ability of diabetes to promote the generation of cells with pro-calcifying properties among others [20], with clear implications in diabetic micro- and macro-angiopathies development. Up to now, few preclinical studies reported that miR-34a up-regulation in the diabetic context impairs vascular function [22]; however, there are no data regarding the involvement of this miRNA in the processes of VC and inflammation associated with T2DM.
Since miRNAs are emerging as promising druggable targets, extending the knowledge of the mechanisms by which miR-34a regulates cell senescence, trans-differentiation and SASP acquisition in different pathological contexts, will help to develop new pharmacological therapies to counteract inflammaging and, eventually, age-related diseases onset.

Author Contributions

A.R. and M.C.V.: Conceptualization and writing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Centro Cardiologico Monzino-IRCCS (Ricerca Corrente 2020) to A.R. and M.C.V.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Franceschi, C.; Bonafe, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254. [Google Scholar] [CrossRef] [PubMed]
  2. Olivieri, F.; Prattichizzo, F.; Grillari, J.; Balistreri, C.R. Cellular Senescence and Inflammaging in Age-Related Diseases. Mediat. Inflamm. 2018, 2018, 9076485. [Google Scholar] [CrossRef] [PubMed]
  3. Tchkonia, T.; Zhu, Y.; van Deursen, J.; Campisi, J.; Kirkland, J.L. Cellular senescence and the senescent secretory phenotype: Therapeutic opportunities. J. Clin. Investig. 2013, 123, 966–972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Badi, I.; Mancinelli, L.; Polizzotto, A.; Ferri, D.; Zeni, F.; Burba, I.; Milano, G.; Brambilla, F.; Saccu, C.; Bianchi, M.E.; et al. miR-34a Promotes Vascular Smooth Muscle Cell Calcification by Downregulating SIRT1 (Sirtuin 1) and Axl (AXL Receptor Tyrosine Kinase). Arter. Thromb. Vasc. Biol. 2018, 38, 2079–2090. [Google Scholar] [CrossRef] [PubMed]
  5. Jaminon, A.; Reesink, K.; Kroon, A.; Schurgers, L. The Role of Vascular Smooth Muscle Cells in Arterial Remodeling: Focus on Calcification-Related Processes. Int. J. Mol. Sci. 2019, 20, 5694. [Google Scholar] [CrossRef] [Green Version]
  6. Johnson, R.C.; Leopold, J.A.; Loscalzo, J. Vascular calcification: Pathobiological mechanisms and clinical implications. Circ. Res. 2006, 99, 1044–1059. [Google Scholar] [CrossRef] [Green Version]
  7. Liu, Y.; Drozdov, I.; Shroff, R.; Beltran, L.E.; Shanahan, C.M. Prelamin A accelerates vascular calcification via activation of the DNA damage response and senescence-associated secretory phenotype in vascular smooth muscle cells. Circ. Res. 2013, 112, e99–e109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Nakano-Kurimoto, R.; Ikeda, K.; Uraoka, M.; Nakagawa, Y.; Yutaka, K.; Koide, M.; Takahashi, T.; Matoba, S.; Yamada, H.; Okigaki, M.; et al. Replicative senescence of vascular smooth muscle cells enhances the calcification through initiating the osteoblastic transition. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H1673–H1684. [Google Scholar] [CrossRef]
  9. Badi, I.; Burba, I.; Ruggeri, C.; Zeni, F.; Bertolotti, M.; Scopece, A.; Pompilio, G.; Raucci, A. MicroRNA-34a Induces Vascular Smooth Muscle Cells Senescence by SIRT1 Downregulation and Promotes the Expression of Age-Associated Pro-inflammatory Secretory Factors. J. Gerontol. A Biol. Sci. Med. Sci. 2015, 70, 1304–1311. [Google Scholar] [CrossRef]
  10. Ito, T.; Yagi, S.; Yamakuchi, M. MicroRNA-34a regulation of endothelial senescence. Biochem. Biophys. Res. Commun. 2010, 398, 735–740. [Google Scholar] [CrossRef]
  11. Zuccolo, E.; Badi, I.; Scavello, F.; Gambuzza, I.; Mancinelli, L.; Macri, F.; Tedesco, C.C.; Veglia, F.; Bonfigli, A.R.; Olivieri, F.; et al. The microRNA-34a-Induced Senescence-Associated Secretory Phenotype (SASP) Favors Vascular Smooth Muscle Cells Calcification. Int. J. Mol. Sci. 2020, 21, 4454. [Google Scholar] [CrossRef] [PubMed]
  12. Prattichizzo, F.; De Nigris, V.; La Sala, L.; Procopio, A.D.; Olivieri, F.; Ceriello, A. “Inflammaging” as a Druggable Target: A Senescence-Associated Secretory Phenotype-Centered View of Type 2 Diabetes. Oxid. Med. Cell. Longev. 2016, 2016, 1810327. [Google Scholar] [CrossRef]
  13. Zhu, Q.; Guo, R.; Liu, C.; Fu, D.; Liu, F.; Hu, J.; Jiang, H. Endoplasmic Reticulum Stress-Mediated Apoptosis Contributing to High Glucose-Induced Vascular Smooth Muscle Cell Calcification. J. Vasc. Res. 2015, 52, 291–298. [Google Scholar] [CrossRef] [PubMed]
  14. Chen, N.X.; Duan, D.; O’Neill, K.D.; Moe, S.M. High glucose increases the expression of Cbfa1 and BMP-2 and enhances the calcification of vascular smooth muscle cells. Nephrol. Dial. Transpl. 2006, 21, 3435–3442. [Google Scholar] [CrossRef] [Green Version]
  15. Gallagher, K.A.; Joshi, A.; Carson, W.F.; Schaller, M.; Allen, R.; Mukerjee, S.; Kittan, N.; Feldman, E.L.; Henke, P.K.; Hogaboam, C.; et al. Epigenetic changes in bone marrow progenitor cells influence the inflammatory phenotype and alter wound healing in type 2 diabetes. Diabetes 2015, 64, 1420–1430. [Google Scholar] [CrossRef] [Green Version]
  16. Bannon, P.; Wood, S.; Restivo, T.; Campbell, L.; Hardman, M.J.; Mace, K.A. Diabetes induces stable intrinsic changes to myeloid cells that contribute to chronic inflammation during wound healing in mice. Dis. Model. Mech. 2013, 6, 1434–1447. [Google Scholar] [CrossRef] [Green Version]
  17. Lowe, G.; Woodward, M.; Hillis, G.; Rumley, A.; Li, Q.; Harrap, S.; Marre, M.; Hamet, P.; Patel, A.; Poulter, N.; et al. Circulating inflammatory markers and the risk of vascular complications and mortality in people with type 2 diabetes and cardiovascular disease or risk factors: The ADVANCE study. Diabetes 2014, 63, 1115–1123. [Google Scholar] [CrossRef] [Green Version]
  18. Yang, M.; Gan, H.; Shen, Q.; Tang, W.; Du, X.; Chen, D. Proinflammatory CD14+CD16+ monocytes are associated with microinflammation in patients with type 2 diabetes mellitus and diabetic nephropathy uremia. Inflammation 2012, 35, 388–396. [Google Scholar] [CrossRef]
  19. Ong, S.M.; Hadadi, E.; Dang, T.M.; Yeap, W.H.; Tan, C.T.; Ng, T.P.; Larbi, A.; Wong, S.C. The pro-inflammatory phenotype of the human non-classical monocyte subset is attributed to senescence. Cell Death Dis. 2018, 9, 266. [Google Scholar] [CrossRef]
  20. Vinci, M.C.; Gambini, E.; Bassetti, B.; Genovese, S.; Pompilio, G. When Good Guys Turn Bad: Bone Marrow’s and Hematopoietic Stem Cells’ Role in the Pathobiology of Diabetic Complications. Int. J. Mol. Sci. 2020, 21, 3864. [Google Scholar] [CrossRef]
  21. Fadini, G.P.; Albiero, M.; Menegazzo, L.; Boscaro, E.; Agostini, C.; de Kreutzenberg, S.V.; Rattazzi, M.; Avogaro, A. Procalcific phenotypic drift of circulating progenitor cells in type 2 diabetes with coronary artery disease. Exp. Diabetes Res. 2012, 2012, 921685. [Google Scholar] [CrossRef]
  22. Li, Q.; Kim, Y.R.; Vikram, A.; Kumar, S.; Kassan, M.; Gabani, M.; Lee, S.K.; Jacobs, J.S.; Irani, K. P66Shc-Induced MicroRNA-34a Causes Diabetic Endothelial Dysfunction by Downregulating Sirtuin1. Arter. Thromb. Vasc. Biol. 2016, 36, 2394–2403. [Google Scholar] [CrossRef] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Raucci, A.; Vinci, M.C. miR-34a: A Promising Target for Inflammaging and Age-Related Diseases. Int. J. Mol. Sci. 2020, 21, 8293. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218293

AMA Style

Raucci A, Vinci MC. miR-34a: A Promising Target for Inflammaging and Age-Related Diseases. International Journal of Molecular Sciences. 2020; 21(21):8293. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218293

Chicago/Turabian Style

Raucci, Angela, and Maria Cristina Vinci. 2020. "miR-34a: A Promising Target for Inflammaging and Age-Related Diseases" International Journal of Molecular Sciences 21, no. 21: 8293. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms21218293

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop