Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case...

9
Distinct roles of resident and nonresident macrophages in nonischemic cardiomyopathy Xudong Liao a,b,1 , Yuyan Shen a,b , Rongli Zhang a,b , Keiki Sugi a,b , Neelakantan T. Vasudevan a,b , M. Amer Alaiti a,b , David R. Sweet a,b , Lin Zhou c , Yulan Qing d,e , Stanton L. Gerson d,e , Chen Fu f , Anthony Wynshaw-Boris f , Rui Hu g , Martin A. Schwartz g , Hisashi Fujioka h , Brian Richardson i , Mark J. Cameron i , Hiroki Hayashi j , Jonathan S. Stamler j,k , and Mukesh K. Jain a,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106; b Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106; c Department of Cardiology, Tongji Hospital of Tongji University, 20065 Shanghai, China; d Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106; e National Center for Regenerative Medicine, Seidman Cancer Center, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH 44106; f Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106; g Yale Cardiovascular Research Center, Yale University, New Haven, CT 06511; h Electron Microscopy Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44106; i Department of Epidemiology and Biostatistics, Case Western Reserve University School of Medicine, Cleveland, OH 44106; j Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106; and k Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106 Edited by Christine E. Seidman, Howard Hughes Medical Institute and Brigham and Womens Hospital and Harvard Medical School, Boston, MA, and approved April 11, 2018 (received for review November 17, 2017) Nonischemic cardiomyopathy (NICM) resulting from long-standing hypertension, valvular disease, and genetic mutations is a major cause of heart failure worldwide. Recent observations suggest that myeloid cells can impact cardiac function, but the role of tissue-intrinsic vs. tissue-extrinsic myeloid cells in NICM remains poorly understood. Here, we show that cardiac resident macro- phage proliferation occurs within the first week following pres- sure overload hypertrophy (POH; a model of heart failure) and is requisite for the hearts adaptive response. Mechanistically, we identify Kruppel-like factor 4 (KLF4) as a key transcription fac- tor that regulates cardiac resident macrophage proliferation and angiogenic activities. Finally, we show that blood-borne macro- phages recruited in late-phase POH are detrimental, and that blockade of their infiltration improves myocardial angiogenesis and preserves cardiac function. These observations demonstrate previously unappre- ciated temporal and spatial roles for resident and nonresident macro- phages in the development of heart failure. cardiac macrophage | angiogenesis | pressure overload hypertrophy C ardiomyopathy is a disease of the heart muscle that de- creases the hearts ability to pump blood and meet the bodys energetic demands. Cardiomyopathy can be broadly classified as ischemic cardiomyopathy (ICM) or nonischemic cardiomyopa- thy (NICM). In ICM, interruption of coronary artery blood flow, as seen in myocardial infarction (MI), is the primary cause of cardiac dysfunction. In NICM, cardiac muscle function can be compromised by either intrinsic gene mutation (hypertrophic cardiomyopathy) or extrinsic stress, such as pressure overload secondary to chronic hypertension or valve diseases [pressure overload hypertrophy (POH)]. With respect to NICM, accu- mulating clinical evidence suggests that small vessel dysfunc- tion is a strong independent predictor of clinical deterioration and death in patients (14). Further, experimental studies sug- gest that inadequate myocardial angiogenesis is a critical de- terminant of the transition from compensatory hypertrophy to heart failure (57). Macrophages are found in virtually every tissue and are critical for homeostasis and stress-induced responses. Macrophages broadly consist of two classes: resident macrophages and blood- borne infiltrating macrophages. Resident macrophages originate from yolk sac-derived erythromyeloid progenitors (EMPs), re- side in tissue, perform homeostatic functions, and self-maintain locally (810). In general, infiltrating macrophages arise from circulating classic Ly6C hi (inflammatory) monocytes and are recruited after inciting pathology through the CCL2CCR2 chemotaxis pathway (8, 11). Nonclassic Ly6C lo (patrolling) mono- cytes also patrol the luminal side of the endothelium and extravasate in response to both septic and aseptic tissue injury, playing a protective or antiinflammatory role during tissue injury (12). The adult heart contains two major subsets of Ly6C lo /CCR2 resident macrophages that express different levels of MHC-II, which are EMP-derived resident macrophages self-maintained through local proliferation at steady state. In addition, there are two minor sub- sets of Ly6C hi macrophages (<2%) that differ in CCR2 levels (13). These Ly6C hi subsets are derived from hematopoietic stem cells, are maintained through monocyte infiltration, and help to resolve Significance Pressure overload triggers responses in cardiomyocytes and noncardiomyocytes, leading to pressure overload hypertrophy (POH). Here, we show that cardiac resident macrophages reg- ulate compensatory myocardial adaptation to POH, while nonresident infiltrating macrophages are detrimental. At early- phase POH, pressure overload induces cardiac resident macro- phage proliferation, which is regulated by Kruppel-like factor 4. At late-phase POH, pressure overload also induces Ly6C hi monocyte infiltration, and its blockade improves myocardial an- giogenesis and preserves cardiac function. Mechanistically, the differential impact of these two macrophage subsets on myocardial angiogenesis may underlie the cardiac phenotype. These findings provide insights regarding the role of cardiac resident and nonresident macrophages, conceptually update the view of myocardial angiogenesis, and identify monocyte infiltration as a therapeutic target for nonischemic cardiomyopathy. Author contributions: X.L., N.T.V., M.A.A., and M.K.J. designed research; X.L., Y.S., R.Z., K.S., N.T.V., M.A.A., D.R.S., L.Z., Y.Q., R.H., H.F., and H.H. performed research; X.L., Y.S., R.Z., K.S., N.T.V., M.A.A., D.R.S., Y.Q., S.L.G., C.F., A.W.-B., M.A.S., H.F., B.R., M.J.C., J.S.S., and M.K.J. analyzed data; and X.L. and M.K.J. wrote the paper. Conflict of interest statement: X.L. and M.K.J. have a patent application (serial no. 62/ 644,792). This article is a PNAS Direct Submission. Published under the PNAS license. Data deposition: The data reported in this paper have been deposited in the Gene Ex- pression Omnibus (GEO) database, https://www.ncbi.nlm.nih.gov/geo (accession no. GSE107016). 1 To whom correspondence may be addressed. Email: [email protected] or mukesh. [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1720065115/-/DCSupplemental. Published online April 30, 2018. www.pnas.org/cgi/doi/10.1073/pnas.1720065115 PNAS | vol. 115 | no. 20 | E4661E4669 MEDICAL SCIENCES PNAS PLUS Downloaded by guest on July 9, 2020

Transcript of Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case...

Page 1: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

Distinct roles of resident and nonresident macrophagesin nonischemic cardiomyopathyXudong Liaoa,b,1, Yuyan Shena,b, Rongli Zhanga,b, Keiki Sugia,b, Neelakantan T. Vasudevana,b, M. Amer Alaitia,b,David R. Sweeta,b, Lin Zhouc, Yulan Qingd,e, Stanton L. Gersond,e, Chen Fuf, Anthony Wynshaw-Borisf, Rui Hug,Martin A. Schwartzg, Hisashi Fujiokah, Brian Richardsoni, Mark J. Cameroni, Hiroki Hayashij, Jonathan S. Stamlerj,k,and Mukesh K. Jaina,b,1

aCase Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH 44106; bHarrington Heart and VascularInstitute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106; cDepartment of Cardiology, Tongji Hospital of Tongji University, 20065Shanghai, China; dCase Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106; eNational Center for Regenerative Medicine,Seidman Cancer Center, University Hospitals Cleveland Medical Center and Case Western Reserve University, Cleveland, OH 44106; fDepartment of Geneticsand Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106; gYale Cardiovascular Research Center, Yale University,New Haven, CT 06511; hElectron Microscopy Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH 44106; iDepartment ofEpidemiology and Biostatistics, Case Western Reserve University School of Medicine, Cleveland, OH 44106; jInstitute for Transformative Molecular Medicine,Case Western Reserve University School of Medicine, Cleveland, OH 44106; and kHarrington Discovery Institute, University Hospitals Cleveland MedicalCenter, Cleveland, OH 44106

Edited by Christine E. Seidman, Howard Hughes Medical Institute and Brigham andWomen’s Hospital and Harvard Medical School, Boston, MA, and approvedApril 11, 2018 (received for review November 17, 2017)

Nonischemic cardiomyopathy (NICM) resulting from long-standinghypertension, valvular disease, and genetic mutations is a majorcause of heart failure worldwide. Recent observations suggestthat myeloid cells can impact cardiac function, but the role oftissue-intrinsic vs. tissue-extrinsic myeloid cells in NICM remainspoorly understood. Here, we show that cardiac resident macro-phage proliferation occurs within the first week following pres-sure overload hypertrophy (POH; a model of heart failure) and isrequisite for the heart’s adaptive response. Mechanistically, weidentify Kruppel-like factor 4 (KLF4) as a key transcription fac-tor that regulates cardiac resident macrophage proliferation andangiogenic activities. Finally, we show that blood-borne macro-phages recruited in late-phase POH are detrimental, and that blockadeof their infiltration improves myocardial angiogenesis and preservescardiac function. These observations demonstrate previously unappre-ciated temporal and spatial roles for resident and nonresident macro-phages in the development of heart failure.

cardiac macrophage | angiogenesis | pressure overload hypertrophy

Cardiomyopathy is a disease of the heart muscle that de-creases the heart’s ability to pump blood and meet the body’s

energetic demands. Cardiomyopathy can be broadly classified asischemic cardiomyopathy (ICM) or nonischemic cardiomyopa-thy (NICM). In ICM, interruption of coronary artery blood flow,as seen in myocardial infarction (MI), is the primary cause ofcardiac dysfunction. In NICM, cardiac muscle function can becompromised by either intrinsic gene mutation (hypertrophiccardiomyopathy) or extrinsic stress, such as pressure overloadsecondary to chronic hypertension or valve diseases [pressureoverload hypertrophy (POH)]. With respect to NICM, accu-mulating clinical evidence suggests that small vessel dysfunc-tion is a strong independent predictor of clinical deteriorationand death in patients (1–4). Further, experimental studies sug-gest that inadequate myocardial angiogenesis is a critical de-terminant of the transition from compensatory hypertrophy toheart failure (5–7).Macrophages are found in virtually every tissue and are critical

for homeostasis and stress-induced responses. Macrophagesbroadly consist of two classes: resident macrophages and blood-borne infiltrating macrophages. Resident macrophages originatefrom yolk sac-derived erythromyeloid progenitors (EMPs), re-side in tissue, perform homeostatic functions, and self-maintainlocally (8–10). In general, infiltrating macrophages arise fromcirculating classic Ly6Chi (inflammatory) monocytes and arerecruited after inciting pathology through the CCL2–CCR2

chemotaxis pathway (8, 11). Nonclassic Ly6Clo (patrolling) mono-cytes also patrol the luminal side of the endothelium and extravasatein response to both septic and aseptic tissue injury, playing aprotective or antiinflammatory role during tissue injury (12). Theadult heart contains two major subsets of Ly6Clo/CCR2− residentmacrophages that express different levels of MHC-II, which areEMP-derived resident macrophages self-maintained through localproliferation at steady state. In addition, there are two minor sub-sets of Ly6Chi macrophages (<2%) that differ in CCR2 levels (13).These Ly6Chi subsets are derived from hematopoietic stem cells,are maintained through monocyte infiltration, and help to resolve

Significance

Pressure overload triggers responses in cardiomyocytes andnoncardiomyocytes, leading to pressure overload hypertrophy(POH). Here, we show that cardiac resident macrophages reg-ulate compensatory myocardial adaptation to POH, whilenonresident infiltrating macrophages are detrimental. At early-phase POH, pressure overload induces cardiac resident macro-phage proliferation, which is regulated by Kruppel-like factor4. At late-phase POH, pressure overload also induces Ly6Chi

monocyte infiltration, and its blockade improves myocardial an-giogenesis and preserves cardiac function. Mechanistically,the differential impact of these two macrophage subsetson myocardial angiogenesis may underlie the cardiac phenotype.These findings provide insights regarding the role of cardiac residentand nonresident macrophages, conceptually update the view ofmyocardial angiogenesis, and identify monocyte infiltrationas a therapeutic target for nonischemic cardiomyopathy.

Author contributions: X.L., N.T.V., M.A.A., and M.K.J. designed research; X.L., Y.S., R.Z., K.S.,N.T.V., M.A.A., D.R.S., L.Z., Y.Q., R.H., H.F., and H.H. performed research; X.L., Y.S., R.Z., K.S.,N.T.V., M.A.A., D.R.S., Y.Q., S.L.G., C.F., A.W.-B., M.A.S., H.F., B.R., M.J.C., J.S.S., and M.K.J.analyzed data; and X.L. and M.K.J. wrote the paper.

Conflict of interest statement: X.L. and M.K.J. have a patent application (serial no. 62/644,792).

This article is a PNAS Direct Submission.

Published under the PNAS license.

Data deposition: The data reported in this paper have been deposited in the Gene Ex-pression Omnibus (GEO) database, https://www.ncbi.nlm.nih.gov/geo (accession no.GSE107016).1To whom correspondence may be addressed. Email: [email protected] or [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1720065115/-/DCSupplemental.

Published online April 30, 2018.

www.pnas.org/cgi/doi/10.1073/pnas.1720065115 PNAS | vol. 115 | no. 20 | E4661–E4669

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

July

9, 2

020

Page 2: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

tissue injury (14). Recent studies suggest that in ICM (i.e., MI),tissue injury induces both local proliferation of resident macro-phages and infiltration of monocytes in the site of injury to regulatewound healing and inflammation in the heart (9, 15). Further,macrophages also accumulate in the remote nonischemic zone ofthe myocardium after MI to mediate post-MI cardiac remodeling(16). By contrast, much less is known in the setting of NICM. Forexample, whether macrophages are involved in disease progressionand, if so, the identity, origin, and function of such cardiac macro-phages remain unknown.

Here, using a classic mouse POH model for NICM, we trackresident and infiltrating macrophages in the myocardium duringthe early and late phases of POH and provide multiple lines ofevidence showing previously unappreciated and distinct time-dependent roles of different subsets of cardiac macrophages inPOH. Using the myeloid-specific Kruppel-like factor 4 (KLF4)mutant mouse, we also shed light on the molecular mechanismthat regulates local proliferation of cardiac resident macro-phages and a potential role that these cells play in regulatingperfusion during POH.

Fig. 1. Pressure overload induces local proliferation of cardiac resident macrophages at early phase and infiltration of monocytes at late phase. (A) Gatingstrategy for flow cytometry. (B) Dynamics of cardiac macrophages following TAC and its correlation with cardiac function. The dashed line indicates baseline.Cardiac macrophages were gated as live (live/dead stain-negative) CD45+CD11b+Ly6G−F4/80+CD64+ cells and are shown as a percentage vs. total heart cells.Cardiac function was expressed as left ventricular fractional shortening. (C) Immunostaining for Mac-3 to reveal macrophages in the myocardium, particularlyin the perivascular region. Brown (DAB-positive) cells are macrophages. Arrowheads indicate vessels. (Scale bars, 100 μm.) (D) LV dysfunction following TAC.(E) Strategy to generate CD45.1-CD45.2 chimeric mice. Recipient CD45.2 mice were irritated with their head-chest shielded by lead block and transplanted viatail vein injection with bone marrow cells from CD45.1 donor mice. The resulting chimera mice carry both CD45.2 and CD45.1 cells in the blood but only haveCD45.2 cardiac resident macrophages. (F) Blood (50 μL) was drawn from the tail vein at 8 wk posttransplantation and analyzed by FACS for CD45.1+ myeloidcells (CD45+CD11b+). (G) FACS analysis for CD45.1+ cardiac macrophages before and after TAC. (H) Corrected monocyte infiltration rate considering that only50% of monocytes were CD45.1+ cells. (I) FACS analysis for Ki-67 expression in cardiac macrophages. (B and D) P values were calculated from one-way ANOVA.(F–I) *P < 0.05 by t test after Bonferroni correction (n = 5–8 for all data points). NS, not significant.

E4662 | www.pnas.org/cgi/doi/10.1073/pnas.1720065115 Liao et al.

Dow

nloa

ded

by g

uest

on

July

9, 2

020

Page 3: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

ResultsPressure Overload Induces Local Proliferation of Cardiac ResidentMacrophages. To understand the role of macrophages in POH,we first assessed macrophage numbers in the heart followingtransverse aortic constriction (TAC), a well-established model ofpressure overload-induced hypertrophy and heart failure (i.e.,POH). Hearts from sham or TAC animals were analyzed by flowcytometry. As shown in Fig. 1A, we found that TAC inducedsignificant accumulation of myeloid cells (CD45+/CD11b+) inthe myocardium. The majority of these cells are macrophages(Ly6G−/F4/80+/CD64+ subset; >95%), and the remainder areLy6G+ neutrophils (<5%) (Fig. 1A). Time course studies showedthat cardiac macrophages increased significantly by 3 d post-TAC, peaked at 7 d, returned to baseline after 2 wk, and thenincreased modestly at 4 wk after TAC (Fig. 1B). The presence ofmacrophages in the myocardium was further confirmed by anti–Mac-3 immunostaining, particularly in the perivascular region(Fig. 1C). This dynamic change in cardiac macrophage numbersis noteworthy as it correlates with two major phases of the car-diac response to TAC, namely, the compensatory cardiac hy-pertrophy with preserved contractile function phase thatcharacterizes the first 7–10 d following TAC, followed by de-compensation and development of heart failure between 2 and4 wk after TAC (Fig. 1D).Next, we undertook multiple complementary approaches to

determine if the increase in cardiac macrophages following TACwas from resident macrophages, blood monocytes recruited tothe myocardium, or both. First, we utilized a recently describedfluorescence-activated cell sorting (FACS) strategy in which thesurface expression of CCR2 is low in resident macrophages buthigh in infiltrating macrophages (9). Importantly, we found thatmacrophages accumulating after 1-wk TAC are CCR2−, in-dicating that they are resident macrophages (SI Appendix, Fig. S1A and B). Second, we showed that blockade of monocyte in-filtration by RS-504393 (RS), a small-molecule CCR2 antagonist(9, 17), had no effect on cardiac macrophage populations (SIAppendix, Fig. S1 A and B). Consistently, most cardiac macro-phages are Ly6Clo before and after 1-wk TAC (SI Appendix, Fig.S1 C and D). Finally, to validate if pressure overload only in-duces resident macrophage expansion, we utilized a CD45.1-CD45.2 chimeric mouse model, in which recipient CD45.2 micewere irradiated utilizing a thorax shield to minimize cardiacmacrophage depletion. Following transplantation, the donorCD45.1+ bone marrow cells replaced circulating CD45.2+ bloodcells without affecting cardiac resident macrophages (Fig. 1E).We chose the chimeric animals with ∼50% CD45.1+ myeloidcells (CD11b+) for the TAC model to ensure effective in vivo celltracking (Fig. 1F). In sham animals, we observed that only 2.3%of cardiac macrophages are CD45.1+, confirming that cardiacmacrophages are maintained locally at steady state (Fig. 1G).After 1-wk TAC, the CD45.1+ cardiac macrophages remainedlow at 5.4%, suggesting that no significant monocyte infiltrationoccurs at this early stage of POH (Fig. 1G). However, after 6-wkTAC, the CD45.1+ cardiac macrophages reached 20% (Fig. 1G),indicating monocyte infiltration in late-stage POH that is asso-ciated with the transition from cardiac compensation to de-compensation. Because the CD45.1-CD45.2 chimeras subjectedto TAC carried both CD45.1+ and CD45.2+ white blood cells incirculation (1:1 by experimental design), the above percentagesof CD45.1+ macrophages only reflect 50% of the actual in-filtrating cells. As such, the corrected rate of monocyte in-filtration into the myocardium is about 5–10% at baseline andearly-phase POH, and it increases to around 40% in late-phasePOH (Fig. 1H).Because TAC increased cardiac macrophage numbers in-

dependent of monocyte infiltration at the early stage, we hy-pothesized that such cells have to be derived from local

proliferation. Indeed, we found TAC significantly induced theexpression of Ki-67 protein, a nuclear proliferation marker, incardiac macrophages (Fig. 1I). The local proliferation of cardiacresident macrophages was further confirmed by in vivo BrdUpulse labeling. Mice undergoing sham or TAC surgery were in-jected i.p. with BrdU for 2 h. Because bone marrow-derived cellsare not released into the circulation within this period (SI Ap-pendix, Fig. S2), BrdU incorporation in the heart is limited tolocally proliferating cells (9). We observed significant cell pro-liferation in cardiac macrophages 3 d post-TAC as demonstratedby BrdU incorporation and DNA S-phase populations by flowcytometry (Fig. 2 A and B). Of note, the pattern of macrophageproliferation (Fig. 2B, blue line) closely aligned with that ofcardiac macrophage count (Fig. 2B, red dotted line) along theTAC time line, with proliferation peaking just before the cellularmaximum at day 7 and subsequently decreasing to baseline byday 14. Collectively, these studies demonstrate that pressureoverload induces local proliferation of cardiac resident macro-phages in early-phase POH.The late-phase monocyte infiltration (Fig. 1H) may result from

an expanded circulating monocyte pool and/or greater in-filtrative capacity of monocytes. To assess circulating monocytenumbers (18), we gated blood for monocytes (CD115+CD11b+)and found that 4-wk TAC (late-phase POH) significantly in-creased monocyte numbers in blood and spleen but not in bonemarrow, indicating TAC-induced extramural monocytosis (Fig. 2C and D). Peripheral blood monocytes are classified into threedifferent populations based on expression of cell surface mole-cules and functions. In humans, these consist of CD14hiCD16−

(classic monocytes), CD14loCD16lo (intermediate monocytes),and CD14loCD16hi (nonclassic monocytes). In mice, the equivalentpopulations are Ly6ChiCD62L+CD43−CCR2+ (classic monocytes),Ly6CintCD62L−CD43+CCR2− (intermediate monocytes), andLy6CloCD62L−CD43+CCR2− (nonclassic monocytes) (12).Given the heterogeneity of these populations, we sought todetermine if one population predominantly contributes to mono-cyte infiltration after TAC. Interestingly, TAC induced an increasein Ly6Chi monocytes in blood but not in spleen or bone marrow(Fig. 2 E and F). These data demonstrate that pressure overloadinduces circulating Ly6Chi classic monocyte infiltration during late-phase POH.

Cardiac Resident Macrophages Are Required for Myocardial Adaptationin POH. We next sought to determine the functional importance ofresident macrophages in the early adaptive response to TAC. Asthere is no specific method to specifically deplete cardiac residentmacrophages, we undertook a stepwise elimination approach todistinguish the unique roles of cardiac resident vs. infiltratingmacrophages. We first treated mice with the CCR2 antagonist RSto block infiltration of blood-borne CCR2+Ly6Chi classic monocytes(15, 17, 19). RS administration had no impact on cardiac macro-phage numbers or cardiac function before and after 1 wk post-TAC(Fig. 3A and SI Appendix, Fig. S1). Next, we subjected animals toclodronate liposomes (CLs), which deplete both circulating andresident macrophages (20). Mice with CL administration for a week(500 μg/kg i.v. every other day) did not exhibit any symptom ofsickness or heart dysfunction as reported previously (21). However,when subjected to TAC, the macrophage-depleted animals (CL-injected) developed acute heart failure following TAC as revealedby serial echocardiography studies, while cardiac function was wellmaintained in PBS-liposome [vehicle (Veh)]–treated animals (Fig.3B). CL-injected animals also exhibited striking postoperativemortality, with ∼80% dead within 6 d, while a 100% survival ratewas observed in all other groups as expected (Fig. 3C). Postmortemautopsy revealed similar heart weight increases after TAC butheavier lungs in the CL-treated TAC group, indicating lung edemasecondary to heart failure (SI Appendix, Fig. S3A). Spleen weightwas similarly reduced by CL in both the sham and TAC groups,

Liao et al. PNAS | vol. 115 | no. 20 | E4663

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

July

9, 2

020

Page 4: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

confirming efficient macrophage/monocyte depletion (SI Appendix,Fig. S3A). Gene expression analysis by quantitative real-time PCR(qPCR) revealed no difference in hypertrophy, fibrosis, or glycolysisgenes (Fig. 3 D–F). However, the expression of hypoxia-induciblegenes, including Adm, Pdk4, and p21 (Cdkn1a), was significantlyhigher in the macrophage-depleted TAC (TAC+CL) group com-pared with the normal TAC (TAC+Veh) group (Fig. 3G), in-dicating that TAC induced more severe hypoxia stress in themacrophage-depleted myocardium. TAC also induced expres-sion of inflammatory markers, including IL-1β, TNF-α, andCCL2 (MCP1), which was abrogated following macrophage de-pletion (Fig. 3H), suggesting that inflammation was not thereason for heart failure in early-phase TAC. Histological studiesconfirmed no significant differences in cardiomyocyte hypertro-phy (wheat germ agglutinin staining), fibrosis (collagen staining),or capillary density (CD31 staining) in the myocardium after CLtreatment (SI Appendix, Fig. S3B). However, we detected pro-found cardiomyocyte death in macrophage-depleted hearts (Fig.3I and SI Appendix, Fig. S4A). The mechanism of cell death of

these cardiomyocytes appeared to be nonapoptotic, as demon-strated by negative caspase-3 activation/cleavage (SI Appendix,Fig. S4B). These data underscore the importance of residentmacrophages in the early-phase cardiac adaptation to POH.While these data underscore the importance of resident

macrophages in early-phase cardiac adaptation to POH, the roleof cardiac resident macrophages in late-phase adaptation re-mains unexplored. To address this, we allowed early compensa-tion to occur after TAC and subsequently depleted macrophagesvia three alternate-day doses of CL beginning at day 14 post-TAC. As with pre-TAC macrophage depletion, an 80% mortalityrate was observed after three injections of CL 3 wk post-TAC (SIAppendix, Fig. S5A). It is noteworthy that CL administrationtrigged a quick transition from cardiac compensation [ejectionfraction (EF) ∼ 60%] to heart failure (EF < 30%) as revealed byechocardiography (SI Appendix, Fig. S5B). CL administration atthis point of TAC exhibited little effect on further developmentof cardiac hypertrophy or fibrosis (SI Appendix, Fig. S5C), butresulted in profound cardiomyocyte death (SI Appendix, Fig. S5

Fig. 2. Local proliferation of cardiac resident macrophages and infiltration of blood-borne Ly6C+ monocytes. (A and B) TAC induced local proliferation ofcardiac resident macrophages as assessed by BrdU pulse-labeling assay. DNA was stained with Hoechst 33342 for cycle analysis, and the S-phase was clearlyvisible in TAC 3-d samples. The time course curve of total cardiac macrophages (dotted red line) was overlaid to show the phase difference. (C and D)Monocytes (CD115+/CD11b+) in blood, spleen, and bone marrow before and after TAC. (E and F) Classic Ly6C+ monocytes and nonclassic Ly6C− monocytesbefore and after TAC. In B, P values were calculated from one-way ANOVA for BrdU data. In D and F, *P < 0.05 by t test (n = 5 for all data points). NS, notsignificant.

E4664 | www.pnas.org/cgi/doi/10.1073/pnas.1720065115 Liao et al.

Dow

nloa

ded

by g

uest

on

July

9, 2

020

Page 5: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

D–F). Because blocking peripheral monocyte infiltration did notpromote cardiac dysfunction in early-phase (Fig. 3A) or late-phase-TAC (see Fig. 5), these late-stage macrophage depletionstudies implicate cardiac resident macrophages in maintainingcardioprotection throughout the POH disease course.

KLF4 Regulates Proliferation of Cardiac Resident Macrophages. Mosttissue resident macrophages, including cardiac resident macro-phages, are maintained by self-renewal (9, 22). While our un-derstanding of how resident macrophages proliferate is limited, arecent seminal study by Sieweke and coworkers (23) providescritical insights. Specifically, it was found that primary macro-phages from MafB/c-Maf double-deficient (Maf-DKO) micedemonstrated indefinite KLF4-dependent proliferative capacitywhen cultured with macrophage colony-stimulating factor(M-CSF) (23). Therefore, we utilized myeloid-specific KLF4-deficient mice (Lyz2-Cre:KLF4flox/flox; designated K4-cKO)(24) to investigate if KLF4 regulates cardiac macrophage pro-liferation in the context of POH.Given that Lyz2-Cre is active in macrophages and neutrophils

but not in monocytes (21), we first sought to explore any po-tential role for neutrophils in this model. The infiltration ofLy6G+ neutrophils was marginal after TAC (Fig. 1A), and de-pletion of neutrophils using the anti-Ly6G antibody (clone 1A8)before TAC demonstrated very little effect on POH (SI Appen-dix, Fig. S6). Therefore, these data supported our decision tofocus on the role of KLF4-deficient macrophages.

We first performed transcriptomic analysis of primarymacrophages from Lyz2-Cre (designated Cre) and K4-cKOmice via RNA-sequencing (RNA-seq) and found 238 genesthat were differentially expressed (false discovery rate < 0.05).Gene ontology (GO) analysis demonstrated enrichment inmultiple pathways of cell proliferation and division (Fig. 4A).Importantly, when treated in vitro with CSF1 and CSF2,KLF4-deficient primary macrophages showed less accumula-tion of Ki-67 protein (Fig. 4B and SI Appendix, Fig. S7A),demonstrating impaired proliferative capacity. Next, we sub-jected the K4-cKO and Cre animals to TAC. FACS analysisdemonstrated that TAC-induced cardiac macrophage accu-mulation was significantly reduced in the K4-cKO groupcompared with that of Cre (Fig. 4C). Consistent with fewercardiac macrophages, myeloid KLF4 deficiency impairedcardiac resident macrophage proliferation in vivo followingTAC as demonstrated by reduced BrdU incorporation (SI Appen-dix, Fig. S7B).Post-TAC serial echocardiography revealed that K4-cKO mice

rapidly develop a dilated cardiomyopathy characterized by re-duced left ventricular EF (Fig. 4D), elevated heart and lungweight, and expression of the cardiac hypertrophy marker geneNppa (Fig. 4E). Histological analyses exhibited enhanced fibrosisand cell death, as well as severe mitochondrial damage in K4-cKO hearts post-TAC (Fig. 4F). Plasma cardiac troponin I levelswere significantly higher in K4-cKO mice post-TAC, indicatingmyocardial injury (Fig. 4G). K4-cKO hearts exhibited significantly

Fig. 3. Cardiac resident macrophages are requisite for cardiac adaptation to POH. (A) Blocking monocyte infiltration by RS had no effect on POH. (B) Leftventricular function following clodronate-mediated macrophage depletion and TAC (n = 8–10 in each group). The P value was calculated from two-wayANOVA. The blue arrow indicates the time of TAC, and red arrows indicate the time of liposome injections. (C) Postoperation survival rates. P values werecalculated from log-rank between TAC+CL and TAC+Veh groups. (D–H) Gene expression in the heart after 4-d TAC ± CL (n = 5). *P < 0.05 by t test withBonferroni correction. (I) Cell death detected by TUNEL staining in the myocardium after 4-d TAC ± CL. Red arrowheads indicate nuclei of dead cells. *P <0.05 by t test (n = 5). NS, not significant. (Scale bars, 200 μm.)

Liao et al. PNAS | vol. 115 | no. 20 | E4665

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

July

9, 2

020

Page 6: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

impaired angiogenesis as demonstrated by CD31 immunostainingand the dysregulation of angiogenesis genes (Fig. 4H and SI Ap-pendix, Fig. S8 A and B). Although not induced by TAC, macro-phage KLF4 appeared to be required for optimal expression of apotent angiogenesis factor, VEGFA, in vitro and in the heart (SIAppendix, Fig. S8 C andD). These data, paired with the observationthat many cardiac macrophages reside in the perivascular region(Fig. 1C), suggest that cardiac macrophages may have a potentialrole in angiogenesis or other vascular responses. In addition toimpaired angiogenesis, we observed that K4-cKO hearts exhibitedenhanced fibrosis after TAC (Fig. 4F). However, qPCR analysis

using a panel of 84 fibrosis-related genes (Qiagen RT2 Profiler PCRarray) revealed no significant difference between Cre and K4-cKOcardiac macrophages (SI Appendix, Fig. S8E). These observations,coupled with the appreciation of reduced KLF4-deficient cardiacmacrophage numbers (Fig. 4C), suggest that enhanced fibrosis seenin the K4-cKO heart was likely secondary to myocardial injury (Fig.4 F and G).Because KLF4 also regulates macrophage M1/M2 polarization

(24, 25) and gene expression in KLF4-deficient macrophagesexhibited enriched inflammation pathways (Fig. 4A), we tested ifthe macrophage polarization state contributes to the phenotype

Fig. 4. Myeloid KLF4 regulates cardiac macrophage proliferation in POH. (A) RNA-seq studies with primary peritoneal macrophages from Cre and K4-cKOmice. A total of 283 genes were differentially regulated between the Cre and K4-cKO groups, and gene ontology (GO) term enrichment analysis revealed ninepathways that were functionally enriched [false discovery rate (FDR) < 0.05]. Arrows indicate pathways of interest. (B) KLF4-deficient peritoneal macrophagesexhibited an impaired proliferative response to CSF1/2 stimulation (48 h). *P < 0.05. (C) KLF4-deficiency impaired TAC-induced cardiac macrophage pro-liferation (n = 10). *P < 0.05. (D) Myeloid KLF4 deficiency impaired cardiac function after TAC, resulting in dilated cardiomyopathy at 2 wk post-TAC (n = 8–10). *P < 0.05. EDV, end-diastolic volume; ESV, end-systolic volume; LVEF, left ventricular ejection fraction. (E) Myeloid KLF4 deficiency accelerated TAC-induced cardiac hypertrophy. Heart weight (HW; milligrams) and lung weight (LW; milligrams) at 2 wk post-TAC were normalized to body weight (grams) (n =6 in each group). *P < 0.05 by t test with Bonferroni correction. (F) TAC induced more fibrosis, cell death, and mitochondrial damage in K4-cKO hearts.Samples were from 1-wk TAC and sham mice. (Black scale bar, 100 μm; white scale bar, 2 μm.) (G) TAC induced higher plasma cardiac troponin I levels in theK4-cKO group (n = 5). *P < 0.05 with Bonferroni correction. (H) Impaired angiogenesis in K4-cKO hearts after TAC as revealed by myocardial capillary densityusing CD31 immunostaining (n = 5). *P < 0.05. (Scale bars, 100 μm.).

E4666 | www.pnas.org/cgi/doi/10.1073/pnas.1720065115 Liao et al.

Dow

nloa

ded

by g

uest

on

July

9, 2

020

Page 7: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

observed after TAC. Consistent with our previous report (24),K4-cKO hearts at 1 wk post-TAC showed lower expression ofmultiple M2 genes (SI Appendix, Fig. S9A). However, the ex-pression of M1 genes in K4-cKO hearts was also lower, in-dicating the absence of excessive inflammation (SI Appendix, Fig.S9B). We attribute this phenomenon to reduced cardiac mac-rophage numbers in K4-cKO hearts after TAC (Fig. 4C). Wefurther studied two classic M2-deficient animals (26), namely,hematopoietic STAT6-KO mice (via transplantation of STAT6-KO bone marrow to WT recipients) and myeloid IL4Ra-KOmice (Lyz2-Cre:IL4Raflox/flox), and found that biased M1/M2polarization had no effect on POH (SI Appendix, Fig. S9 C and D).As such, we conclude that the role of KLF4 in cardiac macrophagesin POH is not simply defined by M1/M2 polarization but is sec-ondary to its regulation of cardiac resident macrophage pro-liferation and their angiogenic potential (Fig. 4C and SI Appendix,Fig. S8D).

Infiltrating Macrophages Are Detrimental to the Heart in Late-PhasePOH. While our studies indicate that blood-borne macrophagesare not required for the early-phase protective compensatoryresponse to POH, they do infiltrate the myocardium during late-stage POH (Fig. 1G). Because heart failure is often associatedwith immune cell infiltration and inflammation (27–29), we asked if

blood-borne macrophages contribute to late-phase decompensationof POH. As shown in Fig. 2F, TAC increased circulating Ly6Chi

monocytes during late-phase POH. These monocytes were furthercharacterized as Ly6ChiCX3CR1+CCR2+ classic monocytes (Fig.5A). To block such monocytes, we utilized the CCR2-KO mouse,which is known to have reduced circulating classic monocytes due toimpaired CCL2/CCR2-mediated trafficking from the bone marrow(30, 31). When subjected to 4-wk TAC, the circulating monocytesremained much lower (∼25%) in CCR2-KO mice than WT controlmice (Fig. 5B). Consistently, overall macrophage numbers in late-phase POH were also lower (∼50%) within CCR2-KO myocardium(Fig. 5C).Serial echocardiography for 8 wk post-TAC demonstrated that

WT mice gradually develop cardiac dysfunction during late-phase POH, while CCR2-KO mice maintain relatively normalcardiac function (EF at 8 wk post-TAC: WT: 52 ± 2.0% vs.CCR2-KO: 65 ± 1.5%; P < 0.0001) (Fig. 5D). Although therewas no difference in cardiac hypertrophy or fibrosis between WTand CCR2-KO groups (Fig. 5 E–H), myocardial capillary densitywas significantly higher in the CCR2-KO group (capillary countsper 1 mm2 of myocardium: WT: 1,872 ± 113 vs. CCR2-KO:2,359 ± 98; ∼26% increase; P < 0.0038) (Fig. 5I), suggestingthat CCR2-KO mice were able to maintain superior angiogenesisduring the 8 wk of TAC. Finally, blocking CCR2 signaling in WT

Fig. 5. Blockade-infiltrating macrophages preserved cardiac function in late-phase POH. (A) Classic Ly6C+CCR2+ monocytes increased in blood after 4 wk ofTAC. A representative FACS plot from five samples is shown. (B) Reduced monocyte numbers in CCR2-KO mice after 4 wk of TAC (n = 5). *P < 0.05 by t test. (C)Reduced cardiac macrophage numbers in CCR2-KO hearts after 4 wk of TAC (n = 5). *P < 0.05 by t test. (D) Left ventricular function assessed by echocar-diography. The P value is shown as P (genotype * time) calculated from two-way ANOVA. (E) Heart weight and lung weight. (F) Expression of hypertrophymarker genes. (G) Cardiomyocyte cross-section area. Cell membranes were outlined by wheat germ agglutinin (WGA) staining. (H) Fibrosis assessed byPicoSirus Red staining, with collagen stained in red. Perivascular and intramuscular areas are shown. Fibrosis was quantified as the percentage of the fibroticarea in total by ImageJ (NIH) software. (I) Myocardial capillary density assessed by CD31 staining. All tissue samples were assessed at 8 wk post-TAC (n = 5–8).*P < 0.05. NS, not significant. (Scale bars, 200 μm.)

Liao et al. PNAS | vol. 115 | no. 20 | E4667

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

July

9, 2

020

Page 8: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

animals using RS preserved cardiac function in an establishedPOH model (RS oral administration 2 wk after TAC) withoutaffecting cardiac hypertrophy (SI Appendix, Fig. S10). Collec-tively, these data demonstrate that blocking monocyte/macro-phage infiltration maintains angiogenesis and preserves cardiacfunction without affecting cardiac hypertrophy or fibrosis in late-phase POH. Taken together, these data demonstrate that late-phase infiltration of Ly6ChiCX3CR1+CCR2+ classic monocytescontributes to cardiac dysfunction in POH and, conversely, thatblockade of infiltrating macrophages (genetically by CCR2-KOor pharmaceutically by CCR2 antagonist) preserves heart func-tion, in part, through improved angiogenesis.

DiscussionThe heart contains resident macrophages and can recruit in-filtrating macrophages upon stress. As cardiac resident andinfiltrating macrophages have separate origins and reside indistinct geographic locations, it is not surprising that they havedifferent functions as well. In the present study using a mousemodel of NICM, we show that resident macrophage proliferationand angiogenic activity in the initial period following pressureoverload stress are critical for cardiac adaptation and function.Furthermore, cardiac resident macrophages are essential tomaintain cardiac function even in the late phase of POH. Bycontrast, in the late phase of POH, infiltrating monocytes/mac-rophages promote decompensation and blockade of this ingressis ameliorative. These findings are aligned with recent observa-tions that in models of acute and chronic post-MI remodeling,resident macrophages are salutary, while infiltrating macro-phages are detrimental (15, 16). Further, in neonatal hearts,tissue macrophages have been shown to regulate cardiac angio-genesis and regeneration after apical resection or MI (15, 32).Collectively, the current work, along with these recent findings,underscores the differential roles of tissue-intrinsic and tissue-extrinsic macrophages in the cardiac response to injury (sum-marized in Fig. 6).An important aspect of our work relates to the identification

of KLF4 as a critical regulator of resident macrophage pro-liferation and angiogenic function. We found that myeloidKLF4 deficiency resulted in a marked reduction in local pro-liferation of cardiac resident macrophages and the total numberof cardiac macrophages 7 d after TAC. Pathway analysis ofRNA-seq studies in KLF4-deficient macrophages highlightedenrichment for pathways that control cellular proliferation.These findings are particularly intriguing considering prior worklinking KLF4 to macrophage proliferation (23). The upstreammechanism following POH that leads to KLF4 induction remainsincompletely understood. However, a recent study by Fujiu et al.(33) reported that kidney-derived CSF2 activated proliferationof cardiac resident macrophages in response to TAC. We ob-served that myeloid KLF4 deficiency significantly diminishedmacrophage proliferation in response to CSF2 (as well as CSF1;Fig. 4B). As such, it is possible that kidney-derived CSF2 inducesKLF4-dependent resident macrophage proliferation. Futurestudies will be needed to delineate the detailed mechanisms bywhich KLF4 regulates proliferation of resident macrophages invivo and whether KLF4 is universally required for residentmacrophage proliferation in other tissues.Another important observation relates to myeloid KLF4

control of angiogenesis. We observed that myeloid KLF4 KOanimals demonstrated a marked susceptibility to heart failure,likely due to reduced angiogenic capacity leading to car-diomyocyte death (Fig. 4). Consistent with this, myeloid KLF4KO mice demonstrated significant cardiac troponin levels inplasma and clear evidence of cardiac mitochondrial damage.The basis for myeloid KLF4 control of angiogenesis remainsincompletely understood, but we note that expression of anumber of proangiogenic factors, such VEGFA, is reduced

and that expression of potent antiangiogenic factors, such asThbs1, is increased in the K4-cKO hearts. The reduced ex-pression of VEGFA is further confirmed in cardiac macro-phages isolated from K4-cKO hearts. An intriguing andunanswered question relates to how the relatively smallnumber of resident macrophages can impact angiogenesisthroughout the heart. As macrophages often cluster aroundblood vessels, it is possible that they may release VEGF intothe coronary circulation to impact the heart more broadly.Alternatively, the acute heart failure (<3 d) observed inmacrophage-depleted animals (Fig. 3B) suggests that cardiacmacrophages may also regulate arteriogenesis in POH asreported previously (34). Additional studies will be requiredto address these considerations. Finally, while we and othershave also shown that KLF4 regulates macrophage polarization(24, 25), the present study excludes the significance of mac-rophage M1/M2 polarization in POH using two additionalM2-deficient mouse models, namely, hematopoietic STAT6-KO and myeloid-specific IL4Ra-KO mice, which exhibitednormal cardiac adaptation to POH despite lack of classic M2macrophages (26, 35). We also note that Aurora et al. (32)have reported that cardiac resident macrophages mainly me-diate angiogenesis rather than inflammation, with no clear biastoward M1 or M2 in a neonatal myocardial injury model.In addition to resident macrophage proliferation in early-

phase POH (<7 d), there is significant infiltration of classicLy6Chi monocytes during late-phase POH (>4 wk). Infiltrationof these tissue-extrinsic monocytes/macrophages takes placeduring the cardiac decompensation phase, suggesting a poten-tially negative role in POH. Consistent with this view, our studiesin CCR2-KO mice showed that blockade of infiltrating macro-phages (monocytes) preserved cardiac function, likely throughimproved myocardial angiogenesis (Fig. 5). How infiltratingmacrophages affect angiogenesis remains to be fully understood.Infiltrating macrophages may trigger inflammation and otherresponses that negatively regulate angiogenesis (32). Alterna-tively, infiltrating macrophages may negatively impact residentmacrophages, which are endowed with angiogenic capacity (15).It is noteworthy that cardiac resident macrophages also persistinto and clearly are requisite for the late-phase POH (SIAppendix, Fig. S5). As such, cardiac resident and infiltrating

Fig. 6. Dynamic changes and distinct roles of cardiac macrophage subsets inPOH. Our studies have demonstrated a two-phase response of cardiacmacrophages to pressure overload. (i) During the early phase of POH, thereis robust local proliferation of resident macrophages that is crucial for car-diac compensation to pressure overload. (ii) During the late phase of POH,the infiltration of inflammatory monocytes increases, which promotes thetransition to cardiac decompensation. Mechanistically, transcription factorKLF4 is required for resident macrophage proliferation and the infiltratingmonocytes are Ly6ChiCX3CR1+CCR2+ classic monocytes. Finally, this modelimplies that these two subsets of cardiac macrophages can be targeted fortherapeutic gain, either by blocking monocytes (i.e., RS administration) or bypromoting resident macrophages (i.e., prolonged proliferation), or by si-multaneously targeting both subsets for a healthy balance.

E4668 | www.pnas.org/cgi/doi/10.1073/pnas.1720065115 Liao et al.

Dow

nloa

ded

by g

uest

on

July

9, 2

020

Page 9: Distinct roles of resident and nonresident macrophages in ... · and Mukesh K. Jaina,b,1 a Case Cardiovascular Research Institute, Case Western Reserve University School of Medicine,

macrophage subsets may regulate POH through temporal andspatial activation and interaction between each other. In additionto their roles in angiogenesis as discussed above, cardiac residentmacrophages have been reported to regulate electrical conduc-tion of the heart (21). Although CL-mediated macrophage de-pletion is insufficient to induce arrhythmia in unstressed baselinehearts (21), it is possible that loss of resident macrophages mayresult in arrhythmia in a more susceptible substrate, such as thehypertrophied heart (36). For instance, some of the post-TACmortality in CL-treated TAC animals might result from suddendeath (Fig. 3C and SI Appendix, Fig. S5A).Heart failure affects more than 26 million people worldwide

and (37, 38) is associated with a devastating ∼50% mortalitywithin 5 y of diagnosis (39). Current therapies aimed at reducingneurohormonal stress and improving hemodynamics are onlypartially effective, underscoring the need for orthogonal strate-gies based on newly identified molecular and cellular mechanisms.The studies here highlight the differential impact of cardiac residentand nonresident macrophages in an experimental model of heartfailure. Further, they suggest that inhibition of blood-borne mac-

rophages through blockade of the CCR2 pathway can be exploitedfor therapeutic gain.

Materials and MethodsDetailed materials and methods can be found in SI Appendix. RNA-seq dataare deposited in the Gene Expression Omnibus (accession no. GSE107016).All experimental procedures were approved by the Institutional Animal Careand Use Committee of Case Western Reserve University School of Medicineand were conducted in accordance with the NIH Guide for the Care and Useof Laboratory Animals (40).

ACKNOWLEDGMENTS. We thank Ajay Chawla for providing the IL4Ra-cKOmice, Simone Edelheit and Alexander Miron for assistance in RNA-seqstudies, Mike Sramkoski for assistance in flow cytometry, and Sarah Liaofor artistic assistance. This work was supported by American Heart Associ-ation (AHA) National Scientist Development Grant 12SDG12070077 (to X.L.),AHA Postdoctoral Fellowship 17POST33650110 (to Y.S.), National NaturalScience Foundation of China Grant 81400347 (to L.Z.), NIH Grants T32GM007250and F30HL139014 (to D.R.S.), NIH Grants R35 HL135789 and R01 DK111468-01 (toM.K.J.), and the Elisabeth Severance Prentiss Foundation Grant (to M.K.J.). Thiswork was also generously supported by Tom F. Peterson.

1. Cecchi F, et al. (2003) Coronary microvascular dysfunction and prognosis in hyper-trophic cardiomyopathy. N Engl J Med 349:1027–1035.

2. Treasure CB, et al. (1993) Hypertension and left ventricular hypertrophy are associ-ated with impaired endothelium-mediated relaxation in human coronary resistancevessels. Circulation 87:86–93.

3. Maron BJ, Wolfson JK, Epstein SE, Roberts WC (1986) Intramural (“small vessel”)coronary artery disease in hypertrophic cardiomyopathy. J Am Coll Cardiol 8:545–557.

4. Frey N, Luedde M, Katus HA (2011) Mechanisms of disease: Hypertrophic cardiomy-opathy. Nat Rev Cardiol 9:91–100.

5. SanoM, et al. (2007) p53-induced inhibition of Hif-1 causes cardiac dysfunction duringpressure overload. Nature 446:444–448.

6. Prosdocimo DA, et al. (2014) Kruppel-like factor 15 is a critical regulator of cardiaclipid metabolism. J Biol Chem 289:5914–5924.

7. Izumiya Y, et al. (2006) Vascular endothelial growth factor blockade promotes thetransition from compensatory cardiac hypertrophy to failure in response to pressureoverload. Hypertension 47:887–893.

8. Hashimoto D, et al. (2013) Tissue-resident macrophages self-maintain locallythroughout adult life with minimal contribution from circulating monocytes.Immunity 38:792–804.

9. Epelman S, et al. (2014) Embryonic and adult-derived resident cardiac macrophagesare maintained through distinct mechanisms at steady state and during in-flammation. Immunity 40:91–104.

10. Gomez Perdiguero E, et al. (2015) Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518:547–551.

11. Deshmane SL, Kremlev S, Amini S, Sawaya BE (2009) Monocyte chemoattractantprotein-1 (MCP-1): An overview. J Interferon Cytokine Res 29:313–326.

12. Misharin AV, et al. (2014) Nonclassical Ly6C(-) monocytes drive the development ofinflammatory arthritis in mice. Cell Rep 9:591–604.

13. Cohen HB, Mosser DM (2014) Cardiac macrophages: How to mend a broken heart.Immunity 40:3–5.

14. Frantz S, Nahrendorf M (2014) Cardiac macrophages and their role in ischaemic heartdisease. Cardiovasc Res 102:240–248.

15. Lavine KJ, et al. (2014) Distinct macrophage lineages contribute to disparate patternsof cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl AcadSci USA 111:16029–16034, and erratum (2016) 113:E1414.

16. Sager HB, et al. (2016) Proliferation and recruitment contribute to myocardial mac-rophage expansion in chronic heart failure. Circ Res 119:853–864.

17. Mirzadegan T, et al. (2000) Identification of the binding site for a novel class of CCR2bchemokine receptor antagonists: Binding to a common chemokine receptor motifwithin the helical bundle. J Biol Chem 275:25562–25571.

18. Terrazas C, et al. (2017) Ly6Chi inflammatory monocytes promote susceptibility toLeishmania donovani infection. Sci Rep 7:14693.

19. Raghu H, et al. (2017) CCL2/CCR2, but not CCL5/CCR5, mediates monocyte recruitment,inflammation and cartilage destruction in osteoarthritis. Ann Rheum Dis 76:914–922.

20. Van Rooijen N, Sanders A (1994) Liposome mediated depletion of macrophages:Mechanism of action, preparation of liposomes and applications. J Immunol Methods174:83–93.

21. Hulsmans M, et al. (2017) Macrophages facilitate electrical conduction in the heart.Cell 169:510–522.e20.

22. Jenkins SJ, et al. (2013) IL-4 directly signals tissue-resident macrophages to proliferatebeyond homeostatic levels controlled by CSF-1. J Exp Med 210:2477–2491.

23. Aziz A, Soucie E, Sarrazin S, Sieweke MH (2009) MafB/c-Maf deficiency enables self-renewal of differentiated functional macrophages. Science 326:867–871.

24. Liao X, et al. (2011) Krüppel-like factor 4 regulates macrophage polarization. J ClinInvest 121:2736–2749.

25. Kapoor N, et al. (2015) Transcription factors STAT6 and KLF4 implement macrophagepolarization via the dual catalytic powers of MCPIP. J Immunol 194:6011–6023.

26. Tugal D, Liao X, Jain MK (2013) Transcriptional control of macrophage polarization.Arterioscler Thromb Vasc Biol 33:1135–1144.

27. Dick SA, Epelman S (2016) Chronic heart failure and inflammation: What do we reallyknow? Circ Res 119:159–176.

28. Fu M (2009) Inflammation in chronic heart failure: What is familiar, what is un-familiar? Eur J Heart Fail 11:111–112.

29. Ismahil MA, et al. (2014) Remodeling of the mononuclear phagocyte network un-derlies chronic inflammation and disease progression in heart failure: Critical im-portance of the cardiosplenic axis. Circ Res 114:266–282.

30. Geissmann F, Jung S, Littman DR (2003) Blood monocytes consist of two principalsubsets with distinct migratory properties. Immunity 19:71–82.

31. Tsou CL, et al. (2007) Critical roles for CCR2 and MCP-3 in monocyte mobilization frombone marrow and recruitment to inflammatory sites. J Clin Invest 117:902–909.

32. Aurora AB, et al. (2014) Macrophages are required for neonatal heart regeneration.J Clin Invest 124:1382–1392.

33. Fuji UK, et al. (2017) A heart-brain-kidney network controls adaptation to cardiacstress through tissue macrophage activation. Nat Med 23:611–622.

34. Takeda Y, et al. (2011) Macrophage skewing by Phd2 haplodeficiency prevents is-chaemia by inducing arteriogenesis. Nature 479:122–126.

35. Nguyen KD, et al. (2011) Alternatively activated macrophages produce catechol-amines to sustain adaptive thermogenesis. Nature 480:104–108.

36. McLenachan JM, Henderson E, Morris KI, Dargie HJ (1987) Ventricular arrhythmias inpatients with hypertensive left ventricular hypertrophy. N Engl J Med 317:787–792.

37. Benjamin EJ, et al.; American Heart Association Statistics Committee and StrokeStatistics Subcommittee (2017) Heart disease and stroke statistics-2017 update: Areport from the American Heart Association. Circulation 135:e146–e603, and erratum(2017) 136:e196.

38. Ambrosy AP, et al. (2014) The global health and economic burden of hospitalizationsfor heart failure: Lessons learned from hospitalized heart failure registries. J Am CollCardiol 63:1123–1133.

39. Mozaffarian D, et al.; Writing Group Members; American Heart Association StatisticsCommittee; Stroke Statistics Subcommittee (2016) Heart disease and stroke statistics-2016 update: A report from the American Heart Association. Circulation 133:e38–e360, and erratum (2016) 133:e599.

40. National Research Council (2011) Guide for the Care and Use of Laboratory Animals(National Academies Press, Washington, DC), 8th Ed.

Liao et al. PNAS | vol. 115 | no. 20 | E4669

MED

ICALSC

IENCE

SPN

ASPL

US

Dow

nloa

ded

by g

uest

on

July

9, 2

020