9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      CA Dreamin’: Carbonic Anhydrase Inhibitors, Macrophages, and Pulmonary Hypertension

      editorial
      , M.S., Ph.D. 1
      American Journal of Respiratory Cell and Molecular Biology
      American Thoracic Society

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Although it is a rare condition, a diagnosis of pulmonary arterial hypertension (PAH) confers a risk of significant morbidity and mortality. Characterized by vascular cell hyperproliferation, formation of occlusive lesions, and extension of smooth muscle to the typically nonmuscular regions of the arterial tree, the vascular remodeling that occurs during PAH is well described, but it is poorly understood from a mechanistic standpoint. Consequently, available treatments for PAH rely on manipulating pathways involved in vasoconstriction, and no current therapies are aimed primarily at slowing or reversing the vascular remodeling. Thus, a major focus in PAH research has been to elucidate pathways involved in the initiation and progression of remodeling, with the goal of identifying druggable targets for which therapies (i.e., small-molecule inhibitors) can be developed. Given the long time frame associated with drug discovery, a more practical and certainly time-effective approach might be to use repurposed drugs, with known safety profiles, to treat PAH. However, with an incomplete understanding of the mechanisms that control the remodeling process, achieving this goal has remained closer to a dream than a reality. Immune cells have been long recognized as being present in vascular lesions of patients with PAH, and recent investigations have moved inflammation from a bystander to a central player in the pathogenesis of PAH (1). Although roles for mast cells, T cells, and B cells have been proposed, accumulating work is focusing on the macrophage as a viable target that modulates vascular remodeling during PAH (2). Macrophages can sense the local tissue microenvironment and transduce that signal via metabolic reprogramming to regulate their functional phenotype (3). In broad strokes, M1 (classically activated) macrophages are proinflammatory, whereas M2 (alternatively activated) macrophages are antiinflammatory. Both phenotypes have been described in PAH (4) and are believed to be driven by metabolic derangements in vascular wall cells that may influence the local milieu. It is against this background that Hudalla and colleagues (pp. 512–524) tested the hypothesis that carbonic anhydrase inhibition (CAI) with acetazolamide could induce metabolic acidosis and ameliorate pulmonary hypertension in the SU5416 plus hypoxia (SuHx) rat model of PAH, as reported in this issue of the Journal (5). CAI was initially developed with primary action on the kidney, interfering with acid–base regulation and causing diuresis. Presently used to treat glaucoma, metabolic alkalosis, epilepsy, and acute mountain sickness, CAI has been shown to have a beneficial effect on pulmonary hypertension in preclinical models with hypoxia as the stimulus (6). The authors of the current report used a straightforward study design in a relevant preclinical model of PAH, and a comprehensive approach that included the use of samples from patients with PAH and mechanistic in vitro studies. When added to the drinking water either early or late in the course of the SuHx model, CAI reduced measures of pulmonary hypertension. Similar results were observed when metabolic acidosis was induced by ammonium chloride. The authors also showed that expression of proinflammatory mediators (i.e., Tnfa and Il-6) were upregulated in both the lungs and macrophages of SuHx rats and in tissues from patients with PAH. Interestingly, macrophages from SuHx animals exhibited both increased proinflammatory markers and increased markers of alternative activation. One of the most intriguing findings of this study is that macrophage mediators, such as TNF-α and IL-1β, promoted dedifferentiation in pulmonary arterial smooth muscle cells (PASMCs), leading to a synthetic (i.e., hyperproliferative) phenotype. Indeed, supernatant from SuHx macrophages promoted PASMC dedifferentiation in vitro, which was prevented if the rats were treated with CAI. These results, coupled with other recent findings (7, 8), firmly position the macrophage as a mediator of vascular remodeling in pulmonary hypertension. Although the current work clearly demonstrates a novel immunomodulatory role for CAI in macrophage function and links macrophage activation with the PASMC phenotype, several issues remain to be addressed. First and foremost is the mechanism by which CAI exerts its effects. The authors found that carbonic anhydrase II (CAII) was abundantly expressed and upregulated in macrophages from SuHx rats and lung tissue from patients with PAH. Given that activation of both M1 and M2 was suppressed by CAI, it is possible that a compensatory upregulation of CAII is required for macrophage metabolic reprogramming. Surprisingly, inducing acidosis with ammonium chloride suppressed only M1 activation. These dichotomous results raise an interesting question about mechanisms associated with CAI that are unrelated to the acid–base balance. It is also not clear that macrophages are the sole or primary target that can alleviate pulmonary hypertension in vivo. For example, acetazolamide directly inhibits pulmonary vasoconstriction, attenuates Ca2+ signaling in PASMCs, and has been suggested to act as an antioxidant or stimulant of nitric oxide production (9–12). Some of these effects are independent of CAI; for example, an acetazolamide analog with a methylation in place of the sulfonamide moiety (N-methylacetazolamide) was shown to lack CAI, yet similarly repressed Ca2+ signaling and pulmonary contraction (9, 10). The non-CAI effects of acetazolamide are not well understood, and whether any of these effects played a role in the findings noted in the current paper awaits further exploration. Regardless of remaining questions regarding the exact mechanisms by which CAI exerts beneficial effects in the SuHx model and modulates macrophage function, this study provides intriguing new pieces in the PAH pathogenesis puzzle and demonstrates improved hemodynamics in the SuHx model. Whether similar effects will be observed in patients with PAH is unknown. A clinical trial (ClinicalTrials.gov Identifier: NCT02755259) examining the acute hemodynamic effects of acetazolamide in PAH is ongoing, but given the short exposure time (60 min) used in this trial, the results are unlikely to determine whether repurposing acetazolamide for treatment of PAH will become a reality. Nonetheless, the results from the current study encourage investigators to keep dreamin’.

          Related collections

          Most cited references12

          • Record: found
          • Abstract: found
          • Article: not found

          Hypoxia-induced pulmonary vascular remodeling requires recruitment of circulating mesenchymal precursors of a monocyte/macrophage lineage.

          Vascular remodeling in chronic hypoxic pulmonary hypertension includes marked fibroproliferative changes in the pulmonary artery (PA) adventitia. Although resident PA fibroblasts have long been considered the primary contributors to these processes, we tested the hypothesis that hypoxia-induced pulmonary vascular remodeling requires recruitment of circulating mesenchymal precursors of a monocyte/macrophage lineage, termed fibrocytes. Using two neonatal animal models (rats and calves) of chronic hypoxic pulmonary hypertension, we demonstrated a dramatic perivascular accumulation of mononuclear cells of a monocyte/macrophage lineage (expressing CD45, CD11b, CD14, CD68, ED1, ED2). Many of these cells produced type I collagen, expressed alpha-smooth muscle actin, and proliferated, thus exhibiting mesenchymal cell characteristics attributed to fibrocytes. The blood-borne origin of these cells was confirmed in experiments wherein circulating monocytes/macrophages of chronically hypoxic rats were in vivo-labeled with DiI fluorochrome via liposome delivery and subsequently identified in the remodeled pulmonary, but not systemic, arterial adventitia. The DiI-labeled cells that appeared in the vessel wall expressed monocyte/macrophage markers and procollagen. Selective depletion of this monocytic cell population, using either clodronate-liposomes or gadolinium chloride, prevented pulmonary adventitial remodeling (ie, production of collagen, fibronectin, and tenascin-C and accumulation of myofibroblasts). We conclude that circulating mesenchymal precursors of a monocyte/macrophage lineage, including fibrocytes, are essential contributors to hypoxia-induced pulmonary vascular remodeling.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Early macrophage recruitment and alternative activation are critical for the later development of hypoxia-induced pulmonary hypertension.

            Lung inflammation precedes the development of hypoxia-induced pulmonary hypertension (HPH); however, its role in the pathogenesis of HPH is poorly understood. We sought to characterize the hypoxic inflammatory response and to elucidate its role in the development of HPH. We also aimed to investigate the mechanisms by which heme oxygenase-1, an anti-inflammatory enzyme, is protective in HPH. We generated bitransgenic mice that overexpress human heme oxygenase-1 under doxycycline control in an inducible, lung-specific manner. Hypoxic exposure of mice in the absence of doxycycline resulted in early transient accumulation of monocytes/macrophages in the bronchoalveolar lavage. Alveolar macrophages acquired an alternatively activated phenotype (M2) in response to hypoxia, characterized by the expression of found in inflammatory zone-1, arginase-1, and chitinase-3-like-3. A brief 2-day pulse of doxycycline delayed, but did not prevent, the peak of hypoxic inflammation, and could not protect against HPH. In contrast, a 7-day doxycycline treatment sustained high heme oxygenase-1 levels during the entire period of hypoxic inflammation, inhibited macrophage accumulation and activation, induced macrophage interleukin-10 expression, and prevented the development of HPH. Supernatants from hypoxic M2 macrophages promoted the proliferation of pulmonary artery smooth muscle cells, whereas treatment with carbon monoxide, a heme oxygenase-1 enzymatic product, abrogated this effect. Early recruitment and alternative activation of macrophages in hypoxic lungs are critical for the later development of HPH. Heme oxygenase-1 may confer protection from HPH by effectively modifying the macrophage activation state in hypoxia.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Contribution of metabolic reprogramming to macrophage plasticity and function.

              Macrophages display a spectrum of functional activation phenotypes depending on the composition of the microenvironment they reside in, including type of tissue/organ and character of injurious challenge they are exposed to. Our understanding of how macrophage plasticity is regulated by the local microenvironment is still limited. Here we review and discuss the recent literature regarding the contribution of cellular metabolic pathways to the ability of the macrophage to sense the microenvironment and to alter its function. We propose that distinct alterations in the microenvironment induce a spectrum of inducible and reversible metabolic programs that might form the basis of the inducible and reversible spectrum of functional macrophage activation/polarization phenotypes. We highlight that metabolic pathways in the bidirectional communication between macrophages and stromals cells are an important component of chronic inflammatory conditions. Recent work demonstrates that inflammatory macrophage activation is tightly associated with metabolic reprogramming to aerobic glycolysis, an altered TCA cycle, and reduced mitochondrial respiration. We review cytosolic and mitochondrial mechanisms that promote initiation and maintenance of macrophage activation as they relate to increased aerobic glycolysis and highlight potential pathways through which anti-inflammatory IL-10 could promote macrophage deactivation. Finally, we propose that in addition to their role in energy generation and regulation of apoptosis, mitochondria reprogram their metabolism to also participate in regulating macrophage activation and plasticity.
                Bookmark

                Author and article information

                Journal
                Am J Respir Cell Mol Biol
                Am. J. Respir. Cell Mol. Biol
                ajrcmb
                American Journal of Respiratory Cell and Molecular Biology
                American Thoracic Society
                1044-1549
                1535-4989
                October 2019
                October 2019
                October 2019
                : 61
                : 4
                : 412-413
                Affiliations
                [ 1 ]Division of Pulmonary and Critical Care Medicine

                Johns Hopkins School of Medicine

                Baltimore, Maryland
                Article
                2019-0122ED
                10.1165/rcmb.2019-0122ED
                6775948
                30973760
                6f8eca29-4ee1-443e-bb05-7dd13e3aaecc
                Copyright © 2019 by the American Thoracic Society

                This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0 ( http://creativecommons.org/licenses/by-nc-nd/4.0/). For commercial usage and reprints please contact Diane Gern ( dgern@ 123456thoracic.org ).

                History
                Page count
                Figures: 0, Tables: 0, Pages: 2
                Categories
                Editorials

                Comments

                Comment on this article