Your browser doesn't support javascript.
loading
Single-cell Transcriptome Analysis Identifies Senescent Osteocytes as Contributors to Bone Destruction in Breast Cancer Metastasis.
Adhikari, Manish; Kaur, Japneet; Sabol, Hayley M; Anloague, Aric; Khan, Sharmin; Kurihara, Noriyoshi; Diaz-delCastillo, Marta; Andreasen, Christina Møller; Barnes, C Lowry; Stambough, Jeffrey B; Palmieri, Michela; Reyes-Castro, Olivia; Ambrogini, Elena; Almeida, Maria; O'Brien, Charles A; Nookaw, Intawat; Delgado-Calle, Jesus.
Afiliación
  • Adhikari M; Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US.
  • Kaur J; Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US.
  • Sabol HM; Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US.
  • Anloague A; Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US.
  • Khan S; Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, US.
  • Kurihara N; Division of Hematology and Oncology, Department of Medicine, Indiana University, Indianapolis, IN, US.
  • Diaz-delCastillo M; Forensic Medicine, University of Aarhus, Aarhus, Denmark.
  • Andreasen CM; Molecular Bone Histology lab, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
  • Barnes CL; Department of Clinical Pathologyogy, Odense University Hospital, Odense University Hospital, Odense, Denmark.
  • Stambough JB; Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US.
  • Palmieri M; Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, US.
  • Reyes-Castro O; Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US.
  • Ambrogini E; Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US.
  • Almeida M; Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US.
  • O'Brien CA; Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US.
  • Nookaw I; Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, Little Rock, AR, US.
  • Delgado-Calle J; Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, US.
Res Sq ; 2024 Mar 14.
Article en En | MEDLINE | ID: mdl-38558984
ABSTRACT
Breast cancer bone metastases increase fracture risk and are a major cause of morbidity and mortality among women. Upon colonization by tumor cells, the bone microenvironment undergoes profound reprogramming to support cancer progression that disrupts the balance between osteoclasts and osteoblasts, leading to bone lesions. Whether such reprogramming affects matrix-embedded osteocytes remains poorly understood. Here, we demonstrate that osteocytes in breast cancer bone metastasis develop premature senescence and a distinctive senescence-associated secretory phenotype (SASP) that favors bone destruction. Single-cell RNA sequencing identified osteocytes from mice with breast cancer bone metastasis enriched in senescence and SASP markers and pro-osteoclastogenic genes. Using multiplex in situ hybridization and AI-assisted analysis, we detected osteocytes with senescence-associated distension of satellites, telomere dysfunction, and p16Ink4a expression in mice and patients with breast cancer bone metastasis. In vitro and ex vivo organ cultures showed that breast cancer cells promote osteocyte senescence and enhance their osteoclastogenic potential. Clearance of senescent cells with senolytics suppressed bone resorption and preserved bone mass in mice with breast cancer bone metastasis. These results demonstrate that osteocytes undergo pathological reprogramming by breast cancer cells and identify osteocyte senescence as an initiating event triggering bone destruction in breast cancer metastases.
Palabras clave

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: Res Sq Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Idioma: En Revista: Res Sq Año: 2024 Tipo del documento: Article País de afiliación: Estados Unidos