Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Can J Physiol Pharmacol ; 102(1): 14-25, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37748207

RESUMO

Cardiovascular disease (CVD) complications have remained a major cause of death among patients with diabetes. Hence, there is a need for effective therapeutics against diabetes-induced CVD complications. Since its discovery, proprotein convertase subtilisin/kexin type 9 (PCSK9) has been reported to be involved in the pathology of various CVDs, with studies showing a positive association between plasma levels of PCSK9, hyperglycemia, and dyslipidemia. PCSK9 regulates lipid homeostasis by interacting with low-density lipoprotein receptors (LDLRs) present in hepatocytes and subsequently induces LDLR degradation via receptor-mediated endocytosis, thereby reducing LDL uptake from circulation. In addition, PCSK9 also induces pro-inflammatory cytokine expression and apoptotic cell death in diabetic-CVD. Furthermore, therapies designed to inhibit PCSK9 effectively reduces diabetic dyslipidemia with clinical studies reporting reduced cardiovascular events in patients with diabetes and no significant adverse effect on glycemic controls. In this review, we discuss the role of PCSK9 in the pathogenesis of diabetes-induced CVD and the potential mechanisms by which PCSK9 inhibition reduces cardiovascular events in diabetic patients.


Assuntos
Doenças Cardiovasculares , Complicações do Diabetes , Diabetes Mellitus , Dislipidemias , Humanos , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/uso terapêutico , Dislipidemias/complicações , Dislipidemias/tratamento farmacológico , Dislipidemias/metabolismo , Doenças Cardiovasculares/etiologia , Subtilisinas/uso terapêutico , Diabetes Mellitus/tratamento farmacológico
2.
Am J Physiol Heart Circ Physiol ; 325(4): H751-H767, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37594487

RESUMO

The use of immunotherapies like pembrolizumab (PEM) is increasingly common for the management of numerous cancer types. The use of PEM to bolster T-cell response against tumor growth is well documented. However, the interactions PEM has on other immune cells to facilitate tumor regression and clearance is unknown and warrants further investigation. In this review, we present literature findings that have reported the interactions of PEM in stimulating innate and adaptive immune cells, which enhance cytotoxic phenotypes. This triggers secretion of cytokines and chemokines, which have both beneficial and detrimental effects. We also describe how this leads to the development of rare but underreported occurrence of PEM-induced immune-related cardiovascular complications that arise suddenly and progress rapidly to debilitating and fatal consequences. This review encourages further research and investigation of PEM-induced cardiovascular complications and other immune cell interactions in patients with cancer. As PEM therapy in treating cancer types is expanding, we expect that this review will inform health care professionals of diverse specializations of medicine like dermatology (melanoma skin cancers), ophthalmology (eye cancers), and pathology (hematological malignancies) about PEM-induced cardiac complications.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Anticorpos Monoclonais Humanizados/efeitos adversos
3.
Am J Physiol Heart Circ Physiol ; 323(3): H403-H420, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35776683

RESUMO

Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, is a global pandemic impacting 254 million people in 190 countries. Comorbidities, particularly cardiovascular disease, diabetes, and hypertension, increase the risk of infection and poor outcomes. SARS-CoV-2 enters host cells through the angiotensin-converting enzyme-2 receptor, generating inflammation and cytokine storm, often resulting in multiorgan failure. The mechanisms and effects of COVID-19 on patients with high-risk diabetes are not yet completely understood. In this review, we discuss the variety of coronaviruses, structure of SARS-CoV-2, mutations in SARS-CoV-2 spike proteins, receptors associated with viral host entry, and disease progression. Furthermore, we focus on possible mechanisms of SARS-CoV-2 in diabetes, leading to inflammation and heart failure. Finally, we discuss existing therapeutic approaches, unanswered questions, and future directions.


Assuntos
COVID-19 , Diabetes Mellitus , Diabetes Mellitus/epidemiologia , Humanos , Inflamação , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , SARS-CoV-2
4.
Can J Physiol Pharmacol ; 100(5): 441-452, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34932406

RESUMO

Diabetic cancer patients were treated with doxorubicin (DOX), a potent chemotherapeutic drug that induces cardiac toxicity; however, molecular mechanisms of cardiac toxicity in this specific disease progression in patients and animal models are completely unknown. Therefore, we designed a study to understand the effects of DOX-induced cardiac toxicity in diabetic animals and the involved pathophysiological mechanisms. C57BL/6 J mice were divided into four DOX- and diabetic (streptozotocin; STZ) - treated groups; control, STZ, DOX, and DOX+STZ. At day 14, animals were sacrificed, echocardiography was used to examine heart function, and heart and blood samples were collected to investigate apoptotic mechanisms (caspase 3, BAX, B-Cell leukemia/lymphoma 2 (Bcl2)), inflammation, and cardiac remodeling. Our data shows a significant (p < 0.05) increase in glucose levels, apoptotic markers, and monocyte infiltration at the site of apoptosis and triggered inflammatory immune response (tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6)), in DOX+STZ animals compared with control and experimental groups. We also observed significant (p < 0.05) increase in myofibrillar area, fibrosis, and significantly decreased (p < 0.05) cardiac function in DOX-treated diabetic animals compared with controls. In conclusion, our data suggest that DOX induces significantly increased apoptosis, fibrosis, and structural alterations in diabetic hearts compared with non-diabetic animals.


Assuntos
Cardiotoxicidade , Diabetes Mellitus , Animais , Apoptose , Doxorrubicina/efeitos adversos , Fibrose , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Monócitos , Estreptozocina/farmacologia , Remodelação Ventricular
5.
Am J Physiol Heart Circ Physiol ; 317(2): H460-H471, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31172809

RESUMO

Doxorubicin (Dox)-induced cardiac side effects are regulated through increased oxidative stress and apoptosis. However, it remains unknown whether Dox induces the specific inflammatory-mediated form of cell death called pyroptosis. The current study is undertaken to determine whether Dox induces pyroptosis in an in vitro model and to test the potential of exosomes derived from embryonic stem cells (ES-Exos) in inhibiting pyroptosis. H9c2 cells were exposed to Dox to generate pyroptosis and then subsequently treated with exosomes to investigate the protective effects of ES-Exos. Mouse embryonic fibroblast-exosomes (MEF-Exos) were used as a cell line control. We confirmed pyroptosis by analyzing the presence of Toll-like receptor 4 (TLR4)-pyrin domain containing-3 (NLRP3) inflammasome that initiates pyroptosis, which was further confirmed with pyroptotic markers caspase-1, IL-1ß, caspase-11, and gasdermin-D. The presence of inflammation was confirmed for proinflammatory cytokines, TNF-α, and IL-6. Our data show that Dox exposure significantly (P < 0.05) increases expression of TLR4, NLRP3, pyroptotic markers (caspase-1, IL-1ß, caspase-11, and gasdermin-D), and proinflammatory cytokines (TNF-α and IL-6) in H9c2 cells. The increased expression of inflammasome, pyroptosis, and inflammation was significantly (P < 0.05) inhibited by ES-Exos. Interestingly, our cell line control, MEF-Exos, did not show any protective effects. Furthermore, our cytokine array data suggest increased anti-inflammatory (IL-4, IL-9, and IL-13) and decreased proinflammatory cytokines (Fas ligand, IL-12, and TNF-α) in ES-Exos, suggesting that anti-inflammatory cytokines might be mediating the protective effects of ES-Exos. In conclusion, our data show that Dox induces pyroptotic cell death in the H9c2 cell culture model and is attenuated via treatment with ES-Exos.NEW & NOTEWORTHY Doxorubicin (Dox)-induced cardiotoxicity is mediated through increased oxidative stress, apoptosis, and necrosis. We report for the first time as per the best of our knowledge that Dox initiates Toll-like receptor 4 and pyrin domain containing-3 inflammasome formation and induces caspase-1-mediated inflammatory pyroptotic cell death in H9c2 cells. Moreover, we establish that inflammation and pyroptosis is inhibited by embryonic stem cell-derived exosomes that could be used as a future therapeutic option to treat Dox-induced cardiotoxicity.


Assuntos
Doxorrubicina/toxicidade , Exossomos/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Piroptose/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Cardiotoxicidade , Linhagem Celular , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos , Transdução de Sinais
6.
Mol Cell Biochem ; 462(1-2): 1-10, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31468244

RESUMO

Diabetic cardiomyopathy is known to involve two forms of cardiac cell death: apoptosis and necrosis. However, it remains unknown whether hyperglycemia-induced apoptosis in the H9c2 cell culture system is inhibited by parasympathetic ganglionic neurons (PGN) derived exosomes (exos). We isolated PGN and sympathetic ganglionic neurons (SGN) from the right stellate ganglion in rats, and derived exos from these sources. H9c2 cells were divided into 4 groups: (1) Control, (2) H9c2 + Glucose (100 mmol/L), (3) H9c2 + Glucose + PGN-exos, and (4) H9c2 + Glucose + SGN-exos. We determined cell proliferation and viability with an MTT assay kit, and assessed apoptotic cell death with TUNEL staining and ELISA. Data were further confirmed by analyzing the presence of pro-apoptotic proteins Caspase-3 and Bax, and anti-apoptotic protein Bcl-2. Glucose exposed H9c2 cells significantly reduced cell viability, which was improved by PGN-exos, but not by SGN-exos. Furthermore, increased apoptosis in hyperglycemia in H9c2 cells was confirmed with TUNEL staining and cell death ELISA which demonstrated significantly (p < 0.05) reduction with PGN-exos treatment, but not with SGN-exos. Moreover, high expression of pro-apoptotic proteins Caspase-3 and Bax was reduced following treatment with PGN-exos; however, SGN-exos were unable to reduce the expression. Significantly reduced anti-apoptotic protein Bcl-2 following glucose treatment was improved with PGN-exos. Therefore, our data suggest that hyperglycemia induces apoptosis in H9c2 cells and decreases cell viability, and that PGN-exos are able to inhibit apoptosis, improve cell viability, and restore levels of anti-apoptotic protein Bcl-2.


Assuntos
Apoptose , Exossomos/metabolismo , Gânglios Parassimpáticos/patologia , Hiperglicemia/patologia , Miócitos Cardíacos/patologia , Neurônios/patologia , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Exossomos/efeitos dos fármacos , Glucose/toxicidade , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/metabolismo
7.
Can J Physiol Pharmacol ; 97(4): 277-286, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30395483

RESUMO

Cardiovascular disease resulting from atypical cardiac structures continues to be a leading health concern despite advancements in diagnostic imaging and surgical techniques. However, the ability to visualize spatial relationships using current technologies remains a challenge. Therefore, 3D modeling has gained significant interest to understand complex and atypical cardiovascular disorders. Moreover, 3D modeling can be personalized and patient-specific. 3D models have been demonstrated to aid surgical planning and simulation, enhance communication among surgeons and patients, optimize medical device design, and can be used as a potential teaching tool in medical schools. In this review, we discuss the key components needed to generate cardiac 3D models. We highlight prevalent structural conditions that have utilized 3D modeling in pre-operative planning. Furthermore, we discuss the current limitations of routine use of 3D models in the clinic as well as future directions for utilization of this technology in the cardiovascular field.


Assuntos
Doenças Cardiovasculares , Imageamento Tridimensional , Impressão Tridimensional , Doenças Cardiovasculares/diagnóstico por imagem , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Humanos
8.
Am J Physiol Heart Circ Physiol ; 315(5): H1236-H1249, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30095997

RESUMO

Doxorubicin (Doxo) is an effective agent commonly used in cancer therapeutics. Unfortunately, Doxo treatment can stimulate cardiomyopathy and subsequent heart failure, limiting the use of this drug. The role of phosphatase and tensin homolog (PTEN) in apoptosis has been documented in Doxo-induced cardiomyopathy (DIC) and heart failure models. However, whether direct inhibition of PTEN attenuates apoptosis, cardiac remodeling, and inflammatory M1 macrophages in the DIC model remains elusive. Therefore, the present study was designed to understand the effects of VO-OHpic (VO), a potent inhibitor of PTEN, in reducing apoptosis and cardiac remodeling. At day 56, echocardiography was performed, which showed that VO treatment significantly ( P < 0.05) improved heart function. Immunohistochemistry, TUNEL, and histological staining were used to determine apoptosis, proinflammatory M1 macrophages, anti-inflammatory M2 macrophages, and cardiac remodeling. Our data show a significant increase in apoptosis, hypertrophy, fibrosis, and proinflammatory M1 macrophages with Doxo treatment, whereas VO treatment significantly reduced apoptosis, adverse cardiac remodeling, and proinflammatory M1 macrophages significantly ( P < 0.05) compared with the Doxo-treated group. Western blot analysis confirmed the reduction of phosphorylated PTEN and increase in phosphorylated AKT protein expression in the Doxo + VO-treated group. Moreover, VO administration increased anti-inflammatory M2 macrophages. Collectively, our data suggest that VO treatment attenuates apoptosis and adverse cardiac remodeling, a process that is mediated through the PTEN/AKT pathway, resulting in improved heart function in DIC. NEW & NOTEWORTHY Doxorubicin-induced cardiomyopathy (DIC) is still a major issue in patients with cancer. These novel findings on the phosphatase and tensin homolog inhibitor VO-OHpic in DIC is the first report, as per the best of our knowledge, that VO-OHpic significantly decreases apoptosis, fibrosis, hypertrophy, adverse cardiac remodeling, and proinflammatory M1 macrophages and increases anti-inflammatory M2 macrophages along with significantly improved cardiac function. VO-OHpic could be a future therapeutic agent for patients with DIC.


Assuntos
Anti-Inflamatórios/farmacologia , Cardiomiopatias/prevenção & controle , Doxorrubicina , Inibidores Enzimáticos/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Compostos Organometálicos/farmacologia , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Função Ventricular Esquerda/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Cardiomegalia/induzido quimicamente , Cardiomegalia/enzimologia , Cardiomegalia/fisiopatologia , Cardiomegalia/prevenção & controle , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/enzimologia , Cardiomiopatias/fisiopatologia , Cardiotoxicidade , Modelos Animais de Doenças , Fibrose , Ventrículos do Coração/enzimologia , Ventrículos do Coração/fisiopatologia , Macrófagos/enzimologia , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Can J Physiol Pharmacol ; 96(7): 647-654, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29842793

RESUMO

Breast cancer is one of the most prevalent forms of cancer in the United States and worldwide. Cancer occurs through the uncontrolled development of new abnormal cell growth. Clinicians and researchers strive to improve diagnostics and treatments in pursuit of remedying breast cancer, while limiting or removing any potential side effects that may arise. Unfortunately, traditional treatments, such as anthracyclines (i.e., doxorubicin), can damage the cardiovascular system. Recent strategies have utilized antibody-based compounds as singular treatments, or in conjunction with other treatments, with the aim to minimize side effects. The human epidermal growth factor receptor 2 (HER2) protein has been the target of numerous antibody-based breast cancer therapies, such as trastuzumab (TZM) and trastuzumab emtansine (T-DM1). This review will discuss the HER2 receptor as a diagnostic marker in targeting breast cancer using the therapeutic agents TZM and T-DM1, as well as discuss the induced cardiac toxicity following TZM and T-DM1 treatments.


Assuntos
Antineoplásicos Imunológicos/efeitos adversos , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Maitansina/análogos & derivados , Receptor ErbB-2/metabolismo , Trastuzumab/efeitos adversos , Ado-Trastuzumab Emtansina , Animais , Biomarcadores Tumorais/antagonistas & inibidores , Neoplasias da Mama/patologia , Cardiotoxicidade/epidemiologia , Cardiotoxicidade/etiologia , Feminino , Coração/efeitos dos fármacos , Humanos , Maitansina/efeitos adversos , Miocárdio/metabolismo , Prognóstico , Receptor ErbB-2/antagonistas & inibidores
10.
Can J Physiol Pharmacol ; 96(3): 304-307, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28926719

RESUMO

Doxorubicin (Dox) is an effective anticancer drug. Unfortunately, it causes cardiac and muscle toxicity due to increased oxidative stress and inflammation; however, it remains unknown whether Dox induces "pyroptosis" - an inflammation-mediated cell death. We investigated whether Dox induces pyroptosis in mouse soleus muscle (Sol 8) cells in vitro and to show the protective effect of embryonic stem cell exosomes (ES-exos) on pyroptosis. Dox and inflammation-induced in vitro model was generated. Pyroptosis was confirmed using immunohistochemistry (with putative markers caspase-1, IL-1ß, and pro-inflammatory cytokine IL-18) and Western blotting of caspase-1 and IL-1ß. The results show significant increase in the expression of caspase-1, IL-1ß, and IL-18 following treatment with Dox, which was inhibited by ES-exos but not mouse embryonic fibroblast exosomes. Moreover, GW4869 compound inhibited functional activity of ES-exos, suggesting these vesicles are key players in the inhibition of pyroptosis. These results suggest that Dox induces inflammatory pyroptosis in Sol 8 cells, which is attenuated by ES-exos in vitro.


Assuntos
Doxorrubicina/efeitos adversos , Células-Tronco Embrionárias/citologia , Exossomos/metabolismo , Células Musculares/citologia , Células Musculares/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Animais , Linhagem Celular , Inflamação/induzido quimicamente , Inflamação/patologia , Camundongos
11.
Mol Cell Biochem ; 425(1-2): 77-84, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27804049

RESUMO

Fibroblast growth factors (FGFs) comprise a large family of signaling molecules that involve cell patterning, mobilization, differentiation, and proliferation. Various FGFs, including FGF-1, FGF-2, and FGF-5, have been shown to play a role in cytoprotection during adverse cardiac events; however, whether FGF-8 is a cytoprotective remains unclear. The current study was designed to evaluate the effect of FGF-8 treatment on oxidative stress-induced apoptosis in H9c2 cells. Cells were divided into three groups: control, H2O2 (400 µm H2O2), and H2O2 + FGF-8 (4 ng/ml FGF-8). Our results suggest apoptosis was significantly (p < 0.05) enhanced in the H2O2 group relative to control. Moreover, a significant (p < 0.05) decline in apoptosis was observed in the H2O2 + FGF-8 group compared to H2O2-treated cells as evidenced by TUNEL staining, a cell death detection ELISA, and cell viability. Levels of downstream apoptotic mediators, caspase-3 and caspase-9, were significantly (p < 0.05) upregulated following H2O2 treatment but were abrogated following FGF-8 application. Expression levels of Forkhead box protein O1 (FoxO-1), MnSOD, catalase, pAKT, and p-mTOR were significantly (p < 0.05) reduced in the H2O2 group (p < 0.05). Notably, these levels were significantly (p < 0.05) reversed following FGF-8 treatment. Our data, for the first time, suggest FGF-8 is an anti-apoptotic mediator in oxidative-stressed H9c2 cells. Furthermore, our data demonstrate that apoptotic inhibition by FGF-8 is consequent to FoxO-1 oxidative detoxification as well as augmentation to the PI3K/AKT cell survival pathway.


Assuntos
Apoptose/efeitos dos fármacos , Fator 8 de Crescimento de Fibroblasto/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
12.
Can J Physiol Pharmacol ; 95(3): 288-294, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28238274

RESUMO

The current study investigates whether inhibiting the Notch-1 signaling pathway in primary human monocytes enhances M2 macrophage differentiation. We generated a primary human monocyte cell culture model to understand the effect of the Notch-1 signaling pathway. Monocytes were treated with Notch-1 inhibitors DAPT or siRNA. Our data show that there was a significant increase in the M1 macrophage population demonstrated by iNOS marker in the primary human monocytes treated with apoptotic-conditioned medium (ACM). Next, the levels of pro-inflammatory cytokines IL-6 and MCP-1, as well as TNF-α, increased in ACM media (p < 0.05). Furthermore, M1 macrophages and pro-inflammatory cytokines were reduced following DAPT or siRNA treatment. Comparatively, there was a significant increase in M2 macrophages, as demonstrated by an increase in CD206 and arginase-1 positive cells treated with DAPT or siRNA (p < 0.05). Furthermore, a significant increase in the associated anti-inflammatory cytokines IL-10 and IL-1RA was also observed with respect to control groups (p < 0.05). We conclude that blocking the Notch-1 pathway with DAPT or siRNA attenuates pro-inflammatory cytokines, enhances M2 macrophage differentiation, and increases anti-inflammatory cytokines in primary human monocytes. As a result, Notch-1 pathway inhibition has potential therapeutic applications of inflammatory disease.


Assuntos
Diferenciação Celular , Macrófagos/metabolismo , Monócitos/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , Apoptose , Arginase/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Diaminas/farmacologia , Humanos , Mediadores da Inflamação/metabolismo , Lectinas Tipo C/metabolismo , Macrófagos/efeitos dos fármacos , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/patologia , Óxido Nítrico Sintase Tipo II/metabolismo , Fenótipo , Cultura Primária de Células , Interferência de RNA , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/genética , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia , Transfecção
13.
Mol Cell Biochem ; 406(1-2): 101-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25920449

RESUMO

Cardiac cell regeneration from endogenous cardiac stem cells (CSCs) following MI is rather low. Therefore, identifying mechanisms to boost endogenous CSC activation and participation in cardiac repair appears to be the most promising strategy for MI patients. We previously engineered tissue inhibitor of metalloproteinases-1 (TIMP-1) overexpressing embryonic stem (ES-TIMP-1) cells and transplanted them into the infarcted murine heart. Collected data demonstrated that TIMP-1 enhanced transplanted ES cell engraftment, survival and differentiation into cardiac myocytes post-transplantation. Therefore, we postulated that there may be a new stem cell population present in the heart that is regulated by extracellular protein TIMP-1. Furthermore, we hypothesized that this cell population has a potential for cell proliferation and differentiation into cardiac cell types. Therefore, we isolated CSCs from 4 weeks old C57BL/6 mice and cultured them in vitro in presence of ESCM, ES-TIMP-1-CM or TIMP-1. Our immunostaining data demonstrated the existence of a novel CSC subpopulation, CD63(+ve)/c-kit(+ve). When treated with TIMP-1, these cells showed significantly (p < 0.05) increased proliferation rates compared to control cells, enhanced TIMP-1 receptor (CD63), along with improved expression of phospho and total ß-catenin proteins as demonstrated by Western blot analysis. Next, we demonstrate significantly (p < 0.05) improved cardiac myocyte, vascular smooth muscle cell, and endothelial cell differentiation. Furthermore, our RT-PCR data shows increase in cardiac gene (GATA-4, Mef2C, and Nkx-2.5) expression when compared to ESCM and control cells. Collectively, these data, for the first time, establish the existence of a new CD63(+ve)/c-kit(+ve) CSC subpopulation that has a significant potential for proliferation and differentiation into cardiac cell types once stimulated with TIMP-1.


Assuntos
Células-Tronco Adultas/fisiologia , Miocárdio/citologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Tetraspanina 30/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Separação Celular , Camundongos Endogâmicos C57BL , Inibidor Tecidual de Metaloproteinase-1/fisiologia , Ativação Transcricional
14.
Am J Physiol Heart Circ Physiol ; 307(5): H762-72, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24993041

RESUMO

The main objective of this study was to determine whether or not monocyte infiltration occurs in the prediabetic (PD) heart and its role in PD cardiomyopathy. We hypothesized that the PD heart is significantly populated with monocytes and that bone morphogenetic protein (BMP)-7, a novel mediator of monocyte polarization, activates infiltrated monocytes into anti-inflammatory M2 macrophages, thereby inhibiting apoptosis and fibrosis and improving cardiac function. C57Bl6 mice were assigned to control, PD, or PD + BMP-7 groups. PD and PD + BMP-7 groups were administered streptozotocin (50 mg/kg), whereas control animals received sodium citrate buffer. Afterward, the PD + BMP-7 group was administered BMP-7 (200 µg/kg) for 3 days. Our data showed significantly increased infiltrated monocytes and associated pro-inflammatory cytokines, adverse cardiac remodeling, and heart dysfunction in the PD group (P < 0.05). Interestingly, M2 macrophage differentiation and associated anti-inflammatory cytokines were enhanced and there were reduced adverse cardiac remodeling and improved cardiac function in the PD + BMP-7 group (P < 0.05). In conclusion, our data suggest that PD cardiomyopathy is associated with increased monocyte infiltration and released proinflammatory cytokines, which contributes to adverse cardiac remodeling and cardiac dysfunction. Moreover, we report that BMP-7 possesses novel therapeutic potential in its ability to differentiate monocytes into M2 macrophages and confer cardiac protection in the PD heart.


Assuntos
Proteína Morfogenética Óssea 7/farmacologia , Cardiomiopatias Diabéticas/tratamento farmacológico , Células Precursoras de Monócitos e Macrófagos/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Apoptose , Proteína Morfogenética Óssea 7/uso terapêutico , Diferenciação Celular , Movimento Celular , Cardiomiopatias Diabéticas/patologia , Fibrose/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Células Precursoras de Monócitos e Macrófagos/citologia , Células Precursoras de Monócitos e Macrófagos/fisiologia
15.
Am J Physiol Heart Circ Physiol ; 307(11): H1634-42, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25260616

RESUMO

Macrophage polarization is emerging as an important area of research for the development of novel therapeutics to treat inflammatory diseases. Within the current study, the role of Notch1R in macrophage differentiation was investigated as well as downstream effects in THP-1 monocytes cultured in "inflammation mimicry" media. Interference of Notch signaling was achieved using either the pharmaceutical inhibitor DAPT or Notch1R small interfering RNA (siRNA), and Notch1R expression, macrophage phenotypes, and anti- and proinflammatory cytokine expression were evaluated. Data presented show that Notch1R expression on M1 macrophages as well as M1 macrophage differentiation is significantly elevated during cellular stress (P < 0.05). However, under identical culture conditions, interference to Notch signaling via Notch1R inhibition mitigated these results as well as promoted M2 macrophage differentiation. Moreover, when subjected to cellular stress, macrophage secretion of proinflammatory cytokines was significantly heightened (P < 0.05). Importantly, Notch1R inhibition not only diminished proinflammatory cytokine secretion but also enhanced anti-inflammatory protein release (P < 0.05). Our data suggest that Notch1R plays a pivotal role in M1 macrophage differentiation and heightened inflammatory responses. Therefore, we conclude that inhibition of Notch1R and subsequent downstream signaling enhances monocyte to M2 polarized macrophage outcomes and promotes anti-inflammatory mediation during cellular stress.


Assuntos
Macrófagos/fisiologia , Monócitos/fisiologia , Receptor Notch1/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Polaridade Celular/fisiologia , Meios de Cultivo Condicionados , Citocinas/metabolismo , Humanos , Mediadores da Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , RNA Interferente Pequeno/genética , Receptor Notch1/antagonistas & inibidores , Receptor Notch1/genética , Transdução de Sinais/efeitos dos fármacos
16.
Front Immunol ; 15: 1348043, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38390337

RESUMO

Introduction: Diabetes is a debilitating disease that leads to complications like cardiac dysfunction and heart failure. In this study, we investigated the pathophysiology of diabetes-induced cardiac dysfunction in mice with dyslipidemia. We hypothesize diabetes in ApoE knockout (ApoE-/-) mice induces cardiac dysfunction by increasing inflammation and necroptosis. Methods: ApoE-/- mice were divided into experimental groups: Control, Streptozotocin (STZ), STZ + MSC-Exo (mesenchymal stem cell-derived exosomes), and STZ+MEF-Exo (Mouse embryonic fibroblast derived exosomes). At Day 42, we assessed cardiac function, collected blood and heart tissues. Heart tissue samples were analyzed for inflammation, necroptosis, signaling mechanism, hypertrophy and adverse structural remodeling using histology, immunohistochemistry, western blotting, RT-PCR, cytokine array and TF array. Results and Discussion: STZ treated ApoE-/- mice developed diabetes, with significantly (p<0.05) increased blood glucose and body weight loss. These mice developed cardiac dysfunction with significantly (p<0.05) increased left ventricular internal diameter end diastole and end systole, and decreased ejection fraction, and fractional shortening. We found significant (p<0.05) increased expression of inflammatory cytokines TNF- a, IL-6, IL-1a, IL-33 and decreased IL-10 expression. Diabetic mice also exhibited significantly (p<0.05) increased necroptosis marker expression and infiltration of inflammatory monocytes and macrophages. MSC-Exos treated mice showed recovery of diabetes associated pathologies with significantly reduced blood glucose, recovered body weight, increased IL-10 secretion and M2 polarized macrophages in the heart. These mice showed reduced TAK1-pJNK-NFKB inflammation associated expression and improved cardiac function with significantly reduced cardiac hypertrophy and fibrosis compared to diabetic mice. Treatment with MEF-Exos did not play a significant role in attenuating diabetes-induced cardiomyopathy as these treatment mice presented with cardiac dysfunction and underlying pathologies observed in STZ mice. Conclusion: Thus, we conclude that cardiac dysfunction develops in diabetic ApoE-/- mice, arising from inflammation, necroptosis, and adverse tissue remodeling, which is ameliorated by MSC-Exos, a potential therapeutic for diabetes-induced cardiomyopathy.


Assuntos
Diabetes Mellitus Experimental , Cardiomiopatias Diabéticas , Exossomos , Cardiopatias , Animais , Camundongos , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Glicemia/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Experimental/metabolismo , Cardiomiopatias Diabéticas/etiologia , Cardiomiopatias Diabéticas/patologia , Exossomos/metabolismo , Fibroblastos/patologia , Cardiopatias/metabolismo , Inflamação/metabolismo , Interleucina-10/metabolismo , Camundongos Knockout para ApoE , Necroptose
17.
Pharmaceuticals (Basel) ; 17(1)2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38256928

RESUMO

Doxorubicin (DOX) is an incessantly used chemotherapeutic drug that can cause detrimental dose-dependent effects such as cardiotoxicity and congestive heart failure. Hence, there is a need to discover innovative therapeutic approaches to counteract DOX-induced cardiotoxicity (DIC). MSC-Exos have shown to reduce apoptosis and cardiac fibrosis and promote cardiomyocyte proliferation in myocardial infracted mice. However, the effect of MSC-Exos on ameliorating DOX-induced pyroptosis has not been investigated. In this current study, H9c2 were first exposed to DOX to stimulate pyroptosis, followed by subsequent treatment with MSC-Exos, with further analysis performed through immunocytochemistry, western blotting, and RT-PCR. Our data depicted that post-treatment with MSC-Exos significantly (p < 0.05) reduced the HMGB1/TLR4 axis, inflammasome formation (NLRP3), pyroptotic markers (caspase-1, IL-1ß, and IL-18), and the pyroptotic executioner (GSDMD) in DOX-treated H9c2 cells. In conclusion, our data show that MSC-Exos attenuates inflammation-induced pyroptosis in our in vitro DIC model. Our findings indicate that MSC-Exos may serve as a promising therapeutic intervention for mitigating DIC, as they maintain the therapeutic capabilities of MSCs while circumventing the drawbacks associated with traditional stem cell therapy.

18.
Mol Pharm ; 10(9): 3425-32, 2013 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-23879836

RESUMO

Recent evidence suggests transplanted stem cells improve left ventricular function in diabetic induced cardiomyopathy (DICM). However, little is known about the mechanisms by which induced pluripotent stem (iPS) cells or factors released from these cells inhibit adverse cardiac remodeling in DICM. The present study was designed to determine molecular mediators and pathways regulated by transplanted iPS cells and their conditioned media (CM) in DICM. Animals were divided into four experimental groups such as control, streptozotocin (STZ), STZ+iPS-CM, and STZ+iPS cells. Experimental diabetes was induced in C57BL/6 mice by intraperitoneal STZ injections (100 mg/kg body weight for 2 consecutive days). Following STZ injections, iPS cells or CM was given intravenously for 3 consecutive days. Animals were humanely killed, and hearts were harvested at D14. Animals transplanted with iPS cells or CM demonstrated a significant reduction in apoptosis, mediated by Akt upregulation and ERK1/2 downregulation, and inhibition of interstitial fibrosis via MMP-9 suppression compared with the STZ group. Oxidative stress was significantly hindered in iPS cell and CM groups as evidenced by diminished pro-oxidant expression and enhanced antioxidant (catalase and MnSOD) concentration. Echocardiography data suggest a significant improvement in cardiac function in cells and CM groups in comparison to STZ. In conclusion, our data strongly suggest that iPS cells and CM attenuate oxidative stress and associated apoptosis and fibrosis. Moreover, we also suggest that increased antioxidant levels, decreased adverse cardiac remodeling, and improved cardiac function is mediated by iPS CM and cells in DICM through multiple autocrine and paracrine mechanisms.


Assuntos
Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/terapia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Animais , Apoptose/fisiologia , Linhagem Celular , Sobrevivência Celular/fisiologia , Ensaio de Imunoadsorção Enzimática , Feminino , Células-Tronco Pluripotentes Induzidas/metabolismo , Peróxidos Lipídicos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo/fisiologia , Ratos
19.
Mol Cell Biochem ; 382(1-2): 113-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23780512

RESUMO

Recent studies suggest that Klf5 is required to maintain embryonic stem (ES) cells in an undifferentiated state. However, whether Klf5 can be inactivated by novel fusion technology of zinc finger nucleases (ZFN) has never before been examined. Therefore, we used ZFN technology to target the Klf5 gene in mouse ES cells, and examined the effects of the Klf5 gene on the expression of pluripotency-related genes, Oct3/4, Nanog, and Sox2 and on the self-renewal of ES cells. In Klf5-ZFN-transfected cells, expression of the Klf5 mRNA was downregulated by ~80% compared to the control. Furthermore, expression of the Oct3/4 and Nanog mRNAs was significantly decreased in the Klf5-ZFN-targeted cells. RT-PCR analysis, however, showed no significant change in the level of Sox2 mRNA, but a decreased trend was evident in the Klf5-ZFN-targeted cells. Moreover, we observed the spontaneous differentiation of Klf5-ZFN-transfected cells and quantitative analysis revealed a significant decrease in colony formation in Klf5-ZFN-transfected cells. In conclusion, our data suggest that ZFN methodology is an effective approach to target the Klf5 gene and that Klf5 plays an important role in the maintenance of ES cell self-renewal.


Assuntos
Regulação para Baixo/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Endonucleases/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Células-Tronco Pluripotentes/metabolismo , Dedos de Zinco , Animais , Diferenciação Celular/genética , Forma Celular/genética , Ensaio de Unidades Formadoras de Colônias , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transfecção
20.
Mol Cell Biochem ; 372(1-2): 267-73, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23054191

RESUMO

Recent studies suggest that disturbed blood flow-induced shear stress can induce atherosclerosis (ATH) in humans and animals without a high fat diet. Therefore, we hypothesize that partial ligation of the left carotid artery can generate disturbed blood flow and shear stress and would lead to ATH in a predisposed genetic model of Apo E(-/-) mice. The partial left carotid artery model was generated by ligating three out of four branches of the left carotid artery compared with controls which experienced similar surgery conditions but no ligation. Animals were sacrificed 2 weeks post-ligation and examined for plaque formation, infiltration of leukocytes, pro-inflammatory immune response, and blood flow velocity. Our findings suggest a significant (p < 0.05) increase in plaque formation and lipid deposition in the partial ligated animals compared with controls, confirmed with hematoxylin and eosin and oil red O staining. Furthermore, there was a significant (p < 0.05) increase in the number of M1 macrophages and release of pro-inflammatory cytokines, IL-6 and TNFα, as compared with the control. Moreover, partial ligated carotid arteries demonstrated disturbed blood flow as their systolic velocity was significantly reduced. In conclusion, our data suggest that partial ligation of the left carotid artery induces disturbed flow and shear stress in the predisposed genetic model of Apo E(-/-) mice and leads to significantly developed ATH. Similarities to clinical patients who develop ATH independent of a high fat diet show that this could be a potential animal model to examine various parameters in ATH.


Assuntos
Aterosclerose/metabolismo , Artéria Carótida Primitiva/metabolismo , Estenose das Carótidas/metabolismo , Metabolismo dos Lipídeos , Macrófagos/imunologia , Monócitos/imunologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/imunologia , Aterosclerose/fisiopatologia , Fenômenos Biomecânicos , Velocidade do Fluxo Sanguíneo , Artéria Carótida Primitiva/imunologia , Artéria Carótida Primitiva/patologia , Estenose das Carótidas/imunologia , Estenose das Carótidas/fisiopatologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/metabolismo , Ligadura , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Monócitos/metabolismo , Placa Aterosclerótica/imunologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/fisiopatologia , Estresse Fisiológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA