Fan2016 Tissue Repair and Regeneration PDF
Fan2016 Tissue Repair and Regeneration PDF
Fan2016 Tissue Repair and Regeneration PDF
REGENERATIVE MEDICINE
Tissue repair and regenerative medicine address the important medical needs to replace damaged tissue with func-
tional tissue. Most regenerative medicine strategies have focused on delivering biomaterials and cells, yet there is
the untapped potential for drug-induced regeneration with good specificity and safety profiles. The Hippo pathway
is a key regulator of organ size and regeneration by inhibiting cell proliferation and promoting apoptosis. Kinases
MST1 and MST2 (MST1/2), the mammalian Hippo orthologs, are central components of this pathway and are, there-
cascade that play a pivotal role in stem cell self-renewal, tissue regenera- the melting temperature (Fig. 1F). Furthermore, with increasing
tion, and organ size control (9–17). Central to this pathway is a kinase ATP concentration, XMU-MP-1 exhibited a proportional increase
cascade formed by kinases MST1/2; a scaffolding protein, Salvador/ in IC50 against MST1/2 (Fig. 1G), as well as an attenuated inhibition
WW45 (Sav); nuclear Dbf2–related family kinases LATS1 and of the MST2-mediated phosphorylation of MOB1 (fig. S3E). Together
LATS2 (LATS1/2); and an adaptor protein, MOB1. MST1/2 phos- with substrate kinetic analyses (fig. S3F), these data suggest that XMU-
phorylates and activates LATS1/2–MOB1, which then phosphorylates MP-1 is an ATP-competitive inhibitor for MST1/2.
Yes-associated protein/transcriptional coactivator with PDZ-binding Kinase selectivity of XMU-MP-1 was determined using KINO-
motif (YAP/TAZ). Phospho-YAP/TAZ is either degraded or sequest- MEscan, which profiled the inhibitor at a concentration of 1 mM
ered in the cytoplasm by the 14-3-3 protein. When the Hippo path- against a panel of 468 diverse kinases using an in vitro ATP-site com-
way is off, YAP/TAZ translocates to the nucleus and forms a functional petition binding assay (20). The KINOMEscan results are reported as
hybrid transcriptional factor with TEA domain transcription factor the percentage of the DMSO negative control signal (ctrl%) set at
(TEAD) to turn on pro-proliferative and prosurvival genes, thereby 100%, and a lower number represents higher-affinity binding. There
enabling cell proliferation. Genetic defects in this pathway in mice lead were 23 kinases with ctrl% ≤10% in total containing MST1-4, indicat-
to sustained tissue growth and can lead to cancer (12, 13, 16). Al- ing very strong inhibition. The dissociation constants (Kd) of XMU-
though Hippo kinase MST1/2 structures have been determined, MP-1 against these kinases were determined (Fig. 1H and tables S1
small-molecule MST1/2 inhibitors have yet been disclosed (18, 19). and S2). A selectivity score [S(10)] of 0.05 was determined for XMU-
Therefore, small molecules targeting Hippo signaling in a reversible MP-1, calculated by dividing the number of kinases with ctrl% ≤10%
(Fig. 2F). Consistently, Western blotting analysis indicated that the washing (Fig. 2, G and H, and fig. S4, E and F). These data establish
XMU-MP-1 treatment greatly reduced the phosphorylation levels of that XMU-MP-1 can potently and reversibly suppress the activities of
MOB1 and YAP, which were restored to normal levels 2 hours after kinases MST1/2 and enhance their downstream YAP activation in cells.
Fig. 2. Inhibition of
MST1/2 signaling by
XMU-MP-1 in vitro in var-
ious cell types. (A) Relative
levels of p-MOB1 and p-
MST in a variety of cell lines
or primary mouse hepato-
cytes stimulated with or
without H2O2 after treat-
ment with various con-
centrations of XMU-MP-1.
(B) Immunoblot analysis
of YAP, poly(ADP-ribose)
polymerase (PARP), and
a-tubulin, and quantifica-
tion of relative levels of
YAP in cytoplasmic (c) and
nuclear (n) fractions of hu-
Cocrystal structure and SAR reveal that XMU-MP-1 is We next explored whether the pharmacological inhibition of MST1/2
on-target to MST2 would similarly potentiate liver repair and regeneration in vivo. The
To study how XMU-MP-1 recognizes the MST1/2 kinases, we pharmacokinetic properties of XMU-MP-1 were first evaluated in
determined the protein/inhibitor complex structure via cocrystallization. Sprague-Dawley rats administered a single intravenous or oral dose.
MST2 in this complex adopted the typical active conformation closely XMU-MP-1 exhibited favorable pharmacokinetics with a half-life of
resembling the wild-type MST1 crystal structure [Protein Data Bank 1.2 hours, an area under the curve of 1035 (h·ng)/ml, and a bio-
(PDB) 3COM; fig. S5A] as evidenced by the extended activation loop availability of 39.5% (table S3). In pharmacodynamic experiments,
(A-loop) and the phosphorylated Thr174 and Thr180 residues (residues the maximal phosphorylation inhibition of MOB1 and YAP was
177 and 183 in MST1) (Fig. 3A). In contrast, the previously reported achieved between 1.5 and 6 hours after intraperitoneal dosing with
apo-bound and AMP-PNP–bound kinase–dead mutant MST2 D146N XMU-MP-1 (1 mg/kg) (Fig. 4A). A dose escalation study of XMU-
structures (PDB 4LG4 and 4LGD) were in the inactive conformation MP-1 revealed that the phosphorylation of MOB1 in liver tissue
(fig. S5B) (18, 19). XMU-MP-1 bound deeply into the ATP-binding was blocked at a minimal dose (1 mg/kg, intraperitoneally) (Fig. 4B).
pocket, sandwiched between the N- and C-lobes, and closely associated Liver-specific deletion of MST1/2 using Alb-Cre (Mst1fl/flMst2fl/fl–
with the hinge of the kinase (Fig. 3B and fig. S5C). The pyrimido[4,5-b] Alb-Cre) resulted in a two- to threefold increase in liver/body weight
thieno[3,2-e][1,4]diazepine scaffold adopted a bent conformation that ratio and overexpansion of bile duct–like liver progenitor cells (oval
optimally fit the hydrophobic Val41 and Leu153 side chains; the phenyl cells) compared to control wild-type mice (Mst1fl/flMst2fl/fl) within
ring of the compound contacted the side chain of Leu33 through a hy- 2 months (Fig. 4C). Wild-type mice treated with XMU-MP-1 daily for
Fig. 4. Inhibition of MST1/2 signaling by XMU-MP-1 in mouse liver. sections (n = 3) (D) or liver/body weight ratio (n = 6) (E) of wild-type mice
(A and B) Immunoblots and quantification of p-MOB1 and p-YAP in lysates treated with a vehicle or XMU-MP-1 (1 mg/kg) daily for 2 months, or after
of livers from wild-type mice treated with XMU-MP-1 (1 mg/kg) over time 2 weeks of XMU-MP-1 withdrawal. ND, not determined. Data are means ±
(0 to 12 hours) (A) or different doses of XMU-MP-1 for 3 hours (B). Data are SD. P values were determined by Student’s t test. Scale bars, 100 mm.
means ± SD relative to DMSO controls (n = 3). *P < 0.05, **P < 0.01, ***P < (F) Ki67 staining in tissue sections from wild-type mice treated with a
0.001 versus control, Student’s t test. GAPDH, glyceraldehyde-3-phosphate vehicle or XMU-MP-1 (3 mg/kg) daily for 7 days. Quantification of the Ki67-
dehydrogenase. (C) Hematoxylin and eosin (H&E) or immunohistochemical positive cells in various tissues. Data are means ± SD (n = 6). P values were
(IHC) staining of the oval cell marker CK19 in liver sections of Mst1fl/flMst2fl/fl determined by Student’s t test. Scale bars, 100 mm. Data are from one ex-
and Mst1fl/flMst2fl/fl–Alb-Cre mice. Scale bars, 100 mm. (D and E) H&E and periment representative of three (A to C) or two (D to F) independent
IHC staining and quantification of CK19- and BrdU-positive cells in liver experiments. Full blots are shown in fig. S11.
NAC (250 mg/kg) administered at 1.5 hours, but not at 2.5 or marily by helping to maintain colonocyte proliferation in damaged
3.5 hours after APAP administration, protected mice from liver cen- mucosa.
trilobular necrosis (Fig. 6A and fig. S10A). Extending this therapeutic
window, XMU-MP-1 administered within 2.5 hours after APAP ad- XMU-MP-1 ameliorates chronic liver injury
ministration effectively prevented the continual expansion of centri- Next, we evaluated the regenerative effect of XMU-MP-1 in chronic
lobular necrotic lesions in APAP-treated mouse livers (Fig. 6B and liver injury, which is of great clinical interest. The two most commonly
fig. S10, B to D). NAC treatment alone at 1.5 hours after APAP ad- used models of experimental chronic liver injury are iterative toxic
ministration treated about 80% of mice, whereas the combination of damage (for example, elicited by CCl4 intoxication) and bile duct
NAC and XMU-MP-1 (1 mg/kg) at 1.5 hours after APAP administra- ligation (35). Chronic liver injury was induced by the administration
tion protected 100% of mice from APAP-induced death (Fig. 6A). The of CCl4 twice per week for 4 weeks (Fig. 7A) or by bile duct ligation
combination of NAC and XMU-MP-1, or XMU-MP-1 treatment (Fig. 7B). Liver fibrosis is the common result of chronic hepatic injury.
alone, effectively extended the protective effects to 2.5 hours after Liver fibrosis and cell death were assessed by Sirius Red and TUNEL
APAP administration, as shown by the increased survival rate from staining, respectively. In the model of CCl4-induced liver chronic in-
~40% (NAC alone or vehicle control) to ~75% (Fig. 6B), as well as jury, mice treated with XMU-MP-1 (1 mg/kg) daily for 10 days
the smaller liver centrilobular necrotic lesions compared with the con- (starting at day 30 after toxic insult) had a small amount of collagen
trol groups (Fig. 6C and fig. S10D). Furthermore, compared with the deposition as well as few dead cells, whereas mice treated with vehicle
vehicle controls, all treatments given at either 1.5 or 2.5 hours after control demonstrated bridging fibrosis and significantly more TUNEL-
the damaged tissue repair and regeneration but not affect intact, Liver fibrosis occurs as an attempt to limit tissue damage in re-
healthy organs. Moreover, we demonstrated that the inhibitory ef- sponse to chronic liver injury. However, progressive fibrosis eventu-
fect of XMU-MP-1 on MST1/2 is reversible. Thus, XMU-MP-1 appears ally results in cirrhosis. Antifibrotic therapies should inhibit matrix
to be safe as a targeted therapeutic to treat tissue injury. deposition, collagen synthesis, and hepatic stellate cell activation. A
Fig. 7. XMU-MP-1
potentiates tissue re-
pair and regeneration
after chronic liver inju-
ry. (A and B) Schematics
for CCl4-induced (A) and
bile duct ligation (BDL)–
induced (B) chronic liver
injury and fibrosis evalu-
ation in mice. (C) Sirius
Red and TUNEL staining
andcorrespondingquan-
tifications in liver sec-
tions 10 days after CCl4
administration. Injured
mice were treated with
a vehicle or XMU-MP-1
(1 mg/kg) for 10 days.
previous study showed that macrophage depletion in mice during in- blinded to genotype and/or treatment arm. For drug treatments, age-
jury resulted in a marked loss of stellate cells and matrix deposition and gender-matched animals were randomly assigned to treatment
(40); thus, macrophages are likely essential for regeneration. Addition- arms with about equivalent numbers in each group. Box-and-whisker
ally, reactive oxygen species could augment fibrogenesis (41). We pre- plots were identified using RStudio-defined outliers (shown as circles),
viously found that MST1/2 kinases are essential for macrophage but all data points were used in statistical analyses.
activation and that the elimination of MST1/2 from the liver resulted
in increased expression of enzymes that scavenge oxidants (25, 42). General synthesis of XMU-MP-1 and its analogs
Another study showed that the Hippo downstream effector protein MST1/2 kinase inhibitor XMU-MP-1 and its analogs were synthesized
YAP promoted stellate cell activation (43). The complex interplay be- from a starting mixture of methyl 3-aminothiophene-2-carboxylate,
tween MST1/2 kinase activity and cellular activity during regeneration, diisopropylethylamine, and 2,4-dichloro-5-nitropyrimidine in isopro-
evidenced by these contradictory findings, might explain why pharma- panol, as described in Supplementary Methods (figs. S2 and S8A). For
cological inhibition of MST1/2 results in only a partial rescue of liver biochemical assays and cellular studies, XMU-MP-1 was dissolved in
fibrosis in mice. Thus, more work is needed to determine the antifi- DMSO (stock concentration, 10 mM). For animal treatments, XMU-
brotic mechanisms of XMU-MP-1 and to further optimize the dosage MP-1 was dissolved in 0.1% citric acid aqueous solution containing
to achieve better antifibrotic efficacy. 20% Kolliphor HS 15.
Together, our results show that XMU-MP-1 is a valuable chemical
probe to study the Hippo signaling pathway, and it provides a starting In vitro and in vivo kinase inhibition assays
(Invitrogen). RT-qPCR was performed using a Bio-Rad iQ SYBR D. Mikkola, J. Naidoo, S. Wei, H.-H. Tai, S. L. Gerson, J. M. Ready, B. Posner, J. K. V. Willson,
S. D. Markowitz, Inhibition of the prostaglandin-degrading enzyme 15-PGDH potentiates
Green Supermix Kit and the Bio-Rad iCycler iQ system (Bio-Rad).
tissue regeneration. Science 348, aaa2340 (2015).
All runs were accompanied by the internal control Gapdh gene. The 5. Y. Zhang, I. Strehin, K. Bedelbaeva, D. Gourevitch, L. Clark, J. Leferovich, P. B. Messersmith,
samples were run in triplicate and normalized to GAPDH using a DD E. Heber-Katz, Drug-induced regeneration in adult mice. Sci. Transl. Med. 7, 290ra92 (2015).
cycle threshold–based algorithm to provide arbitrary units represent- 6. B. D. Polizzotti, B. Ganapathy, S. Walsh, S. Choudhury, N. Ammanamanchi, D. G. Bennett,
ing relative expression levels. The primer sequences for specific genes C. G. dos Remedios, B. J. Haubner, J. M. Penninger, B. Kühn, Neuregulin stimulation of
cardiomyocyte regeneration in mice and human myocardium reveals a therapeutic
are shown in table S6. window. Sci. Transl. Med. 7, 281ra45 (2015).
7. Y. Tian, Y. Liu, T. Wang, N. Zhou, J. Kong, L. Chen, M. Snitow, M. Morley, D. Li, N. Petrenko,
Statistical analysis S. Zhou, M. Lu, E. Gao, W. J. Koch, K. M. Stewart, E. E. Morrisey, A microRNA-Hippo
All statistical analyses were performed using Prism 5 (GraphPad pathway that promotes cardiomyocyte proliferation and cardiac regeneration in mice.
Sci. Transl. Med. 7, 279ra38 (2015).
Software). The liver weights and liver/body weight ratios, BrdU or
8. N. Shyh-Chang, H. Zhu, T. Yvanka de Soysa, G. Shinoda, M. T. Seligson, K. M. Tsanov, L. Nguyen,
TUNEL labeling count, relative mRNA levels, and quantification of J. M. Asara, L. C. Cantley, G. Q. Daley, Lin28 enhances tissue repair by reprogramming cellular
immune blots were tabulated graphically with error bars metabolism. Cell 155, 778–792 (2013).
corresponding to means ± SD and compared using two-tailed Stu- 9. S. J. Rawat, J. Chernoff, Regulation of mammalian Ste20 (Mst) kinases. Trends Biochem. Sci.
dent’s t test. Body weight changes and DAI scores in the DSS model 40, 149–156 (2015).
10. J. Avruch, D. Zhou, J. Fitamant, N. Bardeesy, F. Mou, L. R. Barrufet, Protein kinases of the
with the different treatment arms were compared across the time Hippo pathway: Regulation and substrates. Semin. Cell Dev. Biol. 23, 770–784 (2012).
course of study using two-tailed paired-samples Student’s t test. Sur-
30. R. Yang, K. Miki, X. He, M. E. Killeen, M. P. Fink, Prolonged treatment with N-acetylcystine 42. F. Mou, M. Praskova, F. Xia, D. Van Buren, H. Hock, J. Avruch, D. Zhou, The Mst1 and Mst2
delays liver recovery from acetaminophen hepatotoxicity. Crit. Care 13, R55 (2009). kinases control activation of rho family GTPases and thymic egress of mature thymocytes.
31. Y. Xie, A. Ramachandran, D. G. Breckenridge, J. T. Liles, M. Lebofsky, A. Farhood, H. Jaeschke, J. Exp. Med. 209, 741–759 (2012).
Inhibitor of apoptosis signal-regulating kinase 1 protects against acetaminophen-induced liver 43. I. Mannaerts, S. B. Leite, S. Verhulst, S. Claerhout, N. Eysackers, L. F. R. Thoen, A. Hoorens,
injury. Toxicol. Appl. Pharmacol. 286, 1–9 (2015). H. Reynaert, G. Halder, L. A. van Grunsven, The Hippo pathway effector YAP controls
32. D. Zhou, Y. Zhang, H. Wu, E. Barry, Y. Yin, E. Lawrence, D. Dawson, J. E. Willis, S. D. Markowitz, mouse hepatic stellate cell activation. J. Hepatol. 63, 679–688 (2015).
F. D. Camargo, J. Avruch, Mst1 and Mst2 protein kinases restrain intestinal stem cell prolifer-
ation and colonic tumorigenesis by inhibition of Yes-associated protein (Yap) overabundance. Acknowledgments: We thank J. Avruch for his discussions and comments on the manuscript.
Proc. Natl. Acad. Sci. U.S.A. 108, E1312–E1320 (2011). Funding: This work was supported by the National Basic Research Program (973) of China
33. J. Cai, N. Zhang, Y. Zheng, R. F. de Wilde, A. Maitra, D. Pan, The Hippo signaling pathway [2015CB910502 (to L.C.) and 2012CB917202 (to C.-H.Y.)], the National Natural Science Foundation
restricts the oncogenic potential of an intestinal regeneration program. Genes Dev. 24, of China [31270918, U1505224, and J1310027 (to D.Z.); 81422045, U1405223, and 21272195
2383–2388 (2010). (to X.D.); 81372617, 81422018, and U1405225 (to L.C.); 31270769 (to C.-H.Y.); and 81472229 (to
34. H. S. Cooper, S. N. Murthy, R. S. Shah, D. J. Sedergran, Clinicopathologic study of dextran L.H.)], the 111 Projects (B12001 and B06016), the Fundamental Research Funds for the Central
sulfate sodium experimental murine colitis. Lab. Invest. 69, 238–249 (1993). Universities of China-Xiamen University [CXB2014004 (to Z.J.), 20720140551 (to L.C.),
35. J. P. Iredale, Models of liver fibrosis: Exploring the dynamic nature of inflammation and 20720160064 and 2013121032 (to X.D.), and 2013121034 and 20720140537 (to D.Z.)]. The
repair in a solid organ. J. Clin. Invest. 117, 539–548 (2007). funders had no role in the study design, data collection and analysis, decision to publish, or
36. S. Jiao, H. Wang, Z. Shi, A. Dong, W. Zhang, X. Song, F. He, Y. Wang, Z. Zhang, W. Wang, preparation of the manuscript. Author contributions: D.Z., X.D., C.-H.Y., and L.C. conceived
X. Wang, T. Guo, P. Li, Y. Zhao, H. Ji, L. Zhang, Z. Zhou, A peptide mimicking VGLL4 the project. D.Z., L.C., X.D., and F.F. performed data analysis/statistics. X.D., N.S.G., and Z.H.
function acts as a YAP antagonist therapy against gastric cancer. Cancer Cell 25, 166–180 conceived and performed chemical synthesis of XMU-MP-1 and its analogs and small-molecule
(2014). structure determination. L.-L.K. and C.-H.Y. designed and performed cocrystal structure study of
SUPPLEMENTARY http://stm.sciencemag.org/content/suppl/2016/08/15/8.352.352ra108.DC1
MATERIALS
RELATED http://stm.sciencemag.org/content/scitransmed/8/357/357ra122.full
CONTENT
http://science.sciencemag.org/content/sci/356/6342/1020.full
http://stke.sciencemag.org/content/sigtrans/11/547/eaaj1757.full
REFERENCES This article cites 43 articles, 12 of which you can access for free
http://stm.sciencemag.org/content/8/352/352ra108#BIBL
PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions
Science Translational Medicine (ISSN 1946-6242) is published by the American Association for the Advancement of
Science, 1200 New York Avenue NW, Washington, DC 20005. 2017 © The Authors, some rights reserved; exclusive
licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. The
title Science Translational Medicine is a registered trademark of AAAS.