Altered Macrophage Polarization Induces Experimental Pulmonary Hypertension and Is Observed in Patients With Pulmonary Arterial Hypertension

Arterioscler Thromb Vasc Biol. 2021 Jan;41(1):430-445. doi: 10.1161/ATVBAHA.120.314639. Epub 2020 Nov 5.

Abstract

Objective: To determine whether global reduction of CD68 (cluster of differentiation) macrophages impacts the development of experimental pulmonary arterial hypertension (PAH) and whether this reduction affects the balance of pro- and anti-inflammatory macrophages within the lung. Additionally, to determine whether there is evidence of an altered macrophage polarization in patients with PAH. Approach and Results: Macrophage reduction was induced in mice via doxycycline-induced CD68-driven cytotoxic diphtheria toxin A chain expression (macrophage low [MacLow] mice). Chimeric mice were generated using bone marrow transplant. Mice were phenotyped for PAH by echocardiography and closed chest cardiac catheterization. Murine macrophage phenotyping was performed on lungs, bone marrow-derived macrophages, and alveolar macrophages using immunohistochemical and flow cytometry. Monocyte-derived macrophages were isolated from PAH patients and healthy volunteers and polarization capacity assessed morphologically and by flow cytometry. After 6 weeks of macrophage depletion, male but not female MacLow mice developed PAH. Chimeric mice demonstrated a requirement for both MacLow bone marrow and MacLow recipient mice to cause PAH. Immunohistochemical analysis of lung sections demonstrated imbalance in M1/M2 ratio in male MacLow mice only, suggesting that this imbalance may drive the PAH phenotype. M1/M2 imbalance was also seen in male MacLow bone marrow-derived macrophages and PAH patient monocyte-derived macrophages following stimulation with doxycycline and IL (interleukin)-4, respectively. Furthermore, MacLow-derived alveolar macrophages showed characteristic differences in terms of their polarization and expression of diphtheria toxin A chain following stimulation with doxycycline.

Conclusions: These data further highlight a sex imbalance in PAH and further implicate immune cells into this paradigm. Targeting imbalance of macrophage population may offer a future therapeutic option.

Keywords: heart failure, right; humans; macrophages; models, animal; pulmonary hypertension.

Publication types

  • Observational Study
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adult
  • Aged
  • Animals
  • Antigens, CD / genetics
  • Antigens, Differentiation, Myelomonocytic / genetics
  • Case-Control Studies
  • Cell Proliferation
  • Diphtheria Toxin / genetics
  • Disease Models, Animal
  • Female
  • Humans
  • Hypoxia / complications
  • Macrophage Activation*
  • Macrophages, Alveolar / metabolism
  • Macrophages, Alveolar / pathology*
  • Male
  • Mice, Transgenic
  • Middle Aged
  • Muscle, Smooth, Vascular / metabolism
  • Muscle, Smooth, Vascular / pathology*
  • Myocytes, Smooth Muscle / metabolism
  • Myocytes, Smooth Muscle / pathology
  • Paracrine Communication
  • Peptide Fragments / genetics
  • Phenotype
  • Pulmonary Arterial Hypertension / etiology
  • Pulmonary Arterial Hypertension / metabolism
  • Pulmonary Arterial Hypertension / pathology*
  • Pulmonary Artery / metabolism
  • Pulmonary Artery / pathology
  • Sex Factors
  • Vascular Remodeling*

Substances

  • Antigens, CD
  • Antigens, Differentiation, Myelomonocytic
  • CD68 antigen, human
  • Diphtheria Toxin
  • Peptide Fragments
  • diphtheria toxin fragment A