Your browser doesn't support javascript.
loading
A lactate-SREBP2 signaling axis drives tolerogenic dendritic cell maturation and promotes cancer progression.
Plebanek, Michael P; Xue, Yue; Nguyen, Y-Van; DeVito, Nicholas C; Wang, Xueying; Holtzhausen, Alisha; Beasley, Georgia M; Theivanthiran, Balamayooran; Hanks, Brent A.
Affiliation
  • Plebanek MP; Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27708, USA.
  • Xue Y; Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27708, USA.
  • Nguyen YV; Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27708, USA.
  • DeVito NC; Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27708, USA.
  • Wang X; Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27708, USA.
  • Holtzhausen A; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
  • Beasley GM; Department of Surgery, Division of Surgical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27708, USA.
  • Theivanthiran B; Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27708, USA.
  • Hanks BA; Department of Medicine, Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, NC 27708, USA.
Sci Immunol ; 9(95): eadi4191, 2024 May 10.
Article in En | MEDLINE | ID: mdl-38728412
ABSTRACT
Conventional dendritic cells (DCs) are essential mediators of antitumor immunity. As a result, cancers have developed poorly understood mechanisms to render DCs dysfunctional within the tumor microenvironment (TME). After identification of CD63 as a specific surface marker, we demonstrate that mature regulatory DCs (mregDCs) migrate to tumor-draining lymph node tissues and suppress DC antigen cross-presentation in trans while promoting T helper 2 and regulatory T cell differentiation. Transcriptional and metabolic studies showed that mregDC functionality is dependent on the mevalonate biosynthetic pathway and its master transcription factor, SREBP2. We found that melanoma-derived lactate activates SREBP2 in tumor DCs and drives conventional DC transformation into mregDCs via homeostatic or tolerogenic maturation. DC-specific genetic silencing and pharmacologic inhibition of SREBP2 promoted antitumor CD8+ T cell activation and suppressed melanoma progression. CD63+ mregDCs were found to reside within the lymph nodes of several preclinical tumor models and in the sentinel lymph nodes of patients with melanoma. Collectively, this work suggests that a tumor lactate-stimulated SREBP2-dependent program promotes CD63+ mregDC development and function while serving as a promising therapeutic target for overcoming immune tolerance in the TME.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Dendritic Cells / Signal Transduction / Lactic Acid / Sterol Regulatory Element Binding Protein 2 / Melanoma Limits: Animals / Female / Humans Language: En Journal: Sci Immunol Year: 2024 Document type: Article Affiliation country: Estados Unidos Country of publication: Estados Unidos

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Dendritic Cells / Signal Transduction / Lactic Acid / Sterol Regulatory Element Binding Protein 2 / Melanoma Limits: Animals / Female / Humans Language: En Journal: Sci Immunol Year: 2024 Document type: Article Affiliation country: Estados Unidos Country of publication: Estados Unidos