Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
PLoS One ; 19(1): e0295651, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38271331

RESUMO

BACKGROUND: We have developed a new clinical research approach for the quantification of cellular proliferation in human infants to address unanswered questions about tissue renewal and regeneration. The approach consists of oral 15N-thymidine administration to label cells in S-phase, followed by Multi-isotope Imaging Mass Spectrometry for detection of the incorporated label in cell nuclei. To establish the approach, we performed an observational study to examine uptake and elimination of 15N-thymidine. We compared at-home label administration with in-hospital administration in infants with tetralogy of Fallot, a form of congenital heart disease, and infants with heart failure. METHODS: We examined urine samples from 18 infants who received 15N-thymidine (50 mg/kg body weight) by mouth for five consecutive days. We used Isotope Ratio Mass Spectrometry to determine enrichment of 15N relative to 14N (%) in urine. RESULTS/FINDINGS: 15N-thymidine dose administration produced periodic rises of 15N enrichment in urine. Infants with tetralogy of Fallot had a 3.2-fold increase and infants with heart failure had a 4.3-fold increase in mean peak 15N enrichment over baseline. The mean 15N enrichment was not statistically different between the two patient populations (p = 0.103). The time to peak 15N enrichment in tetralogy of Fallot infants was 6.3 ± 1 hr and in infants with heart failure 7.5 ± 2 hr (mean ± SEM). The duration of significant 15N enrichment after a dose was 18.5 ± 1.7 hr in tetralogy of Fallot and in heart failure 18.2 ± 1.8 hr (mean ± SEM). The time to peak enrichment and duration of enrichment were also not statistically different (p = 0.617 and p = 0.887). CONCLUSIONS: The presented results support two conclusions of significance for future applications: (1) Demonstration that 15N-thymidine label administration at home is equivalent to in-hospital administration. (2) Two different types of heart disease show no differences in 15N-thymidine absorption and elimination. This enables the comparative analysis of cellular proliferation between different types of heart disease.


Assuntos
Insuficiência Cardíaca , Tetralogia de Fallot , Humanos , Tetralogia de Fallot/tratamento farmacológico , Isótopos de Nitrogênio , Administração Oral , Boca , Insuficiência Cardíaca/tratamento farmacológico
2.
Nat Metab ; 5(12): 2184-2205, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37996701

RESUMO

Barth syndrome (BTHS) is a life-threatening genetic disorder with unknown pathogenicity caused by mutations in TAFAZZIN (TAZ) that affect remodeling of mitochondrial cardiolipin (CL). TAZ deficiency leads to accumulation of mono-lyso-CL (MLCL), which forms a peroxidase complex with cytochrome c (cyt c) capable of oxidizing polyunsaturated fatty acid-containing lipids. We hypothesized that accumulation of MLCL facilitates formation of anomalous MLCL-cyt c peroxidase complexes and peroxidation of polyunsaturated fatty acid phospholipids as the primary BTHS pathogenic mechanism. Using genetic, biochemical/biophysical, redox lipidomic and computational approaches, we reveal mechanisms of peroxidase-competent MLCL-cyt c complexation and increased phospholipid peroxidation in different TAZ-deficient cells and animal models and in pre-transplant biopsies from hearts of patients with BTHS. A specific mitochondria-targeted anti-peroxidase agent inhibited MLCL-cyt c peroxidase activity, prevented phospholipid peroxidation, improved mitochondrial respiration of TAZ-deficient C2C12 myoblasts and restored exercise endurance in a BTHS Drosophila model. Targeting MLCL-cyt c peroxidase offers therapeutic approaches to BTHS treatment.


Assuntos
Síndrome de Barth , Animais , Humanos , Síndrome de Barth/genética , Síndrome de Barth/patologia , Citocromos c , Fosfolipídeos , Cardiolipinas , Ácidos Graxos Insaturados , Peroxidases
3.
STAR Protoc ; 4(3): 102552, 2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37651236

RESUMO

Nuclear pore complexes are pathways for nuclear-cytoplasmic communication that participate in chromatin organization. Here, we present a protocol to image and quantify the number of nuclear pore complexes in cells. We describe steps for cell plating and culture, immunofluorescence detection, and confocal microscopy visualization of nuclear pore complexes. We then detail quantification and 3D data analysis. This protocol utilizes digital thresholding under human supervision for quantification of nuclear pore complexes. For complete details on the use and execution of this protocol, please refer to Han et al.1.

4.
Nucleus ; 14(1): 2246310, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37606283

RESUMO

In adult mammals, many heart muscle cells (cardiomyocytes) are polyploid, do not proliferate (post-mitotic), and, consequently, cannot contribute to heart regeneration. In contrast, fetal and neonatal heart muscle cells are diploid, proliferate, and contribute to heart regeneration. We have identified interdependent changes of the nuclear lamina, nuclear pore complexes, and DNA-content (ploidy) in heart muscle cell maturation. These results offer new perspectives on how cells alter their nuclear transport and, with that, their gene regulation in response to extracellular signals. We present how changes of the nuclear lamina alter nuclear pore complexes in heart muscle cells. The consequences of these changes for cellular regeneration and stress response in the heart are discussed.


Assuntos
Lâmina Nuclear , Poro Nuclear , Animais , Ploidias , Diferenciação Celular , Laminas , Mamíferos
5.
Dev Cell ; 57(20): 2397-2411.e9, 2022 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-36283391

RESUMO

Nuclear pores are essential for nuclear-cytoplasmic transport. Whether and how cells change nuclear pores to alter nuclear transport and cellular function is unknown. Here, we show that rat heart muscle cells (cardiomyocytes) undergo a 63% decrease in nuclear pore numbers during maturation, and this changes their responses to extracellular signals. The maturation-associated decline in nuclear pore numbers is associated with lower nuclear import of signaling proteins such as mitogen-activated protein kinase (MAPK). Experimental reduction of nuclear pore numbers decreased nuclear import of signaling proteins, resulting in decreased expression of immediate-early genes. In a mouse model of high blood pressure, reduction of nuclear pore numbers improved adverse heart remodeling and reduced progression to lethal heart failure. The decrease in nuclear pore numbers in cardiomyocyte maturation and resulting functional changes demonstrate how terminally differentiated cells permanently alter their handling of information flux across the nuclear envelope and, with that, their behavior.


Assuntos
Membrana Nuclear , Poro Nuclear , Camundongos , Ratos , Animais , Poro Nuclear/metabolismo , Transporte Ativo do Núcleo Celular , Membrana Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 322(4): H579-H596, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35179974

RESUMO

During the past two decades, the field of mammalian myocardial regeneration has grown dramatically, and with this expanded interest comes increasing claims of experimental manipulations that mediate bona fide proliferation of cardiomyocytes. Too often, however, insufficient evidence or improper controls are provided to support claims that cardiomyocytes have definitively proliferated, a process that should be strictly defined as the generation of two de novo functional cardiomyocytes from one original cardiomyocyte. Throughout the literature, one finds inconsistent levels of experimental rigor applied, and frequently the specific data supplied as evidence of cardiomyocyte proliferation simply indicate cell-cycle activation or DNA synthesis, which do not necessarily lead to the generation of new cardiomyocytes. In this review, we highlight potential problems and limitations faced when characterizing cardiomyocyte proliferation in the mammalian heart, and summarize tools and experimental standards, which should be used to support claims of proliferation-based remuscularization. In the end, definitive establishment of de novo cardiomyogenesis can be difficult to prove; therefore, rigorous experimental strategies should be used for such claims.


Assuntos
Miócitos Cardíacos , Regeneração , Animais , Ciclo Celular , Proliferação de Células , Coração/fisiologia , Mamíferos , Miócitos Cardíacos/fisiologia
7.
Development ; 148(14)2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34897388

RESUMO

Terminally differentiated cells are generally thought to have arrived at their final form and function. Many terminally differentiated cell types are polyploid, i.e. they have multiple copies of the normally diploid genome. Mammalian heart muscle cells, termed cardiomyocytes, are one such example of polyploid cells. Terminally differentiated cardiomyocytes are bi- or multi-nucleated, or have polyploid nuclei. Recent mechanistic studies of polyploid cardiomyocytes indicate that they can limit cellular proliferation and, hence, heart regeneration. In this short Spotlight, we present the mechanisms generating bi- and multi-nucleated cardiomyocytes, and the mechanisms generating polyploid nuclei. Our aim is to develop hypotheses about how these mechanisms might relate to cardiomyocyte proliferation and cardiac regeneration. We also discuss how these new findings could be applied to advance cardiac regeneration research, and how they relate to studies of other polyploid cells, such as cancer cells.


Assuntos
Coração/fisiologia , Miócitos Cardíacos/metabolismo , Poliploidia , Regeneração , Animais , Humanos
8.
PLoS Genet ; 17(11): e1009890, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34723970

RESUMO

In contrast to mammals, the zebrafish maintains its cardiomyocyte proliferation capacity throughout adulthood. However, neither the molecular mechanisms that orchestrate the proliferation of cardiomyocytes during developmental heart growth nor in the context of regeneration in the adult are sufficiently defined yet. We identified in a forward genetic N-ethyl-N-nitrosourea (ENU) mutagenesis screen the recessive, embryonic-lethal zebrafish mutant baldrian (bal), which shows severely impaired developmental heart growth due to diminished cardiomyocyte proliferation. By positional cloning, we identified a missense mutation in the zebrafish histone deacetylase 1 (hdac1) gene leading to severe protein instability and the loss of Hdac1 function in vivo. Hdac1 inhibition significantly reduces cardiomyocyte proliferation, indicating a role of Hdac1 during developmental heart growth in zebrafish. To evaluate whether developmental and regenerative Hdac1-associated mechanisms of cardiomyocyte proliferation are conserved, we analyzed regenerative cardiomyocyte proliferation after Hdac1 inhibition at the wound border zone in cryoinjured adult zebrafish hearts and we found that Hdac1 is also essential to orchestrate regenerative cardiomyocyte proliferation in the adult vertebrate heart. In summary, our findings suggest an important and conserved role of Histone deacetylase 1 (Hdac1) in developmental and adult regenerative cardiomyocyte proliferation in the vertebrate heart.


Assuntos
Coração/fisiologia , Histona Desacetilase 1/metabolismo , Miócitos Cardíacos/citologia , Regeneração/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Proliferação de Células
9.
Int J Cardiol ; 339: 36-42, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34265312

RESUMO

BACKGROUND: Patients with Tetralogy of Fallot with pulmonary stenosis (ToF/PS), the most common form of cyanotic congenital heart disease (CHD), develop adverse right ventricular (RV) remodeling, leading to late heart failure and arrhythmia. We recently demonstrated that overactive ß-adrenergic receptor signaling inhibits cardiomyocyte division in ToF/PS infants, providing a conceptual basis for the hypothesis that treatment with the ß-adrenergic receptor blocker, propranolol, early in life would increase cardiomyocyte division. No data are available in ToF/PS infants on the efficacy of propranolol as a possible novel therapeutic option to increase cardiomyocyte division and potentially reduce adverse RV remodeling. METHODS: Using a randomized, double-blind, placebo-controlled trial, we will evaluate the effect of propranolol administration on reactivating cardiomyocyte proliferation to prevent adverse RV remodeling in 40 infants with ToF/PS. Propranolol administration (1 mg/kg po QID) will begin at 1 month of age and last until surgical repair. The primary endpoint is cardiomyocyte division, quantified after 15N-thymidine administration with Multi-isotope Imaging Mass Spectrometry (MIMS) analysis of resected myocardial specimens. The secondary endpoints are changes in RV myocardial and cardiomyocyte hypertrophy. CONCLUSION: This trial will be the first study in humans to assess whether cardiomyocyte proliferation can be pharmacologically increased. If successful, the results could introduce a paradigm shift in the management of patients with ToF/PS from a purely surgical approach, to synergistic medical and surgical management. It will provide the basis for future multi-center randomized controlled trials of propranolol administration in infants with ToF/PS and other types of CHD with RV hypertension. CLINICAL TRIAL REGISTRATION: The trial protocol was registered at clinicaltrials.gov (NCT04713657).


Assuntos
Estenose da Valva Pulmonar , Tetralogia de Fallot , Humanos , Lactente , Miócitos Cardíacos , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores Adrenérgicos beta 2 , Tetralogia de Fallot/diagnóstico por imagem , Tetralogia de Fallot/cirurgia , Remodelação Ventricular
10.
Nat Protoc ; 16(4): 1995-2022, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33627842

RESUMO

Quantification of cellular proliferation in humans is important for understanding biology and responses to injury and disease. However, existing methods require administration of tracers that cannot be ethically administered in humans. We present a protocol for the direct quantification of cellular proliferation in human hearts. The protocol involves administration of non-radioactive, non-toxic stable isotope 15Nitrogen-enriched thymidine (15N-thymidine), which is incorporated into DNA during S-phase, in infants with tetralogy of Fallot, a common form of congenital heart disease. Infants with tetralogy of Fallot undergo surgical repair, which requires the removal of pieces of myocardium that would otherwise be discarded. This protocol allows for the quantification of cardiomyocyte proliferation in this discarded tissue. We quantitatively analyzed the incorporation of 15N-thymidine with multi-isotope imaging spectrometry (MIMS) at a sub-nuclear resolution, which we combined with correlative confocal microscopy to quantify formation of binucleated cardiomyocytes and cardiomyocytes with polyploid nuclei. The entire protocol spans 3-8 months, which is dependent on the timing of surgical repair, and 3-4.5 researcher days. This protocol could be adapted to study cellular proliferation in a variety of human tissues.


Assuntos
Divisão Celular , Marcação por Isótopo/métodos , Espectrometria de Massas/métodos , Miócitos Cardíacos/citologia , Timidina/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células , Feminino , Feto/citologia , Humanos , Imageamento Tridimensional , Lactente , Leucócitos/citologia , Miocárdio/citologia , Isótopos de Nitrogênio/urina , Ploidias , Gravidez , Sarcômeros/metabolismo , Tetralogia de Fallot/patologia
11.
Methods Mol Biol ; 2158: 125-139, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32857370

RESUMO

Failure to regenerate myocardium after injury is a major cause of mortality and morbidity in humans. Direct differentiation of human induced pluripotent stem cells (iPSCs) into cardiomyocytes provides an invaluable resource to pursue cardiac regeneration based on cellular transplantation. Beyond the potential for clinical therapies, iPSC technology also enables the generation of cardiomyocytes to recapitulate patient-specific phenotypes, thus presenting a powerful in vitro cell-based model to understand disease pathology and guide precision medicine. Here, we describe protocols for reprogramming of human dermal fibroblasts and blood cells into iPSCs using the non-integrative Sendai virus system and for the monolayer differentiation of iPSCs to cardiomyocytes using chemically defined media.


Assuntos
Diferenciação Celular , Reprogramação Celular , Derme/citologia , Fibroblastos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Miócitos Cardíacos/citologia , Regeneração , Humanos
12.
Methods Mol Biol ; 2158: 199-210, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32857375

RESUMO

Procurement and characterization of intact human cells are essential for studies in regenerative medicine and translational medical research. The selection of the currently available approaches to isolate intact cells depends on the age of the hearts. To isolate cardiomyocytes from the fetal or neonatal myocardium, the myocardium can be minced into small tissue blocks followed by enzyme incubation. However, the fetal and neonatal cardiomyocytes are very soft and the morphology changes from long rod or spindle shape to spheres after isolation. Because of the dense packing of the cardiomyocytes and the strong cell-cell connection in adult myocardium, it is difficult to isolate the cardiomyocytes from adult myocardium by enzyme incubation only. A perfusion method is necessary to deliver the enzyme solution to the deep layers of the myocardium. However, intact hearts, which are very rare, are required for the perfusion method. Therefore, lacking methods to efficiently isolate cardiomyocytes from myocardium of various ages builds a barrier between basic research and clinical studies. Here, we describe a method for the isolation of intact cardiomyocytes from fresh or frozen human myocardium or fresh mouse hearts and the quantification of multinucleation, cardiomyocyte size, cell cycle activity, and total cardiomyocyte count per heart. We generalize this fixation-digestion method by isolating cells from a variety of mouse organs, including the liver, lung, and thymus.


Assuntos
Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Coração/crescimento & desenvolvimento , Imagem Molecular/métodos , Miocárdio , Miócitos Cardíacos/citologia , Animais , Células Cultivadas , Humanos , Camundongos , Miócitos Cardíacos/fisiologia , Perfusão
13.
Dev Cell ; 53(1): 42-59.e11, 2020 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-32109383

RESUMO

Heart regeneration requires cardiomyocyte proliferation. It is thought that formation of polyploid nuclei establishes a barrier for cardiomyocyte proliferation, but the mechanisms are largely unknown. Here, we show that the nuclear lamina filament Lamin B2 (Lmnb2), whose expression decreases in mice after birth, is essential for nuclear envelope breakdown prior to progression to metaphase and subsequent division. Inactivating Lmnb2 decreased metaphase progression, which led to formation of polyploid cardiomyocyte nuclei in neonatal mice, which, in turn, decreased myocardial regeneration. Increasing Lmnb2 expression promoted cardiomyocyte M-phase progression and cytokinesis and improved indicators of myocardial regeneration in neonatal mice. Inactivating LMNB2 in human iPS cell-derived cardiomyocytes reduced karyokinesis and increased formation of polyploid nuclei. In primary cardiomyocytes from human infants with heart disease, modifying LMNB2 expression correspondingly altered metaphase progression and ploidy of daughter nuclei. In conclusion, Lmnb2 expression is essential for karyokinesis in mammalian cardiomyocytes and heart regeneration.


Assuntos
Coração/fisiologia , Lamina Tipo B/metabolismo , Miócitos Cardíacos/metabolismo , Regeneração/fisiologia , Animais , Núcleo Celular/metabolismo , Divisão do Núcleo Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Cicatrização/fisiologia
14.
Sci Transl Med ; 11(513)2019 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-31597755

RESUMO

One million patients with congenital heart disease (CHD) live in the United States. They have a lifelong risk of developing heart failure. Current concepts do not sufficiently address mechanisms of heart failure development specifically for these patients. Here, analysis of heart tissue from an infant with tetralogy of Fallot with pulmonary stenosis (ToF/PS) labeled with isotope-tagged thymidine demonstrated that cardiomyocyte cytokinesis failure is increased in this common form of CHD. We used single-cell transcriptional profiling to discover that the underlying mechanism of cytokinesis failure is repression of the cytokinesis gene ECT2, downstream of ß-adrenergic receptors (ß-ARs). Inactivation of the ß-AR genes and administration of the ß-blocker propranolol increased cardiomyocyte division in neonatal mice, which increased the number of cardiomyocytes (endowment) and conferred benefit after myocardial infarction in adults. Propranolol enabled the division of ToF/PS cardiomyocytes in vitro. These results suggest that ß-blockers could be evaluated for increasing cardiomyocyte division in patients with ToF/PS and other types of CHD.


Assuntos
Citocinese/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta/metabolismo , Antagonistas Adrenérgicos beta/farmacologia , Animais , Animais Recém-Nascidos , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Propranolol/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Ratos
15.
Circ Res ; 124(12): 1725-1726, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31170042
16.
Development ; 145(5)2018 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-29444893

RESUMO

Zebrafish regenerate cardiac tissue through proliferation of pre-existing cardiomyocytes and neovascularization. Secreted growth factors such as FGFs, IGF, PDGFs and Neuregulin play essential roles in stimulating cardiomyocyte proliferation. These factors activate the Ras/MAPK pathway, which is tightly controlled by the feedback attenuator Dual specificity phosphatase 6 (Dusp6), an ERK phosphatase. Here, we show that suppressing Dusp6 function enhances cardiac regeneration. Inactivation of Dusp6 by small molecules or by gene inactivation increased cardiomyocyte proliferation, coronary angiogenesis, and reduced fibrosis after ventricular resection. Inhibition of Erbb or PDGF receptor signaling suppressed cardiac regeneration in wild-type zebrafish, but had a milder effect on regeneration in dusp6 mutants. Moreover, in rat primary cardiomyocytes, NRG1-stimulated proliferation can be enhanced upon chemical inhibition of Dusp6 with BCI. Our results suggest that Dusp6 attenuates Ras/MAPK signaling during regeneration and that suppressing Dusp6 can enhance cardiac repair.


Assuntos
Fosfatase 6 de Especificidade Dupla/fisiologia , Coração/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Regeneração/genética , Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Proliferação de Células/genética , Regulação para Baixo/genética , Miócitos Cardíacos/fisiologia , Transdução de Sinais/genética , Proteínas de Peixe-Zebra/fisiologia
17.
J Cell Biol ; 217(2): 635-647, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29187525

RESUMO

Chaperone-mediated autophagy (CMA) serves as quality control during stress conditions through selective degradation of cytosolic proteins in lysosomes. Humanin (HN) is a mitochondria-associated peptide that offers cytoprotective, cardioprotective, and neuroprotective effects in vivo and in vitro. In this study, we demonstrate that HN directly activates CMA by increasing substrate binding and translocation into lysosomes. The potent HN analogue HNG protects from stressor-induced cell death in fibroblasts, cardiomyoblasts, neuronal cells, and primary cardiomyocytes. The protective effects are lost in CMA-deficient cells, suggesting that they are mediated through the activation of CMA. We identified that a fraction of endogenous HN is present at the cytosolic side of the lysosomal membrane, where it interacts with heat shock protein 90 (HSP90) and stabilizes binding of this chaperone to CMA substrates as they bind to the membrane. Inhibition of HSP90 blocks the effect of HNG on substrate translocation and abolishes the cytoprotective effects. Our study provides a novel mechanism by which HN exerts its cardioprotective and neuroprotective effects.


Assuntos
Autofagia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Chaperonas Moleculares/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Citosol/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Lisossomos/metabolismo , Masculino , Camundongos , Células NIH 3T3 , Ratos , Ratos Wistar
18.
Sci Total Environ ; 609: 788-798, 2017 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-28768211

RESUMO

Pesticides can exert lethal and sublethal effects on streams organisms. Field studies have shown that non-polluted upstream patches promote population recovery from such effects. Nevertheless, the dynamics and potential carryover effects on the upstream patch are largely unknown. We used a metapopulation model with 2 patches to simulate lethal and sublethal effects on the downstream population of an insect with one generation per year, which was structured into early and late instars aquatic life stages, and an adult terrestrial life stage. We examined the implications for the recovery time of a range of scenarios covering different pesticide effect combinations, migration and exposure types. We found that recovery time responded most strongly to a reduction in reproduction in terms of the reduction of the intrinsic growth rate during the early instar aquatic life stage. For 60 of 96 scenarios with pesticide exposure in consecutive years, no recovery occurred within one year if the intrinsic growth rate was reduced by 50% or more. Without migration between patches (32 scenarios), the polluted downstream population went extinct in 5 of these scenarios. Migration lead overall to slightly faster recovery, albeit this was scenario dependent, but also to a carryover of the pesticide effect from the polluted downstream to the non-polluted upstream patch (up to 25% reduction in the minimal population size). A sensitivity analysis revealed that recovery time was most sensitive to the parameters length of the intrinsic growth phase during early instar aquatic life stage and to migration mortality of the late instar aquatic larvae, and least sensitive to the adult emigration rate and timing of pesticide application. Our study highlights the important role of sublethal effects for population responses to pesticides and that migration buffers against effects, but also carries effects over to non-polluted patches.


Assuntos
Insetos/efeitos dos fármacos , Praguicidas/efeitos adversos , Poluentes Químicos da Água/efeitos adversos , Animais , Larva , Reprodução
19.
Curr Cardiol Rep ; 19(2): 13, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28185170

RESUMO

PURPOSE OF REVIEW: Congenital heart disease is the most common birth defect and acquired heart disease is the leading cause of death in adults. Understanding the mechanisms that drive cardiomyocyte proliferation and differentiation has the potential to advance the understanding and potentially the treatment of different cardiac pathologies, ranging from myopathies and heart failure to myocardial infarction. This review focuses on studies aimed at elucidating signal transduction pathways and molecular mechanisms that promote proliferation, differentiation, and regeneration of differentiated heart muscle cells, cardiomyocytes. RECENT FINDINGS: There is now significant evidence that demonstrates cardiomyocytes continue to proliferate into adulthood. Potential regulators have been identified, including cell cycle regulators, extracellular ligands such as neuregulin, epigenetic targets, reactive oxygen species, and microRNA. The necessary steps should involve validating and applying the new knowledge about cardiomyocyte regeneration towards the development of therapeutic targets for patients. This will be facilitated by the application of standardized pre-clinical models to study cardiomyocyte regeneration.


Assuntos
Diferenciação Celular , Proliferação de Células , Miócitos Cardíacos/citologia , Regeneração , Doenças Cardiovasculares/fisiopatologia , Ciclo Celular , Humanos , Transdução de Sinais
20.
PLoS One ; 11(5): e0155456, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27175488

RESUMO

BACKGROUND: We previously developed and validated a strategy for stimulating heart regeneration by administration of recombinant neuregulin (rNRG1), a growth factor, in mice. rNRG1 stimulated proliferation of heart muscle cells, cardiomyocytes, and was most effective when administration began during the neonatal period. Our results suggested the use of rNRG1 to treat pediatric patients with heart failure. However, administration in this age group may stimulate growth outside of the heart. METHODS: NRG1 and ErbB receptor expression was determined by RT-PCR. rNRG1 concentrations in serum were quantified by ELISA. Mice that received protocols of recombinant neuregulin1-ß1 administration (rNRG1, 100 ng/g body weight, daily subcutaneous injection for the first month of life), previously shown to induce cardiac regeneration, were examined at pre-determined intervals. Somatic growth was quantified by weighing. Organ growth was quantified by MRI and by weighing. Neoplastic growth was examined by MRI, visual inspection, and histopathological analyses. Phospho-ERK1/2 and S6 kinase were analyzed with Western blot and ELISA, respectively. RESULTS: Lung, spleen, liver, kidney, brain, and breast gland exhibited variable expression of the NRG1 receptors ErbB2, ErbB3, ErbB4, and NRG1. Body weight and tibia length were not altered in mice receiving rNRG1. MRI showed that administration of rNRG1 did not alter the volume of the lungs, liver, kidneys, brain, or spinal cord. Administration of rNRG1 did not alter the weight of the lungs, spleen, liver, kidneys, or brain. MRI, visual inspection, and histopathological analyses showed no neoplastic growth. Follow-up for 6 months showed no alteration of somatic or organ growth. rNRG1 treatment increased the levels of phospho-ERK1/2, but not phospho-S6 kinase. CONCLUSIONS: Administration protocols of rNRG1 for stimulating cardiac regeneration in mice during the first month of life did not induce unwanted growth effects. Further studies may be required to determine whether this is the case in a corresponding human population.


Assuntos
Envelhecimento/fisiologia , Coração/fisiologia , Morfogênese/efeitos dos fármacos , Neoplasias/patologia , Neuregulina-1/administração & dosagem , Neuregulina-1/farmacologia , Regeneração/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Receptores ErbB/genética , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Humanos , Rim/efeitos dos fármacos , Rim/enzimologia , Camundongos , Neuregulina-1/sangue , Neuregulina-1/genética , Tamanho do Órgão/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Recombinantes/farmacologia , Proteínas Quinases S6 Ribossômicas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...