Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

CRISPR editing of the PPARGC1A Gly482ser (rs8192678) polymorphism in human white adipose cells shows differential effects on mitochondrial function and adipogenesis.

Huang, M. LU ; Claussnitzer, M. ; Saadat, A. ; Mulder, Hindrik LU orcid ; Kalamajski, Sebastian LU and Franks, Paul LU (2021) European Association for the Study of Diabetes (EASD) 57th Annual meeting In Diabetologia 64(Suppl 1). p.159-159
Abstract
Background and aims: PPARGC1A encodes PGC-1α (peroxisome proliferator-activated receptor γ coactivator 1-α), a central regulator of energy metabolism and mitochondrial function. A common polymorphism in PPARGC1A (rs8192678, C/T, Gly482Ser) has been associated with obesity and related metabolic disorders, but no published functional studies have investigated direct allele-specific effects in adipocyte biology. Materials and methods: We used CRISPR-Cas9 to perform allele switching (C-to-T or T-to-C) at rs8192678 in isogenic human pre-adipocyte white adipose tissue (hWAT) cell line; we then evaluated the allelic effects at rs8192678 on adipogenic differentiation and mitochondrial function. Accordingly, single-cell clones were expanded and... (More)
Background and aims: PPARGC1A encodes PGC-1α (peroxisome proliferator-activated receptor γ coactivator 1-α), a central regulator of energy metabolism and mitochondrial function. A common polymorphism in PPARGC1A (rs8192678, C/T, Gly482Ser) has been associated with obesity and related metabolic disorders, but no published functional studies have investigated direct allele-specific effects in adipocyte biology. Materials and methods: We used CRISPR-Cas9 to perform allele switching (C-to-T or T-to-C) at rs8192678 in isogenic human pre-adipocyte white adipose tissue (hWAT) cell line; we then evaluated the allelic effects at rs8192678 on adipogenic differentiation and mitochondrial function. Accordingly, single-cell clones were expanded and screened to obtain homozygous T/T (482Ser) and C/C (482Gly) isogenic cell populations. The effect of the allele editing on white adipocyte differentiation and on mitochondrial function was then studied in three cell populations of the respective genotype. In ongoing experiments, CRISPR/Cas9 was also used to append a luciferase tag to C/C and in T/T cells. The luciferase will be used as a reporter for the endogenously expressed PGC-1 protein stability, and will therefore provide insights into mechanisms by which rs8192678 alleles affect PGC-1 activity. Results (see figure): At the end of the differentiation protocol the C/C adipocytes were apparently less Oil-Red-O positive than T/T adipocytes under optical microscopy, they had 78.5% lower triglyceride content (p<0.0001, n=9), and lower expression of adipogenic markers (all markers p<0.0001, n=3). Furthermore, C/C adipocytes had lower mitochondrial content (p<0.001, n=9), which coincided with decreased oxygen consumption rate (OCR) at basal (p<0.0001, n=3) and maximal respiration (p<0.0001, n=3). Also, C/C adipocytes had lower ATP-linked OCR (p<0.0001, n=3). Conclusion: Our data showcases discriminatory causal effects of the two rs8192678 alleles in adipocytes. The C allele confers lower PPARGC1A expression, and consequential impaired adipocyte differentiation, at least in part due to disrupted mitochondrial biosynthesis and function. Our study is the first to give experimental insights into the molecular mechanisms behind observational epidemiological studies the Gly482Ser variant and obesity and metabolic disorders (Less)
Please use this url to cite or link to this publication:
author
; ; ; ; and
organization
publishing date
type
Contribution to journal
publication status
published
subject
in
Diabetologia
volume
64
issue
Suppl 1
article number
306
pages
159 - 159
publisher
Springer
conference name
European Association for the Study of Diabetes (EASD) 57th Annual meeting
conference dates
2021-09-25 - 2021-10-01
external identifiers
  • pmid:34468792
  • scopus:85116955008
ISSN
1432-0428
DOI
10.1007/s00125-021-05519-y
language
English
LU publication?
yes
id
6ca0d892-08f9-4451-90c9-01336a5b83f1
date added to LUP
2022-01-27 11:37:01
date last changed
2022-06-29 14:00:28
@misc{6ca0d892-08f9-4451-90c9-01336a5b83f1,
  abstract     = {{Background and aims: PPARGC1A encodes PGC-1α (peroxisome proliferator-activated receptor γ coactivator 1-α), a central regulator of energy metabolism and mitochondrial function. A common polymorphism in PPARGC1A (rs8192678, C/T, Gly482Ser) has been associated with obesity and related metabolic disorders, but no published functional studies have investigated direct allele-specific effects in adipocyte biology. Materials and methods: We used CRISPR-Cas9 to perform allele switching (C-to-T or T-to-C) at rs8192678 in isogenic human pre-adipocyte white adipose tissue (hWAT) cell line; we then evaluated the allelic effects at rs8192678 on adipogenic differentiation and mitochondrial function. Accordingly, single-cell clones were expanded and screened to obtain homozygous T/T (482Ser) and C/C (482Gly) isogenic cell populations. The effect of the allele editing on white adipocyte differentiation and on mitochondrial function was then studied in three cell populations of the respective genotype. In ongoing experiments, CRISPR/Cas9 was also used to append a luciferase tag to C/C and in T/T cells. The luciferase will be used as a reporter for the endogenously expressed PGC-1 protein stability, and will therefore provide insights into mechanisms by which rs8192678 alleles affect PGC-1 activity. Results (see figure): At the end of the differentiation protocol the C/C adipocytes were apparently less Oil-Red-O positive than T/T adipocytes under optical microscopy, they had 78.5% lower triglyceride content (p&lt;0.0001, n=9), and lower expression of adipogenic markers (all markers p&lt;0.0001, n=3). Furthermore, C/C adipocytes had lower mitochondrial content (p&lt;0.001, n=9), which coincided with decreased oxygen consumption rate (OCR) at basal (p&lt;0.0001, n=3) and maximal respiration (p&lt;0.0001, n=3). Also, C/C adipocytes had lower ATP-linked OCR (p&lt;0.0001, n=3). Conclusion: Our data showcases discriminatory causal effects of the two rs8192678 alleles in adipocytes. The C allele confers lower PPARGC1A expression, and consequential impaired adipocyte differentiation, at least in part due to disrupted mitochondrial biosynthesis and function. Our study is the first to give experimental insights into the molecular mechanisms behind observational epidemiological studies the Gly482Ser variant and obesity and metabolic disorders}},
  author       = {{Huang, M. and Claussnitzer, M. and Saadat, A. and Mulder, Hindrik and Kalamajski, Sebastian and Franks, Paul}},
  issn         = {{1432-0428}},
  language     = {{eng}},
  note         = {{Conference Abstract}},
  number       = {{Suppl 1}},
  pages        = {{159--159}},
  publisher    = {{Springer}},
  series       = {{Diabetologia}},
  title        = {{CRISPR editing of the PPARGC1A Gly482ser (rs8192678) polymorphism in human white adipose cells shows differential effects on mitochondrial function and adipogenesis.}},
  url          = {{http://dx.doi.org/10.1007/s00125-021-05519-y}},
  doi          = {{10.1007/s00125-021-05519-y}},
  volume       = {{64}},
  year         = {{2021}},
}