Your browser doesn't support javascript.
loading
Engineered allele substitution at PPARGC1A rs8192678 alters human white adipocyte differentiation, lipogenesis, and PGC-1α content and turnover.
Huang, Mi; Claussnitzer, Melina; Saadat, Alham; Coral, Daniel E; Kalamajski, Sebastian; Franks, Paul W.
Afiliación
  • Huang M; Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Clinical Research Centre, Lund University, Malmö, Sweden.
  • Claussnitzer M; Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
  • Saadat A; Metabolism Program, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
  • Coral DE; Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Clinical Research Centre, Lund University, Malmö, Sweden.
  • Kalamajski S; Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Clinical Research Centre, Lund University, Malmö, Sweden. sebastian.kalamajski@med.lu.se.
  • Franks PW; Department of Clinical Sciences, Genetic and Molecular Epidemiology Unit, Clinical Research Centre, Lund University, Malmö, Sweden. paul.franks@med.lu.se.
Diabetologia ; 66(7): 1289-1305, 2023 07.
Article en En | MEDLINE | ID: mdl-37171500
ABSTRACT
AIMS/

HYPOTHESIS:

PPARGC1A encodes peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α), 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. We examined whether rs8192678 is a causal variant and reveal its biological function in human white adipose cells.

METHODS:

We used CRISPR-Cas9 genome editing to perform an allelic switch (C-to-T or T-to-C) at rs8192678 in an isogenic human pre-adipocyte white adipose tissue (hWAs) cell line. Allele-edited single-cell clones were expanded and screened to obtain homozygous T/T (Ser482Ser), C/C (Gly482Gly) and heterozygous C/T (Gly482Ser) isogenic cell populations, followed by functional studies of the allele-dependent effects on white adipocyte differentiation and mitochondrial function.

RESULTS:

After differentiation, the C/C adipocytes were visibly less BODIPY-positive than T/T and C/T adipocytes, and had significantly lower triacylglycerol content. The C allele presented a dose-dependent lowering effect on lipogenesis, as well as lower expression of genes critical for adipogenesis, lipid catabolism, lipogenesis and lipolysis. Moreover, C/C adipocytes had decreased oxygen consumption rate (OCR) at basal and maximal respiration, and lower ATP-linked OCR. We determined that these effects were a consequence of a C-allele-driven dysregulation of PGC-1α protein content, turnover rate and transcriptional coactivator activity. CONCLUSIONS/

INTERPRETATION:

Our data show allele-specific causal effects of the rs8192678 variant on adipogenic differentiation. The C allele confers lower levels of PPARGC1A mRNA and PGC-1α protein, as well as disrupted dynamics of PGC-1α turnover and activity, with downstream effects on cellular differentiation and mitochondrial function. Our study provides the first experimentally deduced insights on the effects of rs8192678 on adipocyte phenotype.
Asunto(s)
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Lipogénesis / Adipocitos Blancos Límite: Humans Idioma: En Revista: Diabetologia Año: 2023 Tipo del documento: Article País de afiliación: Suecia

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Lipogénesis / Adipocitos Blancos Límite: Humans Idioma: En Revista: Diabetologia Año: 2023 Tipo del documento: Article País de afiliación: Suecia