Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Nov 19;5(11):101795.
doi: 10.1016/j.xcrm.2024.101795. Epub 2024 Oct 24.

A humanized monoclonal antibody targeting an ectonucleotidase rescues cardiac metabolism and heart function after myocardial infarction

Affiliations

A humanized monoclonal antibody targeting an ectonucleotidase rescues cardiac metabolism and heart function after myocardial infarction

Shen Li et al. Cell Rep Med. .

Abstract

Myocardial infarction (MI) results in aberrant cardiac metabolism, but no therapeutics have been designed to target cardiac metabolism to enhance heart repair. We engineer a humanized monoclonal antibody against the ectonucleotidase ENPP1 (hENPP1mAb) that targets metabolic crosstalk in the infarcted heart. In mice expressing human ENPP1, systemic administration of hENPP1mAb metabolically reprograms myocytes and non-myocytes and leads to a significant rescue of post-MI heart dysfunction. Using metabolomics, single-nuclear transcriptomics, and cellular respiration studies, we show that the administration of the hENPP1mAb induces organ-wide metabolic and transcriptional reprogramming of the heart that enhances myocyte cellular respiration and decreases cell death and fibrosis in the infarcted heart. Biodistribution and safety studies showed specific organ-wide distribution with the antibody being well tolerated. In humanized animals, with drug clearance kinetics similar to humans, we demonstrate that a single "shot" of the hENPP1mAb after MI is sufficient to rescue cardiac dysfunction.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests The intellectual property associated with hENPP1mAb is held by the Regents, University of California.

Figures

None
Graphical abstract
Figure 1
Figure 1
Avidity, potency, homology modeling, and off-target binding assays for hENPP1mAb (A) Dissociation constant of hENPP1mAb calculated with flow cytometry to determine concentration-dependent binding of hENPP1mAb to HEK cells overexpressing human ENPP1. Calculated KD value (measure of avidity) shown (n = 3). (B) Potency curve of hENPP1mAb in inhibiting human ENPP1 catalytic activity in a concentration-dependent manner. Calculated IC50 shown (n = 3). (C and D) Homology modeling and protein-protein docking interactions show the (C) docked model of hENPP1mAb and human ENPP1. The antibody chains (CDR of light and heavy chains) occlude the enzyme active site (dark gray surface) (human ENPP1 antigen in blue, Fab segment of hENPP1mAb in green, CDRs color coded as shown). (D) Magnified image demonstrating several CDRs of hENPP1mAb inserting into human ENPP1 catalytic domain. (E) Flow cytometry to determine binding of hENPP1mAb to other members of human ENPP family and to other phosphatases (n = 5/group). (F–H) Retrogenix membrane array screening by immunoblotting to determine binding of hENPP1mAb to 6,101 human plasma membrane proteins and 396 human heterodimers expressed on HEK cells. (F) hENPP1mAb shows a significant specific interaction with human ENPP1 isoforms (red) (plasma membrane isoforms upper and middle, and tethered secreted form, lower). Note hENPP1mAb also shows binding to IGHG that may serve as IgG receptors. (G) Rituximab is used as a positive control to determine any non-specific binding of a mAb and demonstrates no binding to human ENPP1 and binding to IGHG proteins. (H) PBS is used as a negative control for the entire assay and demonstrates signal against the IGHG proteins as well. Immunoblotting demonstrates representative images of n = 3. Data are represented as mean ± SEM.
Figure 2
Figure 2
Species reactivity of hENPP1mAb and development of humanized ENPP1 mice (A) Flow cytometry to determine binding of hENPP1mAb against mouse, rat, pig, monkey, and human ENPP1 overexpressed in HEK cell line. HEK cells expressing eGFP used as a negative control (n = 3 for eGFP and monkey, n = 5 in mouse, rat, pig, and human). (B) Schematic representation of generation of the humanized ENPP1 mouse. Using CRISPR-Cas9, human ENPP1CDS with a PolyA signal at the 3′ end is inserted to replace the 1st exon of murine ENPP1 gene. (C) Agarose gel electrophoresis of RT-PCR products of heart tissue from humanized ENPP1 mice or wild-type C57BL/6J mice (n = 3 animals/group). Discriminatory PCR primers are used to distinguish murine and human ENPP1 expression. (D) qPCR demonstrating ENPP1 gene expression in the injured region of the heart compared with uninjured region at 7 days after MI (n = 3 animals/group). (E) qPCR on infarcted heart of humanized ENPP1 mouse at day 7 post MI demonstrating the absence of murine ENPP1 and expression of human ENPP1 in the infarcted region (n = 3 animals/group). (F) Immunostaining for ENPP1 (green, arrowheads) and cardiac troponin I (red) in the injured regions at day 7 after MI. Magnified images demonstrate cells in the infarcted region of the inset expressing human ENPP1 (arrowheads). Note that ENPP1 expression is present in troponin-negative regions. Data are represented as mean ± SEM. ∗p < 0.05, Statistical significance was determined using Student’s t test, 2 tailed.
Figure 3
Figure 3
hENPP1mAb attenuates post-infarct cardiac dysfunction in humanized ENPP1 animals (A) Strategy for hENPP1mAb administration in humanized ENPP1 animals subjected to MI. (B) Western blotting for ENPP1 in wild-type mice hearts at 3, 7, and 14 days following MI. (C) Quantitative densitometry of ENPP1 level (n = 3). (D) Extracellular ATP hydrolytic activity in injured and uninjured hearts of animals treated with IgG or hENPP1mAb (n = 4 animals/group). (E) B (top) and M-mode (below) echocardiogram demonstrating superior contractile function in hENPP1mAb-treated animals. Diastolic (green line) and systolic internal dimensions (yellow line) in hearts of hENPP1mAb/IgG-treated animals. (F) Ejection fraction, fractional shortening, and left ventricular (LV) chamber size in systole (LVIDs) and diastole (LVIDd) in IgG or hENPP1mAb-treated animals at 1, 2, and 4 weeks following MI (n = 21/IgG and n = 19/hENPP1mAb). (G) Pie chart illustrating the fraction of animals with mild, moderate, and severe reduction in EF at 4 weeks after injury following IgG or hENPP1mAb administration. (H) 4D gated cardiac CT showing transverse and coronal views of the heart of IgG or hENPP1mAb-injected animals at day 14 post MI (arrowheads point to the thin wall post infarct scar that is decreased in hENPP1mAb-injected groups). (I) Ejection fraction measurement by gated cardiac CT (n = 7 animals/group). (J) Myocardial strain analysis of cardiac segments in longitudinal axis at day 7 post MI in IgG versus hENPP1mAb-treated animals. Heatmap demonstrating wall strain generated with deeper color corresponding to greater contractile force. (K) Myocardial deformation measurements to demonstrate strain forces generated at various cardiac segments between IgG and hENPP1mAb-treated animals. GLS, global longitudinal strain); Post, posterior base; Post. Mid; Post. Apex; Ant., Anterior apex; Ant. Mid and Ant. base. (n = 9 animals/group). Data are expressed as mean ± SEM. ∗∗p < 0.01, ∗p < 0.05, ns: not significant. Statistical significance was determined using ordinary one-way ANOVA with Tukey’s multiple comparison test (C and D), unpaired multiple t test (F), or Student’s t test, 2 tailed (I and K).
Figure 4
Figure 4
Humanized ENPP1 animals treated with hENPP1mAb after MI exhibit histologic evidence of superior cardiac repair (A) Masson trichrome staining to demonstrate scar size as a fraction of LV surface area measured 4 weeks after injury at the apex and mid ventricle in IgG or hENPP1mAb-injected humanized ENPP1 animals. (B) Quantitation of scar surface area (n = 15/IgG and 22/hENPP1mAb) and (C) pie chart illustrating the fraction of animals with mild, moderate, and severe fibrosis following IgG or hENPP1mAb administration. (D) Hematoxylin/eosin staining to demonstrate the thickness of infarcted wall at 4 weeks after MI in IgG or hENPP1mAb-injected animals with quantification of wall thickness (n = 11/IgG and n = 12/hENPP1mAb). (E) Heart weight (HW), body weight (BW), and HW/BW ratio in IgG versus hENPP1mAb-treated animals (n = 21/IgG and n = 19/hENPP1mAb). (F) Immunostaining for cardiac troponin and wheat germ agglutinin to determine myocyte surface area and quantification (surrogate for cardiac muscle hypertrophy) 4 weeks after MI in IgG or hENPP1mAb-injected animals (n = 10/IgG and n = 9/hENPP1mAb). (G) Staining for endothelial cells (CD31) to determine capillary formation (arrowheads) 4 weeks after MI in IgG or hENPP1mAb-treated animals and quantification of capillary formation (n = 13/IgG and n = 16/hENPP1mAb). Data are represented as mean ± SEM. ∗∗p < 0.01, ∗p < 0.05, ns: not significant. Statistical significance was determined using Student’s t test, 2 tailed.
Figure 5
Figure 5
Single-nuclei RNA sequencing of hearts of humanized ENPP1 animals treated with IgG or hENPP1mAb and harvested at 7 days following MI (A) Uniform manifold approximation and projection (UMAP) demonstrating different phenotypes of cell clusters in the infarcted heart and (B) distribution of cells from IgG and hENPP1mAb-treated animals across these clusters (n = 3 animals/group). (C) Fraction of different cell populations in IgG versus hENPP1mAb-injected animals. (D) Gene ontology analysis of main pathways differentially downregulated in cardiac fibroblasts in hENPP1mAb-treated animals versus IgG control animals. (E) UMAP demonstrating subclustering of fibroblast population across IgG and hENPP1mAb groups and (F) distribution of fibroblasts of IgG versus hENPP1mAb groups across these fibroblast subclusters. (G) Fraction of fibroblasts in IgG or hENPP1mAb-treated groups contributing to the fibroblast subclusters with cluster 0 contributed by IgG-injected group and cluster 1 by the hENPP1mAb group. (p < 0.05 in cluster 0, p < 0.01 in cluster 1, and no significance in cluster 2). (H) Expression of ECM and myofibroblast genes (Col1a1, Postn, and Acta2) across these fibroblast subclusters with abundant expression of ECM genes and myofibroblast marker Postn in subcluster 0 compared to subcluster 1. (I) Dot plot demonstrating distribution of abundantly expressed genes representing the fibroblast subclusters (Note: myofibroblast and ECM genes are abundant in subcluster 0 compared to subcluster 1). (J) Dot blot representing expression of myofibroblast and ECM genes in the entire cardiac fibroblast population of IgG versus hENPP1mAb animals. Data are expressed as mean ± SEM. ∗∗p < 0.01. Statistical significance was determined using Student’s t test, 2 tailed.
Figure 6
Figure 6
hENPP1mAb administration in humanized mice leads to the rescue of metabolic pathways and augmented cellular respiration in the infarcted heart (A) Metabolomic analysis of hearts of animals injected with IgG or hENPP1mAb at day 7 post MI demonstrating metabolites that are significantly upregulated (p < 0.05) in hearts of hENPP1mAb-injected animals (arrows point to pyrimidines or metabolites in pentose phosphate pathway, demonstrating rescue of pyrimidines in hENPP1mAb-injected animals compared to IgG-injected animals, n = 5/IgG and 4/hENPP1mAb). (B) KEGG analysis of metabolic pathways that are significantly upregulated in hearts of hENPP1mAb injected animals demonstrating that pyrimidine biosynthetic pathway is the most significant metabolic pathway. (C) Quantification of NAD, NADH, and nicotinamide levels in hearts of animals injected with hENPP1mAb versus IgG as early as 3 days post infarction (n = 6/group). (D) Seahorse cellular respiration on heart homogenates (day 3 post MI) demonstrating oxygen consumption rate (OCR) at mitochondrial electron transport complexes I and II normalized to MitoTracker Deep Red (MTDR) fluorescence (n = 10/group). Data are expressed as mean ± SEM. ∗∗p < 0.01. Statistics was determined using Student’s t test, 2 tailed.
Figure 7
Figure 7
A single dose of hENPP1mAb administered in humanized ENPP1/Tg32 animals after MI is sufficient to significantly rescue post-infarct cardiac function (A) Genetic strategy of generating humanized ENPP1/Tg32 animals and (B) determining the effects of a single dose of hENPP1mAb administered after MI. (C) B mode (top) and M mode (below) echocardiogram demonstrating cardiac contractile function and chamber dilatation in IgG versus hENPP1mAb-injected animals. Green line points to cardiac dimensions in diastole and yellow lines point to dimensions in systole. (D) Ejection fraction, fractional shortening, and LV dimensions in systole (LVIds) and diastole (LVIDd) at 1, 2, and 4 weeks after MI following a single shot of hENPP1mAb or IgG after MI (n = 11/IgG and n = 12/hENPP1mAb). (E) Heart weight, body weight, and heart weight/body weight ratios of hearts harvested at 4 weeks in animals receiving a single dose of hENPP1mAb or IgG after MI (n = 11/IgG and n = 12/hENPP1mAb). (F) Masson trichrome staining to demonstrate fibrosis at 4 weeks post MI in animals receiving a single dose of hENPP1mAb or IgG and quantification of fibrosis (n = 9/IgG and n = 11/hENPP1mAb). Data represented as mean ± SEM, ∗∗p < 0.01, ∗p < 0.05, ns: not significant. Statistical significance was determined using unpaired multiple t test (D) or Student’s t test, 2 tailed (E and F).

References

    1. Frangogiannis N.G. The extracellular matrix in myocardial injury, repair, and remodeling. J. Clin. Invest. 2017;127:1600–1612. doi: 10.1172/JCI87491. - DOI - PMC - PubMed
    1. Ziaeian B., Fonarow G.C. Epidemiology and aetiology of heart failure. Nat. Rev. Cardiol. 2016;13:368–378. doi: 10.1038/nrcardio.2016.25. - DOI - PMC - PubMed
    1. Gulati A., Jabbour A., Ismail T.F., Guha K., Khwaja J., Raza S., Morarji K., Brown T.D.H., Ismail N.A., Dweck M.R., et al. Association of fibrosis with mortality and sudden cardiac death in patients with nonischemic dilated cardiomyopathy. JAMA, J. Am. Med. Assoc. 2013;309:896–908. doi: 10.1001/jama.2013.1363. - DOI - PubMed
    1. Frangogiannis N.G. Pathophysiology of Myocardial Infarction. Compr. Physiol. 2015;5:1841–1875. doi: 10.1002/cphy.c150006. - DOI - PubMed
    1. Tzahor E., Dimmeler S. A coalition to heal-the impact of the cardiac microenvironment. Science. 2022;377 doi: 10.1126/science.abm4443. - DOI - PubMed

MeSH terms

Substances