Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Nat Cardiovasc Res ; 3(3): 343-355, 2024 Mar.
Article in English | MEDLINE | ID: mdl-39183958

ABSTRACT

Heart failure affects millions of people worldwide, with men exhibiting a higher incidence than women. Our previous work has shown that mosaic loss of the Y chromosome (LOY) in leukocytes is causally associated with an increased risk for heart failure. Here, we show that LOY macrophages from the failing hearts of humans with dilated cardiomyopathy exhibit widespread changes in gene expression that correlate with cardiac fibroblast activation. Moreover, we identify the ubiquitously transcribed t et ratricopeptide Y-linked (Uty) gene in leukocytes as a causal locus for an accelerated progression of heart failure in male mice with LOY. We demonstrate that Uty disruption leads to epigenetic alterations in both monocytes and macrophages, increasing the propensity of differentiation into profibrotic macrophages. Treatment with a transforming growth factor-ß-neutralizing antibody prevented the cardiac pathology associated with Uty deficiency in leukocytes. These findings shed light on the mechanisms that contribute to the higher incidence of heart failure in men.


Subject(s)
Chromosomes, Human, Y , Epigenesis, Genetic , Heart Failure , Animals , Male , Heart Failure/genetics , Heart Failure/pathology , Humans , Chromosomes, Human, Y/genetics , Fibrosis/genetics , Fibrosis/pathology , Macrophages/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Disease Models, Animal , Mice , Female , Phenotype , Mice, Inbred C57BL , Cells, Cultured , Mice, Knockout
2.
Heart Fail Rev ; 29(5): 1049-1063, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38985383

ABSTRACT

Improvements in therapies for heart failure with preserved ejection fraction (HFpEF) are crucial for improving patient outcomes and quality of life. Although HFpEF is the predominant heart failure type among older individuals, its prognosis is often poor owing to the lack of effective therapies. The roles of the spleen and bone marrow are often overlooked in the context of HFpEF. Recent studies suggest that the spleen and bone marrow could play key roles in HFpEF, especially in relation to inflammation and immune responses. The bone marrow can increase production of certain immune cells that can migrate to the heart and contribute to disease. The spleen can contribute to immune responses that either protect or exacerbate heart failure. Extramedullary hematopoiesis in the spleen could play a crucial role in HFpEF. Increased metabolic activity in the spleen, immune cell production and mobilization to the heart, and concomitant cytokine production may occur in heart failure. This leads to systemic chronic inflammation, along with an imbalance of immune cells (macrophages) in the heart, resulting in chronic inflammation and progressive fibrosis, potentially leading to decreased cardiac function. The bone marrow and spleen are involved in altered iron metabolism and anemia, which also contribute to HFpEF. This review presents the concept of an interplay between the heart, spleen, and bone marrow in the setting of HFpEF, with a particular focus on extramedullary hematopoiesis in the spleen. The aim of this review is to discern whether the spleen can serve as a new therapeutic target for HFpEF.


Subject(s)
Bone Marrow , Heart Failure , Hematopoiesis, Extramedullary , Spleen , Humans , Heart Failure/physiopathology , Heart Failure/metabolism , Hematopoiesis, Extramedullary/physiology , Spleen/immunology , Spleen/metabolism , Stroke Volume/physiology , Myocardium/metabolism , Myocardium/pathology , Myocardium/immunology , Inflammation
3.
Circulation ; 148(15): 1165-1178, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37681311

ABSTRACT

BACKGROUND: Clonal hematopoiesis (CH), which results from an array of nonmalignant driver gene mutations, can lead to altered immune cell function and chronic disease, and has been associated with worse outcomes in patients with heart failure (HF) with reduced ejection fraction. However, the role of CH in the prognosis of HF with preserved ejection fraction (HFpEF) has been understudied. This study aimed to characterize CH in patients with HFpEF and elucidate its causal role in a murine model. METHODS: Using a panel of 20 candidate CH driver genes and a variant allele fraction cutoff of 0.5%, ultradeep error-corrected sequencing identified CH in a cohort of 81 patients with HFpEF (mean age, 71±6 years; ejection fraction, 63±5%) and 36 controls without a diagnosis of HFpEF (mean age, 74±7 years; ejection fraction, 61.5±8%). CH was also evaluated in a replication cohort of 59 individuals with HFpEF. RESULTS: Compared with controls, there was an enrichment of TET2-mediated CH in the HFpEF patient cohort (12% versus 0%, respectively; P=0.02). In the HFpEF cohort, patients with CH exhibited exacerbated diastolic dysfunction in terms of E/e' (14.9 versus 11.7, respectively; P=0.0096) and E/A (1.69 versus 0.89, respectively; P=0.0206) compared with those without CH. The association of CH with exacerbated diastolic dysfunction was corroborated in a validation cohort of individuals with HFpEF. In accordance, patients with HFpEF, an age ≥70 years, and CH exhibited worse prognosis in terms of 5-year cardiovascular-related hospitalization rate (hazard ratio, 5.06; P=0.042) compared with patients with HFpEF and an age ≥70 years without CH. To investigate the causal role of CH in HFpEF, nonconditioned mice underwent adoptive transfer with Tet2-wild-type or Tet2-deficient bone marrow and were subsequently subjected to a high-fat diet/L-NAME (Nω-nitro-l-arginine methyl ester) combination treatment to induce features of HFpEF. This model of Tet2-CH exacerbated cardiac hypertrophy by heart weight/tibia length and cardiomyocyte size, diastolic dysfunction by E/e' and left ventricular end-diastolic pressure, and cardiac fibrosis compared with the Tet2-wild-type condition. CONCLUSIONS: CH is associated with worse heart function and prognosis in patients with HFpEF, and a murine experimental model of Tet2-mediated CH displays greater features of HFpEF.


Subject(s)
Heart Failure , Ventricular Dysfunction, Left , Humans , Mice , Animals , Aged , Aged, 80 and over , Heart Failure/diagnosis , Heart Failure/genetics , Heart Failure/drug therapy , Stroke Volume , Ventricular Function, Left , Clonal Hematopoiesis/genetics , Ventricular Dysfunction, Left/genetics
4.
Science ; 377(6603): 292-297, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35857592

ABSTRACT

Hematopoietic mosaic loss of Y chromosome (mLOY) is associated with increased risk of mortality and age-related diseases in men, but the causal and mechanistic relationships have yet to be established. Here, we show that male mice reconstituted with bone marrow cells lacking the Y chromosome display increased mortality and age-related profibrotic pathologies including reduced cardiac function. Cardiac macrophages lacking the Y chromosome exhibited polarization toward a more fibrotic phenotype, and treatment with a transforming growth factor ß1-neutralizing antibody ameliorated cardiac dysfunction in mLOY mice. A prospective study revealed that mLOY in blood is associated with an increased risk for cardiovascular disease and heart failure-associated mortality. Together, these results indicate that hematopoietic mLOY causally contributes to fibrosis, cardiac dysfunction, and mortality in men.


Subject(s)
Aging , Chromosome Deletion , Heart Failure , Hematopoietic Stem Cells , Myocardium , Y Chromosome , Aging/genetics , Animals , Antibodies, Neutralizing/pharmacology , Antibodies, Neutralizing/therapeutic use , Fibrosis , Heart Failure/genetics , Heart Failure/therapy , Macrophages , Male , Mice , Mosaicism , Myocardium/pathology , Transforming Growth Factor beta/antagonists & inhibitors , Y Chromosome/genetics
5.
Heart Fail Clin ; 18(3): 349-359, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35718411

ABSTRACT

Clonal hematopoiesis is a precancerous state that is recognized as a new causal risk factor for cardiovascular disease. Therapy-related clonal hematopoiesis is a condition that is often found in cancer survivors. These clonal expansions are caused by mutations in DNA damage-response pathway genes that allow hematopoietic stem cells to undergo positive selection in response to the genotoxic stress. These mutant cells increasingly give rise to progeny leukocytes that display enhanced proinflammatory properties. Recent experimental studies suggest that therapy-related clonal hematopoiesis may contribute to the medium- to long-term risk of genotoxic therapies on the cardiovascular system.


Subject(s)
Cardiovascular Diseases , Neoplasms , Cardiovascular Diseases/etiology , Clonal Hematopoiesis/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Neoplasms/complications
6.
Cardiovasc Res ; 118(6): 1413-1432, 2022 05 06.
Article in English | MEDLINE | ID: mdl-34164655

ABSTRACT

Clonal haematopoiesis (CH) is a phenomenon whereby somatic mutations confer a fitness advantage to haematopoietic stem and progenitor cells (HSPCs) and thus facilitate their aberrant clonal expansion. These mutations are carried into progeny leucocytes leading to a situation whereby a substantial fraction of an individual's blood cells originate from the HSPC mutant clone. Although this condition rarely progresses to a haematological malignancy, circulating blood cells bearing the mutation have the potential to affect other organ systems as they infiltrate into tissues under both homeostatic and disease conditions. Epidemiological and clinical studies have revealed that CH is highly prevalent in the elderly and is associated with an increased risk of cardiovascular disease and mortality. Recent experimental studies in murine models have assessed the most commonly mutated 'driver' genes associated with CH, and have provided evidence for mechanistic connections between CH and cardiovascular disease. A deeper understanding of the mechanisms by which specific CH mutations promote disease pathogenesis is of importance, as it could pave the way for individualized therapeutic strategies targeting the pathogenic CH gene mutations in the future. Here, we review the epidemiology of CH and the mechanistic work from studies using murine disease models, with a particular focus on the strengths and limitations of these experimental systems. We intend for this review to help investigators select the most appropriate models to study CH in the setting of cardiovascular disease.


Subject(s)
Cardiovascular Diseases , Clonal Hematopoiesis , Aged , Animals , Cardiovascular Diseases/epidemiology , Clonal Hematopoiesis/genetics , Disease Models, Animal , Hematopoiesis/genetics , Hematopoietic Stem Cells , Humans , Mice , Mutation
7.
JCI Insight ; 6(13)2021 07 08.
Article in English | MEDLINE | ID: mdl-34236050

ABSTRACT

Therapy-related clonal hematopoiesis (t-CH) is often observed in cancer survivors. This form of clonal hematopoiesis typically involves somatic mutations in driver genes that encode components of the DNA damage response and confer hematopoietic stem and progenitor cells (HSPCs) with resistance to the genotoxic stress of the cancer therapy. Here, we established a model of TP53-mediated t-CH through the transfer of Trp53 mutant HSPCs to mice, followed by treatment with a course of the chemotherapeutic agent doxorubicin. These studies revealed that neutrophil infiltration in the heart significantly contributes to doxorubicin-induced cardiac toxicity and that this condition is amplified in the model of Trp53-mediated t-CH. These data suggest that t-CH could contribute to the elevated heart failure risk that occurs in cancer survivors who have been treated with genotoxic agents.


Subject(s)
Cardiotoxicity , Clonal Hematopoiesis/genetics , DNA Damage/drug effects , Doxorubicin , Neutrophil Infiltration/drug effects , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Disease Models, Animal , Doxorubicin/pharmacology , Doxorubicin/toxicity , Gene Transfer Techniques , Mice
9.
J Vis Exp ; (171)2021 05 26.
Article in English | MEDLINE | ID: mdl-34125083

ABSTRACT

Clonal hematopoiesis is a prevalent age-associated condition that results from the accumulation of somatic mutations in hematopoietic stem and progenitor cells (HSPCs). Mutations in driver genes, that confer cellular fitness, can lead to the development of expanding HSPC clones that increasingly give rise to progeny leukocytes harboring the somatic mutation. Because clonal hematopoiesis has been associated with heart disease, stroke, and mortality, the development of experimental systems that model these processes is key to understanding the mechanisms that underly this new risk factor. Bone marrow transplantation procedures involving myeloablative conditioning in mice, such as total-body irradiation (TBI), are commonly employed to study the role of immune cells in cardiovascular diseases. However, simultaneous damage to the bone marrow niche and other sites of interest, such as the heart and brain, is unavoidable with these procedures. Thus, our lab has developed two alternative methods to minimize or avoid possible side effects caused by TBI: 1) bone marrow transplantation with irradiation shielding and 2) adoptive BMT to non-conditioned mice. In shielded organs, the local environment is preserved allowing for the analysis of clonal hematopoiesis while the function of resident immune cells is unperturbed. In contrast, the adoptive BMT to non-conditioned mice has the additional advantage that both the local environments of the organs and the hematopoietic niche are preserved. Here, we compare three different hematopoietic cell reconstitution approaches and discuss their strengths and limitations for studies of clonal hematopoiesis in cardiovascular disease.


Subject(s)
Bone Marrow Transplantation , Clonal Hematopoiesis , Animals , Bone Marrow , Hematopoiesis , Hematopoietic Stem Cells , Mice , Transplantation Conditioning
10.
Nat Aging ; 1(12): 1096-1106, 2021 12.
Article in English | MEDLINE | ID: mdl-36846190

ABSTRACT

Cellular metabolism generates molecular damage affecting all levels of biological organization. Accumulation of this damage over time is thought to play a central role in the aging process, but damage manifests in diverse molecular forms complicating its assessment. Insufficient attention has been paid to date to the role of molecular damage in aging-related phenotypes, particularly in humans, in part because of the difficulty in measuring its various forms. Recently, omics approaches have been developed that begin to address this challenge, because they are able to assess a sizeable proportion of age-related damage at the level of small molecules, proteins, RNA, DNA, organelles and cells. This review describes the concept of molecular damage in aging and discusses its diverse aspects from theoretical models to experimental approaches. Measurement of multiple types of damage enables studies of the role of damage in human aging outcomes and lays a foundation for testing interventions to reduce the burden of molecular damage, opening new approaches to slowing aging and reducing its consequences.


Subject(s)
Aging , DNA Damage , Humans , DNA Damage/genetics , Aging/genetics , Organelles , DNA/genetics , Models, Biological
11.
JCI Insight ; 5(6)2020 03 26.
Article in English | MEDLINE | ID: mdl-32154790

ABSTRACT

Clonal hematopoiesis of indeterminate potential is prevalent in elderly individuals and associated with increased risks of all-cause mortality and cardiovascular disease. However, mouse models to study the dynamics of clonal hematopoiesis and its consequences on the cardiovascular system under homeostatic conditions are lacking. We developed a model of clonal hematopoiesis using adoptive transfer of unfractionated ten-eleven translocation 2-mutant (Tet2-mutant) bone marrow cells into nonirradiated mice. Consistent with age-related clonal hematopoiesis observed in humans, these mice displayed a progressive expansion of Tet2-deficient cells in multiple hematopoietic stem and progenitor cell fractions and blood cell lineages. The expansion of the Tet2-mutant fraction was also observed in bone marrow-derived CCR2+ myeloid cell populations within the heart, but there was a negligible impact on the yolk sac-derived CCR2- cardiac-resident macrophage population. Transcriptome profiling revealed an enhanced inflammatory signature in the donor-derived macrophages isolated from the heart. Mice receiving Tet2-deficient bone marrow cells spontaneously developed age-related cardiac dysfunction characterized by greater hypertrophy and fibrosis. Altogether, we show that Tet2-mediated hematopoiesis contributes to cardiac dysfunction in a nonconditioned setting that faithfully models human clonal hematopoiesis in unperturbed bone marrow. Our data support clinical findings that clonal hematopoiesis per se may contribute to diminished health span.


Subject(s)
Clonal Hematopoiesis/physiology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Heart Diseases , Proto-Oncogene Proteins/metabolism , Adoptive Transfer , Aging/pathology , Animals , Dioxygenases , Hematopoietic Stem Cells , Macrophages , Mice
12.
JACC Basic Transl Sci ; 5(2): 196-207, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32140625

ABSTRACT

Heart failure is a common disease with poor prognosis that is associated with cardiac immune cell infiltration and dysregulated cytokine expression. Recently, the clonal expansion of hematopoietic cells with acquired (i.e., nonheritable) DNA mutations, a process referred to as clonal hematopoiesis, has been reported to be associated with cardiovascular diseases including heart failure. Mechanistic studies have shown that leukocytes that harbor these somatic mutations display altered inflammatory characteristics that worsen the phenotypes associated with heart failure in experimental models. In this review, we summarize recent epidemiological and experimental evidence that support the hypothesis that clonal hematopoiesis-mediated immune cell dysfunction contributes to heart failure and cardiovascular disease in general.

13.
Cell Rep ; 29(10): 3235-3252.e9, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801086

ABSTRACT

Dopamine (DA) activates mitogen-activated protein kinase (MAPK) via protein kinase A (PKA)/Rap1 in medium spiny neurons (MSNs) expressing the dopamine D1 receptor (D1R) in the nucleus accumbens (NAc), thereby regulating reward-related behavior. However, how MAPK regulates reward-related learning and memory through gene expression is poorly understood. Here, to identify the relevant transcriptional factors, we perform proteomic analysis using affinity beads coated with cyclic AMP response element binding protein (CREB)-binding protein (CBP), a transcriptional coactivator involved in reward-related behavior. We identify more than 400 CBP-interacting proteins, including Neuronal Per Arnt Sim domain protein 4 (Npas4). We find that MAPK phosphorylates Npas4 downstream of PKA, increasing the Npas4-CBP interaction and the transcriptional activity of Npas4 at the brain-derived neurotrophic factor (BDNF) promoter. The deletion of Npas4 in D1R-expressing MSNs impairs cocaine-induced place preference, which is rescued by Npas4-wild-type (WT), but not by a phospho-deficient Npas4 mutant. These observations suggest that MAPK phosphorylates Npas4 in D1R-MSNs and increases transcriptional activity to enhance reward-related learning and memory.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression/physiology , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/physiology , Animals , Brain-Derived Neurotrophic Factor/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Cocaine/pharmacology , Dopamine/metabolism , Female , Gene Expression/drug effects , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Neurons/drug effects , Neurons/metabolism , Phosphorylation/drug effects , Proteomics/methods , Receptors, Dopamine D1/metabolism , Reward , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
14.
JACC Basic Transl Sci ; 4(6): 684-697, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31709318

ABSTRACT

Janus kinase 2 (valine to phenylalanine at residue 617) (JAK2 V617F ) mutations lead to myeloproliferative neoplasms associated with elevated myeloid, erythroid, and megakaryocytic cells. Alternatively these same mutations can lead to the condition of clonal hematopoiesis with no impact on blood cell counts. Here, a model of myeloid-restricted JAK2 V617F expression from lineage-negative bone marrow cells was developed and evaluated. This model displayed greater cardiac inflammation and dysfunction following permanent left anterior descending artery ligation and transverse aortic constriction. These data suggest that JAK2 V617F mutations arising in myeloid progenitor cells may contribute to cardiovascular disease by promoting the proinflammatory properties of circulating myeloid cells.

15.
J Vis Exp ; (152)2019 10 03.
Article in English | MEDLINE | ID: mdl-31633690

ABSTRACT

Manipulating genes in hematopoietic stem cells using conventional transgenesis approaches can be time-consuming, expensive, and challenging. Benefiting from advances in genome editing technology and lentivirus-mediated transgene delivery systems, an efficient and economical method is described here that establishes mice in which genes are manipulated specifically in hematopoietic stem cells. Lentiviruses are used to transduce Cas9-expressing lineage-negative bone marrow cells with a guide RNA (gRNA) targeting specific genes and a red fluorescence reporter gene (RFP), then these cells are transplanted into lethally-irradiated C57BL/6 mice. Mice transplanted with lentivirus expressing non-targeting gRNA are used as controls. Engraftment of transduced hematopoietic stem cells are evaluated by flow cytometric analysis of RFP-positive leukocytes of peripheral blood. Using this method, ~90% transduction of myeloid cells and ~70% of lymphoid cells at 4 weeks after transplantation can be achieved. Genomic DNA is isolated from RFP-positive blood cells, and portions of the targeted site DNA are amplified by PCR to validate the genome editing. This protocol provides a high-throughput evaluation of hematopoiesis-regulatory genes and can be extended to a variety of mouse disease models with hematopoietic cell involvement.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Gene Editing , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Animals , Base Sequence , Bone Marrow/radiation effects , Bone Marrow Cells/cytology , Cell Lineage , Disease Models, Animal , Male , Mice, Inbred C57BL , Polyethyleneimine/chemistry , RNA, Guide, Kinetoplastida/genetics
16.
Circulation ; 140(21): 1737-1752, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31564129

ABSTRACT

BACKGROUND: Heart failure is a complex syndrome that results from structural or functional impairment of ventricular filling or blood ejection. Protein phosphorylation is a major and essential intracellular mechanism that mediates various cellular processes in cardiomyocytes in response to extracellular and intracellular signals. The RHOA-associated protein kinase (ROCK/Rho-kinase), an effector regulated by the small GTPase RHOA, causes pathological phosphorylation of proteins, resulting in cardiovascular diseases. RHOA also activates protein kinase N (PKN); however, the role of PKN in cardiovascular diseases remains unclear. METHODS: To explore the role of PKNs in heart failure, we generated tamoxifen-inducible, cardiomyocyte-specific PKN1- and PKN2-knockout mice by intercrossing the αMHC-CreERT2 line with Pkn1flox/flox and Pkn2flox/flox mice and applied a mouse model of transverse aortic constriction- and angiotensin II-induced heart failure. To identify a novel substrate of PKNs, we incubated GST-tagged myocardin-related transcription factor A (MRTFA) with recombinant GST-PKN-catalytic domain or GST-ROCK-catalytic domain in the presence of radiolabeled ATP and detected radioactive GST-MRTFA as phosphorylated MRTFA. RESULTS: We demonstrated that RHOA activates 2 members of the PKN family of proteins, PKN1 and PKN2, in cardiomyocytes of mice with cardiac dysfunction. Cardiomyocyte-specific deletion of the genes encoding Pkn1 and Pkn2 (cmc-PKN1/2 DKO) did not affect basal heart function but protected mice from pressure overload- and angiotensin II-induced cardiac dysfunction. Furthermore, we identified MRTFA as a novel substrate of PKN1 and PKN2 and found that MRTFA phosphorylation by PKN was considerably more effective than that by ROCK in vitro. We confirmed that endogenous MRTFA phosphorylation in the heart was induced by pressure overload- and angiotensin II-induced cardiac dysfunction in wild-type mice, whereas cmc-PKN1/2 DKO mice suppressed transverse aortic constriction- and angiotensin II-induced phosphorylation of MRTFA. Although RHOA-mediated actin polymerization accelerated MRTFA-induced gene transcription, PKN1 and PKN2 inhibited the interaction of MRTFA with globular actin by phosphorylating MRTFA, causing increased serum response factor-mediated expression of cardiac hypertrophy- and fibrosis-associated genes. CONCLUSIONS: Our results indicate that PKN1 and PKN2 activation causes cardiac dysfunction and is involved in the transition to heart failure, thus providing unique targets for therapeutic intervention for heart failure.


Subject(s)
Actins/metabolism , Heart Failure/enzymology , Myocytes, Cardiac/enzymology , Protein Kinase C/metabolism , Trans-Activators/metabolism , Animals , Disease Models, Animal , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Phosphorylation , Protein Binding , Protein Kinase C/deficiency , Protein Kinase C/genetics , Signal Transduction , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
17.
FASEB J ; 33(12): 14147-14158, 2019 12.
Article in English | MEDLINE | ID: mdl-31647879

ABSTRACT

Glutaredoxin-1 (Glrx) is a small cytosolic enzyme that removes S-glutathionylation, glutathione adducts of protein cysteine residues, thus modulating redox signaling and gene transcription. Although Glrx up-regulation prevented endothelial cell (EC) migration and global Glrx transgenic mice had impaired ischemic vascularization, the effects of cell-specific Glrx overexpression remained unknown. Here, we examined the role of EC-specific Glrx up-regulation in distinct models of angiogenesis; namely, hind limb ischemia and tumor angiogenesis. EC-specific Glrx transgenic (EC-Glrx TG) overexpression in mice significantly impaired EC migration in Matrigel implants and hind limb revascularization after femoral artery ligation. Additionally, ECs migrated less into subcutaneously implanted B16F0 melanoma tumors as assessed by decreased staining of EC markers. Despite reduced angiogenesis, EC-Glrx TG mice unexpectedly developed larger tumors compared with control mice. EC-Glrx TG mice showed higher levels of VEGF-A in the tumors, indicating hypoxia, which may stimulate tumor cells to form vascular channels without EC, referred to as vasculogenic mimicry. These data suggest that impaired ischemic vascularization does not necessarily associate with suppression of tumor growth, and that antiangiogenic therapies may be ineffective for melanoma tumors because of their ability to implement vasculogenic mimicry during hypoxia.-Yura, Y., Chong, B. S. H., Johnson, R. D., Watanabe, Y., Tsukahara, Y., Ferran, B., Murdoch, C. E., Behring, J. B., McComb, M. E., Costello, C. E., Janssen-Heininger, Y. M. W., Cohen, R. A., Bachschmid, M. M., Matsui, R. Endothelial cell-specific redox gene modulation inhibits angiogenesis but promotes B16F0 tumor growth in mice.


Subject(s)
Endothelial Cells/metabolism , Glutaredoxins/metabolism , Melanoma/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/drug effects , Animals , Female , Femoral Artery/surgery , Glutaredoxins/genetics , Hindlimb/blood supply , Hindlimb/surgery , Ischemia , Ligation , Male , Mice , Mice, Transgenic , Neoplasms, Experimental
18.
Circulation ; 140(6): 487-499, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31170826

ABSTRACT

BACKGROUND: Although the complex roles of macrophages in myocardial injury are widely appreciated, the function of neutrophils in nonischemic cardiac pathology has received relatively little attention. METHODS: To examine the regulation and function of neutrophils in pressure overload-induced cardiac hypertrophy, mice underwent treatment with Ly6G antibody to deplete neutrophils and then were subjected to transverse aortic constriction. RESULTS: Neutrophil depletion diminished transverse aortic constriction-induced hypertrophy and inflammation and preserved cardiac function. Myeloid deficiency of Wnt5a, a noncanonical Wnt, suppressed neutrophil infiltration to the hearts of transverse aortic constriction-treated mice and produced a phenotype that was similar to the neutropenic conditions. Conversely, mice overexpressing Wnt5a in myeloid cells displayed greater hypertrophic growth, inflammation, and cardiac dysfunction. Neutrophil depletion reversed the Wnt5a overexpression-induced cardiac pathology and eliminated differences in cardiac parameters between wild-type and myeloid-specific Wnt5a transgenic mice. CONCLUSIONS: These findings reveal that Wnt5a-regulated neutrophil infiltration has a critical role in pressure overload-induced heart failure.


Subject(s)
Hypertrophy, Left Ventricular/physiopathology , Neutrophils/physiology , Wnt-5a Protein/physiology , Animals , Aorta, Thoracic , Chemotaxis, Leukocyte , Constriction , Heart Failure/etiology , Heart Failure/physiopathology , Hypertrophy, Left Ventricular/etiology , Hypertrophy, Left Ventricular/immunology , Inflammation , Leukocyte Reduction Procedures , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Pressure , Recombinant Proteins/metabolism , Specific Pathogen-Free Organisms , Stress, Mechanical , Ventricular Remodeling/genetics , Wnt-5a Protein/biosynthesis , Wnt-5a Protein/deficiency , Wnt-5a Protein/genetics
19.
Nat Commun ; 8(1): 33, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28652571

ABSTRACT

A long-standing question in neurodevelopment is how neurons develop a single axon and multiple dendrites from common immature neurites. Long-range inhibitory signaling from the growing axon is hypothesized to prevent outgrowth of other immature neurites and to differentiate them into dendrites, but the existence and nature of this inhibitory signaling remains unknown. Here, we demonstrate that axonal growth triggered by neurotrophin-3 remotely inhibits neurite outgrowth through long-range Ca2+ waves, which are delivered from the growing axon to the cell body. These Ca2+ waves increase RhoA activity in the cell body through calcium/calmodulin-dependent protein kinase I. Optogenetic control of Rho-kinase combined with computational modeling reveals that active Rho-kinase diffuses to growing other immature neurites and inhibits their outgrowth. Mechanistically, calmodulin-dependent protein kinase I phosphorylates a RhoA-specific GEF, GEF-H1, whose phosphorylation enhances its GEF activity. Thus, our results reveal that long-range inhibitory signaling mediated by Ca2+ wave is responsible for neuronal polarization.Emerging evidence suggests that gut microbiota influences immune function in the brain and may play a role in neurological diseases. Here, the authors offer in vivo evidence from a Drosophila model that supports a role for gut microbiota in modulating the progression of Alzheimer's disease.


Subject(s)
Axon Initial Segment/metabolism , Calcium Signaling , Calcium/metabolism , Growth Cones/metabolism , Neurites/metabolism , rho GTP-Binding Proteins/genetics , Animals , Axon Initial Segment/ultrastructure , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cell Communication , Cell Differentiation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Mammalian , Gene Expression Regulation, Developmental , Growth Cones/ultrastructure , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , Mice , Mice, Inbred ICR , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Neurites/ultrastructure , Neurogenesis/genetics , Optical Imaging , Optogenetics , Primary Cell Culture , Protein Transport , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
20.
Curr Protoc Cell Biol ; 72: 14.16.1-14.16.12, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27580705

ABSTRACT

Identifying the substrates of protein kinases to understand their modes of action has been undertaken by various approaches and remains an ongoing challenge. Phosphoproteomic technologies have accelerated the accumulation of data concerning protein phosphorylation and have uncovered vast numbers of phosphorylation sites in vivo. In this unit, a novel in vitro screening approach for protein kinase substrates is presented, based on protein-protein interaction and mass spectrometry-based phosphoproteomic technology. © 2016 by John Wiley & Sons, Inc.


Subject(s)
Biochemistry/methods , Protein Kinases/metabolism , Ammonium Sulfate/chemistry , Animals , Brain/metabolism , Chemical Precipitation , Chromatography, Liquid , Phosphopeptides/analysis , Protein Binding , Rats , Substrate Specificity , Tandem Mass Spectrometry , Tissue Extracts/metabolism , Trypsin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL