TGF-β Signaling in Cellular Senescence and Aging-Related Pathology
Abstract
:1. Introduction
2. Cellular Senescence and Aging
3. Regulation of Cellular Proliferation and Senescence by TGF-β Signaling
4. Regulation of Stem Cell Aging by TGF-β Signaling
4.1. Mesenchymal Stem Cells
4.2. Hematopoietic Stem Cells
5. TGF-β Signaling in Longevity and Aging-Related Pathology
6. Roles of TGF-β Signaling in Alzheimer’s Disease
7. Roles of TGF-β Signaling in Age-Related Muscle Atrophy
8. Roles of TGF-β Signaling in Obesity
9. Conclusions
Funding
Conflicts of Interest
Abbreviations
4-HNE | 4-hydroxynonenal |
ALK | Activin receptor-like kinase |
ASC | Adipose stromal cell |
AD | Alzheimer’s disease |
ALS | Amyotrophic lateral sclerosis |
Aβ | β-amyloid peptide |
BMP | Bone morphogenetic protein |
BAT | Brown adipose tissue |
KLS | c-Kit+/Lin−/Sca-1+ |
CNS | Central nervous system |
CSF | Cerebrospinal fluid |
co-Smad | Common-partner Smad |
DPMSC | Double positive mesenchymal stem cell |
ECM | Extracellular matrix |
GDF | Growth differentiation factor |
HSC | Hematopoietic stem cell |
IPF | Idiopathic pulmonary fibrosis |
I-Smad | Inhibitory Smad |
ITGB3 | Integrin β3 |
MSC | Mesenchymal stem cell |
MHC | Myosin heavy chain |
PAI-1 | Plasminogen activator inhibitor-1 |
ROS | Reactive oxygen species |
R-Smad | Receptor-regulated Smad |
Rb | Retinoblastoma |
SASP | Senescence-associated secretory phenotype |
SA-β-gal | Senescence-associated β-galactosidase |
SIRT6 | Sirtuin 6 |
TERT | Telomerase reverse transcriptase |
TβRI | TGF-β type I receptor |
TβRII | TGF-β type II receptor |
Tif1γ | Transcriptional intermediary factor 1 γ |
TGF-β | Transforming growth factor-β |
UCP1 | Uncoupling protein-1 |
UPR | Unfolded protein response |
WAT | White adipose tissue |
References
- Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed]
- He, S.; Sharpless, N.E. Senescence in Health and Disease. Cell 2017, 169, 1000–1011. [Google Scholar] [CrossRef] [PubMed]
- Passarino, G.; De Rango, F.; Montesanto, A. Human longevity: Genetics or Lifestyle? It takes two to tango. Immun. Ageing 2016, 13, 12. [Google Scholar] [CrossRef] [PubMed]
- Harman, D. Aging: A theory based on free radical and radiation chemistry. J. Gerontol. 1956, 11, 298–300. [Google Scholar] [CrossRef] [PubMed]
- Cadenas, E.; Davies, K.J. Mitochondrial free radical generation, oxidative stress, and aging. Free Radic. Biol. Med. 2000, 29, 222–230. [Google Scholar] [CrossRef]
- Gidalevitz, T.; Prahlad, V.; Morimoto, R.I. The stress of protein misfolding: From single cells to multicellular organisms. Cold Spring Harb. Perspect. Biol. 2011, 3. [Google Scholar] [CrossRef] [PubMed]
- Kaushik, S.; Cuervo, A.M. Proteostasis and aging. Nat. Med. 2015, 21, 1406–1415. [Google Scholar] [CrossRef] [PubMed]
- Morikawa, M.; Derynck, R.; Miyazono, K. TGF-β and the TGF-β Family: Context-Dependent Roles in Cell and Tissue Physiology. Cold Spring Harb. Perspect. Biol. 2016, 8. [Google Scholar] [CrossRef]
- Miyazono, K.; Katsuno, Y.; Koinuma, D.; Ehata, S.; Morikawa, M. Intracellular and extracellular TGF-β signaling in cancer: Some recent topics. Front. Med. 2018, 12, 387–411. [Google Scholar] [CrossRef]
- Franceschi, C.; Capri, M.; Monti, D.; Giunta, S.; Olivieri, F.; Sevini, F.; Panourgia, M.P.; Invidia, L.; Celani, L.; Scurti, M.; et al. Inflammaging and anti-inflammaging: A systemic perspective on aging and longevity emerged from studies in humans. Mech. Ageing Dev. 2007, 128, 92–105. [Google Scholar] [CrossRef]
- Hayflick, L. Intracellular determinants of cell aging. Mech. Ageing Dev. 1984, 28, 177–185. [Google Scholar] [CrossRef]
- Druelle, C.; Drullion, C.; Desle, J.; Martin, N.; Saas, L.; Cormenier, J.; Malaquin, N.; Huot, L.; Slomianny, C.; Bouali, F.; et al. ATF6α regulates morphological changes associated with senescence in human fibroblasts. Oncotarget 2016, 7, 67699–67715. [Google Scholar] [CrossRef] [PubMed]
- Davalos, A.R.; Coppe, J.P.; Campisi, J.; Desprez, P.Y. Senescent cells as a source of inflammatory factors for tumor progression. Cancer Metastasis Rev. 2010, 29, 273–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coppe, J.P.; Desprez, P.Y.; Krtolica, A.; Campisi, J. The senescence-associated secretory phenotype: The dark side of tumor suppression. Annu. Rev. Pathol. 2010, 5, 99–118. [Google Scholar] [CrossRef]
- Watanabe, S.; Kawamoto, S.; Ohtani, N.; Hara, E. Impact of senescence-associated secretory phenotype and its potential as a therapeutic target for senescence-associated diseases. Cancer Sci. 2017, 108, 563–569. [Google Scholar] [CrossRef] [Green Version]
- Baker, D.J.; Wijshake, T.; Tchkonia, T.; LeBrasseur, N.K.; Childs, B.G.; van de Sluis, B.; Kirkland, J.L.; van Deursen, J.M. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 2011, 479, 232–236. [Google Scholar] [CrossRef]
- Xu, M.; Pirtskhalava, T.; Farr, J.N.; Weigand, B.M.; Palmer, A.K.; Weivoda, M.M.; Inman, C.L.; Ogrodnik, M.B.; Hachfeld, C.M.; Fraser, D.G.; et al. Senolytics improve physical function and increase lifespan in old age. Nat. Med. 2018, 24, 1246–1256. [Google Scholar] [CrossRef]
- Heldin, C.H.; Moustakas, A. Signaling Receptors for TGF-β Family Members. Cold Spring Harb. Perspect. Biol. 2016, 8. [Google Scholar] [CrossRef]
- Derynck, R.; Budi, E.H. Specificity, versatility, and control of TGF-β family signaling. Sci. Signal. 2019, 12. [Google Scholar] [CrossRef]
- Aykul, S.; Martinez-Hackert, E. Transforming Growth Factor-β Family Ligands Can Function as Antagonists by Competing for Type II Receptor Binding. J. Biol. Chem. 2016, 291, 10792–10804. [Google Scholar] [CrossRef]
- Papageorgis, P. Complex Interplay Between Aging and Cancer: Role of TGF-β Signaling. Crit. Rev. Oncog. 2017, 22, 313–321. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Alexander, P.B.; Wang, X.F. TGF-β Family Signaling in the Control of Cell Proliferation and Survival. Cold Spring Harb. Perspect. Biol. 2017, 9. [Google Scholar] [CrossRef] [PubMed]
- Debacq-Chainiaux, F.; Borlon, C.; Pascal, T.; Royer, V.; Eliaers, F.; Ninane, N.; Carrard, G.; Friguet, B.; de Longueville, F.; Boffe, S.; et al. Repeated exposure of human skin fibroblasts to UVB at subcytotoxic level triggers premature senescence through the TGF-β1 signaling pathway. J. Cell Sci. 2005, 118, 743–758. [Google Scholar] [CrossRef] [PubMed]
- Minagawa, S.; Araya, J.; Numata, T.; Nojiri, S.; Hara, H.; Yumino, Y.; Kawaishi, M.; Odaka, M.; Morikawa, T.; Nishimura, S.L.; et al. Accelerated epithelial cell senescence in IPF and the inhibitory role of SIRT6 in TGF-β-induced senescence of human bronchial epithelial cells. Am. J. Physiol. Lung Cell Mol. Physiol. 2011, 300, L391–L401. [Google Scholar] [CrossRef] [PubMed]
- Senturk, S.; Mumcuoglu, M.; Gursoy-Yuzugullu, O.; Cingoz, B.; Akcali, K.C.; Ozturk, M. Transforming growth factor-beta induces senescence in hepatocellular carcinoma cells and inhibits tumor growth. Hepatology 2010, 52, 966–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Albright, C.D.; Salganik, R.I.; Craciunescu, C.N.; Mar, M.H.; Zeisel, S.H. Mitochondrial and microsomal derived reactive oxygen species mediate apoptosis induced by transforming growth factor-β1 in immortalized rat hepatocytes. J. Cell Biochem. 2003, 89, 254–261. [Google Scholar] [CrossRef] [PubMed]
- Yoon, Y.S.; Lee, J.H.; Hwang, S.C.; Choi, K.S.; Yoon, G. TGF β1 induces prolonged mitochondrial ROS generation through decreased complex IV activity with senescent arrest in Mv1Lu cells. Oncogene 2005, 24, 1895–1903. [Google Scholar] [CrossRef]
- Hu, B.; Tack, D.C.; Liu, T.; Wu, Z.; Ullenbruch, M.R.; Phan, S.H. Role of Smad3 in the regulation of rat telomerase reverse transcriptase by TGFβ. Oncogene 2006, 25, 1030–1041. [Google Scholar] [CrossRef]
- Li, H.; Xu, D.; Li, J.; Berndt, M.C.; Liu, J.P. Transforming growth factor β suppresses human telomerase reverse transcriptase (hTERT) by Smad3 interactions with c-Myc and the hTERT gene. J. Biol. Chem. 2006, 281, 25588–25600. [Google Scholar] [CrossRef]
- Suzuki, H.I. MicroRNA Control of TGF-β Signaling. Int. J. Mol. Sci. 2018, 19, 1901. [Google Scholar] [CrossRef]
- Suzuki, H.I.; Miyazono, K. Emerging complexity of microRNA generation cascades. J. Biochem. 2011, 149, 15–25. [Google Scholar] [CrossRef] [PubMed]
- Lyu, G.; Guan, Y.; Zhang, C.; Zong, L.; Sun, L.; Huang, X.; Huang, L.; Zhang, L.; Tian, X.L.; Zhou, Z.; et al. TGF-β signaling alters H4K20me3 status via miR-29 and contributes to cellular senescence and cardiac aging. Nat. Commun. 2018, 9, 2560. [Google Scholar] [CrossRef] [PubMed]
- Okumura, N.; Hashimoto, K.; Kitahara, M.; Okuda, H.; Ueda, E.; Watanabe, K.; Nakahara, M.; Sato, T.; Kinoshita, S.; Tourtas, T.; et al. Activation of TGF-β signaling induces cell death via the unfolded protein response in Fuchs endothelial corneal dystrophy. Sci. Rep. 2017, 7, 6801. [Google Scholar] [CrossRef] [PubMed]
- Kiyono, K.; Suzuki, H.I.; Matsuyama, H.; Morishita, Y.; Komuro, A.; Kano, M.R.; Sugimoto, K.; Miyazono, K. Autophagy is activated by TGF-β and potentiates TGF-β-mediated growth inhibition in human hepatocellular carcinoma cells. Cancer Res. 2009, 69, 8844–8852. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, H.I.; Kiyono, K.; Miyazono, K. Regulation of autophagy by transforming growth factor-β (TGF-β) signaling. Autophagy 2010, 6, 645–647. [Google Scholar] [CrossRef] [PubMed]
- Rapisarda, V.; Borghesan, M.; Miguela, V.; Encheva, V.; Snijders, A.P.; Lujambio, A.; O’Loghlen, A. Integrin Beta 3 Regulates Cellular Senescence by Activating the TGF-β Pathway. Cell Rep. 2017, 18, 2480–2493. [Google Scholar] [CrossRef] [PubMed]
- Forsey, R.J.; Thompson, J.M.; Ernerudh, J.; Hurst, T.L.; Strindhall, J.; Johansson, B.; Nilsson, B.O.; Wikby, A. Plasma cytokine profiles in elderly humans. Mech. Ageing Dev. 2003, 124, 487–493. [Google Scholar] [CrossRef]
- Carrieri, G.; Marzi, E.; Olivieri, F.; Marchegiani, F.; Cavallone, L.; Cardelli, M.; Giovagnetti, S.; Stecconi, R.; Molendini, C.; Trapassi, C.; et al. The G/C915 polymorphism of transforming growth factor β1 is associated with human longevity: A study in Italian centenarians. Aging Cell 2004, 3, 443–448. [Google Scholar] [CrossRef] [PubMed]
- Vaughan, D.E.; Rai, R.; Khan, S.S.; Eren, M.; Ghosh, A.K. Plasminogen Activator Inhibitor-1 Is a Marker and a Mediator of Senescence. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 1446–1452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morrison, S.J.; Kimble, J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 2006, 441, 1068–1074. [Google Scholar] [CrossRef]
- Eming, S.A.; Martin, P.; Tomic-Canic, M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci. Transl. Med. 2014, 6, 265sr266. [Google Scholar] [CrossRef] [PubMed]
- Feige, P.; Brun, C.E.; Ritso, M.; Rudnicki, M.A. Orienting Muscle Stem Cells for Regeneration in Homeostasis, Aging, and Disease. Cell Stem Cell 2018, 23, 653–664. [Google Scholar] [CrossRef] [PubMed]
- Oh, J.; Lee, Y.D.; Wagers, A.J. Stem cell aging: Mechanisms, regulators and therapeutic opportunities. Nat. Med. 2014, 20, 870–880. [Google Scholar] [CrossRef] [PubMed]
- Robinson, N.J.; Taylor, D.J.; Schiemann, W.P. Stem cells, immortality, and the evolution of metastatic properties in breast cancer: Telomere maintenance mechanisms and metastatic evolution. J. Cancer Metastasis Treat. 2019, 5. [Google Scholar] [CrossRef]
- Ren, R.; Ocampo, A.; Liu, G.H.; Izpisua Belmonte, J.C. Regulation of Stem Cell Aging by Metabolism and Epigenetics. Cell Metab. 2017, 26, 460–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, X.; Zheng, L.; Yuan, Q.; Zhen, G.; Crane, J.L.; Zhou, X.; Cao, X. Transforming growth factor-β in stem cells and tissue homeostasis. Bone Res. 2018, 6, 2. [Google Scholar] [CrossRef] [PubMed]
- Spees, J.L.; Lee, R.H.; Gregory, C.A. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res. Ther. 2016, 7, 125. [Google Scholar] [CrossRef] [PubMed]
- Patel, S.A.; Meyer, J.R.; Greco, S.J.; Corcoran, K.E.; Bryan, M.; Rameshwar, P. Mesenchymal stem cells protect breast cancer cells through regulatory T cells: Role of mesenchymal stem cell-derived TGF-β. J. Immunol. 2010, 184, 5885–5894. [Google Scholar] [CrossRef] [PubMed]
- Alt, E.U.; Senst, C.; Murthy, S.N.; Slakey, D.P.; Dupin, C.L.; Chaffin, A.E.; Kadowitz, P.J.; Izadpanah, R. Aging alters tissue resident mesenchymal stem cell properties. Stem Cell Res. 2012, 8, 215–225. [Google Scholar] [CrossRef] [Green Version]
- Stolzing, A.; Jones, E.; McGonagle, D.; Scutt, A. Age-related changes in human bone marrow-derived mesenchymal stem cells: Consequences for cell therapies. Mech. Ageing Dev. 2008, 129, 163–173. [Google Scholar] [CrossRef]
- Maredziak, M.; Marycz, K.; Tomaszewski, K.A.; Kornicka, K.; Henry, B.M. The Influence of Aging on the Regenerative Potential of Human Adipose Derived Mesenchymal Stem Cells. Stem Cells Int. 2016, 2016, 2152435. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.K.; Ogando, C.R.; Wang See, C.; Chang, T.Y.; Barabino, G.A. Changes in phenotype and differentiation potential of human mesenchymal stem cells aging in vitro. Stem Cell Res. Ther. 2018, 9, 131. [Google Scholar] [CrossRef] [PubMed]
- Bethel, M.; Chitteti, B.R.; Srour, E.F.; Kacena, M.A. The changing balance between osteoblastogenesis and adipogenesis in aging and its impact on hematopoiesis. Curr Osteoporos. Rep. 2013, 11, 99–106. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Niu, J.; Li, X.; Wang, X.; Guo, Z.; Zhang, F. TGF-β1 induces senescence of bone marrow mesenchymal stem cells via increase of mitochondrial ROS production. BMC Dev. Biol. 2014, 14, 21. [Google Scholar] [CrossRef] [PubMed]
- Gurung, S.; Werkmeister, J.A.; Gargett, C.E. Inhibition of Transforming Growth Factor-β Receptor signaling promotes culture expansion of undifferentiated human Endometrial Mesenchymal Stem/stromal Cells. Sci. Rep. 2015, 5, 15042. [Google Scholar] [CrossRef] [PubMed]
- Jian, H.; Shen, X.; Liu, I.; Semenov, M.; He, X.; Wang, X.F. Smad3-dependent nuclear translocation of β-catenin is required for TGF-β1-induced proliferation of bone marrow-derived adult human mesenchymal stem cells. Genes Dev. 2006, 20, 666–674. [Google Scholar] [CrossRef] [PubMed]
- Salkin, H.; Gonen, Z.B.; Ergen, E.; Bahar, D.; Cetin, M. Effects of TGF-β1 Overexpression on Biological Characteristics of Human Dental Pulp-derived Mesenchymal Stromal Cells. Int. J. Stem Cells 2019, 12, 170–182. [Google Scholar] [CrossRef] [PubMed]
- Walenda, G.; Abnaof, K.; Joussen, S.; Meurer, S.; Smeets, H.; Rath, B.; Hoffmann, K.; Frohlich, H.; Zenke, M.; Weiskirchen, R.; et al. TGF-beta1 does not induce senescence of multipotent mesenchymal stromal cells and has similar effects in early and late passages. PLoS ONE 2013, 8, e77656. [Google Scholar] [CrossRef]
- Muraglia, A.; Cancedda, R.; Quarto, R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J. Cell Sci. 2000, 113, 1161–1166. [Google Scholar] [PubMed]
- Chang, H.Y.; Chi, J.T.; Dudoit, S.; Bondre, C.; van de Rijn, M.; Botstein, D.; Brown, P.O. Diversity, topographic differentiation, and positional memory in human fibroblasts. Proc. Natl. Acad. Sci. USA 2002, 99, 12877–12882. [Google Scholar] [CrossRef] [Green Version]
- Horie, M.; Miyashita, N.; Mikami, Y.; Noguchi, S.; Yamauchi, Y.; Suzukawa, M.; Fukami, T.; Ohta, K.; Asano, Y.; Sato, S.; et al. TBX4 is involved in the super-enhancer-driven transcriptional programs underlying features specific to lung fibroblasts. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 314, L177–L191. [Google Scholar] [CrossRef] [PubMed]
- Horie, M.; Yamaguchi, Y.; Saito, A.; Nagase, T.; Lizio, M.; Itoh, M.; Kawaji, H.; Lassmann, T.; Carninci, P.; Forrest, A.R.; et al. Transcriptome analysis of periodontitis-associated fibroblasts by CAGE sequencing identified DLX5 and RUNX2 long variant as novel regulators involved in periodontitis. Sci. Rep. 2016, 6, 33666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roelen, B.A.; Dijke, P. Controlling mesenchymal stem cell differentiation by TGFβ family members. J. Orthop. Sci. 2003, 8, 740–748. [Google Scholar] [CrossRef] [PubMed]
- Matsuoka, Y.; Nakatsuka, R.; Sumide, K.; Kawamura, H.; Takahashi, M.; Fujioka, T.; Uemura, Y.; Asano, H.; Sasaki, Y.; Inoue, M.; et al. Prospectively Isolated Human Bone Marrow Cell-Derived MSCs Support Primitive Human CD34-Negative Hematopoietic Stem Cells. Stem Cells 2015, 33, 1554–1565. [Google Scholar] [CrossRef] [PubMed]
- Kawamura, H.; Nakatsuka, R.; Matsuoka, Y.; Sumide, K.; Fujioka, T.; Asano, H.; Iida, H.; Sonoda, Y. TGF-β Signaling Accelerates Senescence of Human Bone-Derived CD271 and SSEA-4 Double-Positive Mesenchymal Stromal Cells. Stem Cell Rep. 2018, 10, 920–932. [Google Scholar] [CrossRef] [PubMed]
- Blank, U.; Karlsson, S. TGF-β signaling in the control of hematopoietic stem cells. Blood 2015, 125, 3542–3550. [Google Scholar] [CrossRef]
- Scandura, J.M.; Boccuni, P.; Massague, J.; Nimer, S.D. Transforming growth factor β-induced cell cycle arrest of human hematopoietic cells requires p57KIP2 up-regulation. Proc. Natl. Acad. Sci. USA 2004, 101, 15231–15236. [Google Scholar] [CrossRef]
- Yamazaki, S.; Iwama, A.; Takayanagi, S.; Eto, K.; Ema, H.; Nakauchi, H. TGF-β as a candidate bone marrow niche signal to induce hematopoietic stem cell hibernation. Blood 2009, 113, 1250–1256. [Google Scholar] [CrossRef]
- Wang, X.; Dong, F.; Zhang, S.; Yang, W.; Yu, W.; Wang, Z.; Zhang, S.; Wang, J.; Ma, S.; Wu, P.; et al. TGF-β1 Negatively Regulates the Number and Function of Hematopoietic Stem Cells. Stem Cell Rep. 2018, 11, 274–287. [Google Scholar] [CrossRef]
- Capron, C.; Lacout, C.; Lecluse, Y.; Jalbert, V.; Chagraoui, H.; Charrier, S.; Galy, A.; Bennaceur-Griscelli, A.; Cramer-Borde, E.; Vainchenker, W. A major role of TGF-β1 in the homing capacities of murine hematopoietic stem cell/progenitors. Blood 2010, 116, 1244–1253. [Google Scholar] [CrossRef]
- Yamazaki, S.; Ema, H.; Karlsson, G.; Yamaguchi, T.; Miyoshi, H.; Shioda, S.; Taketo, M.M.; Karlsson, S.; Iwama, A.; Nakauchi, H. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 2011, 147, 1146–1158. [Google Scholar] [CrossRef] [PubMed]
- Brenet, F.; Kermani, P.; Spektor, R.; Rafii, S.; Scandura, J.M. TGFβ restores hematopoietic homeostasis after myelosuppressive chemotherapy. J. Exp. Med. 2013, 210, 623–639. [Google Scholar] [CrossRef] [PubMed]
- Rorby, E.; Hagerstrom, M.N.; Blank, U.; Karlsson, G.; Karlsson, S. Human hematopoietic stem/progenitor cells overexpressing Smad4 exhibit impaired reconstitution potential in vivo. Blood 2012, 120, 4343–4351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Larsson, J.; Blank, U.; Helgadottir, H.; Bjornsson, J.M.; Ehinger, M.; Goumans, M.J.; Fan, X.; Leveen, P.; Karlsson, S. TGF-β signaling-deficient hematopoietic stem cells have normal self-renewal and regenerative ability in vivo despite increased proliferative capacity in vitro. Blood 2003, 102, 3129–3135. [Google Scholar] [CrossRef] [PubMed]
- Larsson, J.; Blank, U.; Klintman, J.; Magnusson, M.; Karlsson, S. Quiescence of hematopoietic stem cells and maintenance of the stem cell pool is not dependent on TGF-β signaling in vivo. Exp. Hematol. 2005, 33, 592–596. [Google Scholar] [CrossRef] [PubMed]
- De Haan, G.; Lazare, S.S. Aging of hematopoietic stem cells. Blood 2018, 131, 479–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gekas, C.; Graf, T. CD41 expression marks myeloid-biased adult hematopoietic stem cells and increases with age. Blood 2013, 121, 4463–4472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, D.; Luo, M.; Jeong, M.; Rodriguez, B.; Xia, Z.; Hannah, R.; Wang, H.; Le, T.; Faull, K.F.; Chen, R.; et al. Epigenomic profiling of young and aged HSCs reveals concerted changes during aging that reinforce self-renewal. Cell Stem Cell 2014, 14, 673–688. [Google Scholar] [CrossRef]
- Quere, R.; Saint-Paul, L.; Carmignac, V.; Martin, R.Z.; Chretien, M.L.; Largeot, A.; Hammann, A.; Pais de Barros, J.P.; Bastie, J.N.; Delva, L. Tif1γ regulates the TGF-β1 receptor and promotes physiological aging of hematopoietic stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, 10592–10597. [Google Scholar] [CrossRef]
- Langer, J.C.; Henckaerts, E.; Orenstein, J.; Snoeck, H.W. Quantitative trait analysis reveals transforming growth factor-β2 as a positive regulator of early hematopoietic progenitor and stem cell function. J. Exp. Med. 2004, 199, 5–14. [Google Scholar] [CrossRef]
- Challen, G.A.; Boles, N.C.; Chambers, S.M.; Goodell, M.A. Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-β1. Cell Stem Cell 2010, 6, 265–278. [Google Scholar] [CrossRef] [PubMed]
- Cavallone, L.; Bonafe, M.; Olivieri, F.; Cardelli, M.; Marchegiani, F.; Giovagnetti, S.; Di Stasio, G.; Giampieri, C.; Mugianesi, E.; Stecconi, R.; et al. The role of IL-1 gene cluster in longevity: A study in Italian population. Mech. Ageing Dev. 2003, 124, 533–538. [Google Scholar] [CrossRef]
- Awad, M.R.; El-Gamel, A.; Hasleton, P.; Turner, D.M.; Sinnott, P.J.; Hutchinson, I.V. Genotypic variation in the transforming growth factor-β1 gene: Association with transforming growth factor-β1 production, fibrotic lung disease, and graft fibrosis after lung transplantation. Transplantation 1998, 66, 1014–1020. [Google Scholar] [CrossRef] [PubMed]
- Goumans, M.J.; Ten Dijke, P. TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb. Perspect. Biol. 2018, 10. [Google Scholar] [CrossRef] [PubMed]
- Saito, A.; Horie, M.; Nagase, T. TGF-β Signaling in Lung Health and Disease. Int. J. Mol. Sci. 2018, 19, 2460. [Google Scholar] [CrossRef] [PubMed]
- Saito, A.; Horie, M.; Micke, P.; Nagase, T. The Role of TGF-β Signaling in Lung Cancer Associated with Idiopathic Pulmonary Fibrosis. Int. J. Mol. Sci. 2018, 19, 3611. [Google Scholar] [CrossRef]
- Van der Kraan, P.M. The changing role of TGFβ in healthy, ageing and osteoarthritic joints. Nat. Rev. Rheumatol. 2017, 13, 155–163. [Google Scholar] [CrossRef]
- Buckwalter, M.S.; Wyss-Coray, T. Modelling neuroinflammatory phenotypes in vivo. J. Neuroinflammation 2004, 1, 10. [Google Scholar] [CrossRef]
- Buckwalter, M.S.; Yamane, M.; Coleman, B.S.; Ormerod, B.K.; Chin, J.T.; Palmer, T.; Wyss-Coray, T. Chronically increased transforming growth factor-β1 strongly inhibits hippocampal neurogenesis in aged mice. Am. J. Pathol. 2006, 169, 154–164. [Google Scholar] [CrossRef]
- Caraci, F.; Spampinato, S.; Sortino, M.A.; Bosco, P.; Battaglia, G.; Bruno, V.; Drago, F.; Nicoletti, F.; Copani, A. Dysfunction of TGF-β1 signaling in Alzheimer’s disease: Perspectives for neuroprotection. Cell Tissue Res. 2012, 347, 291–301. [Google Scholar] [CrossRef]
- Bye, N.; Zieba, M.; Wreford, N.G.; Nichols, N.R. Resistance of the dentate gyrus to induced apoptosis during ageing is associated with increases in transforming growth factor-β1 messenger RNA. Neuroscience 2001, 105, 853–862. [Google Scholar] [CrossRef]
- Bondi, M.W.; Edmonds, E.C.; Salmon, D.P. Alzheimer’s Disease: Past, Present, and Future. J. Int. Neuropsychol. Soc. 2017, 23, 818–831. [Google Scholar] [CrossRef] [PubMed]
- Flanders, K.C.; Lippa, C.F.; Smith, T.W.; Pollen, D.A.; Sporn, M.B. Altered expression of transforming growth factor-beta in Alzheimer’s disease. Neurology 1995, 45, 1561–1569. [Google Scholar] [CrossRef] [PubMed]
- Tarkowski, E.; Issa, R.; Sjogren, M.; Wallin, A.; Blennow, K.; Tarkowski, A.; Kumar, P. Increased intrathecal levels of the angiogenic factors VEGF and TGF-β in Alzheimer’s disease and vascular dementia. Neurobiol. Aging 2002, 23, 237–243. [Google Scholar] [CrossRef]
- Zetterberg, H.; Andreasen, N.; Blennow, K. Increased cerebrospinal fluid levels of transforming growth factor-β1 in Alzheimer’s disease. Neurosci. Lett. 2004, 367, 194–196. [Google Scholar] [CrossRef] [PubMed]
- Rota, E.; Bellone, G.; Rocca, P.; Bergamasco, B.; Emanuelli, G.; Ferrero, P. Increased intrathecal TGF-β1, but not IL-12, IFN-γ and IL-10 levels in Alzheimer’s disease patients. Neurol. Sci. 2006, 27, 33–39. [Google Scholar] [CrossRef]
- Mocali, A.; Cedrola, S.; Della Malva, N.; Bontempelli, M.; Mitidieri, V.A.; Bavazzano, A.; Comolli, R.; Paoletti, F.; La Porta, C.A. Increased plasma levels of soluble CD40, together with the decrease of TGFβ1, as possible differential markers of Alzheimer disease. Exp. Gerontol. 2004, 39, 1555–1561. [Google Scholar] [CrossRef]
- Juraskova, B.; Andrys, C.; Holmerova, I.; Solichova, D.; Hrnciarikova, D.; Vankova, H.; Vasatko, T.; Krejsek, J. Transforming growth factor beta and soluble endoglin in the healthy senior and in Alzheimer’s disease patients. J. Nutr. Health Aging 2010, 14, 758–761. [Google Scholar] [CrossRef]
- Chao, C.C.; Hu, S.; Frey, W.H., 2nd; Ala, T.A.; Tourtellotte, W.W.; Peterson, P.K. Transforming growth factor β in Alzheimer’s disease. Clin. Diagn. Lab. Immunol. 1994, 1, 109–110. [Google Scholar]
- Malaguarnera, L.; Motta, M.; Di Rosa, M.; Anzaldi, M.; Malaguarnera, M. Interleukin-18 and transforming growth factor-beta 1 plasma levels in Alzheimer’s disease and vascular dementia. Neuropathology 2006, 26, 307–312. [Google Scholar] [CrossRef]
- Motta, M.; Imbesi, R.; Di Rosa, M.; Stivala, F.; Malaguarnera, L. Altered plasma cytokine levels in Alzheimer’s disease: Correlation with the disease progression. Immunol. Lett. 2007, 114, 46–51. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Rodriguez, E.; Sanchez-Juan, P.; Mateo, I.; Llorca, J.; Infante, J.; Garcia-Gorostiaga, I.; Berciano, J.; Combarros, O. Serum levels and genetic variation of TGF-β1 are not associated with Alzheimer’s disease. Acta Neurol. Scand. 2007, 116, 409–412. [Google Scholar] [CrossRef] [PubMed]
- Swardfager, W.; Lanctot, K.; Rothenburg, L.; Wong, A.; Cappell, J.; Herrmann, N. A meta-analysis of cytokines in Alzheimer’s disease. Biol. Psychiatry 2010, 68, 930–941. [Google Scholar] [CrossRef] [PubMed]
- Ueberham, U.; Ueberham, E.; Gruschka, H.; Arendt, T. Altered subcellular location of phosphorylated Smads in Alzheimer’s disease. Eur. J. Neurosci. 2006, 24, 2327–2334. [Google Scholar] [CrossRef] [PubMed]
- Chalmers, K.A.; Love, S. Neurofibrillary tangles may interfere with Smad 2/3 signaling in neurons. J. Neuropathol. Exp. Neurol. 2007, 66, 158–167. [Google Scholar] [CrossRef] [PubMed]
- Tesseur, I.; Zou, K.; Esposito, L.; Bard, F.; Berber, E.; Can, J.V.; Lin, A.H.; Crews, L.; Tremblay, P.; Mathews, P.; et al. Deficiency in neuronal TGF-β signaling promotes neurodegeneration and Alzheimer’s pathology. J. Clin. Invest. 2006, 116, 3060–3069. [Google Scholar] [CrossRef] [PubMed]
- Arosio, B.; Bergamaschini, L.; Galimberti, L.; La Porta, C.; Zanetti, M.; Calabresi, C.; Scarpini, E.; Annoni, G.; Vergani, C. +10 T/C polymorphisms in the gene of transforming growth factor-β1 are associated with neurodegeneration and its clinical evolution. Mech. Ageing Dev. 2007, 128, 553–557. [Google Scholar] [CrossRef]
- Wyss-Coray, T.; Lin, C.; Yan, F.; Yu, G.Q.; Rohde, M.; McConlogue, L.; Masliah, E.; Mucke, L. TGF-β1 promotes microglial amyloid-β clearance and reduces plaque burden in transgenic mice. Nat. Med. 2001, 7, 612–618. [Google Scholar] [CrossRef]
- Hu, Y.; Chen, W.; Wu, L.; Jiang, L.; Liang, N.; Tan, L.; Liang, M.; Tang, N. TGF-β1 Restores Hippocampal Synaptic Plasticity and Memory in Alzheimer Model via the PI3K/Akt/Wnt/β-Catenin Signaling Pathway. J. Mol. Neurosci. 2019, 67, 142–149. [Google Scholar] [CrossRef]
- Wyss-Coray, T.; Masliah, E.; Mallory, M.; McConlogue, L.; Johnson-Wood, K.; Lin, C.; Mucke, L. Amyloidogenic role of cytokine TGF-β1 in transgenic mice and in Alzheimer’s disease. Nature 1997, 389, 603–606. [Google Scholar] [CrossRef]
- Gaertner, R.F.; Wyss-Coray, T.; Von Euw, D.; Lesne, S.; Vivien, D.; Lacombe, P. Reduced brain tissue perfusion in TGF-β1 transgenic mice showing Alzheimer’s disease-like cerebrovascular abnormalities. Neurobiol. Dis. 2005, 19, 38–46. [Google Scholar] [CrossRef] [PubMed]
- Ongali, B.; Nicolakakis, N.; Lecrux, C.; Aboulkassim, T.; Rosa-Neto, P.; Papadopoulos, P.; Tong, X.K.; Hamel, E. Transgenic mice overexpressing APP and transforming growth factor-β1 feature cognitive and vascular hallmarks of Alzheimer’s disease. Am. J. Pathol. 2010, 177, 3071–3080. [Google Scholar] [CrossRef] [PubMed]
- Town, T.; Laouar, Y.; Pittenger, C.; Mori, T.; Szekely, C.A.; Tan, J.; Duman, R.S.; Flavell, R.A. Blocking TGF-β-Smad2/3 innate immune signaling mitigates Alzheimer-like pathology. Nat. Med. 2008, 14, 681–687. [Google Scholar] [CrossRef] [PubMed]
- Grammas, P.; Ovase, R. Cerebrovascular transforming growth factor-β contributes to inflammation in the Alzheimer’s disease brain. Am. J. Pathol. 2002, 160, 1583–1587. [Google Scholar] [CrossRef]
- Chen, P.Y.; Qin, L.; Li, G.; Wang, Z.; Dahlman, J.E.; Malagon-Lopez, J.; Gujja, S.; Kauffman, K.J.; Sun, L.; Sun, H.; et al. Endothelial TGF-β signalling drives vascular inflammation and atherosclerosis. Nat. Metab. 2019, 1, 912–926. [Google Scholar] [CrossRef] [PubMed]
- Hodge, R.D.; Bakken, T.E.; Miller, J.A.; Smith, K.A.; Barkan, E.R.; Graybuck, L.T.; Close, J.L.; Long, B.; Johansen, N.; Penn, O.; et al. Conserved cell types with divergent features in human versus mouse cortex. Nature 2019. [Google Scholar] [CrossRef] [PubMed]
- Burks, T.N.; Cohn, R.D. Role of TGF-β signaling in inherited and acquired myopathies. Skelet Muscle 2011, 1, 19. [Google Scholar] [CrossRef]
- Chen, J.L.; Colgan, T.D.; Walton, K.L.; Gregorevic, P.; Harrison, C.A. The TGF-β Signalling Network in Muscle Development, Adaptation and Disease. Adv. Exp. Med. Biol. 2016, 900, 97–131. [Google Scholar] [CrossRef]
- Hepple, R.T. Muscle atrophy is not always sarcopenia. J. Appl. Physiol. (1985) 2012, 113, 677–679. [Google Scholar] [CrossRef] [Green Version]
- Carlson, M.E.; Hsu, M.; Conboy, I.M. Imbalance between pSmad3 and Notch induces CDK inhibitors in old muscle stem cells. Nature 2008, 454, 528–532. [Google Scholar] [CrossRef] [Green Version]
- Leger, B.; Derave, W.; De Bock, K.; Hespel, P.; Russell, A.P. Human sarcopenia reveals an increase in SOCS-3 and myostatin and a reduced efficiency of Akt phosphorylation. Rejuvenation Res. 2008, 11, 163–175B. [Google Scholar] [CrossRef] [PubMed]
- Carlson, M.E.; Conboy, M.J.; Hsu, M.; Barchas, L.; Jeong, J.; Agrawal, A.; Mikels, A.J.; Agrawal, S.; Schaffer, D.V.; Conboy, I.M. Relative roles of TGF-β1 and Wnt in the systemic regulation and aging of satellite cell responses. Aging Cell 2009, 8, 676–689. [Google Scholar] [CrossRef] [PubMed]
- Rathbone, C.R.; Yamanouchi, K.; Chen, X.K.; Nevoret-Bell, C.J.; Rhoads, R.P.; Allen, R.E. Effects of transforming growth factor-beta (TGF-β1) on satellite cell activation and survival during oxidative stress. J. Muscle Res. Cell Motil. 2011, 32, 99–109. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.; Wang, J.L.; Lu, J.; Song, Y.; Kwak, K.S.; Jiao, Q.; Rosenfeld, R.; Chen, Q.; Boone, T.; Simonet, W.S.; et al. Reversal of cancer cachexia and muscle wasting by ActRIIB antagonism leads to prolonged survival. Cell 2010, 142, 531–543. [Google Scholar] [CrossRef] [PubMed]
- Baumann, A.P.; Ibebunjo, C.; Grasser, W.A.; Paralkar, V.M. Myostatin expression in age and denervation-induced skeletal muscle atrophy. J. Musculoskelet. Neuronal Interact. 2003, 3, 8–16. [Google Scholar] [PubMed]
- Biernacka, A.; Frangogiannis, N.G. Aging and Cardiac Fibrosis. Aging Dis. 2011, 2, 158–173. [Google Scholar]
- Kim, J.; Lee, J. Role of transforming growth factor-β in muscle damage and regeneration: Focused on eccentric muscle contraction. J. Exerc. Rehabil. 2017, 13, 621–626. [Google Scholar] [CrossRef]
- Jemal, A.; Bray, F.; Center, M.M.; Ferlay, J.; Ward, E.; Forman, D. Global cancer statistics. CA Cancer J. Clin. 2011, 61, 69–90. [Google Scholar] [CrossRef] [Green Version]
- Vincent, H.K.; Raiser, S.N.; Vincent, K.R. The aging musculoskeletal system and obesity-related considerations with exercise. Ageing Res. Rev. 2012, 11, 361–373. [Google Scholar] [CrossRef] [Green Version]
- Lee, M.J. Transforming growth factor beta superfamily regulation of adipose tissue biology in obesity. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 1160–1171. [Google Scholar] [CrossRef]
- Ambrosi, T.H.; Scialdone, A.; Graja, A.; Gohlke, S.; Jank, A.M.; Bocian, C.; Woelk, L.; Fan, H.; Logan, D.W.; Schurmann, A.; et al. Adipocyte Accumulation in the Bone Marrow during Obesity and Aging Impairs Stem Cell-Based Hematopoietic and Bone Regeneration. Cell Stem Cell 2017, 20, 771–784.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choy, L.; Skillington, J.; Derynck, R. Roles of autocrine TGF-β receptor and Smad signaling in adipocyte differentiation. J. Cell Biol. 2000, 149, 667–682. [Google Scholar] [CrossRef] [PubMed]
- Yadav, H.; Quijano, C.; Kamaraju, A.K.; Gavrilova, O.; Malek, R.; Chen, W.; Zerfas, P.; Zhigang, D.; Wright, E.C.; Stuelten, C.; et al. Protection from obesity and diabetes by blockade of TGF-β/Smad3 signaling. Cell Metab. 2011, 14, 67–79. [Google Scholar] [CrossRef] [PubMed]
- Tan, C.K.; Leuenberger, N.; Tan, M.J.; Yan, Y.W.; Chen, Y.; Kambadur, R.; Wahli, W.; Tan, N.S. Smad3 deficiency in mice protects against insulin resistance and obesity induced by a high-fat diet. Diabetes 2011, 60, 464–476. [Google Scholar] [CrossRef] [PubMed]
- Yan, J.; Zhang, H.; Yin, Y.; Li, J.; Tang, Y.; Purkayastha, S.; Li, L.; Cai, D. Obesity- and aging-induced excess of central transforming growth factor-β potentiates diabetic development via an RNA stress response. Nat. Med. 2014, 20, 1001–1008. [Google Scholar] [CrossRef] [PubMed]
- Fain, J.N.; Tichansky, D.S.; Madan, A.K. Transforming growth factor β1 release by human adipose tissue is enhanced in obesity. Metabolism 2005, 54, 1546–1551. [Google Scholar] [CrossRef] [PubMed]
- Moerman, E.J.; Teng, K.; Lipschitz, D.A.; Lecka-Czernik, B. Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: The role of PPAR-γ2 transcription factor and TGF-β/BMP signaling pathways. Aging Cell 2004, 3, 379–389. [Google Scholar] [CrossRef]
- David, C.J.; Massague, J. Contextual determinants of TGFβ action in development, immunity and cancer. Nat. Rev. Mol. Cell Biol. 2018, 19, 419–435. [Google Scholar] [CrossRef]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Tominaga, K.; Suzuki, H.I. TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. Int. J. Mol. Sci. 2019, 20, 5002. https://doi.org/10.3390/ijms20205002
Tominaga K, Suzuki HI. TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. International Journal of Molecular Sciences. 2019; 20(20):5002. https://doi.org/10.3390/ijms20205002
Chicago/Turabian StyleTominaga, Kana, and Hiroshi I. Suzuki. 2019. "TGF-β Signaling in Cellular Senescence and Aging-Related Pathology" International Journal of Molecular Sciences 20, no. 20: 5002. https://doi.org/10.3390/ijms20205002
APA StyleTominaga, K., & Suzuki, H. I. (2019). TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. International Journal of Molecular Sciences, 20(20), 5002. https://doi.org/10.3390/ijms20205002