Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 7555, 2023 Nov 20.
Article in English | MEDLINE | ID: mdl-37985764

ABSTRACT

Macrophages sense changes in the extracellular matrix environment through the integrins and play a central role in regulation of the reparative response after myocardial infarction. Here we show that macrophage integrin α5 protects the infarcted heart from adverse remodeling and that the protective actions are associated with acquisition of an angiogenic macrophage phenotype. We demonstrate that myeloid cell- and macrophage-specific integrin α5 knockout mice have accentuated adverse post-infarction remodeling, accompanied by reduced angiogenesis in the infarct and border zone. Single cell RNA-sequencing identifies an angiogenic infarct macrophage population with high Itga5 expression. The angiogenic effects of integrin α5 in macrophages involve upregulation of Vascular Endothelial Growth Factor A. RNA-sequencing of the macrophage transcriptome in vivo and in vitro followed by bioinformatic analysis identifies several intracellular kinases as potential downstream targets of integrin α5. Neutralization assays demonstrate that the angiogenic actions of integrin α5-stimulated macrophages involve activation of Focal Adhesion Kinase and Phosphoinositide 3 Kinase cascades.


Subject(s)
Integrin alpha5 , Myocardial Infarction , Mice , Animals , Integrin alpha5/metabolism , Vascular Endothelial Growth Factor A/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Macrophages/metabolism , Mice, Knockout , RNA/metabolism
2.
Circulation ; 148(11): 882-898, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37350296

ABSTRACT

BACKGROUND: Pericytes have been implicated in tissue repair, remodeling, and fibrosis. Although the mammalian heart contains abundant pericytes, their fate and involvement in myocardial disease remains unknown. METHODS: We used NG2Dsred;PDGFRαEGFP pericyte:fibroblast dual reporter mice and inducible NG2CreER mice to study the fate and phenotypic modulation of pericytes in myocardial infarction. The transcriptomic profile of pericyte-derived cells was studied using polymerase chain reaction arrays and single-cell RNA sequencing. The role of transforming growth factor-ß (TGF-ß) signaling in regulation of pericyte phenotype was investigated in vivo using pericyte-specific TGF-ß receptor 2 knockout mice and in vitro using cultured human placental pericytes. RESULTS: In normal hearts, neuron/glial antigen 2 (NG2) and platelet-derived growth factor receptor α (PDGFRα) identified distinct nonoverlapping populations of pericytes and fibroblasts, respectively. After infarction, a population of cells expressing both pericyte and fibroblast markers emerged. Lineage tracing demonstrated that in the infarcted region, a subpopulation of pericytes exhibited transient expression of fibroblast markers. Pericyte-derived cells accounted for ~4% of PDGFRα+ infarct fibroblasts during the proliferative phase of repair. Pericyte-derived fibroblasts were overactive, expressing higher levels of extracellular matrix genes, integrins, matricellular proteins, and growth factors, when compared with fibroblasts from other cellular sources. Another subset of pericytes contributed to infarct angiogenesis by forming a mural cell coat, stabilizing infarct neovessels. Single-cell RNA sequencing showed that NG2 lineage cells diversify after infarction and exhibit increased expression of matrix genes, and a cluster with high expression of fibroblast identity markers emerges. Trajectory analysis suggested that diversification of infarct pericytes may be driven by proliferating cells. In vitro and in vivo studies identified TGF-ß as a potentially causative mediator in fibrogenic activation of infarct pericytes. However, pericyte-specific TGF-ß receptor 2 disruption had no significant effects on infarct myofibroblast infiltration and collagen deposition. Pericyte-specific TGF-ß signaling was involved in vascular maturation, mediating formation of a mural cell coat investing infarct neovessels and protecting from dilative remodeling. CONCLUSIONS: In the healing infarct, cardiac pericytes upregulate expression of fibrosis-associated genes, exhibiting matrix-synthetic and matrix-remodeling profiles. A fraction of infarct pericytes exhibits expression of fibroblast identity markers. Pericyte-specific TGF-ß signaling plays a central role in maturation of the infarct vasculature and protects from adverse dilative remodeling, but it does not modulate fibrotic remodeling.


Subject(s)
Myocardial Infarction , Pericytes , Pregnancy , Mice , Female , Humans , Animals , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Placenta/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Fibrosis , Mice, Knockout , Phenotype , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Mammals
3.
Am J Physiol Cell Physiol ; 323(4): C1304-C1324, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36094436

ABSTRACT

The heart contains a population of resident macrophages that markedly expands following injury through recruitment of monocytes and through proliferation of macrophages. In myocardial infarction, macrophages have been implicated in both injurious and reparative responses. In coronary atherosclerotic lesions, macrophages have been implicated in disease progression and in the pathogenesis of plaque rupture. Following myocardial infarction, resident macrophages contribute to initiation and regulation of the inflammatory response. Phagocytosis and efferocytosis are major functions of macrophages during the inflammatory phase of infarct healing, and mediate phenotypic changes, leading to acquisition of an anti-inflammatory macrophage phenotype. Infarct macrophages respond to changes in the cytokine content and extracellular matrix composition of their environment and secrete fibrogenic and angiogenic mediators, playing a central role in repair of the infarcted heart. Macrophages may also play a role in scar maturation and may contribute to chronic adverse remodeling of noninfarcted segments. Single cell studies have revealed a remarkable heterogeneity of macrophage populations in infarcted hearts; however, the relations between transcriptomic profiles and functional properties remain poorly defined. This review manuscript discusses the fate, mechanisms of expansion and activation, and role of macrophages in the infarcted heart. Considering their critical role in injury, repair, and remodeling, macrophages are important, but challenging, targets for therapeutic interventions in myocardial infarction.


Subject(s)
Myocardial Infarction , Cytokines , Humans , Macrophages/pathology , Monocytes/pathology , Myocardial Infarction/pathology , Phagocytosis
4.
Sci Rep ; 12(1): 4542, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35296717

ABSTRACT

Reliable tools for macrophage identification in mouse tissues are critical for studies investigating inflammatory and reparative responses. Transgenic reporter mice and anti-macrophage antibodies have been used as "specific pan-macrophage" markers in many studies; however, organ-specific patterns of expression and non-specific labeling of other cell types, such as fibroblasts, may limit their usefulness. Our study provides a systematic comparison of macrophage labeling patterns in normal and injured mouse tissues, using the CX3CR1 and CSF1R macrophage reporter lines and anti-macrophage antibodies. Moreover, we tested the specificity of macrophage antibodies using the fibroblast-specific PDGFR[Formula: see text] reporter line. Mouse macrophages exhibit organ-specific differences in expression of macrophage markers. Hepatic macrophages are labeled for CSF1R, Mac2 and F4/80, but lack CX3CR1 expression, whereas in the lung, the CSF1R+/Mac2+/Mac3+ macrophage population is not labeled with F4/80. In the splenic red pulp, subpopulations of CSF1R+/F4/80+/Mac3+cells were labeled with Mac2, CX3CR1 and lysozyme M. In the kidney, Mac2, Mac3 and lysozyme M labeled a fraction of the CSF1R+ and CX3CR1+ macrophages, but also stained tubular epithelial cells. In normal hearts, the majority of CSF1R+ and CX3CR1+ cells were not detected with anti-macrophage antibodies. Myocardial infarction was associated with marked expansion of the CSF1R+ and CX3CR1+ populations that peaked during the proliferative phase of cardiac repair, and also expressed Mac2, Mac3 and lysozyme M. In normal mouse tissues, a small fraction of cells labeled with anti-macrophage antibodies were identified as PDGFR[Formula: see text]+ fibroblasts, using a reporter system. The population of PDGFR[Formula: see text]+ cells expressing macrophage markers expanded following injury, likely reflecting emergence of cellular phenotypes with both fibroblast and macrophage characteristics. In conclusion, mouse macrophages exhibit remarkable heterogeneity. Selection of the most appropriate markers for identification of macrophages in mouse tissues is dependent on the organ and the pathologic condition studied.


Subject(s)
Macrophages , Muramidase , Animals , Biomarkers/metabolism , Kupffer Cells/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muramidase/metabolism , Receptor Protein-Tyrosine Kinases/metabolism
5.
J Mol Cell Cardiol ; 159: 48-61, 2021 10.
Article in English | MEDLINE | ID: mdl-34144051

ABSTRACT

Ly6Clow macrophages promote scar formation and prevent early infarct expansion after myocardial infarction (MI). Although CD4+ T cells influence the regulation of Ly6Clow macrophages after MI, the mechanism remains largely unknown. Based on the hypothesis that some molecule(s) secreted by CD4+ T cells act on Ly6Clow macrophages, we searched for candidate molecules by focusing on cytokine receptors expressed on Ly6Clow macrophages. Comparing the transcriptome between Ly6Chigh macrophages and Ly6Clow macrophages harvested from the infarcted heart, we found that Ly6Clow macrophages highly expressed the receptor for interleukin (IL)-21, a pleiotropic cytokine which is produced by several types of CD4+ T cells, compared with Ly6Chigh macrophages. Indeed, CD4+ T cells harvested from the infarcted heart produce IL-21 upon stimulation. Importantly, the survival rate and cardiac function after MI were significantly improved in IL-21-deficient (il21-/-) mice compared with those in wild-type (WT) mice. Transcriptome analysis of infarcted heart tissue from WT mice and il21-/- mice at 5 days after MI demonstrated that inflammation is persistent in WT mice compared with il21-/- mice. Consistent with the transcriptome analysis, the number of neutrophils and matrix metalloproteinase (MMP)-9 expression were significantly decreased, whereas the number of Ly6Clow macrophages and MMP-12 expression were significantly increased in il21-/- mice. In addition, collagen deposition and the number of myofibroblasts in the infarcted area were significantly increased in il21-/- mice. Consistently, IL-21 enhanced the apoptosis of Ly6Clow macrophages. Finally, administration of neutralizing IL-21 receptor Fc protein increased the number of Ly6Clow macrophages in the infarcted heart and improved the survival and cardiac function after MI. Thus, IL-21 decreases the survival after MI, possibly through the delay of wound healing by inducing the apoptosis of Ly6Clow macrophages.


Subject(s)
Interleukins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Wound Healing/physiology , Animals , Cicatrix/metabolism , Inflammation/metabolism , Macrophages/metabolism , Male , Matrix Metalloproteinase 12/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Ventricular Remodeling/physiology
6.
J Mol Cell Cardiol ; 131: 41-52, 2019 06.
Article in English | MEDLINE | ID: mdl-31009606

ABSTRACT

BACKGROUND: Various immune cells are involved in different phases of cardiac repair after myocardial infarction (MI). Especially, Ly6Clow M2-like macrophages (Ly6Clo macrophages) are vital for cardiac repair after MI. However, the molecular mechanisms how Ly6Clo macrophages promote wound healing after MI are still largely unknown. METHODS AND RESULTS: Transcriptome analysis of Ly6Clo macrophages and Ly6Chigh M1-like macrophages (Ly6Chi macrophages) harvested from the infarcted heart revealed that Ly6Clo macrophages highly expressed matrix metalloproteinase (MMP)-12 mRNA compared to Ly6Chi macrophages. MMP-12 expression was enhanced in the infarcted heart and preferentially observed in Ly6Clo macrophages. Importantly, the survival rate and cardiac function after MI were significantly impaired in MMP-12-deficient (mmp12-/-) mice compared with those in wild-type mice. In addition, the extent of myocardial fibrosis and the number of myofibroblasts in the infarct area were decreased in mmp12-/- mice. MMP-9 expression and neutrophils, which are the major cellular source of MMP-9, in the infarcted heart were increased in mmp12-/- mice. Moreover, mRNA expression of neutrophil-attracting chemokines including CXCL1, CXCL2, and CXCL5 was significantly higher in mmp12-/- mice. Consistently, treatment with anti-CXCR2 antibody significantly decreased neutrophil numbers and MMP-9 expression in the infarcted heart in mmp12-/- mice. Finally, the administration of recombinant MMP-12 into the infarcted heart decreased neutrophil numbers in the infarcted heart and promoted wound healing in both wild-type mice and mmp12-/- mice. CONCLUSION: MMP-12 produced by Ly6Clo macrophages improves the survival after MI possibly through the promotion of wound healing by reducing neutrophil infiltration.


Subject(s)
Macrophages/metabolism , Matrix Metalloproteinase 12/therapeutic use , Myocardial Infarction/drug therapy , Animals , Antibodies, Neutralizing/therapeutic use , Echocardiography , Flow Cytometry , Inflammation/metabolism , Macrophages/drug effects , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardium/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Sequence Analysis, RNA
7.
Circ J ; 80(9): 1971-9, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27396441

ABSTRACT

BACKGROUND: Inflammatory responses, especially by CD4(+)T cells activated by dendritic cells, are known to be important in the pathophysiology of cardiac repair after myocardial infarction (MI). Although co-stimulatory signals through B7 (CD80/86) and CD28 are necessary for CD4(+)T cell activation and survival, the roles of these signals in cardiac repair after MI are still unclear. METHODS AND RESULTS: C57BL/6 (Control) mice and CD28 knockout (CD28KO) mice were subjected to left coronary artery permanent ligation. The ratio of death by cardiac rupture within 5 days after MI was significantly higher in CD28KO mice compared with Control mice. Although there were no significant differences in the infarct size between the 2 groups, left ventricular end-diastolic and end-systolic diameters were significantly increased, and fractional shortening was significantly decreased in CD28KO mice compared with Control mice. Electron microscopic observation revealed that the extent of extracellular collagen fiber was significantly decreased in CD28KO mice compared with Control mice. The number of α-smooth muscle actin-positive myofibroblasts was significantly decreased, and matrix metalloproteinase-9 activity and the mRNA expression of interleukin-1ß were significantly increased in CD28KO mice compared with Control mice. CONCLUSIONS: Deletion of CD28 co-stimulatory signals exacerbates left ventricular remodeling and increases cardiac rupture after MI through prolongation of the inflammatory period and reduction of collagen fiber in the infarct scars. (Circ J 2016; 80: 1971-1979).


Subject(s)
CD28 Antigens/deficiency , Gene Deletion , Heart Rupture, Post-Infarction/metabolism , Myocardial Infarction/metabolism , Signal Transduction , Ventricular Remodeling , Animals , CD28 Antigens/metabolism , Gene Expression Regulation , Heart Rupture, Post-Infarction/genetics , Heart Rupture, Post-Infarction/pathology , Heart Rupture, Post-Infarction/physiopathology , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Male , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myofibroblasts/metabolism , Myofibroblasts/ultrastructure
8.
J Mol Cell Cardiol ; 91: 72-80, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26739213

ABSTRACT

AIMS: Dipeptidyl peptidase-4 (DPP-4) inhibitors are reported to have protective effects on various cells but it is unclear how DPP-4 inhibitors have cardioprotective effects. Our aim was to study the mechanisms of cardioprotective effects by DPP-4 inhibition. METHODS AND RESULTS: C57BL/6 mice and DPP-4 knockout (DPP-4KO) mice were subjected to left coronary artery ligation to produce acute myocardial infarction (MI). C57BL/6 mice were then treated with vehicle or DPP-4 inhibitor. Left ventricular function, infarct size, the number of vessels, and myocardial ischemia were assessed at 5days after MI. The treatment with DPP-4 inhibitor significantly improved cardiac function and decreased the infarct size. DPP-4 inhibitor increased the ratio of endothelial cell numbers to a cardiomyocyte. The extent of myocardial ischemia and the number of TUNEL-positive cells in the border area were significantly decreased by DPP-4 inhibitor. Stromal cell-derived factor-1α (SDF-1α) level in myocardium was significantly increased by DPP-4 inhibitor. Those cardioprotective effects after MI were also recognized in DPP-4KO mice. DPP-4 protein was expressed on rat neonatal cardiomyocytes and DPP-4 inhibitor significantly reduced hypoxia-induced apoptosis in the cardiomyocytes. However, this effect was abolished by the pretreatment with a CXCR4 antagonist or a signal transducer and activator of transcription 3 (STAT3) inhibitor. The beneficial effects of DPP-4 inhibitor on heart failure after MI were abolished by cardiomyocyte-specific deletion of STAT3. CONCLUSIONS: DPP-4 inhibition may have direct protective effects on the post-MI heart by inducing an antiapoptotic effect and inhibiting a decrease in vessel number through the SDF-1α/CXCR4-mediated STAT3 signaling pathway.


Subject(s)
Chemokine CXCL12/genetics , Dipeptidyl Peptidase 4/genetics , Heart Failure/prevention & control , Myocardial Infarction/drug therapy , Receptors, CXCR4/genetics , STAT3 Transcription Factor/genetics , Animals , Animals, Newborn , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Chemokine CXCL12/agonists , Chemokine CXCL12/metabolism , Dipeptidyl Peptidase 4/deficiency , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction , Ventricular Function, Left/drug effects
9.
Hinyokika Kiyo ; 60(9): 431-4, 2014 Sep.
Article in Japanese | MEDLINE | ID: mdl-25293796

ABSTRACT

The patient was a 78-year-old female. At the age of 76, the patient had undergone computed tomographic scanning for left lower abdominal pain. A retroperitoneal mass was detected on the right side of the abdomen. The patient had been subject to right nephrectomy due to renal calculus at the age of 33. With Doppler echocardiography and magnetic resonance imaging (MRI), based on the nephrectomy the patient was diagnosed with a renal arteriovenous fistula developed in the right renal bed. Following the wishes of the patient a follow-up examination was conducted. Because of poor heart condition and renal dysfunction, percutaneous arterial embolization was performed. After the embolization, the heart condition and renal dysfunction indicators improved and the blood flow in the arteriovenous fistula disappeared. Cases of renal arteriovenous fistulae after nephrectomy are rare, with only 90 reported worldwide, and percutaneous arterial embolization has been used as the first choice of treatment in recent years.


Subject(s)
Arteriovenous Fistula/therapy , Embolization, Therapeutic , Kidney/blood supply , Renal Artery/diagnostic imaging , Aged , Arteriovenous Fistula/diagnostic imaging , Embolization, Therapeutic/methods , Female , Humans , Nephrectomy , Tomography, X-Ray Computed
10.
Circ J ; 76(5): 1159-68, 2012.
Article in English | MEDLINE | ID: mdl-22361916

ABSTRACT

BACKGROUND: 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins), which are widely used to lower plasma cholesterol levels, have been reported to have various pleiotropic effects such as protective effect of endothelial cells, angiogenic effect, antioxidant effect and anti-inflammatory effect. It is unclear, however, whether statins have any effects on the progression from left ventricular (LV) hypertrophy to heart failure in the established hypertrophied heart. METHODS AND RESULTS: C57BL/6 mice were treated with pitavastatin (pitava) or vehicle (control) from 2 weeks (established hypertrophy stage) after transverse aortic constriction (TAC) and the treatment was continued for 4 weeks. Pitavastatin significantly inhibited the progression from LV hypertrophy to heart failure as assessed on echocardiography. The cardiomyocyte cross-sectional area was significantly increased in the control group compared to the sham-operated mice (sham group), but it was not significantly different between the control group and the pitava group at 6 weeks after TAC. Moreover, pitavastatin induced myocardial angiogenesis (ratio of number of endothelial cells to cardiomyocytes) and decreased the myocardial fibrosis and oxidative stress. The expression of angiopoietin-1 in the heart was significantly increased by pitavastatin at 6 weeks after TAC. CONCLUSIONS: Pitavastatin has preventive effects on the progression of heart failure even in the hypertrophied heart.


Subject(s)
Blood Pressure , Heart Failure/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Myocardium/metabolism , Quinolines/pharmacokinetics , Angiopoietin-1/biosynthesis , Animals , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Gene Expression Regulation/drug effects , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/pathology , Hypertrophy, Left Ventricular/complications , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Mice , Myocardium/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Oxidative Stress/drug effects , Time Factors
11.
Clin Exp Hypertens ; 34(2): 86-91, 2012.
Article in English | MEDLINE | ID: mdl-22251314

ABSTRACT

The Candesartan Cooperative Research of Therapy Design for Early Morning Hypertension in CHIBA was designed to investigate whether switching from angiotensin II receptor blockers (ARBs) except candesartan to candesartan might be effective in Japanese patients with morning hypertension. Seventy-eight mild to moderate hypertensive patients, who were treated with the standard doses of ARBs except candesartan (losartan, 50 mg; valsartan, 80 mg; telmisartan, 40 mg; or olmesartan, 20 mg), were entered into 12-week treatment period with candesartan 8 mg according to a multicenter, open-label design. Morning and office blood pressures (BPs) were significantly reduced (morning, -10.1 ± 10.5/-4.5 ± 8.4 mm Hg; office, -13.1 ± 17.3/-6.2 ± 11.3 mm Hg) after medication change. Target BPs (morning BPs ≤ 135/85 mm Hg and office BPs ≤ 140/90 mm Hg) achievement rates were 42.9% in the morning and 64.3% at office. No adverse events were recognized in all patients. Candesartan treatment significantly reduced the morning and office BPs compared with other ARBs in Japanese patients with morning hypertension.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antihypertensive Agents/therapeutic use , Benzimidazoles/therapeutic use , Hypertension/drug therapy , Tetrazoles/therapeutic use , Aged , Asian People , Benzoates/therapeutic use , Biphenyl Compounds , Blood Pressure Monitoring, Ambulatory , Circadian Rhythm , Female , Humans , Hypertension/physiopathology , Imidazoles/therapeutic use , Japan , Losartan/therapeutic use , Male , Middle Aged , Prospective Studies , Telmisartan , Valine/analogs & derivatives , Valine/therapeutic use , Valsartan
SELECTION OF CITATIONS
SEARCH DETAIL
...