Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Exp Physiol ; 108(8): 1003-1010, 2023 08.
Article in English | MEDLINE | ID: mdl-37093202

ABSTRACT

NEW FINDINGS: What is the topic of this review? Wound healing is a general response of the body to injury and can be divided into three phases: inflammation, inflammation resolution and repair. In this review, we compare the wound-healing response of the skin after an injury and the wound-healing response of the heart after a myocardial infarction. What advances does it highlight? We highlight differences and similarities between skin and cardiac wound healing and summarize how skin can be used to provide information about the heart. ABSTRACT: Wound healing is a general response of the body to injury. All organs share in common three response elements to wound healing: inflammation to prevent infection and stimulate the removal of dead cells, active anti-inflammatory signalling to turn off the inflammatory response, and a repair phase characterized by extracellular matrix scar formation. The extent of scar formed depends on the ability of endogenous cells that populate each organ to regenerate. The skin has keratinocytes that have regenerative capacity, and in general, wounds are fully re-epithelialized. Heart, in contrast, has cardiac myocytes that have little to no regenerative capacity, and necrotic myocytes are entirely replaced by scars. Despite differences in tissue regeneration, the skin and heart share many wound-healing properties that can be exploited to predict the cardiac response to pathology. We summarize in this review article our current understanding of how the response of the skin to a wounding event can inform us about the ability of the myocardium to respond to a myocardial infarction.


Subject(s)
Cicatrix , Myocardial Infarction , Humans , Cicatrix/pathology , Skin , Wound Healing/physiology , Myocardial Infarction/pathology , Inflammation/pathology
2.
WIREs Mech Dis ; 15(1): e1584, 2023 01.
Article in English | MEDLINE | ID: mdl-36634913

ABSTRACT

Myocardial infarction (MI) is defined as evidence of myocardial necrosis consistent with prolonged ischemia. In response to MI, the myocardium undergoes a series of wound healing events that initiate inflammation and shift to anti-inflammation before transitioning to tissue repair that culminates in scar formation to replace the region of the necrotic myocardium. The overall response to MI is determined by two major steps, the first of which is the secretion of proteases by infiltrating leukocytes to breakdown extracellular matrix (ECM) components, a necessary step to remove necrotic cardiomyocytes. The second step is the generation of new ECM that comprises the scar; and this step is governed by the cardiac fibroblasts as the major source of new ECM synthesis. The leukocyte component resides in the middle of the two-step process, contributing to both sides as the leukocytes transition from pro-inflammatory to anti-inflammatory and reparative cell phenotypes. The balance between the two steps determines the final quantity and quality of scar formed, which in turn contributes to chronic outcomes following MI, including the progression to heart failure. This review will summarize our current knowledge regarding the cardiac wound healing response to MI, primarily focused on experimental models of MI in mice. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Immune System Diseases > Molecular and Cellular Physiology.


Subject(s)
Cicatrix , Myocardial Infarction , Mice , Animals , Cicatrix/metabolism , Wound Healing/physiology , Myocardial Infarction/genetics , Myocardium , Myocytes, Cardiac/metabolism
3.
J Cardiovasc Transl Res ; 16(1): 3-16, 2023 02.
Article in English | MEDLINE | ID: mdl-36197585

ABSTRACT

To identify plasma proteins that mirror current and predict future remodeling after myocardial infarction (MI), we retrospectively interrogated plasma proteomes of day (D)0 control (n = 16) and D3 MI (n = 15) from C57BL/6 J mice (20 ± 1 months). A total of 165 unique proteins were correlated with cardiac physiology variables. We prospectively tested the hypothesis that candidates identified retrospectively would predict cardiac physiology at an extended timepoint (D7 MI) in a second cohort of mice (n = 4 ± 1 months). We also examined human plasma from healthy controls (n = 18) and patients 48 h after presentation for MI (n = 41). Retrospectively, we identified 5 strong reflectors of remodeling (all r ≥ 0.60 and p < 0.05). Prospectively, ApoA1, IgA, IL-17E, and TIMP-1 mirrored current and predicted future remodeling. In humans, cytokine-cytokine receptor signaling was the top enriched KEGG pathway for all candidates. In summary, we identified plasma proteins that serve as useful prognostic indicators of adverse remodeling and progression to heart failure.


Subject(s)
Myocardial Infarction , Proteome , Humans , Mice , Animals , Ventricular Remodeling/physiology , Retrospective Studies , Mice, Inbred C57BL , Myocardial Infarction/metabolism
4.
J Proteomics ; 264: 104636, 2022 07 30.
Article in English | MEDLINE | ID: mdl-35661763

ABSTRACT

While macrophages are well-known to polarize across the inflammatory spectrum, neutrophils have only recently been found to activate in a similar fashion in response to pro- or anti-inflammatory stimuli. Matrix metalloproteinase (MMP)-12 mediates neutrophil physiology with direct signaling mechanisms yet to be investigated. We hypothesized MMP-12 may modify neutrophil signaling. Bone marrow neutrophils were stimulated with interleukin (IL-1ß; pro-inflammatory), IL-4 (anti-inflammatory), or MMP-12. The secretome was mapped by multi-analyte profiling and intracellular signaling evaluated by array. IL-1ß induced a cytokine-mediated inflammatory LPS-like signalome, with upregulation of pro-inflammatory cytokines such as interferon gamma (IFNγ,15.2-fold,p = 0.001), chemokine (C-X-C motif) ligand 1 (CXCL1,8.4-fold,p = 0.005), and tumor necrosis factor alpha (TNFα,11.2-fold,p = 0.004). IL-4 induced strong intracellular signaling with upregulation of mitogen-activated protein kinase kinase (MEK1;1.9-fold,p = 0.0005) and downregulation of signal transducer and activator of transcription 4 (STAT4;0.77-fold,0.001). MMP-12 increased IL-4 secretion 20-fold and induced a robust apoptotic neutrophil signalome with upregulation of forkhead box O1 (FOXO1;1.4-fold,p < 0.0001) and downregulation of WNT signaling with MMP-12 cleavage of the adherens junction components ß-catenin, cahderin-3, and catenin-α2. In conclusion, neutrophils shifted phenotype by stimuli, with MMP-12 inducing a unique apoptotic signalome with higher resemblance to the anti-inflammatory signalome. SIGNIFICANCE: This study revealed that neutrophils demonstrate unique polarization signaling responses to specific stimuli, with the matrix metalloproteinase (MMP)-12 signalome showing similarity to the IL-4 signalome. MMP-12 polarized neutrophils towards a strong apoptotic signature by upregulating FOXO1 and downregulating WNT signaling. Our results highlight that neutrophils display more plasticity than previously appreciated.


Subject(s)
Matrix Metalloproteinase 12 , Neutrophils , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Interferon-gamma/pharmacology , Interleukin-4/metabolism , Interleukin-4/pharmacology , Matrix Metalloproteinase 12/metabolism , Matrix Metalloproteinase 12/pharmacology , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism , Wnt Signaling Pathway
5.
Mol Omics ; 18(3): 186-195, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35230372

ABSTRACT

Inflammation presides early after myocardial infarction (MI) as a key event in cardiac wound healing. Ischemic cardiomyocytes secrete inflammatory cues to stimulate infiltration of leukocytes, predominantly macrophages and neutrophils. Infiltrating neutrophils degranulate to release a series of proteases including matrix metalloproteinase (MMP)-9 to break down extracellular matrix and remove necrotic myocytes to create space for the infarct scar to form. While neutrophil to macrophage communication has been explored, the reverse has been understudied. We used a proteomics approach to catalogue the macrophage secretome at MI day 1. Murinoglobulin-1 (MUG1) was the highest-ranked secreted protein (4.1-fold upregulated at MI day 1 vs. day 0 pre-MI cardiac macrophages, p = 0.004). By transcriptomics evaluation, galectin-3 (Lgals3) was 2.2-fold upregulated (p = 0.008) in MI day 1 macrophages. We explored the direct roles of MUG1 and Lgals3 on neutrophil degranulation. MUG1 blunted while Lgals3 amplified neutrophil degranulation in response to phorbol 12-myristate 13-acetate or interleukin-1ß, as measured by MMP-9 secretion. Lgals3 itself also stimulated MMP-9 secretion. To determine if MUG1 regulated Lgals3, we co-stimulated neutrophils with MUG1 and Lgals3. MUG1 limited degranulation stimulated by Lgals3 by 64% (p < 0.001). In vivo, MUG1 was elevated in the infarct region at MI days 1 and 3, while Lgals3 increased at MI day 7. The ratio of MUG1 to Lgals3 positively correlated with infarct wall thickness, revealing that MUG1 attenuated infarct wall thinning. In conclusion, macrophages at MI day 1 secrete MUG1 to limit and Lgals3 to accentuate neutrophil degranulation to regulate infarct wall thinning.


Subject(s)
Galectin 3 , Matrix Metalloproteinase 9 , Myocardial Infarction , Animals , Galectin 3/genetics , Galectin 3/metabolism , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Myocardial Infarction/metabolism , Neutrophils/metabolism , Serum Globulins
6.
Am J Physiol Heart Circ Physiol ; 322(4): H537-H548, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35089808

ABSTRACT

Both skin wound healing and the cardiac response to myocardial infarction (MI) progress through similar pathways involving inflammation, resolution, tissue repair, and scar formation. Due to the similarities, we hypothesized that the healing response to skin wounding would predict future response to MI. Mice were given a 3-mm skin wound using a disposable biopsy punch and the skin wound was imaged daily until closure. The same set of animals was given MI by permanent coronary artery ligation 28 days later and followed for 7 days. Cardiac physiology was measured by echocardiography at baseline and MI days 3 and 7. Animals that survived until day 7 were grouped as survivors, and animals that died from MI were grouped as nonsurvivors. Survivors had faster skin wound healing than nonsurvivors. Faster skin wound healing predicted MI survival better than commonly used cardiac functional variables (e.g., infarct size, fractional shortening, and end diastolic dimension). N-glycoproteome profiling of MI day 3 plasma revealed α2-macroglobulin and ELL-associated factor 1 as strong predictors of future MI death and progression to heart failure. A second cohort of MI mice validated these findings. To investigate the clinical relevance of α2-macroglobulin, we mapped the plasma glycoproteome in patients with MI 48 h after admission and in healthy controls. In patients, α2-macroglobulin was increased 48 h after MI. Apolipoprotein D, another plasma glycoprotein, detrimentally regulated both skin and cardiac wound healing in male but not female mice by promoting inflammation. Our results reveal that the skin is a mirror to the heart and common pathways link wound healing across organs.NEW & NOTEWORTHY Faster skin wound healers had more efficient cardiac healing after myocardial infarction (MI). Two plasma proteins at D3 MI, EAF1 and A2M, predicted MI death in 66% of cases. ApoD regulated both skin and cardiac wound healing in male mice by promoting inflammation. The skin was a mirror to the heart and common pathways linked wound healing across organs.


Subject(s)
Myocardial Infarction , Ventricular Remodeling , Animals , Humans , Inflammation/metabolism , Macroglobulins/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Transcription Factors/metabolism , Wound Healing/physiology
7.
Am J Physiol Heart Circ Physiol ; 322(2): H145-H155, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34890276

ABSTRACT

Neutrophils infiltrate into the left ventricle (LV) early after myocardial infarction (MI) and launch a proinflammatory response. Along with neutrophil infiltration, LV wall thinning due to cardiomyocyte necrosis also peaks at day 1 in the mouse model of MI. To understand the correlation, we examined a previously published data set that included day 0 (n = 10) and MI day (D) 1 (n = 10) neutrophil proteome and echocardiography assessments. Out of 123 proteins, 4 proteins positively correlated with the infarct wall thinning index (1/wall thickness): histone 1.2 (r = 0.62, P = 0.004), S100A9 (r = 0.60, P = 0.005), histone 3.1 (r = 0.55, P = 0.01), and fibrinogen (r = 0.47, P = 0.04). As S100A9 was the highest ranked secreted protein, we hypothesized that S100A9 is a functional effector of infarct wall thinning. We exogenously administered S100A8/A9 at the time of MI to mice [C57BL/6J, male, 3-6 mo of age, n = 7 M (D1), and n = 5 M (D3)] and compared with saline vehicle control-treated mice [n = 6 M (D1) and n = 6 M (D3)] at MI days 1 and 3. At MI day 3, the S100A8/A9 group showed a 22% increase in the wall thinning index compared with saline (P = 0.02), along with higher dilation and lower ejection fraction. The decline in cardiac physiology occurred subsequent to increased neutrophil and macrophage infiltration at MI day 1 and increased macrophage infiltration at D3. Our results reveal that S100A9 is a functional effector of infarct wall thinning.NEW & NOTEWORTHY S100A9 is a functional marker of infarct wall thinning.


Subject(s)
Calgranulin B/metabolism , Myocardial Infarction/metabolism , Animals , Calgranulin B/genetics , Cells, Cultured , Heart Ventricles/metabolism , Heart Ventricles/pathology , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Neutrophils/metabolism , Proteome/genetics , Proteome/metabolism
9.
Physiol Rep ; 9(23): e15142, 2021 12.
Article in English | MEDLINE | ID: mdl-34889077

ABSTRACT

Acute kidney injury (AKI) is a common perioperative complication that is associated with increased mortality. This study investigates the renal gene expression in male Long-Evans rats after prolonged anesthesia and surgery to detect molecular mechanisms that could predispose the kidneys to injury upon further insults. Healthy and streptozotocin diabetic rats that underwent autoregulatory investigation in an earlier study were compared to rats that were sacrificed quickly for mRNA quantification in the same study. Prolonged surgery caused massive changes in renal mRNA expression by microarray analysis, which was validated by quantitative real-time PCR with good correlation. Furthermore, bioinformatics analysis using gene ontology and pathway analysis identified biological processes involved in immune system activation, such as immune system processes (p = 1.3 × 10-80 ), immune response (p = 1.3 × 10-60 ), and regulation of cytokine production (p = 1.7 × 10-52 ). PCR analysis of specific cell type markers indicated that the gene activation in kidneys was most probably macrophages, while granulocytes and T cell appeared less activated. Immunohistochemistry was used to quantify immune cell infiltration and showed no difference between groups indicating that the genetic activation depends on the activation of resident cells, or infiltration of a relatively small number of highly activated cells. In follow-up experiments, surgery was performed on healthy rats under standard and sterile condition showing similar expression of immune cell markers, which suggests that the inflammation was indeed caused by the surgical trauma rather than by bacterial infection. In conclusion, surgical trauma is associated with rapid activation of immune cells, most likely macrophages in rat kidneys.


Subject(s)
Acute Kidney Injury/metabolism , Cytokines/metabolism , Diabetes Mellitus, Experimental/metabolism , Immunity/genetics , Kidney/metabolism , Acute Kidney Injury/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Homeostasis/physiology , Inflammation/genetics , Inflammation/metabolism , Male , Microarray Analysis , Rats , Rats, Long-Evans
10.
JVS Vasc Sci ; 2: 33-42, 2021.
Article in English | MEDLINE | ID: mdl-34617056

ABSTRACT

OBJECTIVE: Metformin is associated with a reduced incidence and growth of abdominal aortic aneurysms (AAAs). The aim of the present study was to investigate the inhibitory effects of metformin on AAA development and possible underlying mechanisms in experimentally induced AAAs in mice, along with the possible synergistic effects of metformin and imatinib. METHODS: Angiotensin II was used to induce AAAs in apolipoprotein E knockout (ApoE -/- ) mice for 28 days. The mice were treated with metformin (n = 11), metformin combined with imatinib (n = 7), or vehicle (n = 12), starting 3 days before angiotensin II infusion. Ultrasound examination was used to analyze aneurysm formation. Cholesterol and blood pressure levels were measured at the start and end of the study. Gene array and quantitative polymerase chain reaction were used to analyze the changes in gene expression in the aorta. Wire myography was used to study vascular function. RESULTS: Metformin (n = 11) suppressed the formation and progression of AAAs by 50% compared with the vehicle controls (n = 12), with no further effects from imatinib (n = 7). Metformin reduced total cholesterol and mRNA expression of SPP1 (encoding osteopontin), MMP12, and the glycoprotein genes Gpnmb and Clec7a. Furthermore, metformin inhibited blood pressure increases and reduced vascular contractions, as determined by wire myography, and restored the anticontractile function of perivascular adipose tissue. CONCLUSION: Metformin inhibited aneurysm formation and progression and normalized vascular function in ApoE -/- mice with no additional effect of imatinib. This might be mediated by the protective effects on vascular endothelial function and perivascular adipose tissue via reduced expression of genes promoting inflammation, including SPP1, MMP12, Gpnmb, and Clec7a. CLINICAL RELEVANCE: Retrospective studies of the effects of metformin in patients with aneurysm have so far only been performed of those with type 2 diabetes. The present study shows that metformin has effects on nondiabetic mice and revealed the mechanistic effects mediated by the drug that could also be important to study as outcomes in humans. Future clinical trials using metformin are warranted in patients without diabetes with abdominal aortic aneurysms.

11.
J Am Heart Assoc ; 10(17): e020231, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34420357

ABSTRACT

Background While numerous interventions effectively interfered with abdominal aortic aneurysm (AAA) formation/progression in preclinical models, none of the successes translated into clinical success. Hence, a systematic exploration of parallel and divergent processes in clinical AAA disease and its 2 primary models (the porcine pancreatic elastase and angiotensin-II infusion [AngII] murine model) was performed to identify mechanisms relevant for aneurysm disease. Methods and Results This study combines Movat staining and pathway analysis for histological and genomic comparisons between clinical disease and its models. The impact of a notable genomic signal for metabolic reprogramming was tested in a rescue trial (AngII model) evaluating the impact of 1-(4-pyridinyl)-3-(2-quinolinyl)-2-propen-1-one (PFK15)-mediated interference with main glycolytic switch PFKFB3. Histological evaluation characterized the AngII model as a dissection model that is accompanied by adventitial fibrosis. The porcine pancreatic elastase model showed a transient inflammatory response and aortic dilatation, followed by stabilization and fibrosis. Normalization of the genomic responses at day 14 confirmed the self-limiting nature of the porcine pancreatic elastase model. Clear parallel genomic responses with activated adaptive immune responses, and particularly strong signals for metabolic switching were observed in human AAA and the AngII model. Rescue intervention with the glycolysis inhibitor PFK15 in the AngII model showed that interference with the glycolytic switching quenches aneurysm formation. Conclusions Despite clear morphological contrasts, remarkable genomic parallels exist for clinical AAA disease and the AngII model. The metabolic response appears causatively involved in AAA progression and provides a novel therapeutic target. The clear transient genomic response classifies the porcine pancreatic elastase model as a disease initiation model.


Subject(s)
Aortic Aneurysm, Abdominal , Angiotensin II , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Disease Models, Animal , Fibrosis , Genomics , Humans , Mice , Mice, Inbred C57BL , Pancreatic Elastase , Swine
12.
Biomolecules ; 11(4)2021 03 25.
Article in English | MEDLINE | ID: mdl-33805901

ABSTRACT

Over the past three decades, numerous studies have shown a strong connection between matrix metalloproteinase 9 (MMP-9) levels and myocardial infarction (MI) mortality and left ventricle remodeling and dysfunction. Despite this fact, clinical trials using MMP-9 inhibitors have been disappointing. This review focuses on the roles of MMP-9 in MI wound healing. Infiltrating leukocytes, cardiomyocytes, fibroblasts, and endothelial cells secrete MMP-9 during all phases of cardiac repair. MMP-9 both exacerbates the inflammatory response and aids in inflammation resolution by stimulating the pro-inflammatory to reparative cell transition. In addition, MMP-9 has a dual effect on neovascularization and prevents an overly stiff scar. Here, we review the complex role of MMP-9 in cardiac wound healing, and highlight the importance of targeting MMP-9 only for its detrimental actions. Therefore, delineating signaling pathways downstream of MMP-9 is critical.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Myocardial Infarction/pathology , Extracellular Matrix/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/cytology , Macrophages/metabolism , Myocardial Infarction/metabolism , Neovascularization, Physiologic , Signal Transduction , Ventricular Remodeling
13.
Curr Opin Physiol ; 242021 Dec.
Article in English | MEDLINE | ID: mdl-35664861

ABSTRACT

Myocardial infarction (MI) initiates an intense inflammatory response that induces neutrophil infiltration into the infarct region. Neutrophils commence the pro-inflammatory response that includes upregulation of cytokines and chemokines (e.g., interleukin-1 beta) and degranulation of pre-formed proteases (e.g., matrix metalloproteinases -8 and -9) that degrade existing extracellular matrix to clear necrotic tissue. An increase or complete depletion of neutrophils both paradoxically impair MI resolution, indicating a complex role of neutrophils in cardiac wound healing. Following pro-inflammation, the neutrophil shifts to a reparative phenotype that promotes inflammation resolution and aids in scar formation. Across the shifts in phenotype, the neutrophil communicates with other cells to coordinate repair and scar formation. This review summarizes our current understanding of neutrophil crosstalk with cardiomyocytes and macrophages during MI wound healing.

14.
Cell Signal ; 77: 109816, 2021 01.
Article in English | MEDLINE | ID: mdl-33122000

ABSTRACT

Neutrophils are key effector cells of the innate immune system, serving as a first line of defense in the response to injury and playing essential roles in the wound healing process. Following myocardial infarction (MI), neutrophils infiltrate into the infarct region to propagate inflammation and begin the initial phase of cardiac wound repair. Pro-inflammatory neutrophils release proteases to degrade extracellular matrix (ECM), a necessary step for the removal of necrotic myocytes as a prelude for scar formation. Neutrophils transition their phenotype over time to regulate MI inflammation resolution and stabilize scar formation. Neutrophils contribute to the evolution from inflammation to resolution and scar formation by serving anti-inflammatory and repair functions. As anti-inflammatory cells, neutrophils contribute ECM proteins during scar formation, in particular fibronectin, galectin-3, and vimentin. The diverse and polarizing functions that contribute to MI wound repair make this innate immune cell a viable target to improve MI outcomes. Thus, understanding the signaling involved in neutrophil physiology in the context of MI may help to identify novel therapeutic targets.


Subject(s)
Myocardial Infarction/pathology , Neutrophils/metabolism , Wound Healing , Chemokine CXCL12/metabolism , Extracellular Matrix Proteins/metabolism , Humans , Neutrophil Infiltration , Neutrophils/immunology , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
15.
Am J Physiol Heart Circ Physiol ; 319(1): H235-H241, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32469635

ABSTRACT

To maximize data obtainment from valuable cardiac tissue, we hypothesized that myocardium fixed in optimal cutting temperature (OCT) medium for histology could also be used to investigate the function of myofilament proteins in situ. We compared tissue prepared via conventional liquid nitrogen (LN) snap freezing with tissue fixed in OCT and then sectioned in fiber-parallel orientation. We found that actin-myosin Ca2+ sensitivity, activation rate by Ca2+, cooperativity along the thin filament, as well as cross-bridge cycling rate were unaffected by OCT storage and could reliably be interpreted after sectioning. Absolute values in maximum force generation per cross-sectional area, as well as passive strain, are difficult to investigate after sectioning, as myofibrillar continuity along the preparation cannot be guaranteed. We have shown that myocardial tissue stored in OCT and sectioned before analysis is available for functional analysis, a valuable means of maximizing usage of precious cardiac biopsies.NEW & NOTEWORTHY Myocardial tissue in optimal cutting temperature (OCT) fixation and cryostat sectioning was tested as a means of storing and preparing tissue for myofilament function analysis in relation to conventional liquid nitrogen freezing and dissection. Actomyosin interaction, Ca2+ force activation, and passive compliance were tested. The study concluded that OCT storage and cryostat sectioning do not interfere with the actomyosin cross-bridge dynamics or Ca2+ activation but that absolute tension values suffer and may not be investigated by this method.


Subject(s)
Cryopreservation/methods , Frozen Sections/methods , Myocardium/cytology , Myofibrils/metabolism , Paraffin Embedding/methods , Animals , Cryopreservation/standards , Frozen Sections/standards , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myofibrils/ultrastructure , Paraffin Embedding/standards
16.
Physiol Genomics ; 51(7): 279-289, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31125294

ABSTRACT

Genetic background of different mouse strains determines their susceptibility to disease. We have previously shown that Balb/CJ and C57BL/6J mice develop cardiac hypertrophy to the same degree when treated with a combination of angiotensin II and high-salt diet (ANG II+Salt), but only Balb/CJ show impaired cardiac function associated with edema development and substantial mortality. We hypothesized that the different response to ANG II+Salt is due to the different genetic backgrounds of Balb/CJ and C57BL/6J. To address this we performed quantitative trait locus (QTL) mapping of second filial generation (F2) of mice derived from a backcross between Balb/CJ and first filial generation (F1) of mice. Cardiac function was measured with echocardiography, glomerular filtration rate using FITC-inulin clearance, fluid and electrolyte balance in metabolic cages, and blood pressure with tail-cuff at baseline and on the fourth day of treatment with ANG II+Salt. A total of nine QTLs were found to be linked to different phenotypes in ANG II+Salt-treated F2 mice. A QTL on chromosome 3 was linked to cardiac output, and a QTL on chromosome 12 was linked to isovolumic relaxation time. QTLs on chromosome 2 and 3 were linked to urine excretion and sodium excretion. Eight genes located at the different QTLs contained coding nonsynonymous SNPs published in the mouse genome database that differ between Balb/CJ and C57BL/6J. In conclusion, ANG II+Salt-induced acute decompensation in Balb/CJ is genetically linked to several QTLs, indicating a multifaceted phenotype. The present study identified potential candidate genes that may represent important pathways in acute decompensated heart failure.


Subject(s)
Angiotensin II/genetics , Heart Failure/genetics , Heart Failure/metabolism , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Sodium Chloride, Dietary/adverse effects , Animals , Blood Pressure/physiology , Cardiomegaly/genetics , Chromosome Mapping , Diet , Disease Models, Animal , Echocardiography , Female , Genetic Linkage , Genetic Predisposition to Disease , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Oligonucleotide Array Sequence Analysis , Phenotype , Species Specificity , Thiobarbituric Acid Reactive Substances
17.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R563-R570, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30840486

ABSTRACT

The genetic background of a mouse strain determines its susceptibility to disease. C57BL/6J and Balb/CJ are two widely used inbred mouse strains that we found react dramatically differently to angiotensin II and high-salt diet (ANG II + Salt). Balb/CJ show increased mortality associated with anuria and edema formation while C57BL/6J develop arterial hypertension but do not decompensate and die. Clinical symptoms of heart failure in Balb/CJ mice gave the hypothesis that ANG II + Salt impairs cardiac function and induces cardiac remodeling in male Balb/CJ but not in male C57BL/6J mice. To test this hypothesis, we measured cardiac function using echocardiography before treatment and every day for 7 days during treatment with ANG II + Salt. Interestingly, pulsed wave Doppler of pulmonary artery flow indicated increased pulmonary vascular resistance and right ventricle systolic pressure in Balb/CJ mice, already 24 h after ANG II + Salt treatment was started. In addition, Balb/CJ mice showed abnormal diastolic filling indicated by reduced early and late filling and increased isovolumic relaxation time. Furthermore, Balb/CJ exhibited lower cardiac output compared with C57BL/6J even though they retained more sodium and water, as assessed using metabolic cages. Left posterior wall thickness increased during ANG II + Salt treatment but did not differ between the strains. In conclusion, ANG II + Salt treatment causes early restriction of pulmonary flow and reduced left ventricular filling and cardiac output in Balb/CJ, which results in fluid retention and peripheral edema. This makes Balb/CJ a potential model to study the adaptive capacity of the heart for identifying new disease mechanisms and drug targets.


Subject(s)
Angiotensin II/metabolism , Cardio-Renal Syndrome/physiopathology , Diet , Hypertension/physiopathology , Animals , Blood Pressure/physiology , Cardio-Renal Syndrome/complications , Heart Failure/physiopathology , Hypertension/complications , Hypertension, Pulmonary/complications , Male , Mice, Inbred BALB C , Myocardium/metabolism , Sodium Chloride, Dietary/metabolism , Sodium Chloride, Dietary/pharmacology , Time Factors , Water-Electrolyte Imbalance/drug therapy , Water-Electrolyte Imbalance/metabolism
18.
Am J Physiol Renal Physiol ; 316(5): F914-F933, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30785350

ABSTRACT

Balb/CJ mice are more sensitive to treatment with angiotensin II (ANG II) and high-salt diet compared with C57BL/6J mice. Together with higher mortality, they develop edema, signs of heart failure, and acute kidney injury. The aim of the present study was to identify differences in renal gene regulation that may affect kidney function and fluid balance, which could contribute to decompensation in Balb/CJ mice after ANG II + salt treatment. Male Balb/CJ and C57BL/6J mice were divided into the following five different treatment groups: control, ANG II, salt, ANG II + salt, and ANG II + salt + N-acetylcysteine. Gene expression microarrays were used to explore differential gene expression after treatment and between the strains. Published data from the Mouse Genome Database were used to identify the associated genomic differences. The glomerular filtration rate (GFR) was measured using inulin clearance, and fluid balance was measured using metabolic cages. Gene ontology enrichment analysis of gene expression microarrays identified glutathione transferase (antioxidant system) as highly enriched among differentially expressed genes. Balb/CJ mice had similar GFR compared with C57BL/6J mice but excreted less Na+ and water, although net fluid and electrolyte balance did not differ, suggesting that Balb/CJ mice may be inherently more prone to decompensation. Interestingly, C57BL/6J mice had higher urinary oxidative stress despite their relative protection from decompensation. In addition, treatment with the antioxidant N-acetylcysteine decreased oxidative stress in C57BL/6J mice, reduced urine excretion, and increased mortality. Balb/CJ mice are more sensitive than C57BL/6J to ANG II + salt, in part mediated by lower oxidative stress, which favors fluid and Na+ retention.


Subject(s)
Angiotensin II , Glomerular Filtration Rate , Kidney/physiopathology , Oxidative Stress , Sodium Chloride, Dietary , Water-Electrolyte Balance , Water-Electrolyte Imbalance/physiopathology , Animals , Blood Pressure , Disease Models, Animal , Female , Gene Expression Regulation , Glomerular Filtration Rate/genetics , Kidney/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy , Sex Factors , Species Specificity , Water-Electrolyte Balance/genetics , Water-Electrolyte Imbalance/etiology , Water-Electrolyte Imbalance/genetics , Water-Electrolyte Imbalance/metabolism
19.
Am J Physiol Renal Physiol ; 316(1): F101-F112, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30332317

ABSTRACT

The aim was to identify new targets that regulate gene expression at the posttranscriptional level in angiotensin II (ANGII)-mediated hypertension. Heparin affinity chromatography was used to enrich nucleic acid-binding proteins from kidneys of two-kidney, one-clip (2K1C) hypertensive Wistar rats. The experiment was repeated with 14-day ANGII infusion using Alzet osmotic mini pumps, with or without ANGII receptor AT1a inhibition using losartan in the drinking water. Mean arterial pressure increased after 2K1C or ANGII infusion and was inhibited with losartan. Heparin affinity chromatography and mass spectrometry were used to identify Annexin-A2 (ANXA2) as having differential nucleic acid-binding activity. Total Annexin-A2 protein expression was unchanged, whereas nucleic acid-binding activity was increased in both kidneys of 2K1C and after ANGII infusion through AT1a stimulation. Costaining of Annexin-A2 with α-smooth muscle actin and aquaporin 2 showed prominent expression in the endothelia of larger arteries and the cells of the inner medullary collecting duct. The nuclear factor of activated T cells (NFAT) transcription factor was identified as a likely Annexin-A2 target using enrichment analysis on a 2K1C microarray data set and identifying several binding sites in the regulatory region of the mRNA. Expression analysis showed that ANGII increases NFAT5 protein but not mRNA level and, thus, indicated that NFAT5 is regulated by posttranscriptional regulation, which correlates with activation of the RNA-binding protein Annexin-A2. In conclusion, we show that ANGII increases Annexin-A2 nucleic acid-binding activity that correlates with elevated protein levels of the NFAT5 transcription factor. NFAT signaling appears to be a major contributor to renal gene regulation in high-renin states.


Subject(s)
Angiotensin II , Annexin A2/metabolism , Arterial Pressure , Hypertension/metabolism , Kidney/metabolism , RNA Processing, Post-Transcriptional , Transcription Factors/metabolism , Animals , Annexin A2/genetics , Disease Models, Animal , Hypertension/genetics , Hypertension/pathology , Hypertension/physiopathology , Kidney/pathology , Kidney/physiopathology , Male , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , Transcription Factors/genetics , Up-Regulation
20.
Am J Physiol Renal Physiol ; 315(2): F241-F246, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29667909

ABSTRACT

Renin-angiotensin-system blockers are thought to increase the risk of acute kidney injury after surgery and hemorrhage. We found that losartan does not cause renal cortical hypoxia after hemorrhage in rats because of decreased renal vascular resistance, but we did not evaluate resuscitation. We aimed to study losartan's effect on renal cortical and medullary oxygenation, as well as norepinephrine's vasopressor effect in a model of resuscitated hemorrhage. After 7 days of losartan (60 mg·kg-1·day-1) or control treatment, male Wistar rats were hemorrhaged 20% of their blood volume and resuscitated with Ringer's acetate. Mean arterial pressure, renal blood flow, and kidney tissue oxygenation were measured at baseline and after resuscitation. Finally, the effect of norepinephrine on mean arterial pressure and renal blood flow was investigated. As expected, losartan lowered mean arterial pressure but not renal blood flow. Losartan did not affect renal oxygen consumption and oxygen tension. Mean arterial pressure and renal blood flow were lower after resuscitated hemorrhage. A smaller increase of renal vascular resistance in the losartan group translated to a smaller decrease in cortical oxygen tension, but no significant difference was seen in medullary oxygen tension, either between groups or after hemorrhage. The effect of norepinephrine on mean arterial pressure and renal blood flow was similar in control- and losartan-treated rats. Losartan does not decrease renal oxygenation after resuscitated hemorrhage because of a smaller increase in renal vascular resistance. Further, losartan does not decrease the efficiency of norepinephrine as a vasopressor, indicating that blood pressure may be managed effectively during losartan treatment.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Hemodynamics/drug effects , Hemorrhage/therapy , Kidney/blood supply , Kidney/drug effects , Losartan/pharmacology , Norepinephrine/pharmacology , Oxygen Consumption/drug effects , Renal Circulation/drug effects , Resuscitation/methods , Vasoconstrictor Agents/pharmacology , Animals , Arterial Pressure/drug effects , Disease Models, Animal , Hemorrhage/metabolism , Hemorrhage/physiopathology , Kidney/metabolism , Male , Rats, Wistar , Vascular Resistance/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...