메뉴 건너뛰기




Volumn 111, Issue , 2016, Pages 152-154

Perspective: Targeting the JAK/STAT pathway to fight age-related dysfunction

Author keywords

Cellular senescence; Frailty; Insulin resistance; Senolytics

Indexed keywords

CHEMOKINE; CYTOKINE; GROWTH FACTOR; JANUS KINASE; JANUS KINASE INHIBITOR; PROTEINASE; STAT PROTEIN;

EID: 84975840886     PISSN: 10436618     EISSN: 10961186     Source Type: Journal    
DOI: 10.1016/j.phrs.2016.05.015     Document Type: Review
Times cited : (59)

References (51)
  • 1
    • 84874603171 scopus 로고    scopus 로고
    • Cellular senescence and the senescent secretory phenotype: therapeutic opportunities
    • [1] Tchkonia, T., Zhu, Y., van Deursen, J., Campisi, J., Kirkland, J.L., Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J. Clin. Invest. 123:3 (2013), 966–972.
    • (2013) J. Clin. Invest. , vol.123 , Issue.3 , pp. 966-972
    • Tchkonia, T.1    Zhu, Y.2    van Deursen, J.3    Campisi, J.4    Kirkland, J.L.5
  • 2
    • 0344622606 scopus 로고
    • The serial cultivation of human diploid cell strains
    • [2] Hayflick, L., Moorhead, P.S., The serial cultivation of human diploid cell strains. Exp. Cell Res. 25 (1961), 585–621.
    • (1961) Exp. Cell Res. , vol.25 , pp. 585-621
    • Hayflick, L.1    Moorhead, P.S.2
  • 3
    • 84904702784 scopus 로고    scopus 로고
    • Cellular senescence: from physiology to pathology
    • [3] Munoz-Espin, D., Serrano, M., Cellular senescence: from physiology to pathology. Nat. Rev. Mol. Cell Biol. 15:7 (2014), 482–496.
    • (2014) Nat. Rev. Mol. Cell Biol. , vol.15 , Issue.7 , pp. 482-496
    • Munoz-Espin, D.1    Serrano, M.2
  • 5
    • 84962079683 scopus 로고    scopus 로고
    • Cellular senescence in type 2 diabetes: a therapeutic opportunity
    • [5] Palmer, A.K., et al. Cellular senescence in type 2 diabetes: a therapeutic opportunity. Diabetes 64:7 (2015), 2289–2298.
    • (2015) Diabetes , vol.64 , Issue.7 , pp. 2289-2298
    • Palmer, A.K.1
  • 6
    • 0029047362 scopus 로고
    • A biomarker that identifies senescent human cells in culture and in aging skin in vivo
    • [6] Dimri, G.P., et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. U. S. A. 92:20 (1995), 9363–9367.
    • (1995) Proc. Natl. Acad. Sci. U. S. A. , vol.92 , Issue.20 , pp. 9363-9367
    • Dimri, G.P.1
  • 7
    • 0035079864 scopus 로고    scopus 로고
    • Replicative senescence in normal liver, chronic hepatitis C, and hepatocellular carcinomas
    • [7] Paradis, V., et al. Replicative senescence in normal liver, chronic hepatitis C, and hepatocellular carcinomas. Hum. Pathol. 32:3 (2001), 327–332.
    • (2001) Hum. Pathol. , vol.32 , Issue.3 , pp. 327-332
    • Paradis, V.1
  • 8
    • 66149167336 scopus 로고    scopus 로고
    • DNA damage response and cellular senescence in tissues of aging mice
    • [8] Wang, C., et al. DNA damage response and cellular senescence in tissues of aging mice. Aging Cell 8:3 (2009), 311–323.
    • (2009) Aging Cell , vol.8 , Issue.3 , pp. 311-323
    • Wang, C.1
  • 9
    • 0037850928 scopus 로고    scopus 로고
    • Cell senescence in rat kidneys in vivo increases with growth and age despite lack of telomere shortening
    • [9] Melk, A., et al. Cell senescence in rat kidneys in vivo increases with growth and age despite lack of telomere shortening. Kidney Int. 63:6 (2003), 2134–2143.
    • (2003) Kidney Int. , vol.63 , Issue.6 , pp. 2134-2143
    • Melk, A.1
  • 10
    • 24644497236 scopus 로고    scopus 로고
    • Cellular senescence in vivo: its relevance in aging and cardiovascular disease
    • [10] Erusalimsky, J.D., Kurz, D.J., Cellular senescence in vivo: its relevance in aging and cardiovascular disease. Exp. Gerontol. 40:8–9 (2005), 634–642.
    • (2005) Exp. Gerontol. , vol.40 , Issue.8–9 , pp. 634-642
    • Erusalimsky, J.D.1    Kurz, D.J.2
  • 11
    • 78349299099 scopus 로고    scopus 로고
    • Fat tissue, aging, and cellular senescence
    • [11] Tchkonia, T., et al. Fat tissue, aging, and cellular senescence. Aging Cell 9:5 (2010), 667–684.
    • (2010) Aging Cell , vol.9 , Issue.5 , pp. 667-684
    • Tchkonia, T.1
  • 12
    • 84905705301 scopus 로고    scopus 로고
    • Growth hormone action predicts age-related white adipose tissue dysfunction and senescent cell burden in mice
    • [12] Stout, M.B., et al. Growth hormone action predicts age-related white adipose tissue dysfunction and senescent cell burden in mice. Aging (Albany NY) 6:7 (2014), 575–586.
    • (2014) Aging (Albany NY) , vol.6 , Issue.7 , pp. 575-586
    • Stout, M.B.1
  • 13
    • 84891713034 scopus 로고    scopus 로고
    • Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor
    • [13] Coppe, J.P., et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6:12 (2008), 2853–2868.
    • (2008) PLoS Biol. , vol.6 , Issue.12 , pp. 2853-2868
    • Coppe, J.P.1
  • 14
    • 44649101304 scopus 로고    scopus 로고
    • Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network
    • [14] Kuilman, T., et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 133:6 (2008), 1019–1031.
    • (2008) Cell , vol.133 , Issue.6 , pp. 1019-1031
    • Kuilman, T.1
  • 15
    • 80855138775 scopus 로고    scopus 로고
    • Clearance of p16Ink4a-positive senescent cells delays aging-associated disorders
    • [15] Baker, D.J., et al. Clearance of p16Ink4a-positive senescent cells delays aging-associated disorders. Nature 479:7372 (2011), 232–236.
    • (2011) Nature , vol.479 , Issue.7372 , pp. 232-236
    • Baker, D.J.1
  • 16
    • 84986218395 scopus 로고    scopus 로고
    • Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice
    • [16] Roos, C.M., et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell, 2016.
    • (2016) Aging Cell
    • Roos, C.M.1
  • 17
    • 84928243456 scopus 로고    scopus 로고
    • The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs
    • [17] Zhu, Y., et al. The Achilles’ heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 14:4 (2015), 644–658.
    • (2015) Aging Cell , vol.14 , Issue.4 , pp. 644-658
    • Zhu, Y.1
  • 18
    • 84881399206 scopus 로고    scopus 로고
    • A complex secretory program orchestrated by the inflammasome controls paracrine senescence
    • [18] Acosta, J.C., et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell Biol. 15:8 (2013), 978–990.
    • (2013) Nat. Cell Biol. , vol.15 , Issue.8 , pp. 978-990
    • Acosta, J.C.1
  • 19
    • 84862794254 scopus 로고    scopus 로고
    • A senescent cell bystander effect: senescence-induced senescence
    • [19] Nelson, G., et al. A senescent cell bystander effect: senescence-induced senescence. Aging Cell 11:2 (2012), 345–349.
    • (2012) Aging Cell , vol.11 , Issue.2 , pp. 345-349
    • Nelson, G.1
  • 20
    • 84988603034 scopus 로고    scopus 로고
    • Targeting senescent cells enhances adipogenesis and metabolic function in old age
    • [20] Xu, M., et al. Targeting senescent cells enhances adipogenesis and metabolic function in old age. Elife, 4, 2015, e12997.
    • (2015) Elife , vol.4 , pp. e12997
    • Xu, M.1
  • 21
    • 84966397603 scopus 로고    scopus 로고
    • JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age
    • [21] Xu, M., et al. JAK inhibition alleviates the cellular senescence-associated secretory phenotype and frailty in old age. Proc. Natl. Acad. Sci. U. S. A. 112:46 (2015), E6301–6310.
    • (2015) Proc. Natl. Acad. Sci. U. S. A. , vol.112 , Issue.46 , pp. E6301-6310
    • Xu, M.1
  • 22
    • 84898990827 scopus 로고    scopus 로고
    • Molecular pathways: molecular basis for sensitivity and resistance to JAK kinase inhibitors
    • [22] Meyer, S.C., Levine, R.L., Molecular pathways: molecular basis for sensitivity and resistance to JAK kinase inhibitors. Clin. Cancer Res. 20:8 (2014), 2051–2059.
    • (2014) Clin. Cancer Res. , vol.20 , Issue.8 , pp. 2051-2059
    • Meyer, S.C.1    Levine, R.L.2
  • 23
    • 70350500225 scopus 로고    scopus 로고
    • STATs in cancer inflammation and immunity: a leading role for STAT3
    • [23] Yu, H., Pardoll, D., Jove, R., STATs in cancer inflammation and immunity: a leading role for STAT3. Nat. Rev. Cancer 9:11 (2009), 798–809.
    • (2009) Nat. Rev. Cancer , vol.9 , Issue.11 , pp. 798-809
    • Yu, H.1    Pardoll, D.2    Jove, R.3
  • 24
    • 79960913754 scopus 로고    scopus 로고
    • Emerging roles of JAK-STAT signaling pathways in adipocytes
    • [24] Richard, A.J., Stephens, J.M., Emerging roles of JAK-STAT signaling pathways in adipocytes. Trends Endocrinol. Metab. 22:8 (2011), 325–332.
    • (2011) Trends Endocrinol. Metab. , vol.22 , Issue.8 , pp. 325-332
    • Richard, A.J.1    Stephens, J.M.2
  • 25
    • 0028206119 scopus 로고
    • JAK3: a novel JAK kinase associated with terminal differentiation of hematopoietic cells
    • [25] Rane, S.G., Reddy, E.P., JAK3: a novel JAK kinase associated with terminal differentiation of hematopoietic cells. Oncogene 9:8 (1994), 2415–2423.
    • (1994) Oncogene , vol.9 , Issue.8 , pp. 2415-2423
    • Rane, S.G.1    Reddy, E.P.2
  • 26
    • 61849086101 scopus 로고    scopus 로고
    • Janus kinases in immune cell signaling
    • [26] Ghoreschi, K., Laurence, A., O'Shea, J.J., Janus kinases in immune cell signaling. Immunol. Rev. 228:1 (2009), 273–287.
    • (2009) Immunol. Rev. , vol.228 , Issue.1 , pp. 273-287
    • Ghoreschi, K.1    Laurence, A.2    O'Shea, J.J.3
  • 27
    • 0033696404 scopus 로고    scopus 로고
    • Partial impairment of cytokine responses in Tyk2-deficient mice
    • [27] Karaghiosoff, M., et al. Partial impairment of cytokine responses in Tyk2-deficient mice. Immunity 13:4 (2000), 549–560.
    • (2000) Immunity , vol.13 , Issue.4 , pp. 549-560
    • Karaghiosoff, M.1
  • 28
    • 0033711641 scopus 로고    scopus 로고
    • Tyk2 plays a restricted role in IFN alpha signaling, although it is required for IL-12-mediated T cell function
    • [28] Shimoda, K., et al. Tyk2 plays a restricted role in IFN alpha signaling, although it is required for IL-12-mediated T cell function. Immunity 13:4 (2000), 561–571.
    • (2000) Immunity , vol.13 , Issue.4 , pp. 561-571
    • Shimoda, K.1
  • 29
    • 17644424955 scopus 로고    scopus 로고
    • A gain-of-function mutation of JAK2 in myeloproliferative disorders
    • [29] Kralovics, R., et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N. Engl. J. Med. 352:17 (2005), 1779–1790.
    • (2005) N. Engl. J. Med. , vol.352 , Issue.17 , pp. 1779-1790
    • Kralovics, R.1
  • 30
    • 84878951826 scopus 로고    scopus 로고
    • Safety and efficacy of CYT387, a JAK1 and JAK2 inhibitor, in myelofibrosis
    • [30] Pardanani, A., et al. Safety and efficacy of CYT387, a JAK1 and JAK2 inhibitor, in myelofibrosis. Leukemia 27:6 (2013), 1322–1327.
    • (2013) Leukemia , vol.27 , Issue.6 , pp. 1322-1327
    • Pardanani, A.1
  • 31
    • 76949083372 scopus 로고    scopus 로고
    • Lestaurtinib, a multitargeted tyrosine kinase inhibitor: from bench to bedside
    • [31] Shabbir, M., Stuart, R., Lestaurtinib, a multitargeted tyrosine kinase inhibitor: from bench to bedside. Expert Opin. Invest. Drugs 19:3 (2010), 427–436.
    • (2010) Expert Opin. Invest. Drugs , vol.19 , Issue.3 , pp. 427-436
    • Shabbir, M.1    Stuart, R.2
  • 32
    • 80755140046 scopus 로고    scopus 로고
    • SB1518, a novel macrocyclic pyrimidine-based JAK2 inhibitor for the treatment of myeloid and lymphoid malignancies
    • [32] Hart, S., et al. SB1518, a novel macrocyclic pyrimidine-based JAK2 inhibitor for the treatment of myeloid and lymphoid malignancies. Leukemia 25:11 (2011), 1751–1759.
    • (2011) Leukemia , vol.25 , Issue.11 , pp. 1751-1759
    • Hart, S.1
  • 33
    • 77954531991 scopus 로고    scopus 로고
    • Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050
    • [33] Fridman, J.S., et al. Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J. Immunol. 184:9 (2010), 5298–5307.
    • (2010) J. Immunol. , vol.184 , Issue.9 , pp. 5298-5307
    • Fridman, J.S.1
  • 34
    • 79955027327 scopus 로고    scopus 로고
    • Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550)
    • [34] Ghoreschi, K., et al. Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J. Immunol. 186:7 (2011), 4234–4243.
    • (2011) J. Immunol. , vol.186 , Issue.7 , pp. 4234-4243
    • Ghoreschi, K.1
  • 35
    • 84907998102 scopus 로고    scopus 로고
    • Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity
    • [35] Toso, A., et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 9:1 (2014), 75–89.
    • (2014) Cell Rep. , vol.9 , Issue.1 , pp. 75-89
    • Toso, A.1
  • 36
    • 85042055496 scopus 로고    scopus 로고
    • Pharmacologic inhibition of JAK-STAT signaling promotes hair growth
    • [36] Harel, S., et al. Pharmacologic inhibition of JAK-STAT signaling promotes hair growth. Sci. Adv., 1(9), 2015, e1500973.
    • (2015) Sci. Adv. , vol.1 , Issue.9 , pp. e1500973
    • Harel, S.1
  • 37
    • 84912099331 scopus 로고    scopus 로고
    • Inhibition of JAK-STAT signaling stimulates adult satellite cell function
    • [37] Price, F.D., et al. Inhibition of JAK-STAT signaling stimulates adult satellite cell function. Nat. Med. 20:10 (2014), 1174–1181.
    • (2014) Nat. Med. , vol.20 , Issue.10 , pp. 1174-1181
    • Price, F.D.1
  • 38
    • 77950684805 scopus 로고    scopus 로고
    • Preclinical characterization of the selective JAK1/2 inhibitor INCB04: therapeutic implications for the treatment of myeloproliferative neoplasms
    • [38] Quintas-Cardama, A., Preclinical characterization of the selective JAK1/2 inhibitor INCB04: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood 115:15 (2010), 3109–3117.
    • (2010) Blood , vol.115 , Issue.15 , pp. 3109-3117
    • Quintas-Cardama, A.1
  • 39
    • 84958093401 scopus 로고    scopus 로고
    • Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan
    • [39] Baker, D.J., et al. Naturally occurring p16(Ink4a)-positive cells shorten healthy lifespan. Nature 530:7589 (2016), 184–189.
    • (2016) Nature , vol.530 , Issue.7589 , pp. 184-189
    • Baker, D.J.1
  • 40
    • 84877822086 scopus 로고    scopus 로고
    • Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-kappaB activation
    • [40] Moiseeva, O., et al. Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-kappaB activation. Aging Cell 12:3 (2013), 489–498.
    • (2013) Aging Cell , vol.12 , Issue.3 , pp. 489-498
    • Moiseeva, O.1
  • 41
    • 84938751873 scopus 로고    scopus 로고
    • MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation
    • [41] Laberge, R.M., et al. MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation. Nat. Cell Biol. 17:8 (2015), 1049–1061.
    • (2015) Nat. Cell Biol. , vol.17 , Issue.8 , pp. 1049-1061
    • Laberge, R.M.1
  • 42
    • 84864010194 scopus 로고    scopus 로고
    • Glucocorticoids suppress selected components of the senescence-associated secretory phenotype
    • [42] Laberge, R.M., et al. Glucocorticoids suppress selected components of the senescence-associated secretory phenotype. Aging Cell 11:4 (2012), 569–578.
    • (2012) Aging Cell , vol.11 , Issue.4 , pp. 569-578
    • Laberge, R.M.1
  • 43
    • 67650944993 scopus 로고    scopus 로고
    • Rapamycin fed late in life extends lifespan in genetically heterogeneous mice
    • [43] Harrison, D.E., et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 460:7253 (2009), 392–395.
    • (2009) Nature , vol.460 , Issue.7253 , pp. 392-395
    • Harrison, D.E.1
  • 44
    • 84881247539 scopus 로고    scopus 로고
    • Rapamycin extends murine lifespan but has limited effects on aging
    • [44] Neff, F., et al. Rapamycin extends murine lifespan but has limited effects on aging. J. Clin. Invest. 123:8 (2013), 3272–3291.
    • (2013) J. Clin. Invest. , vol.123 , Issue.8 , pp. 3272-3291
    • Neff, F.1
  • 45
    • 84881347302 scopus 로고    scopus 로고
    • Metformin improves healthspan and lifespan in mice
    • [45] Martin-Montalvo, A., et al. Metformin improves healthspan and lifespan in mice. Nat. Commun., 4, 2013, 2192.
    • (2013) Nat. Commun. , vol.4 , pp. 2192
    • Martin-Montalvo, A.1
  • 46
    • 84863393110 scopus 로고    scopus 로고
    • A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis
    • [46] Verstovsek, S., et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N. Engl. J. Med. 366:9 (2012), 799–807.
    • (2012) N. Engl. J. Med. , vol.366 , Issue.9 , pp. 799-807
    • Verstovsek, S.1
  • 47
    • 84896985687 scopus 로고    scopus 로고
    • Proinflammatory cytokines, aging, and age-related diseases
    • [47] Michaud, M., et al. Proinflammatory cytokines, aging, and age-related diseases. J. Am. Med. Dir. Assoc. 14:12 (2013), 877–882.
    • (2013) J. Am. Med. Dir. Assoc. , vol.14 , Issue.12 , pp. 877-882
    • Michaud, M.1
  • 48
    • 77956696835 scopus 로고    scopus 로고
    • Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis
    • [48] Verstovsek, S., et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N. Engl. J. Med. 363:12 (2010), 1117–1127.
    • (2010) N. Engl. J. Med. , vol.363 , Issue.12 , pp. 1117-1127
    • Verstovsek, S.1
  • 49
    • 84941264776 scopus 로고    scopus 로고
    • Clinical strategies and animal models for developing senolytic agents
    • [49] Kirkland, J.L., Tchkonia, T., Clinical strategies and animal models for developing senolytic agents. Exp. Gerontol. 68 (2015), 19–25.
    • (2015) Exp. Gerontol. , vol.68 , pp. 19-25
    • Kirkland, J.L.1    Tchkonia, T.2
  • 50
    • 84919480323 scopus 로고    scopus 로고
    • An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA
    • [50] Demaria, M., et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev. Cell 31:6 (2014), 722–733.
    • (2014) Dev. Cell , vol.31 , Issue.6 , pp. 722-733
    • Demaria, M.1
  • 51
    • 84960156528 scopus 로고    scopus 로고
    • p16-induced senescence of pancreatic beta cells enhances insulin secretion
    • [51] Helman, A., et al. p16-induced senescence of pancreatic beta cells enhances insulin secretion. Nat. Med., 2016.
    • (2016) Nat. Med.
    • Helman, A.1


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.