Hand2 loss-of-function in Hand1-expressing cells reveals distinct roles in epicardial and coronary vessel development.
Journal: 2011/June - Circulation Research
ISSN: 1524-4571
Abstract:
BACKGROUND
The basic helix-loop-helix (bHLH) transcription factors Hand1 and Hand2 are essential for embryonic development. Given their requirement for cardiogenesis, it is imperative to determine their impact on cardiovascular function.
OBJECTIVE
To deduce the role of Hand2 within the epicardium.
RESULTS
We engineered a Hand1 allele expressing Cre recombinase. Cardiac Hand1 expression is largely limited to cells of the primary heart field, overlapping little with Hand2 expression. Hand1 is expressed within the septum transversum, and the Hand1 lineage marks the proepicardial organ and epicardium. To examine Hand factor functional overlap, we conditionally deleted Hand2 from Hand1-expressing cells. Hand2 mutants display defective epicardialization and fail to form coronary arteries, coincident with altered extracellular matrix deposition and Pdgfr expression.
CONCLUSIONS
These data demonstrate a hierarchal relationship whereby transient Hand1 septum transversum expression defines epicardial precursors that are subsequently dependent on Hand2 function.
Relations:
Content
Citations
(21)
References
(54)
Chemicals
(1)
Genes
(20)
Organisms
(4)
Processes
(2)
Anatomy
(4)
Similar articles
Articles by the same authors
Discussion board
Circ Res 108(8): 940-949

<em>Hand2</em> loss-of-function in <em>Hand1</em>-expressing Cells Reveals Distinct Roles In Epicardial And Coronary Vessel Development

Rationale

The bHLH transcription factors Hand1 and Hand2 are essential for embryonic development. Given their requirement for cardiogenesis, it is imperative to determine their impact on cardiovascular function.

Objective

Deduce the role of Hand2 within the epicardium.

Method &amp; Results

We engineered a Hand1 allele expressing Cre recombinase. Cardiac Hand1 expression is largely limited to cells of the primary heart field, overlapping little with Hand2 expression. Hand1 is expressed within the septum transversum (ST) and the Hand1-lineage marks the proepicardial organ and epicardium. To examine Hand factor functional overlap, we conditionally deleted Hand2 from Hand1-expressing cells. Hand2 mutants display defective epicardialization and fail to form coronary arteries, coincident with altered ECM deposition and Pdgfr expression.

Conclusion

These data demonstrate a hierarchal relationship whereby transient Hand1 ST expression defines epicardial precursors that are subsequently dependent upon Hand2 function.

Introduction

When cardiac gene programs are hobbled, there is rarely an ablation of the cardiac lineage, but more often defective cell differentiation and/or tissue morphogenesis, which manifests as congenital heart defects (CHDs). Multiple cell lineages collectively form the heart 1. While myocyte derivatives ultimately provide the cardiac musculature that powers circulation, it is the extra-cardiac cell lineages (the cardiac neural crest cells (cNCC), coronary endothelium, and epicardium) that define the framework upon which these cardiomyocytes develop 25. Although the heart functions as an integrated whole, our understanding of its individual components is not uniform. For example, the second heart field (SHF) and cNCC have been extensively studied 6, while the differentiation programs of the primary heart field (PHF) and coronary vasculature are less well defined.

Both cNCC and epicardial cell lineages undergo Epithelial-to-Mesenchymal Transition (EMT) and directly interact with differentiating cardiomyocytes. cNCCs enable septation of the outflow track (OFT) into the aorta and pulmonary trunk. Epicardial mesothelium migrates from the proepicardial organ (PE) to cover the surface of the heart 7. Subsequently, an epicardial subpopulation undergoes secondary EMT, invades the heart, and differentiates into coronary smooth muscle and cardiac fibroblasts 811. Cardiac morphogenesis requires communication between cardiac and extra-cardiac cell populations 2, 3, 12. The signaling cascades through which these cell lineages communicate to are just coming to light.

Expression of the Twist-family basic Helix-loop-Helix (bHLH) transcription factors Hand1 and Hand213, 14 partially overlaps within the developing heart and cNCC 1518. Hand2 expression marks the cardiac crescent and linear heart tube and is downregulated within the left ventricle at the onset of cardiac looping 18. At comparable developmental stages, the left ventricle and myocardial cuff express Hand115, 16, 19. Understanding where and when Hand1 and Hand2 function during embryonic development is a challenge due to their dynamic spatiotemporal expression profiles 13.

Hand factors are critical for cardiac morphogenesis 13, 14. Systemic ablation of Hand2 results in a single ventricle, increased apoptosis, and lethality by E9.5 18, 20. Hand1 knockout mice die by E9.5 from extraembryonic and vascular defects 16, 19, 21. Conditional NCC-specific Hand2 ablation (Hand2Δ) causes OFT defects, associated ventricular septal defects (VSD), Double Outlet Right Ventricle (DORV) and decreased SHF myocardial proliferation 22, 23. Myocardial-specific Hand2 deletion causes right ventricular hypoplasia and death by E12.5 23. Reciprocally, myocardial-specific Hand1 deletion causes left-ventricular hypoplasia and although embryos survive until birth, Hand2 haploinsufficiency increases phenotypic severity suggesting genetic and functional overlap 24, 25.

Here, we investigate the contributions of Hand1-expressing cells to cardiogenesis using a Cre recombinase-expressing Hand1 allele 26. The Hand1 lineage robustly marks the left ventricular myocardium, a cNCC sub-population, and, unexpectedly, the epicardium and its derivatives.

To deduce functional/genetic interactions between Hand factors during cardiogenesis, we conditionally deleted Hand2 within the Hand1 lineage (H2CKO). H2CKOs embryos die by E14.5 and present Persistent Truncus Arteriosis (PTA), DORV and associated VSDs. Significantly, Hand2 expression lies temporally downstream of Hand1 within the septum transversum- (ST) derived cells that migrate into the proepicardial organ (PE), and is thus deleted from the PE, epicardium and the epicardial derivatives of H2CKOs. H2CKOs display defective epicardial EMT, decreased cardiac fibroblasts, and a nonfunctional coronary vasculature. Hand2 ablation within the WT1-lineage phenocopies Hand1Cre-mediated Hand2 ablation. Gene expression analyses reveal an altered ratio of Pdgfrα:Pdgfrβ mRNA, a decreased profile of fibroblast markers, and disorganized ECM. These changes are associated with increased epicardial cell apoptosis. Together, our data shows that Hand2 performs an essential role during epicardialization, which directly impacts epicardial cell differentiation, and formation of the coronary vasculature.

Methods

See the online supplement for detailed methods.

Targeting Hand1 and Generation of Mice

The construction of the targeting vector, generation of targeted ES cells, chimera and germline mice is described in detail in 26. Tamoxifen was administered for the as described 4.

In Situ Hybridization

Section and Wholemount in situ hybridization (ISH) was performed essentially as previously described 26, 28,29.

Primary Epicardial Culture

Primary epicardial cultures were isolated and cultured as previously described 31.

Immunohistochemistry

Embryos were fixed in 4% PFA overnight then embedded in paraffin or cryoprotected and sectioned at 7mm as previously reported 26.

Targeting Hand1 and Generation of Mice

The construction of the targeting vector, generation of targeted ES cells, chimera and germline mice is described in detail in 26. Tamoxifen was administered for the as described 4.

In Situ Hybridization

Section and Wholemount in situ hybridization (ISH) was performed essentially as previously described 26, 28,29.

Primary Epicardial Culture

Primary epicardial cultures were isolated and cultured as previously described 31.

Immunohistochemistry

Embryos were fixed in 4% PFA overnight then embedded in paraffin or cryoprotected and sectioned at 7mm as previously reported 26.

Results

Hand1 expression within the ST marks the progenitors of the PE

As Hand1-lineage cells, but not Hand1-expressing cells, are observed within the epicardium at E10.5 (See online supplement Fig. I), we sought the Hand1-expressing progenitors of these cells. The PE is derived from the anterior ST and gives rise to the epicardium 33. ISH was performed for both Hand1 and Tbx18, a marker of the PE, at E9.5 3, 34 (Fig. 1A-E). Tbx18 is expressed throughout the PE but is not detectable within the ST. In contrast, Hand1 expression is not detectable within the PE but is expressed robustly throughout the ST, thus identifying the source of the Hand1-marked epicardium. X-Gal staining of Hand1LacZ/+ embryos confirms expression within the LV and the ST (Fig. 1F). X-Gal staining of the Hand1-lineage shows Hand1-marked cells dispersed throughout the ST and within the more proximal Tbx18-expressing region, supporting established models that PE cells derive from migratory ST cells (Fig. 1G-I) 3, 34. These results establish that the Hand1-lineage cells observed throughout the cardiac fibroblasts, coronary vasculature, epicardium and PE originate from a Hand1-expressing ST cell population.

An external file that holds a picture, illustration, etc.
Object name is nihms280562f1.jpg

The Hand1 lineage gives rise to epicardial progenitors. (A) E9.5 illustration showing Hand1 expression (blue) and depicting the planes of section in B-E (B). Hand1 and Tbx18 section ISH of E9.5 embryos on adjacent sections through the ST and the PE (B-E). Hand1 expression is not detected within the PE but is expressed in the ST. X-gal staining of Hand1LacZ (F) and Hand1 R26R activation (G-I) shows Hand1-lineage cells within the ST and in the PE.. lv, left ventricle; pe, proepicardium; st, septum transversum.

Distinct Hand factor expression during heart morphogenesis

E8.5 Hand1-lineage analysis reveals that the linear heart tube is not completely derived from Hand1-expressing cells, indicating that Hand1 expression initiates after heart tube fusion. To account for a possible temporal delay of Cre expression, we performed wholemount ISH for Hand1 at E7.5 (Fig. 2). Hand1 is detected throughout the extra embryonic mesoderm, chorion, and allantoic rudiment (Fig. 2A). As the chorion is directly adjacent to the cardiac crescent (Fig. 2A,B white arrow), we performed a double label ISH for both Hand1 and the cardiac marker Mlc2a. Hand1 expression does not directly overlap with Mlc2a expression at E7.5, indicating that, consistent with the Hand1-lineage analysis, Hand1 cardiomyocyte expression initiates subsequent to linear heart tube fusion (Fig. 2B).

An external file that holds a picture, illustration, etc.
Object name is nihms280562f2.jpg

Hand factor expression is distinct. Single and double-labeled Mlc2a and Hand1 wholemount RNA ISH of E7.5 embryos show that Hand1 is not expressed within the cardiac crescent (white arrow) but is expressed within extra embryonic mesoderm, allantoic rudiment, and the chorion (A, B). E8.5 wholemounts of Hand1 and Hand2 reveal complimentary expression during cardiac development (C-F). Hand2 expression is confined to the SHF and endocardium (black arrow). cc, cardiac crescent; eem, extraembryonic mesoderm; ht, heart tube; lm, lateral mesoderm; oft, outflow tract; v, ventricle.

Wholemount ISH at E8.5 (Fig. 2C-F) shows Hand1 expression within the posterior heart tube while Hand2 expression is observed throughout the entire cardiac field. Sagittal sections of wholemount-stained embryos confirm that Hand1 expression is restricted to the early ventricular chamber and cuff myocardium. Hand2 expression is robust throughout the endocardium, SHF and OFT. Interestingly, Hand2 expression is not detected within the ventricular myocardium at E8.5 and, in wholemount view, the Hand2 endocardial expression is visible through the thin myocardial wall (Fig. 2F, red arrow). At E9.5, Hand1 and Hand2 expression overlaps within the SHF-derived myocardial cuff and the left ventriclular myocardium (not shown). Collectively, these data reaffirm that Hand factor expression is dynamically regulated and that, during early heart morphogenesis, this expression overlaps solely within the SHF-derived myocardial cuff.

Hand1Cre-mediated ablation of Hand2 within the, PE, and epicardium

To explore potential Hand factor functional overlap, we intercrossed Hand2fx23 and Hand1Cre mice to ablate Hand2 within the Hand1-lineage (H2CKO). Hand2 expression is robust throughout the PE and detectable in both E10.5 epicardial cells and isolated primary epicardial cell cultures (arrow, Fig. 3A,C; Online Fig. II). Hand2 expression was not detected within the epicardium or the PE of H2CKOs, confirming that Hand1-positive ST cells ultimately populate the PE and epicardium (Fig. 3B,D). Hand1 and Hand2 are not coexpressed within the ST or PE; rather, Hand1 expression within the ST precedes PE/epicardial upregulation of Hand2. Thus, Hand1Cre conditionally ablates Hand2 function within the PE and epicardium.

An external file that holds a picture, illustration, etc.
Object name is nihms280562f3.jpg

Inactivation of Hand2 within the Hand1 lineage RNA ISH at E9.5 (A, B) shows Hand2 expression throughout the PE and in the epicardium (red arrow) at E10.5 (C, D). Consequently, Hand2 expression is ablated in the presumptive epicardial mesothelium in H2CKOs; pe, proepicardium.

Hand1-lineage H2CKO reveals novel phenotypes and embryonic lethality

H2CKO embryos were recovered at slightly below expected Mendelian ratios through E12.5 (Fig. 4). At E14.5, roughly 50% of H2CKOs collected were dead. E12.5 H2CKOs appeared phenotypically normal (data not shown). Cardiac patterning, and both Hand1 and cardiomyocyte marker expression were normal (Online Fig. IIIA-P).

An external file that holds a picture, illustration, etc.
Object name is nihms280562f4.jpg

Conditional Hand2 deletion within the Hand1 lineage results in extensive embryonic and cardiovascular defects. Histological (A, B, E, F, I-N) and wholemount (C, D, G, H) analysis of E14.5.H2CKO embryos. Mutants display pericardial hemorrhaging (C) and anasarca (D). Compared to WT mice (G), H2CKOs have hypoplastic livers with extensive hemorrhaging (H).. H2CKOs display either PTA or DORV (A, B, E, F, I, J). All H2CKOs exhibit VSDs (K, L). Myocardial examination shows abnormal trabeculae and non-compaction (M, N). ao, dorsal aorta; cz, compact zone; pt, pulmonary trunk; pta, persistent truncus arteriousus; tr, trabeculae.

By E14.5, H2CKOs displayed pericardial hemorrhaging, anasarca, and liver hypoplasia (Fig. 4C, D, G, H). OFT septation defects, including PTA and DORV, were also observed, reflecting cNCC and/or SHF-derived myocardial defects (Fig. 4A, B, E, F, I-J). Hypertrabeculation/noncompaction and VSDs were also observed (Fig. 4K-N), although, H2CKO sarcomeric ultra-structure was indistinguishable from controls (Online Fig. IIIQ, R).

Hand2 die from a sympathetic nervous system (SNS) norepinepherine deficit 22, 23. As Hand1 and Hand2 are coexpressed in some NCCs, H2CKOs possibly die from SNS defects. Hand2 lethality can be rescued by the β-adrenergic agonist isoproterenol 22, 23. Unlike Hand2NCC embryos, isoproterenol treated Hand1Cre-generated H2CKOs fail to survive to birth (Fig. 4), indicating that SNS defects do not account for embryonic lethality.

Hand2 is required for a functional epicardium

Given the temporal relationship observed between Hand1 expression in the ST and Hand2 within the PE/epicardium, we examined H2CKO epicardial defects (Fig. 5A, B). E14.5 H2CKO display abnormal compaction, an absence of epicardium, and a lack of coronary lumens, indicating a possible epicardial phenotype (Fig. 5A,B). ISH analyses of epicardial markers at E10.5 (data not shown) and at E12.5 for Tcf21 suggest that specification and formation of the early epicardium occurs normally (Fig. 5C,D). To confirm direct disruption of H2CKO epicardial development, we generated a WT1 H2CKO4, in which Hand2 is ablated in the ST, PE and epicardium, but not the myocardium and NCC lineages (Fig. 5E-H). (See Online Fig. IV for a WT1ERT2Cre, Hand1Cre, and Hand2 cardiac lineage/expression comparison). Potentially complicating these analyses, WT1ERT2Cre marks lateral mesoderm, where Hand2 is expressed, and early allelic WT1ERT2Cre induction (E8.5) can produce ectopic Cre activity 4. HoxB6-Cre-mediated Hand2 deletion in lateral mesoderm generates viable neonates 23. Tamoxifen-mediated WT1ERT2Cre allelic induction at E9.5 resulted in no ectopic myocardial R26R reporter activity (Online Fig. V). At E13.5, Hand2fx/−, WT1ERT2Cre mutant embryos phenocopied H2CKO embryos, displaying a poorly organized epicardium and reduced WT1-marked cell lineage (Fig. 5E-H). Myocardial non-compaction was also evident, further suggesting that this phenotype is non-cell autonomous. These mutants appear to phenocopy the epicardial defects and E14.5 embryonic lethality associated with H2CKOs (Fig. 5A, B, G, H). These two independent epicardial Hand2 ablations indicate that Hand2 may play multiple roles during epicardiogenesis, the initiation of secondary epicardial EMT, and the terminal differentiation of post EMT epicardial cell populations.

An external file that holds a picture, illustration, etc.
Object name is nihms280562f5.jpg

H2CKOs display epicardial phenotypes. Histological examination at E14.5 shows a lack of epicardium and compaction abnormalities in H2CKOs (A, B). RNA ISH for Tcf21 at E12.5 shows initial establishment of the epicardium (C, D). X-gal staining and conditional deletion of Hand2 within the WT1 lineage at E13.5 photocopies H2CKO epicardial defects (E-H).

Loss of Hand2 disrupts epicardial gene expression and coronary vasculature patency

As conditional Hand2 loss-of-function leads to epicardial defects, we sought to further identify the mechanistic role for Hand2 within the epicardium. Platelet Derived Growth Factor Receptors (Pdgfr) play essential roles in cell fate and specification of the mature epicardium 35, 36. E12.5 ISH reveals decreased Pdgfrα expression within the H2CKO epicardium (Fig. 6A,B). The number of cells expressing Periostin (Postn), a marker of the cardiac fibroblast cell fate 32, is visibly decreased in the myocardium of E12.5 H2CKO mutants (Fig. 6C-D). Together these data suggest that Hand2 functions in epicardial cell fate determination during or after secondary EMT.

An external file that holds a picture, illustration, etc.
Object name is nihms280562f6.jpg

Fibroblast lineage and secondary coronary defects in H2CKOs. RNA ISH at E12.5 for Pdgfrα and Postn (A-D). A decrease in cardiac fibroblasts is observed within the compact zone of H2CKOs. Immunohistochemistry for Flk1 (green) and F-actin (red) at E12.5 (E, F). A decrease in patent coronary vasculature is observed in H2CKOs.

We performed CD31 (Online supplement; Fig. VIA, B) and Flk1 immunohistochemistry (Fig. 6E, F) at E12.5 to examine coronary vessel formation. Flk1-positive cells running through the epicardium are absent. Phalloidin counterstaining indicates that the epicardium is intact, though absent of potential Flk1-positive lumens (Fig. 6F). The Hand1-lineage does not contribute to coronary endothelial cell populations (Online Fig. VIC-F) 26, and therefore the absence of coronary vasculature within H2CKOs likely results from non-cell autonomous mechanism(s). To take advantage of induction, we administered tamoxifen at E11.5 CD31-positive coronary lumens are visible within H2CKOs when WT1ERT2Cre is activated at E11.5, although they appear less prevalent than in embryos that lack the WT1ERT2Cre allele (Online Fig. VC-D), suggesting that critical Hand2 epricardial function occurs between E9.5 and 11.5.

To further address the impaired function of H2CKO epicardium, we generated Epicardial Primary Cultures (EPCs) from wild type and H2CKOs and isolated total RNA for microarray analyses (Online Fig. IIA-D). Gene ontology from our microarray analysis indicates significant differences in developmental, morphological, and cardiovascular gene programs within EPCs (Online Fig. VIG). Quantitative RT-PCR on RNA isolated from wild type and H2CKO EPCs was performed to validate changes in gene expression observed in the microarray (Online Fig. VIH). Quantitative RT-PCR confirms expression of Hand2 within the epicardium and its ablation within the H2CKO EPCs. Importantly, the ratio of Pdgfrα to Pdgfrβ is greatly altered in H2CKO EPCs. Expression of Pdgfrα is significantly reduced, whereas, Pdgfrβ expression is significantly upregulated (Online Fig. VIH). Hand2 regulation of Pdgfrα is direct (Online supplement; Fig. VI,I). In epicardial cells, Pdgfrβ-mediated signaling promotes a smooth muscle fate, while the role of Pdgfrα signaling is currently unclear 35. The decrease in Postn-positive myofibroblasts within the compact zone suggests that Pdgfrα impacts fibroblast differentiation. Together, these data suggest that Hand2 directly impacts epicardial cell fate through a Pdgfr-dependent mechanism.

Hand2 alters Fn1 fibril assembly and organization

As H2CKO epicardial defects appear direct, we looked at the impact of a loss of Hand2 upon epicardial mesothelium integrity. Fn1 is downregulated in zebrafish hand2 mutants 37. Fn1 fibril assembly regulates the organization and stability of ECM proteins, is capable of promoting EMT and adhesion-dependent growth, and is associated with integrin-mediated cell signaling 3840. Additionally, defects in Fn1 underlie integrin-mediated valve leaflet defects in the lymphatic system 41. Immunohistochemistry shows that Fn1 is expressed in the E12.5 epicardium and is neatly organized around the developing coronaries (Fig. 7A). H2CKOs retain epicardial Fn1; however, it appears disorganized (Fig. 7B). To look at Fn1 organization more closely, we compared EPCs from wild type and H2CKOs (Fig. 7C-F). Immunohistochemistry reveals that Fn1 fibrils form an organized lattice in wild type EPCs. Fn1 deposition appears abnormally uniform and sheet-like throughout the H2CKO explant (Fig. 7C-F). Fn1 dysfunction suggests a role for Hand2 in ECM assembly and epicardium homeostasis. Increased Alcian Blue staining indicates that ECM organization and/or deposition is altered in the H2CKO epicardium (Fig. 7G, H).

An external file that holds a picture, illustration, etc.
Object name is nihms280562f7.jpg

Abnormal Fn1 matrix assembly and function results in ECM related defects and impaired epicardial development. Immunohistochemistry at E12.5 for Fn1 (green) and F-actin (red) showing Fn1 expression in the epicardium and absence of patent coronary vessels (A, B). Primary epicardial cells isolated from H2CKOs show a disorganized Fn1 localization, where Fn1 appears to be distributed uniformly throughout the cells, further suggesting migratory defects in H2CKO epicardial cells (D, F). In contrast, wild type primary epicardial cells show Fn1 distributed in a series of well-organized bundles (C, E). An increase in Alcian Blue staining within the epicardium indicates increased deposition of ECM (G, H). Gene ontology analysis of microarray data from primary epicardial cells showing affected gene pathways within H2CKO isolated epicardium (I). qRT-PCR on wild type (blue bar) and H2CKO (black bar) RNA isolated from primary epicardial cells; n=4 (J). Cell proliferation (K) and cell apoptosis (L) on wild type and H2CKO embryos at E12.5; n=3. Asterisks indicate statistical significance: *p < 0.05; **p <0.01.

Gene ontology from microarray analysis to identify enriched biological processes indicates enrichment in ECM based processes, such as cell-cell signaling, assembly, connective tissue development, and motility (Fig. 7I). qRT-PCR on wild type and H2CKO EPCs detected no changes in Fn1 expression, as has been reported in mice (Fig. 7J) 42.

In addition to its role as an ECM component, Fn1 promotes intracellular signaling via interactions with cell surface integrins 43. To see if the observed H2CKO Fn1 disorganization may alter cell signaling, we assessed integrin expression by qRT-PCR (Fig. 7J). Indeed, expression of the Fn1 receptor, Itga4, is significantly upregulated within H2CKO EPCs. Itgα4 and Itgβ1 together form an Fn1 receptor pair. Itgα4 influences epicardial Fn1 polymerization. Itgα4 overexpression impairs incorporation of new Fn1 into preexisting polymer structures 44, while Itgα4 deletion causes embryonically lethal epicardial defects in mice 45, 46. As Itgα4 and Fn organization are essential for epicardiogenesis, the observed dysregulation of these ECM components suggests that Hand2 plays an important role in maintaining a normal epicardial ECM environment.

Gene ontology data indicates enrichment for cell cycle regulation genes. Loss-of-function of Hand2 has been implicated in apoptosis 20. Moreover, ECM disorganization and deposition are thought to be pro-apoptotic 47. Activated-Caspase3 and Phospho-Histone H3 immunohistochemistry at E12.5 to assess H2CKO epicardial apoptosis and proliferation (Fig. 7K, L; Online Fig. VIIA-D) reveals no change in proliferation, but a significant increase in epicardial apoptosis, These data suggest that the process of epicardial EMT is not affected in H2CKO as EMT is linked with proliferation 48, but the impaired function of the ECM in H2CKOs maybe tied both to the function, integrity, and survival of the epicardium.

Hand1 expression within the ST marks the progenitors of the PE

As Hand1-lineage cells, but not Hand1-expressing cells, are observed within the epicardium at E10.5 (See online supplement Fig. I), we sought the Hand1-expressing progenitors of these cells. The PE is derived from the anterior ST and gives rise to the epicardium 33. ISH was performed for both Hand1 and Tbx18, a marker of the PE, at E9.5 3, 34 (Fig. 1A-E). Tbx18 is expressed throughout the PE but is not detectable within the ST. In contrast, Hand1 expression is not detectable within the PE but is expressed robustly throughout the ST, thus identifying the source of the Hand1-marked epicardium. X-Gal staining of Hand1LacZ/+ embryos confirms expression within the LV and the ST (Fig. 1F). X-Gal staining of the Hand1-lineage shows Hand1-marked cells dispersed throughout the ST and within the more proximal Tbx18-expressing region, supporting established models that PE cells derive from migratory ST cells (Fig. 1G-I) 3, 34. These results establish that the Hand1-lineage cells observed throughout the cardiac fibroblasts, coronary vasculature, epicardium and PE originate from a Hand1-expressing ST cell population.

An external file that holds a picture, illustration, etc.
Object name is nihms280562f1.jpg

The Hand1 lineage gives rise to epicardial progenitors. (A) E9.5 illustration showing Hand1 expression (blue) and depicting the planes of section in B-E (B). Hand1 and Tbx18 section ISH of E9.5 embryos on adjacent sections through the ST and the PE (B-E). Hand1 expression is not detected within the PE but is expressed in the ST. X-gal staining of Hand1LacZ (F) and Hand1 R26R activation (G-I) shows Hand1-lineage cells within the ST and in the PE.. lv, left ventricle; pe, proepicardium; st, septum transversum.

Distinct Hand factor expression during heart morphogenesis

E8.5 Hand1-lineage analysis reveals that the linear heart tube is not completely derived from Hand1-expressing cells, indicating that Hand1 expression initiates after heart tube fusion. To account for a possible temporal delay of Cre expression, we performed wholemount ISH for Hand1 at E7.5 (Fig. 2). Hand1 is detected throughout the extra embryonic mesoderm, chorion, and allantoic rudiment (Fig. 2A). As the chorion is directly adjacent to the cardiac crescent (Fig. 2A,B white arrow), we performed a double label ISH for both Hand1 and the cardiac marker Mlc2a. Hand1 expression does not directly overlap with Mlc2a expression at E7.5, indicating that, consistent with the Hand1-lineage analysis, Hand1 cardiomyocyte expression initiates subsequent to linear heart tube fusion (Fig. 2B).

An external file that holds a picture, illustration, etc.
Object name is nihms280562f2.jpg

Hand factor expression is distinct. Single and double-labeled Mlc2a and Hand1 wholemount RNA ISH of E7.5 embryos show that Hand1 is not expressed within the cardiac crescent (white arrow) but is expressed within extra embryonic mesoderm, allantoic rudiment, and the chorion (A, B). E8.5 wholemounts of Hand1 and Hand2 reveal complimentary expression during cardiac development (C-F). Hand2 expression is confined to the SHF and endocardium (black arrow). cc, cardiac crescent; eem, extraembryonic mesoderm; ht, heart tube; lm, lateral mesoderm; oft, outflow tract; v, ventricle.

Wholemount ISH at E8.5 (Fig. 2C-F) shows Hand1 expression within the posterior heart tube while Hand2 expression is observed throughout the entire cardiac field. Sagittal sections of wholemount-stained embryos confirm that Hand1 expression is restricted to the early ventricular chamber and cuff myocardium. Hand2 expression is robust throughout the endocardium, SHF and OFT. Interestingly, Hand2 expression is not detected within the ventricular myocardium at E8.5 and, in wholemount view, the Hand2 endocardial expression is visible through the thin myocardial wall (Fig. 2F, red arrow). At E9.5, Hand1 and Hand2 expression overlaps within the SHF-derived myocardial cuff and the left ventriclular myocardium (not shown). Collectively, these data reaffirm that Hand factor expression is dynamically regulated and that, during early heart morphogenesis, this expression overlaps solely within the SHF-derived myocardial cuff.

Hand1Cre-mediated ablation of Hand2 within the, PE, and epicardium

To explore potential Hand factor functional overlap, we intercrossed Hand2fx23 and Hand1Cre mice to ablate Hand2 within the Hand1-lineage (H2CKO). Hand2 expression is robust throughout the PE and detectable in both E10.5 epicardial cells and isolated primary epicardial cell cultures (arrow, Fig. 3A,C; Online Fig. II). Hand2 expression was not detected within the epicardium or the PE of H2CKOs, confirming that Hand1-positive ST cells ultimately populate the PE and epicardium (Fig. 3B,D). Hand1 and Hand2 are not coexpressed within the ST or PE; rather, Hand1 expression within the ST precedes PE/epicardial upregulation of Hand2. Thus, Hand1Cre conditionally ablates Hand2 function within the PE and epicardium.

An external file that holds a picture, illustration, etc.
Object name is nihms280562f3.jpg

Inactivation of Hand2 within the Hand1 lineage RNA ISH at E9.5 (A, B) shows Hand2 expression throughout the PE and in the epicardium (red arrow) at E10.5 (C, D). Consequently, Hand2 expression is ablated in the presumptive epicardial mesothelium in H2CKOs; pe, proepicardium.

Hand1-lineage H2CKO reveals novel phenotypes and embryonic lethality

H2CKO embryos were recovered at slightly below expected Mendelian ratios through E12.5 (Fig. 4). At E14.5, roughly 50% of H2CKOs collected were dead. E12.5 H2CKOs appeared phenotypically normal (data not shown). Cardiac patterning, and both Hand1 and cardiomyocyte marker expression were normal (Online Fig. IIIA-P).

An external file that holds a picture, illustration, etc.
Object name is nihms280562f4.jpg

Conditional Hand2 deletion within the Hand1 lineage results in extensive embryonic and cardiovascular defects. Histological (A, B, E, F, I-N) and wholemount (C, D, G, H) analysis of E14.5.H2CKO embryos. Mutants display pericardial hemorrhaging (C) and anasarca (D). Compared to WT mice (G), H2CKOs have hypoplastic livers with extensive hemorrhaging (H).. H2CKOs display either PTA or DORV (A, B, E, F, I, J). All H2CKOs exhibit VSDs (K, L). Myocardial examination shows abnormal trabeculae and non-compaction (M, N). ao, dorsal aorta; cz, compact zone; pt, pulmonary trunk; pta, persistent truncus arteriousus; tr, trabeculae.

By E14.5, H2CKOs displayed pericardial hemorrhaging, anasarca, and liver hypoplasia (Fig. 4C, D, G, H). OFT septation defects, including PTA and DORV, were also observed, reflecting cNCC and/or SHF-derived myocardial defects (Fig. 4A, B, E, F, I-J). Hypertrabeculation/noncompaction and VSDs were also observed (Fig. 4K-N), although, H2CKO sarcomeric ultra-structure was indistinguishable from controls (Online Fig. IIIQ, R).

Hand2 die from a sympathetic nervous system (SNS) norepinepherine deficit 22, 23. As Hand1 and Hand2 are coexpressed in some NCCs, H2CKOs possibly die from SNS defects. Hand2 lethality can be rescued by the β-adrenergic agonist isoproterenol 22, 23. Unlike Hand2NCC embryos, isoproterenol treated Hand1Cre-generated H2CKOs fail to survive to birth (Fig. 4), indicating that SNS defects do not account for embryonic lethality.

Hand2 is required for a functional epicardium

Given the temporal relationship observed between Hand1 expression in the ST and Hand2 within the PE/epicardium, we examined H2CKO epicardial defects (Fig. 5A, B). E14.5 H2CKO display abnormal compaction, an absence of epicardium, and a lack of coronary lumens, indicating a possible epicardial phenotype (Fig. 5A,B). ISH analyses of epicardial markers at E10.5 (data not shown) and at E12.5 for Tcf21 suggest that specification and formation of the early epicardium occurs normally (Fig. 5C,D). To confirm direct disruption of H2CKO epicardial development, we generated a WT1 H2CKO4, in which Hand2 is ablated in the ST, PE and epicardium, but not the myocardium and NCC lineages (Fig. 5E-H). (See Online Fig. IV for a WT1ERT2Cre, Hand1Cre, and Hand2 cardiac lineage/expression comparison). Potentially complicating these analyses, WT1ERT2Cre marks lateral mesoderm, where Hand2 is expressed, and early allelic WT1ERT2Cre induction (E8.5) can produce ectopic Cre activity 4. HoxB6-Cre-mediated Hand2 deletion in lateral mesoderm generates viable neonates 23. Tamoxifen-mediated WT1ERT2Cre allelic induction at E9.5 resulted in no ectopic myocardial R26R reporter activity (Online Fig. V). At E13.5, Hand2fx/−, WT1ERT2Cre mutant embryos phenocopied H2CKO embryos, displaying a poorly organized epicardium and reduced WT1-marked cell lineage (Fig. 5E-H). Myocardial non-compaction was also evident, further suggesting that this phenotype is non-cell autonomous. These mutants appear to phenocopy the epicardial defects and E14.5 embryonic lethality associated with H2CKOs (Fig. 5A, B, G, H). These two independent epicardial Hand2 ablations indicate that Hand2 may play multiple roles during epicardiogenesis, the initiation of secondary epicardial EMT, and the terminal differentiation of post EMT epicardial cell populations.

An external file that holds a picture, illustration, etc.
Object name is nihms280562f5.jpg

H2CKOs display epicardial phenotypes. Histological examination at E14.5 shows a lack of epicardium and compaction abnormalities in H2CKOs (A, B). RNA ISH for Tcf21 at E12.5 shows initial establishment of the epicardium (C, D). X-gal staining and conditional deletion of Hand2 within the WT1 lineage at E13.5 photocopies H2CKO epicardial defects (E-H).

Loss of Hand2 disrupts epicardial gene expression and coronary vasculature patency

As conditional Hand2 loss-of-function leads to epicardial defects, we sought to further identify the mechanistic role for Hand2 within the epicardium. Platelet Derived Growth Factor Receptors (Pdgfr) play essential roles in cell fate and specification of the mature epicardium 35, 36. E12.5 ISH reveals decreased Pdgfrα expression within the H2CKO epicardium (Fig. 6A,B). The number of cells expressing Periostin (Postn), a marker of the cardiac fibroblast cell fate 32, is visibly decreased in the myocardium of E12.5 H2CKO mutants (Fig. 6C-D). Together these data suggest that Hand2 functions in epicardial cell fate determination during or after secondary EMT.

An external file that holds a picture, illustration, etc.
Object name is nihms280562f6.jpg

Fibroblast lineage and secondary coronary defects in H2CKOs. RNA ISH at E12.5 for Pdgfrα and Postn (A-D). A decrease in cardiac fibroblasts is observed within the compact zone of H2CKOs. Immunohistochemistry for Flk1 (green) and F-actin (red) at E12.5 (E, F). A decrease in patent coronary vasculature is observed in H2CKOs.

We performed CD31 (Online supplement; Fig. VIA, B) and Flk1 immunohistochemistry (Fig. 6E, F) at E12.5 to examine coronary vessel formation. Flk1-positive cells running through the epicardium are absent. Phalloidin counterstaining indicates that the epicardium is intact, though absent of potential Flk1-positive lumens (Fig. 6F). The Hand1-lineage does not contribute to coronary endothelial cell populations (Online Fig. VIC-F) 26, and therefore the absence of coronary vasculature within H2CKOs likely results from non-cell autonomous mechanism(s). To take advantage of induction, we administered tamoxifen at E11.5 CD31-positive coronary lumens are visible within H2CKOs when WT1ERT2Cre is activated at E11.5, although they appear less prevalent than in embryos that lack the WT1ERT2Cre allele (Online Fig. VC-D), suggesting that critical Hand2 epricardial function occurs between E9.5 and 11.5.

To further address the impaired function of H2CKO epicardium, we generated Epicardial Primary Cultures (EPCs) from wild type and H2CKOs and isolated total RNA for microarray analyses (Online Fig. IIA-D). Gene ontology from our microarray analysis indicates significant differences in developmental, morphological, and cardiovascular gene programs within EPCs (Online Fig. VIG). Quantitative RT-PCR on RNA isolated from wild type and H2CKO EPCs was performed to validate changes in gene expression observed in the microarray (Online Fig. VIH). Quantitative RT-PCR confirms expression of Hand2 within the epicardium and its ablation within the H2CKO EPCs. Importantly, the ratio of Pdgfrα to Pdgfrβ is greatly altered in H2CKO EPCs. Expression of Pdgfrα is significantly reduced, whereas, Pdgfrβ expression is significantly upregulated (Online Fig. VIH). Hand2 regulation of Pdgfrα is direct (Online supplement; Fig. VI,I). In epicardial cells, Pdgfrβ-mediated signaling promotes a smooth muscle fate, while the role of Pdgfrα signaling is currently unclear 35. The decrease in Postn-positive myofibroblasts within the compact zone suggests that Pdgfrα impacts fibroblast differentiation. Together, these data suggest that Hand2 directly impacts epicardial cell fate through a Pdgfr-dependent mechanism.

Hand2 alters Fn1 fibril assembly and organization

As H2CKO epicardial defects appear direct, we looked at the impact of a loss of Hand2 upon epicardial mesothelium integrity. Fn1 is downregulated in zebrafish hand2 mutants 37. Fn1 fibril assembly regulates the organization and stability of ECM proteins, is capable of promoting EMT and adhesion-dependent growth, and is associated with integrin-mediated cell signaling 3840. Additionally, defects in Fn1 underlie integrin-mediated valve leaflet defects in the lymphatic system 41. Immunohistochemistry shows that Fn1 is expressed in the E12.5 epicardium and is neatly organized around the developing coronaries (Fig. 7A). H2CKOs retain epicardial Fn1; however, it appears disorganized (Fig. 7B). To look at Fn1 organization more closely, we compared EPCs from wild type and H2CKOs (Fig. 7C-F). Immunohistochemistry reveals that Fn1 fibrils form an organized lattice in wild type EPCs. Fn1 deposition appears abnormally uniform and sheet-like throughout the H2CKO explant (Fig. 7C-F). Fn1 dysfunction suggests a role for Hand2 in ECM assembly and epicardium homeostasis. Increased Alcian Blue staining indicates that ECM organization and/or deposition is altered in the H2CKO epicardium (Fig. 7G, H).

An external file that holds a picture, illustration, etc.
Object name is nihms280562f7.jpg

Abnormal Fn1 matrix assembly and function results in ECM related defects and impaired epicardial development. Immunohistochemistry at E12.5 for Fn1 (green) and F-actin (red) showing Fn1 expression in the epicardium and absence of patent coronary vessels (A, B). Primary epicardial cells isolated from H2CKOs show a disorganized Fn1 localization, where Fn1 appears to be distributed uniformly throughout the cells, further suggesting migratory defects in H2CKO epicardial cells (D, F). In contrast, wild type primary epicardial cells show Fn1 distributed in a series of well-organized bundles (C, E). An increase in Alcian Blue staining within the epicardium indicates increased deposition of ECM (G, H). Gene ontology analysis of microarray data from primary epicardial cells showing affected gene pathways within H2CKO isolated epicardium (I). qRT-PCR on wild type (blue bar) and H2CKO (black bar) RNA isolated from primary epicardial cells; n=4 (J). Cell proliferation (K) and cell apoptosis (L) on wild type and H2CKO embryos at E12.5; n=3. Asterisks indicate statistical significance: *p < 0.05; **p <0.01.

Gene ontology from microarray analysis to identify enriched biological processes indicates enrichment in ECM based processes, such as cell-cell signaling, assembly, connective tissue development, and motility (Fig. 7I). qRT-PCR on wild type and H2CKO EPCs detected no changes in Fn1 expression, as has been reported in mice (Fig. 7J) 42.

In addition to its role as an ECM component, Fn1 promotes intracellular signaling via interactions with cell surface integrins 43. To see if the observed H2CKO Fn1 disorganization may alter cell signaling, we assessed integrin expression by qRT-PCR (Fig. 7J). Indeed, expression of the Fn1 receptor, Itga4, is significantly upregulated within H2CKO EPCs. Itgα4 and Itgβ1 together form an Fn1 receptor pair. Itgα4 influences epicardial Fn1 polymerization. Itgα4 overexpression impairs incorporation of new Fn1 into preexisting polymer structures 44, while Itgα4 deletion causes embryonically lethal epicardial defects in mice 45, 46. As Itgα4 and Fn organization are essential for epicardiogenesis, the observed dysregulation of these ECM components suggests that Hand2 plays an important role in maintaining a normal epicardial ECM environment.

Gene ontology data indicates enrichment for cell cycle regulation genes. Loss-of-function of Hand2 has been implicated in apoptosis 20. Moreover, ECM disorganization and deposition are thought to be pro-apoptotic 47. Activated-Caspase3 and Phospho-Histone H3 immunohistochemistry at E12.5 to assess H2CKO epicardial apoptosis and proliferation (Fig. 7K, L; Online Fig. VIIA-D) reveals no change in proliferation, but a significant increase in epicardial apoptosis, These data suggest that the process of epicardial EMT is not affected in H2CKO as EMT is linked with proliferation 48, but the impaired function of the ECM in H2CKOs maybe tied both to the function, integrity, and survival of the epicardium.

Discussion

Collectively, these data reveal a novel and essential function for Hand factors in epicardiogenesis (Online Fig. VIIIA-B). Transient ST expression of Hand1 marks cells that successively populate the PE, epicardium and their derivatives. Hand1 ST expression temporally precedes Hand2 PE expression. Hand2 ablation causes significant epicardial defects including directly impaired Pdgfrα regulation, abnormal differentiation and Fn1 organization, and Itga4 upregulation. Also evident in H2CKOs is increased epicardial apoptosis, potentially reflecting altered Fn-mediated signaling and/or cell migration. Fn1 and Itgα4, have been associated with similar mesodermal, epicardial, and cardiovascular decline 45, 46, 49. Epicardiogenesis initiates normally in Itga4 mice; however, the epicardium subsequently detaches from the myocardium and degrades 45, 46. Itga4 overexpression alters Fn/integrins interactions and disrupts Fn deposition, demonstrating that integrin levels significantly impact Fn organization and function 44. Our observations suggest that Hand2 is necessary to maintain the balance between this receptor/ligand pair. Ultimately, Hand2 ablation leads to compromised epicardial function and a failure to form a patent coronary vasculature, both phenotypes observed in other mouse models presenting coronary malformations 50.

Fn1 is a multifunctional ECM protein that establishes cytoskeletal organization, motility, and cell signaling pathways required for proliferation and growth. As epicardial cells must migrate, alter morphology, and differentiate into functional cell types, the epicardial phenotypes observed in H2CKOs mechanistically fit a model of altered Fn1 function. Defects in Fn1 deposition are associated with increased fibrosis and apoptosis 51, all characteristic of the H2CKO epicardium. Fn1 mRNA expression is not altered within H2CKO epicardium, demonstrating that Hand2 regulation of Fn1 is indirect. Zebrafish hand regulates fn1 deposition and influences ECM deposition during lateral mesoderm remodeling 37. Consistent with our data, hand regulates fn1 indirectly through ECM organization, rather than by directly regulating gene expression 52. These observations suggest a critical, evolutionarily conserved role for Hand2 during maturation of epicardial-derivatives.

Previous studies suggest that Hand factors have partially overlapping expression domains and are functionally redundant during cardiac patterning 25. Detailed Hand1 and Hand2 expression profiles show less spatiotemporal myocardial overlap than previously indicated. Hand2 deletion within the Hand1 expression domain does not cause significant myocardial patterning or differentiation defects (Online Fig. III), suggesting that the H2CKO myocardial phenotypes are non-cell autonomous. Hand2 deletion using cardiac-specific Cre-drivers causes SHF defects and early embryonic lethality 23. To completely rule out cell autonomous Hand2 function within PHF-derived cardiomyocytes, PHF-specific Hand2 deletion would be required.

WT1- and Hand1Cre-mediated Hand2 deletions produce epicardial phenocopies, indicating a novel Hand2 function during epicardial development. While the defects in the epicardium appear direct, WT1 is expressed in lateral mesoderm derivatives in addition to the epicardium 50. Lineage analysis following E9.5 WT1ERT2Cre induction detects no cardiomyocyte expression (Online Fig. V). Hand2 deletion using a lateral mesoderm and extraembryonic tissue-specific HoxB6Cre23, which, as Hand2 is not detected within extraembryonic tissues 18, effectively generates a lateral mesoderm-specific Hand2 deletion, results in viable embryos 23. Hand2 function in the lateral mesoderm is therefore not critical to embryonic survival, and we conclude that PE/epicardium-specific Hand2 ablation disrupts coronary vasculature maturation and thus contributes to H2CKO lethality.

The Hand1-lineage does not contribute to coronary vascular endothelium 26. Current understanding identifies the origin of the coronary vessel endothelium as the sinus venosus 9, and presumes that the functional epicardium interacts with the coronary endothelium to both establish and maintain coronary vessel patency. H2CKOs display epicardial lineage-specific defects that impact both fibroblast and smooth muscle cell fates. No Hand1-lineage independent cells are detectable in the epicardium (Online Fig. I) 26. Hand2 ISH similarly marks the entire epicardium, suggesting that Hand2 is not enriched within a subset of epicardial cells. Pdgfrs govern specific lineage subsets during epicardial development 35. Pdgfrα is downregulated and, conversely, Pdgfrβ is upregulated within the H2CKO epicardium. Luciferase transactivation data suggests direct Pdgfrα regulation (Online Fig. VII,I). Although Pdgfrα’s epicardial function is unknown, it is essential in other EMT-derived cell populations 36. This altered ratio could reflect a Pdgfrβ compensation for decreased Pdgfrα expression; however, Pdgfrβ drives epicardially-derived cell differentiation to smooth muscle cell 35. As the epicardium gives rise to both fibroblast and coronary smooth muscle, and Postn is expressed in cardiac fibroblasts invading the myocardium, (Fig. 6), Pdgfrα may govern the fibroblast cell fate 53. Hand2 appears to modulate the Pdgfr ratios that govern these divergent cell programs. Recently, it has been shown that Hand2 overexpression causes a significant increase in fibroblast marker expression, further implicating Hand2 in the cardiac fibroblast cell fate program 54.

Hand1Cre/+;Hand2fx/− mutants also display cardiac OFT defects. Wnt1-Cre-mediated NCC-specific Hand2 deletion results in aortic arch defects, DORV, and associated VSDs 22, 23. As Hand1Cre– mediated cNCC Hand2 ablation is temporally later and spatially more restricted then Hand2Δ but entails heterozygosity of Hand1, the high penetrance of a more severe NCC-dependent PTA phenotype suggests that genetic and, possibly, functional interactions between Hand1 and Hand2 are critical for OFT septation. Established genetic interactions between Hand1 and Hand2 support this model 24. It is also consistent with established dimer regulation mechanisms governing the biological output of Twist-family bHLH factors. Indeed, dysregulation of Twist1 dimerization causes the human disease Saethre Chotzen Syndrome and directly reflects molecular antagonism between Twist1 and Hand2 55. Thus, Hand1 and Hand2 dimer choice may prove crucial to OFT morphogenesis. Experiments to explore this possibility mechanistically are currently underway.

Our data defines unique Hand1 and Hand2 expression domains within the developing murine heart. Hand1 is largely restricted the left ventricle. SHF expression is restricted to the myocardial cuff 26, and is the sole domain of co-expression with Hand2. At the onset of cardiac looping, Hand1 expression/lineage is detectable within the forming left ventricle. Although we observe a thin compact zone and hypertrabeculation within H2CKOs, the cell autonomy of these defects cannot be deduced. Cardiac-specific Hand2 ablation causes SHF defects and early embryonic death 23. Although Hand1Cre mice allow insight into a possible role for Hand2 in the PFH myocardium, a PHF-specific Cre will be required to address this directly. In summary, these studies demonstrate that, in addition to its established functions within the cNCC and myocardium, Hand2, modulates cell signaling mechanisms that dictate epicardial cell fates and ECM organization, thus playing a novel and critical role in the function and differentiation of the epicardium and, consequently, proper cardiac function.

Supplementary Material

1

1

Click here to view.(9.9M, pdf)

Acknowledgments

We thank Danny Carney for technical assistance. We thank Henry Sucov and William Pu for providing Wnt1-Cre and WT1ERT2Cre mice, respectively. We thank the Herman B Wells Center Cardiac Developmental Biology Group for helpful discussions. Infrastructural support at the Herman B Wells Center is partially supported by the Riley Children’s Foundation and Division of Pediatric Cardiology.

Funding: This work was supported by an AHA predoctoral fellowship AHA 0815426G (RMB) and NIH 2 RO1 HI061677 (ABF) and NIH 1P01HL085098 (ABF SJC).

Riley Heart Research Center, Wells Center for Pediatric Research, Division of Pediatric Cardiology, Departments of Anatomy and Medical and Molecular Genetics, Indiana Medical School, 1044 W. Walnut St., Indianapolis, IN 46202-5225, USA
Department of Cell and Molecular Biology, Tulane University, 2000 Percival Stern Hall, New Orleans, LA 70118, USA
Department of Pathology and Cell Biology, Columbia University, 630 W 168 Street, New York, NY 10032, USA
To whom correspondence should be addressed ude.iupui@illurift (317) 278-5814

Abstract

Rationale

The bHLH transcription factors Hand1 and Hand2 are essential for embryonic development. Given their requirement for cardiogenesis, it is imperative to determine their impact on cardiovascular function.

Objective

Deduce the role of Hand2 within the epicardium.

Method &amp; Results

We engineered a Hand1 allele expressing Cre recombinase. Cardiac Hand1 expression is largely limited to cells of the primary heart field, overlapping little with Hand2 expression. Hand1 is expressed within the septum transversum (ST) and the Hand1-lineage marks the proepicardial organ and epicardium. To examine Hand factor functional overlap, we conditionally deleted Hand2 from Hand1-expressing cells. Hand2 mutants display defective epicardialization and fail to form coronary arteries, coincident with altered ECM deposition and Pdgfr expression.

Conclusion

These data demonstrate a hierarchal relationship whereby transient Hand1 ST expression defines epicardial precursors that are subsequently dependent upon Hand2 function.

Keywords: Hand1, Hand2, bHLH, epicardium
Abstract

Footnotes

Disclosures

The authors have nothing to disclose.

Footnotes

References

  • 1. Buckingham M, Meilhac S, Zaffran SBuilding the mammalian heart from two sources of myocardial cells. Nat Rev Genet. 2005;6:826–835.[PubMed][Google Scholar]
  • 2. Jiang X, Rowitch DH, Soriano P, McMahon AP, Sucov HMFate of the mammalian cardiac neural crest. Development. 2000;127:1607–1616.[PubMed][Google Scholar]
  • 3. Cai CL, Martin JC, Sun Y, Cui L, Wang L, Ouyang K, Yang L, Bu L, Liang X, Zhang X, Stallcup WB, Denton CP, McCulloch A, Chen J, Evans SMA myocardial lineage derives from Tbx18 epicardial cells. Nature. 2008;454:104–108.[Google Scholar]
  • 4. Zhou B, Ma Q, Rajagopal S, Wu SM, Domian I, Rivera-Feliciano J, Jiang D, von Gise A, Ikeda S, Chien KR, Pu WTEpicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature. 2008;454:109–113.[Google Scholar]
  • 5. Wada AM, Willet SG, Bader DCoronary vessel development: a unique form of vasculogenesis. Arterioscler Thromb Vasc Biol. 2003;23:2138–2145.[PubMed][Google Scholar]
  • 6. Black BLTranscriptional pathways in second heart field development. Semin Cell Dev Biol. 2007;18:67–76.[Google Scholar]
  • 7. Snarr BS, Kern CB, Wessels AOrigin and fate of cardiac mesenchyme. Dev Dyn. 2008;237:2804–2819.[PubMed][Google Scholar]
  • 8. Olivey HE, Svensson ECEpicardial-myocardial signaling directing coronary vasculogenesis. Circ Res. 2010;106:818–832.[Google Scholar]
  • 9. Red-Horse K, Ueno H, Weissman IL, Krasnow MACoronary arteries form by developmental reprogramming of venous cells. Nature. 2010;464:549–553.[Google Scholar]
  • 10. Snider P, Standley KN, Wang J, Azhar M, Doetschman T, Conway SJOrigin of cardiac fibroblasts and the role of periostin. Circ Res. 2009;105:934–947.[Google Scholar]
  • 11. Souders CA, Bowers SL, Baudino TACardiac fibroblast: the renaissance cell. Circ Res. 2009;105:1164–1176.[Google Scholar]
  • 12. Zhou B, Pu WTMore than a cover: epicardium as a novel source of cardiac progenitor cells. Regen Med. 2008;3:633–635.[PubMed][Google Scholar]
  • 13. Barnes RM, Firulli ABA twist of insight - the role of Twist-family bHLH factors in development. Int J Dev Biol. 2009;53:909–24.[Google Scholar]
  • 14. Firulli ABA HANDful of questions: the molecular biology of the heart and neural crest derivatives (HAND)-subclass of basic helix-loop-helix transcription factors. Gene. 2003;312:27–40.[PubMed][Google Scholar]
  • 15. Cserjesi P, Brown D, Lyons GE, Olson ENExpression of the Novel Basic Helix-Loop-Helix Gene eHAND in Neural Crest Derivatives and Extraembryonic Membranes during Mouse Development. Developmental biology. 1995;170:664–678.[PubMed][Google Scholar]
  • 16. Firulli AB, McFadden DG, Lin Q, Srivastava D, Olson ENHeart and extra-embryonic mesodermal defects in mouse embryos lacking the bHLH transcription factor Hand1. Nat Genet. 1998;18:266–270.[PubMed][Google Scholar]
  • 17. Srivastava D, Cserjesi P, Olson ENA subclass of bHLH proteins required for cardiac morphogenesis. Science. 1995;270:1995–1999.[PubMed][Google Scholar]
  • 18. Srivastava D, Thomas T, Lin Q, Kirby ML, Brown D, Olson ENRegulation of cardiac mesodermal and neural crest development by the bHLH transcription factor, dHAND. Nat Genet. 1997;16:154–160.[PubMed][Google Scholar]
  • 19. Riley P, Anson-Cartwright L, Cross JCThe Hand1 bHLH transcription factor is essential for placentation and cardiac morphogenesis. Nat Genet. 1998;18:271–275.[PubMed][Google Scholar]
  • 20. Thomas T, Yamagishi H, Overbeek P, Olson E, Srivastava DThe bHLH factors, dHAND and eHAND, specifiy pulmonary and systemic cardiac ventricles independent of left-right sidedness. Developmental Biology. 1998;196:228–236.[PubMed][Google Scholar]
  • 21. Morikawa Y, Cserjesi PExtra-embryonic vasculature development is regulated by the transcription factor HAND1. Development. 2004;131:2195–2204.[PubMed][Google Scholar]
  • 22. Holler KL, Hendershot TJ, Troy SE, Vincentz JW, Firulli AB, Howard MJTargeted deletion of Hand2 in cardiac neural crest-derived cells influences cardiac gene expression and outflow tract development. Dev Biol. 2010;341:291–304.[Google Scholar]
  • 23. Morikawa Y, Cserjesi PCardiac neural crest expression of Hand2 regulates outflow and second heart field development. Circ Res. 2008;103:1422–1429.[PubMed][Google Scholar]
  • 24. Barbosa AC, Funato N, Chapman S, McKee MD, Richardson JA, Olson EN, Yanagisawa HHand transcription factors cooperatively regulate development of the distal midline mesenchyme. Dev Biol. 2007;310:154–168.[Google Scholar]
  • 25. McFadden DG, Barbosa AC, Richardson JA, Schneider MD, Srivastava D, Olson ENThe Hand1 and Hand2 transcription factors regulate expansion of the embryonic cardiac ventricles in a gene dosage-dependent manner. Development. 2005;132:189–201.[PubMed][Google Scholar]
  • 26. Barnes RM, Firulli B, Conway SJ, Vincentz JW, Firulli ABAnalysis of the Hand1 Cell Lineage Reveals Novel Contributions to Cardiovascular,Neural Crest, Extra-Embryonic, and Lateral Mesoderm Derivatives. Dev Dyn. 2010;239:3086–3097.[Google Scholar]
  • 27. Soriano PGeneralized lacZ expression with the ROSA26 Cre reporter strain. Nat Genet. 1999;21:70–71.[PubMed][Google Scholar]
  • 28. Vincentz JW, Barnes RM, Rodgers R, Firulli BA, Conway SJ, Firulli ABAn absence of Twist1 results in aberrant cardiac neural crest morphogenesis. Dev Biol. 2008;320:131–139.[Google Scholar]
  • 29. Nagy A Manipulating the mouse embryo : a laboratory manual. 3. Cold Spring Harbor, N.Y: Cold Spring Harbor Laboratory Press; 2003. [PubMed][Google Scholar]
  • 30. Ishii Y, Langberg JD, Hurtado R, Lee S, Mikawa TInduction of proepicardial marker gene expression by the liver bud. Development. 2007;134:3627–3637.[PubMed][Google Scholar]
  • 31. Rhee DY, Zhao XQ, Francis RJ, Huang GY, Mably JD, Lo CWConnexin 43 regulates epicardial cell polarity and migration in coronary vascular development. Development. 2009;136:3185–3193.[Google Scholar]
  • 32. Snider P, Hinton RB, Moreno-Rodriguez RA, Wang J, Rogers R, Lindsley A, Li F, Ingram DA, Menick D, Field L, Firulli AB, Molkentin JD, Markwald R, Conway SJPeriostin is required for maturation and extracellular matrix stabilization of noncardiomyocyte lineages of the heart. Circ Res. 2008;102:752–760.[Google Scholar]
  • 33. Watt AJ, Battle MA, Li J, Duncan SAGATA4 is essential for formation of the proepicardium and regulates cardiogenesis. Proc Natl Acad Sci U S A. 2004;101:12573–12578.[Google Scholar]
  • 34. Christoffels VM, Grieskamp T, Norden J, Mommersteeg MT, Rudat C, Kispert ATbx18 and the fate of epicardial progenitors. Nature. 2009;458:E8–9. E9–10.[PubMed][Google Scholar]
  • 35. Mellgren AM, Smith CL, Olsen GS, Eskiocak B, Zhou B, Kazi MN, Ruiz FR, Pu WT, Tallquist MDPlatelet-derived growth factor receptor beta signaling is required for efficient epicardial cell migration and development of two distinct coronary vascular smooth muscle cell populations. Circ Res. 2008;103:1393–1401.[Google Scholar]
  • 36. Tallquist MD, Soriano PCell autonomous requirement for PDGFRalpha in populations of cranial and cardiac neural crest cells. Development. 2003;130:507–518.[PubMed][Google Scholar]
  • 37. Trinh LA, Yelon D, Stainier DYHand2 regulates epithelial formation during myocardial diferentiation. Curr Biol. 2005;15:441–446.[PubMed][Google Scholar]
  • 38. Sottile J, Hocking DC, Swiatek PJFibronectin matrix assembly enhances adhesion-dependent cell growth. J Cell Sci. 1998;111:2933–2943.[PubMed][Google Scholar]
  • 39. Leiss M, Beckmann K, Giros A, Costell M, Fassler RThe role of integrin binding sites in fibronectin matrix assembly in vivo. Curr Opin Cell Biol. 2008;20:502–507.[PubMed][Google Scholar]
  • 40. Liao YF, Gotwals PJ, Koteliansky VE, Sheppard D, Van De Water LThe EIIIA segment of fibronectin is a ligand for integrins alpha 9beta 1 and alpha 4beta 1 providing a novel mechanism for regulating cell adhesion by alternative splicing. J Biol Chem. 2002;277:14467–14474.[PubMed][Google Scholar]
  • 41. Bazigou E, Xie S, Chen C, Weston A, Miura N, Sorokin L, Adams R, Muro AF, Sheppard D, Makinen TIntegrin-alpha9 is required for fibronectin matrix assembly during lymphatic valve morphogenesis. Dev Cell. 2009;17:175–186.[Google Scholar]
  • 42. Yin C, Kikuchi K, Hochgreb T, Poss KD, Stainier DYHand2 regulates extracellular matrix remodeling essential for gut-looping morphogenesis in zebrafish. Dev Cell. 2010;18:973–984.[Google Scholar]
  • 43. Ieda M, Tsuchihashi T, Ivey KN, Ross RS, Hong TT, Shaw RM, Srivastava DCardiac fibroblasts regulate myocardial proliferation through beta1 integrin signaling. Dev Cell. 2009;16:233–244.[Google Scholar]
  • 44. Pae SH, Dokic D, Dettman RWCommunication between integrin receptors facilitates epicardial cell adhesion and matrix organization. Developmental Dynamics. 2008;237:962–978.[PubMed][Google Scholar]
  • 45. Yang JT, Rayburn H, Hynes ROCell adhesion events mediated by alpha 4 integrins are essential in placental and cardiac development. Development. 1995;121:549–560.[PubMed][Google Scholar]
  • 46. Sengbusch JK, He W, Pinco KA, Yang JTDual functions of [alpha]4[beta]1 integrin in epicardial development: initial migration and long-term attachment. Journal of Cell Biology. 2002;157:873–882.[Google Scholar]
  • 47. Frisch SM, Francis HDisruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol. 1994;124:619–626.[Google Scholar]
  • 48. Wu M, Smith CL, Hall JA, Lee I, Luby-Phelps K, Tallquist MDEpicardial spindle orientation controls cell entry into the myocardium. Dev Cell. 2010;19:114–125.[Google Scholar]
  • 49. George EL, Baldwin HS, Hynes ROFibronectins are essential for heart and blood vessel morphogenesis but are dispensable for initial specification of precursor cells. Blood. 1997;90:3073–3081.[PubMed][Google Scholar]
  • 50. Moore AW, McInnes L, Kreidberg J, Hastie ND, Schedl AYAC complementation shows a requirement for Wt1 in the development of epicardium, adrenal gland and throughout nephrogenesis. Development. 1999;126:1845–1857.[PubMed][Google Scholar]
  • 51. Tomita M, Okuyama T, Katsuyama H, Miura Y, Nishimura Y, Hidaka K, Otsuki T, Ishikawa TMouse model of paraquat-poisoned lungs and its gene expression profile. Toxicology. 2007;231:200–209.[PubMed][Google Scholar]
  • 52. Garavito-Aguilar ZV, Riley HE, Yelon DHand2 ensures an appropriate environment for cardiac fusion by limiting Fibronectin function. Development. 2010;137:3215–3220.[Google Scholar]
  • 53. Richarte AM, Mead HB, Tallquist MDCooperation between the PDGF receptors in cardiac neural crest cell migration. Dev Biol. 2007;306:785–796.[Google Scholar]
  • 54. Ieda M, ji-dong F, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG, Srivastava DDirect Reprogramming of Fibroblasts into Functional Cardiomyocytes by Defined Factors. Cell. 2010;142:375–386.[Google Scholar]
  • 55. Firulli BA, Krawchuk D, Centonze VE, Virshup DE, Conway SJ, Cserjesi P, Laufer E, Firulli ABAltered Twist1 and Hand2 dimerization is associated with Saethre-Chotzen syndrome and limb abnormalities. Nat Genet. 2005;37:373–381.[Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.