研究生: |
鍾易翰 Patrick, Choong |
---|---|
論文名稱: |
系統生物學方法探討MSC分化與中心體的細胞間機制 A systems biology study on intercellular mechanism of MSCs differentiation in correlation with centrosome |
指導教授: |
林澤
Lin, Che |
口試委員: |
郭津岑
Kuo, Jean-Cheng 李佳霖 Lee, Jia-Lin 王仰高 Wang, Yang-Kao |
學位類別: |
碩士 Master |
系所名稱: |
電機資訊學院 - 通訊工程研究所 Communications Engineering |
論文出版年: | 2018 |
畢業學年度: | 106 |
語文別: | 英文 |
論文頁數: | 59 |
中文關鍵詞: | 系統生物 、中心體 、蛋白質互動網絡 、骨髓間質幹細胞 |
外文關鍵詞: | proteins-proteins-interactions, MSCs, systems-biology |
相關次數: | 點閱:3 下載:0 |
分享至: |
查詢本校圖書館目錄 查詢臺灣博碩士論文知識加值系統 勘誤回報 |
背景:
骨髓間質乾細胞(MSCs)是首先在骨髓中發現的非造血幹細胞群。各種外部因素有助於MSCs分化的微妙平衡。近年來,人們發現在人工製造的不同硬度的基質上培養MSCs可以推動MSCs分化為特定的細胞類型。此外,已知微管(或與微管相關的蛋白質)似乎在MSC成骨分化過程中具有調節作用,並且中心體是微管組織中心之一。這促使我們研究中心體在MSCs分化成不同細胞類型中的作用。為此,我們在MSC上進行了兩組14天的實驗:對照(control)組和加入0.5μM centronone以抑制培養基中的中心體生長的(treatment)組。為了更深入地了解我們的實驗,還在我們的進一步計算研究中對RNA序列進行了取樣和分析。新的證據表明系統生物學中的動態建模和網絡構建可以幫助研究複雜生物系統內的複雜相互作用和機制。我們將系統生物學方法應用於我們的MSCs研究,以探索在中心體存在和不存在期間如何誘導蛋白質 - 蛋白質相互作用(PPI)和途徑。我們的主要目標是研究找出MSCs分化中複雜的細胞內機制。具體而言,我們要找出在中心體存在與否的情況下關鍵生物途徑的交互作用。
結果:
在對照實驗組中,我們觀察到MSC主要分化為成骨細胞,而觀察到處理組主要分化為脂肪細胞。根據時間序列RNA序列數據,我們構建了兩個PPI網絡,捕獲在具有或不具有中心體下MSC分化期間被激活的關鍵途徑和蛋白質。在建構出的MSC control PPI網絡由2396種蛋白質和3997種相互作用組成; MSC treatment PPI網絡由2734種蛋白質和4704種相互作用組成。然後,我們通過PANTHER途徑分類數據庫分析我們建構的PPI網絡。我們的結果顯示,眾所周知的MSCs成骨轉錄因子RUNX2周遭連接到的蛋白多與硬骨發育有關。而MSCs脂肪轉錄因子PPARG與脂肪形成和脂肪相關的蛋白質,例如BMP4,SMAD3和NLK有密切的連接。此外,我們推理了TGF-β/ BMP信號傳導和JAK / STAT信號傳導途徑,是在中心體存在期間觸發以促進MSCs成骨分化。我們還發現據IGF信號通路在沒有中心體的情況下被促進MSCs脂肪形成分化。
結論:
基於我們構建的PPI網絡,途徑分析,我們研究了MSCs分化相對於中心體存在/不存在下的潛在細胞內機制。我們相信,我們的研究結果不僅可以提供對MSCs分化的深入了解,還可以為未來的再生醫學提供更好的組織修復和再生治療策略。
Background:
Mesenchymal stem cells (MSCs) are a non-hematopoietic stem cell population first discovered in bone marrow. A variety of external factors contributes to the delicate balance of MSCs differentiation. In recent years, it was discovered that growing MSCs on artificially made substrates of different stiffness can drive MSCs differentiation into specific cell types over time. Moreover, it is known that microtubules (or proteins associated to microtubules) appears to have a regulatory role on the process of MSCs osteogenic differentiation and centrosome is one of the microtubule-organizing centres. This motivates us to study the role of centrosome in MSCs differentiation into different cell types. For this, we conducted two sets of 14 days experiments on MSCs: the control set and the set in which 0.5μM centrinone was added to inhibit centrosome growth in the culture medium (treatment). To have a deeper understanding into our experiments, RNA-sequence was also sampled and analysed in our further computational studies.
There have been emerging evidences indicating that dynamic modeling and network constructions in systems biology can assist the investigation of complex interactions and mechanisms within complicated biological systems. In this thesis, we applied a systems biology approach into our study of MSCs to explore how protein-protein interactions (PPIs) and pathways are induced during the presence and absence of centrosome. Our main goal is to investigate the complex intracellular mechanisms that dictate the differentiation linages of MSCs. Specifically, we seek to study the crucial signalling pathways and possible cross-talks between these pathways that are involved in the presence and absence of centrosome.
Result:
On the control experiment set, the MSCs were observed to differentiate into mainly osteoblasts, while the treatment set was observed to differentiate mainly into adipocytes. Based on time-course RNA sequence data, we have constructed two refined PPI networks, which capture the crucial pathways and proteins activated during MSCs differentiation with/without centrosome. The resulting MSC control PPI network consists of 2396 proteins and 3997 interactions; the MSC treatment PPI network consists of 2734 proteins and 4704 interactions. We then analyzed the constructed PPI networks through Protein ANalysis THrough Evolutionary Relationships (PANTHER) online pathway classification database. Our results showed that the well-known MSCs osteogenesis marker, RUNX2, were identified to interact with proteins highly related to osteogenic differentiation. MSCs adipogenesis marker, PPARG, was also identified to interact with proteins related to adipogenic and fats related proteins, such as BMP4, SMAD3 and NLK. Furthermore, we also identified TGF-β/BMP signaling and JAK/STAT signaling pathways that may be triggered during the presence of centrosome to promote MSCs osteogenic differentiation. We also identified evidences suggesting IGF signaling pathway is triggered to promote MSCs adipogenic differentiation in the absence of centrosome.
Conclusion:
Based on our constructed PPI networks, pathway analysis, we investigated the underlying mechanism of MSCs differentiation in relation to the presence/absence of centrosome. We believe that our findings could not only provide insight into the differentiation of MSCs but also shed lights towards a better tissue repair and regeneration treatment strategies for future regenerative medicine.
[1] V. Alonso-Goulart et al., ‘Mesenchymal stem cells from human adipose tissue and bone repair: a literature review’, Biotechnol. Res. Innov., Nov. 2017.
[2] M. A. Vodyanik et al., ‘A mesoderm-derived precursor for mesenchymal stem and endothelial cells’, Cell Stem Cell, vol. 7, no. 6, pp. 718–729, Dec. 2010.
[3] C. M. Kolf, E. Cho, and R. S. Tuan, ‘Mesenchymal stromal cells. Biology of adult mesenchymal stem cells: regulation of niche, self-renewal and differentiation’, Arthritis Res. Ther., vol. 9, no. 1, p. 204, 2007.
[4] G. Sheng, ‘The developmental basis of mesenchymal stem/stromal cells (MSCs)’, BMC Dev. Biol., vol. 15, Nov. 2015.
[5] Q. Chen et al., ‘Fate decision of mesenchymal stem cells: adipocytes or osteoblasts?’, Cell Death Differ., vol. 23, no. 7, pp. 1128–1139, Jul. 2016.
[6] M. F. Pittenger et al., ‘Multilineage Potential of Adult Human Mesenchymal Stem Cells’, Science, vol. 284, no. 5411, pp. 143–147, Apr. 1999.
[7] M. A. Scott, V. T. Nguyen, B. Levi, and A. W. James, ‘Current Methods of Adipogenic Differentiation of Mesenchymal Stem Cells’, Stem Cells Dev., vol. 20, no. 10, pp. 1793–1804, Oct. 2011.
[8] S. R. K. Meka, L. A. Chacko, A. Ravi, K. Chatterjee, and V. Ananthanarayanan, ‘Role of Microtubules in Osteogenic Differentiation of Mesenchymal Stem Cells on 3D Nanofibrous Scaffolds’, ACS Biomater. Sci. Eng., vol. 3, no. 4, pp. 551–559, Apr. 2017.
[9] W. Shin, N.-K. Yu, B.-K. Kaang, and K. Rhee, ‘The microtubule nucleation activity of centrobin in both the centrosome and cytoplasm’, Cell Cycle, vol. 14, no. 12, pp. 1925–1931, Jun. 2015.
[10] S. Sankaran, L. M. Starita, A. C. Groen, M. J. Ko, and J. D. Parvin, ‘Centrosomal microtubule nucleation activity is inhibited by BRCA1-dependent ubiquitination’, Mol. Cell. Biol., vol. 25, no. 19, pp. 8656–8668, Oct. 2005.
[11] Y. L. Wong et al., ‘Reversible centriole depletion with an inhibitor of Polo-like kinase 4’, Science, vol. 348, no. 6239, pp. 1155–1160, Jun. 2015.
[12] K. R. Kukurba and S. B. Montgomery, ‘RNA Sequencing and Analysis’, Cold Spring Harb. Protoc., vol. 2015, no. 11, pp. 951–969, Apr. 2015.
[13] R. Morin et al., ‘Profiling the HeLa S3 transcriptome using randomly primed cDNA and massively parallel short-read sequencing’, BioTechniques, vol. 45, no. 1, pp. 81–94, Jul. 2008.
[14] Z. Wang, M. Gerstein, and M. Snyder, ‘RNA-Seq: a revolutionary tool for transcriptomics’, Nat. Rev. Genet., vol. 10, no. 1, pp. 57–63, Jan. 2009.
[15] T. Ideker, T. Galitski, and L. Hood, ‘A NEW APPROACH TO DECODING LIFE: Systems Biology’, Annu. Rev. Genomics Hum. Genet., vol. 2, no. 1, pp. 343–372, 2001.
[16] H. Kitano, ‘Computational systems biology’, Nature, 14-Nov-2002. [Online]. Available: https://www.nature.com/articles/nature01254. [Accessed: 16-Apr-2018].
[17] S. Draghici et al., ‘A systems biology approach for pathway level analysis’, Genome Res., vol. 17, no. 10, pp. 1537–1545, Oct. 2007.
[18] C. Lin et al., ‘The role of TGF-β signaling and apoptosis in innate and adaptive immunity in zebrafish: a systems biology approach’, BMC Syst. Biol., vol. 8, p. 116, Oct. 2014.
[19] F.-Y. Liu et al., ‘Uncovering the regeneration strategies of zebrafish organs: a comprehensive systems biology study on heart, cerebellum, fin, and retina regeneration’, BMC Syst. Biol., vol. 12, no. Suppl 2, Mar. 2018.
[20] C. Lin et al., ‘Robustness analysis on interspecies interaction network for iron and glucose competition between Candida albicans and zebrafish during infection’, BMC Syst. Biol., vol. 8 Suppl 5, p. S6, 2014.
[21] U. H. Trivedi et al., ‘Quality control of next-generation sequencing data without a reference’, Front. Genet., vol. 5, May 2014.
[22] B. Langmead, C. Trapnell, M. Pop, and S. L. Salzberg, ‘Ultrafast and memory-efficient alignment of short DNA sequences to the human genome’, Genome Biol., vol. 10, no. 3, p. R25, 2009.
[23] B. Langmead, ‘Aligning short sequencing reads with Bowtie’, Curr. Protoc. Bioinforma., vol. Chapter 11, p. Unit 11.7, Dec. 2010.
[24] C. Trapnell et al., ‘Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks’, Nat. Protoc., vol. 7, no. 3, pp. 562–578, Mar. 2012.
[25] C. Trapnell et al., ‘Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation’, Nat. Biotechnol., vol. 28, no. 5, pp. 511–515, May 2010.
[26] J. S. Kang, T. Alliston, R. Delston, and R. Derynck, ‘Repression of Runx2 function by TGF-β through recruitment of class II histone deacetylases by Smad3’, EMBO J., vol. 24, no. 14, pp. 2543–2555, Jul. 2005.
[27] L. Wang, L. Yang, M. Debidda, D. Witte, and Y. Zheng, ‘Cdc42 GTPase-activating protein deficiency promotes genomic instability and premature aging-like phenotypes’, Proc. Natl. Acad. Sci., vol. 104, no. 4, pp. 1248–1253, Jan. 2007.
[28] J. Bricambert et al., ‘Impaired histone deacetylases 5 and 6 expression mimics the effects of obesity and hypoxia on adipocyte function’, Mol. Metab., vol. 5, no. 12, pp. 1200–1207, Dec. 2016.
[29] S. H. Raun et al., ‘Rac1 muscle knockout exacerbates the detrimental effect of high-fat diet on insulin-stimulated muscle glucose uptake independently of Akt’, J. Physiol., vol. 596, no. 12, pp. 2283–2299.
[30] ‘Cell Shape, Cytoskeletal Tension, and RhoA Regulate Stem Cell Lineage Commitment - ScienceDirect’. [Online]. Available: https://www.sciencedirect.com/science/article/pii/S1534580704000759. [Accessed: 27-Jun-2018].
[31] J. Li, ‘JAK-STAT and bone metabolism’, JAK-STAT, vol. 2, no. 3, Jul. 2013.
[32] G. Chen, C. Deng, and Y.-P. Li, ‘TGF-β and BMP Signaling in Osteoblast Differentiation and Bone Formation’, Int. J. Biol. Sci., vol. 8, no. 2, pp. 272–288, Jan. 2012.
[33] M. Aouadi, K. Laurent, M. Prot, Y. L. Marchand-Brustel, B. Binétruy, and F. Bost, ‘Inhibition of p38MAPK Increases Adipogenesis From Embryonic to Adult Stages’, Diabetes, vol. 55, no. 2, pp. 281–289, Feb. 2006.
[34] J. Zhu, E. Shimizu, X. Zhang, N. C. Partridge, and L. Qin, ‘EGFR signaling suppresses osteoblast differentiation and inhibits expression of master osteoblastic transcription factors Runx2 and Osterix’, J. Cell. Biochem., vol. 112, no. 7, pp. 1749–1760, Jul. 2011.
[35] F. Atashi, A. Modarressi, and M. S. Pepper, ‘The role of reactive oxygen species in mesenchymal stem cell adipogenic and osteogenic differentiation: a review’, Stem Cells Dev., vol. 24, no. 10, pp. 1150–1163, May 2015.
[36] J. Xu, Z. Li, Y. Hou, and W. Fang, ‘Potential mechanisms underlying the Runx2 induced osteogenesis of bone marrow mesenchymal stem cells’, Am. J. Transl. Res., vol. 7, no. 12, pp. 2527–2535, Dec. 2015.
[37] A. W. James, ‘Review of Signaling Pathways Governing MSC Osteogenic and Adipogenic Differentiation’, Scientifica, 2013. [Online]. Available: https://www.hindawi.com/journals/scientifica/2013/684736/. [Accessed: 21-Apr-2018].
[38] C. Bruedigam et al., ‘A new concept underlying stem cell lineage skewing that explains the detrimental effects of thiazolidinediones on bone’, Stem Cells Dayt. Ohio, vol. 28, no. 5, pp. 916–927, May 2010.
[39] B. A. Hug, ‘HDAC4: a corepressor controlling bone development’, Cell, vol. 119, no. 4, pp. 448–449, Nov. 2004.
[40] T. Lei et al., ‘HES1 inhibits adipogenesis of porcine mesenchymal stem cells via transcriptional repression of FAD24’, Domest. Anim. Endocrinol., vol. 45, no. 1, pp. 28–32, Jul. 2013.
[41] M. Wang, H. Jin, D. Tang, S. Huang, M. J. Zuscik, and D. Chen, ‘Smad1 plays an essential role in bone development and postnatal bone formation’, Osteoarthritis Cartilage, vol. 19, no. 6, pp. 751–762, Jun. 2011.
[42] B. Aranda-Orgilles et al., ‘MED12 Regulates HSC-Specific Enhancers Independently of Mediator Kinase Activity to Control Hematopoiesis’, Cell Stem Cell, vol. 19, no. 6, pp. 784–799, Dec. 2016.
[43] J.-E. Huh and S. Y. Lee, ‘IL-6 is produced by adipose-derived stromal cells and promotes osteogenesis’, Biochim. Biophys. Acta BBA - Mol. Cell Res., vol. 1833, no. 12, pp. 2608–2616, Dec. 2013.
[44] S. Itoh et al., ‘A critical role for interleukin-6 family-mediated Stat3 activation in osteoblast differentiation and bone formation’, Bone, vol. 39, no. 3, pp. 505–512, Sep. 2006.
[45] O. Levy, E. Ruvinov, T. Reem, Y. Granot, and S. Cohen, ‘Highly efficient osteogenic differentiation of human mesenchymal stem cells by eradication of STAT3 signaling’, Int. J. Biochem. Cell Biol., vol. 42, no. 11, pp. 1823–1830, Nov. 2010.
[46] Y. Tsurutani et al., ‘The roles of transforming growth factor-β and Smad3 signaling in adipocyte differentiation and obesity’, Biochem. Biophys. Res. Commun., vol. 407, no. 1, pp. 68–73, Apr. 2011.
[47] T. Alliston, L. Choy, P. Ducy, G. Karsenty, and R. Derynck, ‘TGF-β-induced repression of CBFA1 by Smad3 decreases cbfa1 and osteocalcin expression and inhibits osteoblast differentiation’, EMBO J., vol. 20, no. 9, pp. 2254–2272, May 2001.
[48] K. Hino et al., ‘Master Regulator for Chondrogenesis, Sox9, Regulates Transcriptional Activation of the Endoplasmic Reticulum Stress Transducer BBF2H7/CREB3L2 in Chondrocytes’, J. Biol. Chem., vol. 289, no. 20, pp. 13810–13820, May 2014.
[49] S. Stöckl, R. J. Bauer, A. K. Bosserhoff, C. Göttl, J. Grifka, and S. Grässel, ‘Sox9 modulates cell survival and adipogenic differentiation of multipotent adult rat mesenchymal stem cells’, J. Cell Sci., vol. 126, no. Pt 13, pp. 2890–2902, Jul. 2013.
[50] P. Hallenborg et al., ‘MDM2 facilitates adipocyte differentiation through CRTC-mediated activation of STAT3’, Cell Death Dis., vol. 7, no. 6, p. e2289, Jun. 2016.
[51] K. Nishikawa et al., ‘Maf promotes osteoblast differentiation in mice by mediating the age-related switch in mesenchymal cell differentiation’, J. Clin. Invest., vol. 120, no. 10, pp. 3455–3465, Oct. 2010.
[52] T. Komori, ‘Regulation of osteoblast differentiation by Runx2’, Adv. Exp. Med. Biol., vol. 658, pp. 43–49, 2010.
[53] V. Schönitzer et al., ‘Sox2 is a potent inhibitor of osteogenic and adipogenic differentiation in human mesenchymal stem cells’, Cell. Reprogramming, vol. 16, no. 5, pp. 355–365, Oct. 2014.
[54] ‘Distinct functions of Sox2 control self-renewal and differentiation in the osteoblast lineage. - PubMed - NCBI’. [Online]. Available: https://www.ncbi.nlm.nih.gov/pubmed/21930787. [Accessed: 06-Feb-2018].
[55] X. Wei et al., ‘Effects of bone morphogenetic protein-4 (BMP-4) on adipocyte differentiation from mouse adipose-derived stem cells’, Cell Prolif., vol. 46, no. 4, pp. 416–424, Aug. 2013.
[56] M. Kortenjann et al., ‘Abnormal bone marrow stroma in mice deficient for nemo-like kinase, Nlk’, Eur. J. Immunol., vol. 31, no. 12, pp. 3580–3587, Dec. 2001.
[57] ‘PPAR- Signaling Crosstalk in Mesenchymal Stem Cells’. [Online]. Available: https://www.hindawi.com/journals/ppar/2010/341671/. [Accessed: 06-Feb-2018].
[58] I. Takada, Y. Yogiashi, and S. Kato, ‘Signaling Crosstalk between PPARγ and BMP2 in Mesenchymal Stem Cells’, PPAR Research, 2012. [Online]. Available: https://www.hindawi.com/journals/ppar/2012/607141/. [Accessed: 24-Mar-2018].
[59] Y.-H. Lee, W. G. Tharp, R. L. Maple, S. Nair, P. A. Permana, and R. E. Pratley, ‘Amyloid precursor protein expression is upregulated in adipocytes in obesity’, Obes. Silver Spring Md, vol. 16, no. 7, pp. 1493–1500, Jul. 2008.
[60] A. Kumar, B. P. Salimath, M. Schieker, G. B. Stark, and G. Finkenzeller, ‘Inhibition of metastasis-associated gene 1 expression affects proliferation and osteogenic differentiation of immortalized human mesenchymal stem cells’, Cell Prolif., vol. 44, no. 2, pp. 128–138, Apr. 2011.
[61] V. Nicolaidou et al., ‘Monocytes Induce STAT3 Activation in Human Mesenchymal Stem Cells to Promote Osteoblast Formation’, PLoS ONE, vol. 7, no. 7, Jul. 2012.
[62] J. M. Stephens, R. F. Morrison, and P. F. Pilch, ‘The expression and regulation of STATs during 3T3-L1 adipocyte differentiation’, J. Biol. Chem., vol. 271, no. 18, pp. 10441–10444, May 1996.
[63] J. B. Harp, D. Franklin, A. A. Vanderpuije, and J. M. Gimble, ‘Differential expression of signal transducers and activators of transcription during human adipogenesis’, Biochem. Biophys. Res. Commun., vol. 281, no. 4, pp. 907–912, Mar. 2001.
[64] D. Wang et al., ‘Signal transducer and activator of transcription 3 (STAT3) regulates adipocyte differentiation via peroxisome-proliferator-activated receptor gamma (PPARgamma)’, Biol. Cell, vol. 102, no. 1, pp. 1–12, Sep. 2009.
[65] A. J. Richard and J. M. Stephens, ‘Emerging roles of JAK–STAT signaling pathways in adipocytes’, Trends Endocrinol. Metab. TEM, vol. 22, no. 8, pp. 325–332, Aug. 2011.
[66] H. H. Luu et al., ‘Distinct roles of bone morphogenetic proteins in osteogenic differentiation of mesenchymal stem cells’, J. Orthop. Res., vol. 25, no. 5, pp. 665–677.
[67] K. Miyazono, Y. Kamiya, and M. Morikawa, ‘Bone morphogenetic protein receptors and signal transduction’, J. Biochem. (Tokyo), vol. 147, no. 1, pp. 35–51, Jan. 2010.
[68] T. Matsubara et al., ‘BMP2 Regulates Osterix through Msx2 and Runx2 during Osteoblast Differentiation’, J. Biol. Chem., vol. 283, no. 43, pp. 29119–29125, Oct. 2008.
[69] G. L. Lin and K. D. Hankenson, ‘Integration of BMP, Wnt, and notch signaling pathways in osteoblast differentiation’, J. Cell. Biochem., vol. 112, no. 12, pp. 3491–3501, Dec. 2011.
[70] M. Wu, G. Chen, and Y.-P. Li, ‘TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease’, Bone Res., vol. 4, p. 16009, 2016.
[71] L. Choy and R. Derynck, ‘Transforming Growth Factor-β Inhibits Adipocyte Differentiation by Smad3 Interacting with CCAAT/Enhancer-binding Protein (C/EBP) and Repressing C/EBP Transactivation Function’, J. Biol. Chem., vol. 278, no. 11, pp. 9609–9619, Mar. 2003.
[72] S. Blüher, J. Kratzsch, and W. Kiess, ‘Insulin-like growth factor I, growth hormone and insulin in white adipose tissue’, Best Pract. Res. Clin. Endocrinol. Metab., vol. 19, no. 4, pp. 577–587, Dec. 2005.
[73] J. Boucher et al., ‘Differential Roles of Insulin and IGF-1 Receptors in Adipose Tissue Development and Function’, Diabetes, vol. 65, no. 8, pp. 2201–2213, Aug. 2016.
[74] ‘Review of Signaling Pathways Governing MSC Osteogenic and Adipogenic Differentiation’. [Online]. Available: https://www.hindawi.com/journals/scientifica/2013/684736/. [Accessed: 27-Mar-2018].
[75] G. H. Reference, ‘PIK3CA gene’, Genetics Home Reference. [Online]. Available: https://ghr.nlm.nih.gov/gene/PIK3CA. [Accessed: 27-Mar-2018].
[76] K. Tawaramoto et al., ‘Ablation of 3-phosphoinositide-dependent protein kinase 1 (PDK1) in vascular endothelial cells enhances insulin sensitivity by reducing visceral fat and suppressing angiogenesis’, Mol. Endocrinol. Baltim. Md, vol. 26, no. 1, pp. 95–109, Jan. 2012.