リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「HSP47 levels determine the degree of body adiposity」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

HSP47 levels determine the degree of body adiposity

Shin, Jihoon 大阪大学

2023.11.11

概要

Title

HSP47 levels determine the degree of body
adiposity

Author(s)

Shin, Jihoon; Toyoda, Shinichiro; Okuno, Yosuke
et al.

Citation

Nature Communications. 2023, 14, p. 7319

Version Type VoR
URL
rights

https://hdl.handle.net/11094/93378
This article is licensed under a Creative
Commons Attribution 4.0 International License.

Note

Osaka University Knowledge Archive : OUKA
https://ir.library.osaka-u.ac.jp/
Osaka University

Article

https://doi.org/10.1038/s41467-023-43080-x

HSP47 levels determine the degree of body
adiposity
Received: 9 March 2023
Accepted: 31 October 2023

Jihoon Shin 1,2,3 , Shinichiro Toyoda1, Yosuke Okuno1, Reiko Hayashi1,
Shigeki Nishitani1, Toshiharu Onodera1,4, Haruyo Sakamoto1, Shinya Ito5,
Sachiko Kobayashi 1, Hirofumi Nagao 1, Shunbun Kita 1,6, Michio Otsuki1,7,
Atsunori Fukuhara 1,6, Kazuhiro Nagata8,9 & Iichiro Shimomura1

Adiposity varies among individuals with the influence of diverse physiological,
pathological, environmental, hormonal, and genetic factors, but a unified
molecular basis remains elusive. Here, we identify HSP47, a collagen-specific
chaperone, as a key determinant of body adiposity. HSP47 expression is
abundant in adipose tissue; increased with feeding, overeating, and obesity;
decreased with fasting, exercise, calorie restriction, bariatric surgery, and
cachexia; and correlated with fat mass, BMI, waist, and hip circumferences.
Insulin and glucocorticoids, respectively, up- and down-regulate HSP47
expression. In humans, the increase of HSP47 gene expression by its intron or
synonymous variants is associated with higher body adiposity traits. In mice,
the adipose-specific knockout or pharmacological inhibition of HSP47 leads to
lower body adiposity compared to the control. Mechanistically, HSP47 promotes collagen dynamics in the folding, secretion, and interaction with
integrin, which activates FAK signaling and preserves PPARγ protein from
proteasomal degradation, partly related to MDM2. The study highlights the
significance of HSP47 in determining the amount of body fat individually and
under various circumstances.

1234567890():,;

1234567890():,;

Check for updates

Adipose tissue, composed of adipocytes, is a specialized organ to store
energy as fat, thus commonly referred to as fat tissue1. It is distributed
throughout the body, including subcutaneous fat beneath the skin,
visceral fat around internal organs, and brown fat in the back, neck,
and shoulder area2. Fat tissue can dynamically change its size and mass
in response to nutritional and hormonal states. During eating or
overeating conditions, increased blood insulin levels stimulate glucose
uptake and lipid accumulation in fat cells, promoting the expansion of
fat tissue3. Conversely, fasting or starving conditions induce glucocorticoid hormone, which triggers the lipolysis of stored fat, leading to

a reduction in fat tissue4. The peroxisome proliferator-activated
receptor-γ (PPARγ) is a master regulator of fat cells5. Defects in
PPARγ result in the loss of fat tissue6–9, while its activation by agonizts,
such as pioglitazone, promotes lipid storage and the expansion of fat
tissue10,11. The unique properties of fat tissue and its regulation by
various nutritional, hormonal, and molecular factors have been well
established, yet the intricate connections between these elements is
not fully understood.
Adiposity, referring to the amount or percentage of body fat,
varies greatly between individuals, ranging from as low as 5% in very

1

Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan. 2Department of Diabetes Care Medicine, Graduate
School of Medicine, Osaka University, Suita, Osaka, Japan. 3Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and
Harvard Medical School, Boston, MA, USA. 4Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical
Center, Dallas, USA. 5Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan. 6Department of Adipose Management, Graduate School of Medicine,
Osaka University, Suita, Osaka, Japan. 7Department of Endocrinology, Graduate School of Medical Science, Tokyo Women’s Medical University, Tokyo, Japan.
8
Institute for Protein Dynamics, Kyoto Sangyo University, Kyoto, Japan. 9JT Biohistory Research Hall, Osaka, Japan.
e-mail: shinjihoon0209@gmail.com

Nature Communications | (2023)14:7319

1

Article
thin/lean people to over 40% in the cases of morbid obesity12. Additionally, each person exhibits a unique susceptibility to gain body fat
under regular eating or overeating conditions13. These individual variations are influenced by a combination of physiological, environmental, pathological, and genetic factors. Physiologically, eating habits
and patterns, which affect food calorie intake, fasting duration, and
hormone levels, have a significant impact on body adiposity14,15.
Environmentally, engaging in high physical activity or exercise can also
contribute to lower body adiposity14,15. Pathologically, obesity is characterized by high levels of body fat tissue15, while cachexia, a wasting
disorder, leads to a significant loss of fat tissue16. Bariatric surgery has
been shown effective results in reducing body adiposity in patients
with obesity17. Genetic factors, such as single nucleotide polymorphisms (SNPs), also play a role in determining body adiposity traits, such
as fat mass, BMI, waist circumference, and hip circumferences18.
Despite the comprehensive contribution of physiological, pathological, environmental, and genetic backgrounds to body adiposity, a
unified molecular basis for its regulation remains elusive.
In this study, through a series of in silico, in vivo, and in vitro
experiments, we identify HSP47, a collagen-specific molecular
chaperone19,20, as a significant determinant of body adiposity. Our
transcriptomic and genetic analyses indicate that HSP47 is prominently expressed in adipose tissue and adipocytes, with its levels
tightly linked to the degree of body adiposity in humans. Adiposespecific and pharmacological ablation models in mice provide compelling evidence of the causal impact of HSP47 on body fat deposition.
Biochemical assays reveal that HSP47-meidated enhancement of collagen protein dynamics, such as its folding, secretion, and binding in
the extracellular matrix, induce focal adhesion signaling, stabilize
PPARγ protein, and ultimately lead to the expansion of fat tissue. This
research not only deepens our understanding of the individual and
contextual factors that determine body adiposity but also provides a
robust scientific rationale for these mechanisms.

Results
Collagen matrix, focal adhesion, and PPARγ characterize
fat tissue
Each organ/tissue has its unique characteristics, which are defined by
their specific gene expression patterns21. To understand the characteristics of fat tissue in transcription levels, we compared the gene
expression patterns of adipose tissue with those of other 52 different
organs and tissues. We scored the adipose enrichment of each transcript and plotted it against the gene expression level. As expected,
ADIPOQ (Adiponectin), LEP (Leptin), and FABP4 (aP2) genes were
found to be highly enriched and expressed in adipose tissue (Fig. 1a).
We performed gene ontology (GO) analysis on the top 500 genes and
found that components of the collagen-containing ECM and focal
adhesion were enriched in fat tissue (Fig. 1b). Pathway analysis further
revealed that these genes are involved in the PPAR signaling pathway,
ECM-receptor interaction, and focal adhesion (Fig. 1c). Focal adhesion
is subcellular structures composed of multiple proteins, including
Integrins and focal adhesion kinase (FAK), that serves as a link between
the extracellular environment and intracellular compartments22,23.
Therefore, the interactions between extracellular collagen matrix,
subcellular focal adhesion, and intracellular PPARγ may play a crucial
role in shaping the characteristics of adipose tissue.

https://doi.org/10.1038/s41467-023-43080-x

tissue, (3) induced by overeating (8 weeks of high-fat diet) in human
subcutaneous adipose tissue, (4) upregulated by obesity in human
adipocytes, and (5) associated with various adiposity traits in human
genome-wide association study (GWAS), such as waist circumferences,
hip circumferences, waist/hip ratio, obesity, and BMI. Through this
comparative analysis, we identified HSP47, a collagen-specific chaperone encoded by SERPINH1 gene19,20, as a potential determinant of
body adiposity.
Detailed expression profiles revealed that HSP47 gene and protein
are highly expressed in adipose tissues and adipocytes compared with
other tissues, organs, and cells in humans (Fig. 1e, f). Similar expression
patterns were observed in mice (Supplementary Fig. 1a–c). The gene
expression of HSP47 increased in human adipose tissues with feeding
(Fig. 1g) and overeating (Fig. 1h). Individuals with obesity exhibited
higher gene expression of HSP47 in adipocytes (Fig. 1i). There were no
significant sexual differences of HSP47 expression in subcutaneous
and visceral adipose tissues (Supplementary Fig. 2a); both male and
female subjects displayed equivalent induction of HSP47 in obesity
(Supplementary Fig. 2b). We also analyzed adipose tissues from young
adult (22- to 36-year-old) monozygotic twin-pairs with discordant BMI
(inter-pair difference ΔBMI > 3 kg/m2), which showed significant differences in various body adiposity traits such as body weight, fat mass,
and volume26. The gene expression of HSP47 was significantly higher in
the adipose tissue of high-adiposity monozygotic twin pairs compared
to that of low-adiposity twin pairs (Fig. 1j), indicating that HSP47 gene
expression associates with body adiposity independent of genetic
backgrounds. In contrast, HSP47 gene expression was significantly
lower in adipose tissues under low-adiposity conditions, such as
exercise training (Fig. ...

この論文で使われている画像

参考文献

1.

Cohen, P. & Spiegelman, B. M. Cell biology of fat storage. Mol. Biol.

Cell 27, 2523–2527 (2016).

2. Cohen, P. & Kajimura, S. The cellular and functional complexity of

thermogenic fat. Nat. Rev. Mol. Cell Biol. 22, 393–409 (2021).

3. Cheatham, B. & Kahn, C. R. Insulin action and the insulin signaling

network. Endocr. Rev. 16, 117–142 (1995).

4. Djurhuus, C. B. et al. Effects of cortisol on lipolysis and regional

interstitial glycerol levels in humans. Am. J. Physiol. Endocrinol.

Metab. 283, E172–E177 (2002).

5. Rosen, E. D. et al. PPARγ is required for the differentiation of adipose

tissue in vivo and in vitro. Mol. Cell 4, 611–617 (1999).

6. Gilardi, F. et al. Systemic PPARγ deletion in mice provokes lipoatrophy, organomegaly, severe type 2 diabetes and metabolic

inflexibility. Metabolism 95, 8–20 (2019).

7. He, W. et al. Adipose-specific peroxisome proliferator-activated

receptor γ knockout causes insulin resistance in fat and liver but not

in muscle. Proc. Natl Acad. Sci. 100, 15712–15717 (2003).

8. Hegele, R. A., Cao, H., Frankowski, C., Mathews, S. T. & Leff, T.

PPARG F388L, a transactivation-deficient mutant, in familial partial

lipodystrophy. Diabetes 51, 3586–3590 (2002).

9. Stalin, A. et al. Computational analysis of single nucleotide polymorphisms (SNPs) in PPAR gamma associated with obesity, diabetes and cancer. J. Biomol. Struct. Dyn. 40, 1843–1857 (2022).

10. Miyazaki, Y. et al. Effect of pioglitazone on abdominal fat distribution and insulin sensitivity in type 2 diabetic patients. J. Clin.

Endocrinol. Metab. 87, 2784–2791 (2002).

11. Rasouli, N. et al. Pioglitazone improves insulin sensitivity through

reduction in muscle lipid and redistribution of lipid into adipose

tissue. Am. J. Physiol. Endocrinol. Metab. 288, E930–E934

(2005).

12. Gallagher, D. et al. Healthy percentage body fat ranges: an

approach for developing guidelines based on body mass index.

Am. J. Clin. Nutr. 72, 694–701 (2000).

Nature Communications | (2023)14:7319

https://doi.org/10.1038/s41467-023-43080-x

13. Blundell, J. E. et al. Resistance and susceptibility to weight gain:

individual variability in response to a high-fat diet. Physiol. Behav.

86, 614–622 (2005).

14. Marti, A., Moreno-Aliaga, M., Hebebrand, J. & Martinez, J. Genes,

lifestyles and obesity. Int. J. Obes. 28, S29–S36 (2004).

15. Hill, J. O., Wyatt, H. R., Reed, G. W. & Peters, J. C. Obesity and the

environment: where do we go from here. Science 299,

853–855 (2003).

16. Argilés, J. M., Busquets, S., Stemmler, B. & López-Soriano, F. J.

Cancer cachexia: understanding the molecular basis. Nat. Rev.

Cancer 14, 754–762 (2014).

17. Miras, A. D. & le Roux, C. W. Mechanisms underlying weight loss

after bariatric surgery. Nat. Rev. Gastroenterol. Hepatol. 10,

575–584 (2013).

18. Shungin, D. et al. New genetic loci link adipose and insulin biology

to body fat distribution. Nature 518, 187–196 (2015).

19. Nagata, K. Hsp47: a collagen-specific molecular chaperone. Trends

Biochem. Sci. 21, 23–26 (1996).

20. Ito, S. & Nagata, K. Biology of Hsp47 (Serpin H1), a collagen-specific

molecular chaperone. Seminars in cell & developmental biology,

142–151 (2017).

21. Lonsdale, J. et al. The genotype-tissue expression (GTEx) project.

Nat. Genet. 45, 580–585 (2013).

22. Wozniak, M. A., Modzelewska, K., Kwong, L. & Keely, P. J. Focal

adhesion regulation of cell behavior. Biochim. Biophys. Acta Mol.

Cell Res. 1692, 103–119 (2004).

23. Parsons, J. T. Focal adhesion kinase: the first ten years. J. cell Sci.

116, 1409–1416 (2003).

24. Barrett, T. et al. NCBI GEO: archive for functional genomics data sets

—update. Nucleic Acids Res. 41, D991–D995 (2012).

25. Welter, D. et al. The NHGRI GWAS Catalog, a curated resource of

SNP-trait associations. Nucleic Acids Res. 42, D1001–D1006

(2014).

26. Heinonen, S. et al. Mitochondria-related transcriptional signature is

downregulated in adipocytes in obesity: a study of young healthy

MZ twins. Diabetologia 60, 169–181 (2017).

27. Koza, R. A. et al. Changes in gene expression foreshadow dietinduced obesity in genetically identical mice. PLoS Genet. 2,

e81 (2006).

28. Civelek, M. et al. Genetic regulation of adipose gene expression and

cardio-metabolic traits. Am. J. Hum. Genet. 100, 428–443

(2017).

29. Siva, N. 1000 Genomes project. Nat. Biotechnol. 26,

256–257 (2008).

30. Ito, S. et al. A small-molecule compound inhibits a collagenspecific molecular chaperone and could represent a potential

remedy for fibrosis. J. Biol. Chem. 292, 20076–20085 (2017).

31. Cai, H. et al. Identification of HSP47 binding site on native collagen

and its implications for the development of HSP47 inhibitors. Biomolecules 11, 983 (2021).

32. Softic, S. et al. Lipodystrophy due to adipose tissue–specific insulin

receptor knockout results in progressive NAFLD. Diabetes 65,

2187–2200 (2016).

33. Polyzos, S. A., Perakakis, N. & Mantzoros, C. S. Fatty liver in lipodystrophy: a review with a focus on therapeutic perspectives of

adiponectin and/or leptin replacement. Metabolism 96,

66–82 (2019).

34. Shimomura, I., Hammer, R. E., Ikemoto, S., Brown, M. S. & Goldstein,

J. L. Leptin reverses insulin resistance and diabetes mellitus in mice

with congenital lipodystrophy. Nature 401, 73–76 (1999).

35. Stark, C. et al. BioGRID: a general repository for interaction datasets.

Nucleic Acids Res. 34, D535–D539 (2006).

36. Sun, K., Tordjman, J., Clément, K. & Scherer Philipp, E. Fibrosis and

adipose tissue dysfunction. Cell Metab. 18, 470–477 (2013).

13

Article

37. Bosman, F. T. & Stamenkovic, I. Functional structure and composition of the extracellular matrix. J. Pathol. 200, 423–428 (2003).

38. Petäistö, T. et al. Lack of collagen XVIII leads to lipodystrophy and

perturbs hepatic glucose and lipid homeostasis. J. Physiol. 598,

3373–3393 (2020).

39. Yang, J. et al. Collagen β(1-O) galactosyltransferase 2 deficiency

contributes to lipodystrophy and aggravates NAFLD related to

HMW adiponectin in mice. Metabolism 120, 154777 (2021).

40. Ruiz-Ojeda, F. J. et al. Active integrins regulate white adipose tissue

insulin sensitivity and brown fat thermogenesis. Mol. Metab. 45,

101147 (2021).

41. Luk, C. T. et al. FAK signalling controls insulin sensitivity through

regulation of adipocyte survival. Nat. Commun. 8, 14360 (2017).

42. Blüher, M. et al. Role of insulin action and cell size on protein

expression patterns in adipocytes. J. Biol. Chem. 279,

31902–31909 (2004).

43. Hotamisligil, G. S., Shargill, N. S. & Spiegelman, B. M. Adipose

expression of tumor necrosis factor-α: direct role in obesity-linked

insulin resistance. Science 259, 87–91 (1993).

44. Furukawa, S. et al. Increased oxidative stress in obesity and its

impact on metabolic syndrome. J. Clin. Investig. 114,

1752–1761 (2017).

45. Friedman, J. M. & Halaas, J. L. Leptin and the regulation of body

weight in mammals. Nature 395, 763–770 (1998).

46. Hayashi, R. et al. Adipocyte GR inhibits healthy adipose expansion

through multiple mechanisms in cushing syndrome. Endocrinology

160, 504–521 (2019).

47. Masago, Y. et al. The molecular chaperone Hsp47 is essential for

cartilage and endochondral bone formation. J. Cell Sci. 125,

1118–1128 (2012).

Acknowledgements

This work was partly supported by the Japan Society for the Promotion of

Science Grant-in-aid for Scientific Research (grant no. 22K16413) and

Front Runner of Future Diabetes Research (FFDR). We thank all members

of the department of Metabolic Medicine at Graduate School of Medicine, Osaka University.

Author contributions

All authors made direct and/or intellectual contributions to the work and

approved it for publication. J.S. designed the study and researched the

data. J.S. and S.T. performed mouse experiments. J.S. and I.S. wrote the

manuscript. Y.O. and R.H generated and managed AGRKO mice. S.I. and

K.N. provided AdHSP47KO mice and discussed the research. H.S.

Nature Communications | (2023)14:7319

https://doi.org/10.1038/s41467-023-43080-x

performed cell preparation and mouse genotyping. S.T., Y.O, R.H., H.S.,

S.N., T.O., S.K. (Sachiko Kobayashi), H.N., S.K. (Shunbun Kita), O.M., and

A.F. contributed to the discussion and reviewed the manuscript. J.S. is

the guarantor of this work and, as such, had full access to all the data in

the study and takes responsibility for the integrity of the data and the

accuracy of the data analysis.

Competing interests

The authors declare no competing interests.

Additional information

Supplementary information The online version contains

supplementary material available at

https://doi.org/10.1038/s41467-023-43080-x.

Correspondence and requests for materials should be addressed to

Jihoon Shin.

Peer review information Nature Communications thanks the anonymous reviewer(s) for their contribution to the peer review of this work. A

peer review file is available.

Reprints and permissions information is available at

http://www.nature.com/reprints

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons

Attribution 4.0 International License, which permits use, sharing,

adaptation, distribution and reproduction in any medium or format, as

long as you give appropriate credit to the original author(s) and the

source, provide a link to the Creative Commons license, and indicate if

changes were made. The images or other third party material in this

article are included in the article’s Creative Commons license, unless

indicated otherwise in a credit line to the material. If material is not

included in the article’s Creative Commons license and your intended

use is not permitted by statutory regulation or exceeds the permitted

use, you will need to obtain permission directly from the copyright

holder. To view a copy of this license, visit http://creativecommons.org/

licenses/by/4.0/.

© The Author(s) 2023

14

...

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る