Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 183(2): 490-502.e18, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-33002410

RESUMO

The non-receptor protein tyrosine phosphatase (PTP) SHP2, encoded by PTPN11, plays an essential role in RAS-mitogen-activated protein kinase (MAPK) signaling during normal development. It has been perplexing as to why both enzymatically activating and inactivating mutations in PTPN11 result in human developmental disorders with overlapping clinical manifestations. Here, we uncover a common liquid-liquid phase separation (LLPS) behavior shared by these disease-associated SHP2 mutants. SHP2 LLPS is mediated by the conserved well-folded PTP domain through multivalent electrostatic interactions and regulated by an intrinsic autoinhibitory mechanism through conformational changes. SHP2 allosteric inhibitors can attenuate LLPS of SHP2 mutants, which boosts SHP2 PTP activity. Moreover, disease-associated SHP2 mutants can recruit and activate wild-type (WT) SHP2 in LLPS to promote MAPK activation. These results not only suggest that LLPS serves as a gain-of-function mechanism involved in the pathogenesis of SHP2-associated human diseases but also provide evidence that PTP may be regulated by LLPS that can be therapeutically targeted.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Células A549 , Animais , Criança , Pré-Escolar , Feminino , Mutação com Ganho de Função/genética , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Células-Tronco Embrionárias Murinas , Mutação/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Transdução de Sinais , Domínios de Homologia de src/genética
2.
Helicobacter ; 27(4): e12895, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35437862

RESUMO

BACKGROUND: Macrophages, as innate immune cells, were reported to participate in the pathogenesis of Helicobacter pylori (H. pylori)-induced gastritis. However, the role and mechanism of macrophage dysfunction in H. pylori-associated pediatric gastritis remain unclear. MATERIALS AND METHODS: An RNA-sequencing assay was used to examine the differential gene expression in normal gastric antrum, non-H. pylori-infected tissue, and H. pylori-infected pediatric gastritis tissue. qPCR assays were applied to verify the expression of target genes. HE staining was performed to identify the occurrence of inflammation in the normal gastric antrum, non-H. pylori-infected tissue, and H. pylori-infected pediatric gastritis tissue. Western blotting was used to measure the expression of SHP2 in pediatric gastritis tissue. The metabolic profile of macrophages was determined via Seahorse metabolic analysis. Flow cytometry analysis was used to examine the level of reactive oxygen species (ROS). RESULTS: We found that H. pylori -infected gastritis tissue exhibited many differentially expressed genes (DEGs) compared to gastritis tissue without H. pylori infection. Moreover, H. pylori -infected gastritis tissue showed many DEGs annotated with an overactive immune response. We identified that tyrosine-protein phosphatase nonreceptor type 11 (PTPN11), which encodes SHP2, was significantly increased in macrophages of H. pylori -infected gastritis tissue. Furthermore, we revealed that SHP2 could activate the glycolytic function of macrophages to promote H. pylori -induced inflammation. The transcription factor SPI1 , as the downstream molecule of SHP2, could be responsible for the regulation of metabolism-associated gene expression and inflammation. CONCLUSION: Our study illustrated the molecular landscape of H. pylori-infected gastritis tissue in children and suggested that the SHP2/SPI1axis could be a novel therapeutic target in H. pylori-induced pediatric gastritis.


Assuntos
Gastrite , Infecções por Helicobacter , Helicobacter pylori , Criança , Mucosa Gástrica/patologia , Gastrite/patologia , Glicólise , Humanos , Inflamação/patologia , Macrófagos/metabolismo
3.
Cancer Cell Int ; 21(1): 337, 2021 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-34217295

RESUMO

BACKGROUND: Additional epidermal growth factor receptor (EGFR) mutations confer the drug resistance to generations of EGFR targeted tyrosine kinase inhibitor (EGFR-TKI), posing a major challenge to developing effective treatment of lung adenocarcinoma (LUAD). The strategy of combining EGFR-TKI with other synergistic or sensitizing therapeutic agents are considered a promising approach in the era of precision medicine. Moreover, the role and mechanism of SHP2, which is involved in cell proliferation, cytokine production, stemness maintenance and drug resistance, has not been carefully explored in lung adenocarcinoma (LUAD). METHODS: To evaluate the impact of SHP2 on the efficacy of EGFR T790M mutant LUAD cells to Osimertinib, SHP2 inhibition was tested in Osimertinib treated LUAD cells. Cell proliferation and stemness were tested in SHP2 modified LUAD cells. RNA sequencing was performed to explore the mechanism of SHP2 promoted stemness. RESULTS: This study demonstrated that high SHP2 expression level correlates with poor outcome of LUAD patients, and SHP2 expression is enriched in Osimertinib resistant LUAD cells. SHP2 inhibition suppressed the cell proliferation and damaged the stemness of EGFR T790M mutant LUAD. SHP2 facilitates the secretion of CXCL8 cytokine from the EGFR T790M mutant LUAD cells, through a CXCL8-CXCR1/2 positive feedback loop that promotes stemness and tumorigenesis. Our results further show that SHP2 mediates CXCL8-CXCR1/2 feedback loop through ERK-AKT-NFκB and GSK3ß-ß-Catenin signaling in EGFR T790M mutant LUAD cells. CONCLUSIONS: Our data revealed that SHP2 inhibition enhances the anti-cancer effect of Osimertinib in EGFR T790M mutant LUAD by blocking CXCL8-CXCR1/2 loop mediated stemness, which may help provide an alternative therapeutic option to enhance the clinical efficacy of osimertinib in EGFR T790M mutant LUAD patients.

4.
Am J Emerg Med ; 40: 148-158, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32063427

RESUMO

OBJECTIVE: To develop a novel model for predicting Emergency Department (ED) prolonged length of stay (LOS) patients upon triage completion, and further investigate the benefit of a targeted intervention for patients with prolonged ED LOS. MATERIALS AND METHODS: A two-step model to predict patients with prolonged ED LOS (>16 h) was constructed. This model was initially used to predict ED resource usage and was subsequently adapted to predict patient ED LOS based on the number of ED resources using binary logistic regressions and was validated internally with accuracy. Finally, a discrete event simulation was used to move patients with predicted prolonged ED LOS directly to a virtual Clinical Decision Unit (CDU). The changes of ED crowding status (Overcrowding, Crowding, and Not-Crowding) and savings of ED bed-hour equivalents were estimated as the measures of the efficacy of this intervention. RESULTS: We screened a total of 123,975 patient visits with final enrollment of 110,471 patient visits. The overall accuracy of the final model predicting prolonged patient LOS was 67.8%. The C-index of this model ranges from 0.72 to 0.82. By implementing the proposed intervention, the simulation showed a 12% (1044/8760) reduction of ED overcrowded status - an equivalent savings of 129.3 ED bed-hours per day. CONCLUSIONS: Early prediction of prolonged ED LOS patients and subsequent (simulated) early CDU transfer could lead to more efficiently utilization of ED resources and improved efficacy of ED operations. This study provides evidence to support the implementation of this novel intervention into real healthcare practice.


Assuntos
Serviço Hospitalar de Emergência , Tempo de Internação/estatística & dados numéricos , Adolescente , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Estudos Retrospectivos , Medição de Risco , Triagem
5.
Sensors (Basel) ; 21(12)2021 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204575

RESUMO

Advanced heart monitors, especially those enabled by the Internet of Health Things (IoHT), provide a great opportunity for continuous collection of the electrocardiogram (ECG), which contains rich information about underlying cardiac conditions. Realizing the full potential of IoHT-enabled cardiac monitoring hinges, to a great extent, on the detection of disease-induced anomalies from collected ECGs. However, challenges exist in the current literature for IoHT-based cardiac monitoring: (1) Most existing methods are based on supervised learning, which requires both normal and abnormal samples for training. This is impractical as it is generally unknown when and what kind of anomalies will occur during cardiac monitoring. (2) Furthermore, it is difficult to leverage advanced machine learning approaches for information processing of 1D ECG signals, as most of them are designed for 2D images and higher-dimensional data. To address these challenges, a new sensor-based unsupervised framework is developed for IoHT-based cardiac monitoring. First, a high-dimensional tensor is generated from the multi-channel ECG signals through the Gramian Angular Difference Field (GADF). Then, multi-linear principal component analysis (MPCA) is employed to unfold the ECG tensor and delineate the disease-altered patterns. Obtained principal components are used as features for anomaly detection using machine learning models (e.g., deep support vector data description (deep SVDD)) as well as statistical control charts (e.g., Hotelling T2 chart). The developed framework is evaluated and validated using real-world ECG datasets. Comparing to the state-of-the-art approaches, the developed framework with deep SVDD achieves superior performances in detecting abnormal ECG patterns induced by various types of cardiac disease, e.g., an F-score of 0.9771 is achieved for detecting atrial fibrillation, 0.9986 for detecting right bundle branch block, and 0.9550 for detecting ST-depression. Additionally, the developed framework with the T2 control chart facilitates personalized cycle-to-cycle monitoring with timely detected abnormal ECG patterns. The developed framework has a great potential to be implemented in IoHT-enabled cardiac monitoring and smart management of cardiac health.


Assuntos
Fibrilação Atrial , Eletrocardiografia , Coração , Humanos , Internet , Monitorização Fisiológica
6.
Cancer Cell Int ; 19: 354, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31889906

RESUMO

BACKGROUND: miR-100 has been reported to closely associate with gastric cancer (GC) initiation and progression. However, the underlying mechanism of miR-100-3p in GC is still largely unclear. In this study, we intend to study how miR-100-3p regulates GC malignancy. METHODS: The expression levels of miR-100-3p in vitro (GES-1 and GC cell lines) and in vivo (cancerous and normal gastric tissues) were examined by quantitative real-time PCR (qRT-PCR). MTT and PE/Annexin V analyses were responsible for measurement of the effects of miR-100-3p on GC cell proliferation and apoptosis. Transwell assay with or without matrigel was used to examine the capacity of migration and invasion in GC cells. The interaction of miR-100-3p with bone morphogenetic protein receptor 2 (BMPR2) was confirmed through transcriptomics analysis and luciferase reporter assay. qRT-PCR and Western blot analyses were applied to determine the expression of ERK/AKT and Bax/Bcl2/Caspase3, which were responsible for the dysfunction of miR-100-3p. RESULTS: miR-100-3p was down-regulated in GC cell lines and cancerous tissues, and was negatively correlated with BMPR2. Loss of miR-100-3p promoted tumor growth and BMPR2 expression. Consistently, the effects of miR-100-3p inhibition on GC cells were partially neutralized by knockdown of BMPR2. Over-expression of miR-100-3p simultaneously inhibited tumor growth and down-regulated BMPR2 expression. Consistently, over-expression of BMPR2 partially neutralized the effects of miR-100-3p over-expression. Further study demonstrated that BMPR2 mediated the effects downstream of miR-100-3p, which might indirectly regulate ERK/AKT and Bax/Bcl2/Caspase3 signaling pathways. CONCLUSION: miR-100-3p acted as a tumor-suppressor miRNA that down-regulated BMPR2, which consequently inhibited the ERK/AKT signaling and activated Bax/Bcl2/Caspase3 signaling. This finding provided novel insights into GC and could contribute to identify a new diagnostic and therapeutic target.

7.
Am J Physiol Renal Physiol ; 315(6): F1592-F1600, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30089032

RESUMO

Myogenic contraction of renal arterioles is an important regulatory mechanism for renal blood flow autoregulation. We have previously demonstrated that integrin-mediated mechanical force increases the occurrence of Ca2+ sparks in freshly isolated renal vascular smooth muscle cells (VSMCs). To further test whether the generation of Ca2+ sparks is a downstream signal of mechanotransduction in pressure-induced myogenic constriction, the relationship between Ca2+ sparks and transmural perfusion pressure was investigated in intact VSMCs of pressurized rat afferent arterioles. Spontaneous Ca2+ sparks were found in VSMCs when afferent arterioles were perfused at 80 mmHg. The spark frequency was significantly increased when perfusion pressure was increased to 120 mmHg. A similar increase of spark frequency was also observed in arterioles stimulated with ß1-integrin-activating antibody. Moreover, spark frequency was significantly higher in arterioles of spontaneous hypertensive rats at 80 and 120 mmHg. Spontaneous membrane current recorded using whole cell perforated patch in renal VSMCs showed predominant activity of spontaneous transient inward currents instead of spontaneous transient outward currents when holding potential was set close to physiological resting membrane potential. Real-time PCR and immunohistochemistry confirmed the expression of Ca2+-activated Cl- channel (ClCa) TMEM16A in renal VSMCs. Inhibition of TMEM16A with T16Ainh-A01 impaired the pressure-induced myogenic contraction in perfused afferent arterioles. Our study, for the first time to our knowledge, detected Ca2+ sparks in VSMCs of intact afferent arterioles, and their frequencies were positively modulated by the perfusion pressure. Our results suggest that Ca2+ sparks may couple to ClCa channels and trigger pressure-induced myogenic constriction via membrane depolarization.


Assuntos
Anoctamina-1/metabolismo , Pressão Arterial , Sinalização do Cálcio , Hipertensão/metabolismo , Rim/irrigação sanguínea , Mecanotransdução Celular , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Vasoconstrição , Animais , Anoctamina-1/genética , Arteríolas/metabolismo , Arteríolas/fisiopatologia , Modelos Animais de Doenças , Hipertensão/genética , Hipertensão/fisiopatologia , Masculino , Potenciais da Membrana , Músculo Liso Vascular/fisiopatologia , Ratos Endogâmicos SHR , Ratos Sprague-Dawley
8.
Am J Transl Res ; 16(1): 255-271, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38322576

RESUMO

OBJECTIVES: Heterotopic ossification (HO), whether hereditary or traumatic, refers to the abnormal formation of bone in extraskeletal sites, often triggered by inflammation or flare-ups. Unfortunately, there are currently no effective treatments for HO. Metformin is well-known for its anti-diabetic, anti-inflammatory, anti-aging, and anti-cancer effects. However, its potential role in treating HO remains uncertain. METHODS: Metformin was dissolved into water and given to mice. All the mice in this study were examined by microCT and myeloid cell quantification using flow cytometry. Complex activity kit was used to examine the activity of mitochondrial complexes of myeloid cells. RESULTS: In this study, we discovered that metformin effectively inhibits genetic and traumatic HO formation and progression. Additionally, we observed a significant increase in myeloid cells in the genetic and traumatic HO mouse model compared to uninjured mice. Notably, metformin specifically reduced the infiltration of myeloid cells into the injured sites of the genetic and traumatic HO model mice. Further investigations revealed that metformin targets mitochondrial complex I and suppresses mitochondrial metabolism in myeloid cells. CONCLUSION: These findings suggest that metformin suppresses HO development by potentially downregulating the mitochondrial metabolism of myeloid cells, offering a promising therapeutic option for HO treatment.

9.
JCI Insight ; 9(8)2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-38451719

RESUMO

Mesenchymal stem cells (MSCs), suffering from diverse gene hits, undergo malignant transformation and aberrant osteochondral differentiation. Src homology region 2-containing protein tyrosine phosphatase 2 (SHP2), a nonreceptor protein tyrosine phosphatase, regulates multicellular differentiation, proliferation, and transformation. However, the role of SHP2 in MSC fate determination remains unclear. Here, we showed that MSCs bearing the activating SHP2E76K mutation underwent malignant transformation into sarcoma stem-like cells. We revealed that the SHP2E76K mutation in mouse MSCs led to hyperactive mitochondrial metabolism by activating mitochondrial complexes I and III. Inhibition of complexes I and III prevented hyperactive mitochondrial metabolism and malignant transformation of SHP2E76K MSCs. Mechanistically, we verified that SHP2 underwent liquid-liquid phase separation (LLPS) in SHP2E76K MSCs. SHP2 LLPS led to its dissociation from complexes I and III, causing their hyperactivation. Blockade of SHP2 LLPS by LLPS-defective mutations or allosteric inhibitors suppressed complex I and III hyperactivation as well as malignant transformation of SHP2E76K MSCs. These findings reveal that complex I and III hyperactivation driven by SHP2 LLPS promotes malignant transformation of SHP2E76K MSCs and suggest that inhibition of SHP2 LLPS could be a potential therapeutic target for the treatment of activated SHP2-associated cancers.


Assuntos
Transformação Celular Neoplásica , Células-Tronco Mesenquimais , Mitocôndrias , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Células-Tronco Mesenquimais/metabolismo , Animais , Camundongos , Mitocôndrias/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Humanos , Mutação , Diferenciação Celular , Separação de Fases
10.
Oncol Lett ; 25(5): 176, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37033103

RESUMO

Breast cancer is the most common malignancy and ranks second among the causes of tumor-associated death in females. The recurrence and drug resistance of breast cancer are intractable due to the presence of breast cancer stem cells (BCSCs), which are adequate to initiate tumor formation and refractory to conventional remedies. Runt-related transcription factor 2 (RUNX2), a pivotal transcription factor in mammary gland and bone development, has also been related to metastatic cancer and BCSCs. State-of-the-art research has indicated the retention of RUNX2 expression in a more invasive subtype of breast cancer, and in particular, triple-negative breast cancer development and drug resistance are associated with estrogen receptor signaling pathways. The present review mainly focused on the latest updates on RUNX2 in BCSCs and their roles in breast cancer progression and drug resistance, providing insight that may aid the development of RUNX2-based diagnostics and treatments for breast cancer in clinical practice.

11.
Biomedicines ; 11(4)2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37189851

RESUMO

Hepatocellular carcinoma (HCC) is an aggressive and challenging disease to treat. Due to the lack of effective early diagnosis and therapy for the illness, it is crucial to identify novel biomarkers that can predict tumor behavior in HCC. In such cases, family with sequence similarity 210 member B (FAM210B) is abundant in various human tissues, but its regulatory mechanisms and role in various tissues remain unclear. In this study, we analyzed the expression pattern of FAM210B in HCC using public gene expression databases and clinical tissue samples. Our results confirmed that FAM210B was dysregulated in both HCC cell lines and HCC paraffin section samples. FAM210B depletion significantly increased the capacity of cells to grow, migrate, and invade in vitro, while overexpression of FAM210B suppressed tumor growth in a xenograft tumor model. Furthermore, we identified FAM210B's involvement in MAPK signaling and p-AKT signaling pathways, both of which are known oncogenic signaling pathways. In summary, our study provides a rational basis for the further investigation of FAM210B as a valuable biological marker for diagnosing and predicting the prognosis of HCC patients.

12.
Front Immunol ; 13: 868813, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35514975

RESUMO

Breast cancer development and progression rely not only on the proliferation of neoplastic cells but also on the significant heterogeneity in the surrounding tumor microenvironment. Its unique microenvironment, including tumor-infiltrating lymphocytes, complex myeloid cells, lipid-associated macrophages, cancer-associated fibroblasts (CAFs), and other molecules that promote the growth and migration of tumor cells, has been shown to play a crucial role in the occurrence, growth, and metastasis of breast cancer. However, a detailed understanding of the complex microenvironment in breast cancer remains largely unknown. The unique pattern of breast cancer microenvironment cells has been poorly studied, and neither has the supportive role of these cells in pathogenesis been assessed. Single-cell multiomics biotechnology, especially single-cell RNA sequencing (scRNA-seq) reveals single-cell expression levels at much higher resolution, finely dissecting the molecular characteristics of tumor microenvironment. Here, we review the recent literature on breast cancer microenvironment, focusing on scRNA-seq studies and analyzing heterogeneity and spatial location of different cells, including T and B cells, macrophages/monocytes, neutrophils, and stromal cells. This review aims to provide a more comprehensive perception of breast cancer microenvironment and annotation for their clinical classification, diagnosis, and treatment. Furthermore, we discuss the impact of novel single-cell omics technologies, such as abundant omics exploration strategies, multiomics conjoint analysis mode, and deep learning network architecture, on the future research of breast cancer immune microenvironment.


Assuntos
Neoplasias da Mama , Fibroblastos Associados a Câncer , Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/metabolismo , Feminino , Humanos , Linfócitos do Interstício Tumoral , Análise de Célula Única , Microambiente Tumoral
13.
Front Cell Dev Biol ; 10: 857045, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35756991

RESUMO

Bone marrow microenvironment (BMM) has been proven to have benefits for both normal hematopoietic stem cell niche and pathological leukemic stem cell niche. In fact, the pathological leukemia microenvironment reprograms bone marrow niche cells, especially mesenchymal stem cells for leukemia progression, chemoresistance and relapse. The growth and differentiation of MSCs are modulated by leukemia stem cells. Moreover, chromatin abnormality of mesenchymal stem cells is sufficient for leukemia initiation. Here, we summarize the detailed relationship between MSC and leukemia. MSCs can actively and passively regulate the progression of myelogenous leukemia through cell-to-cell contact, cytokine-receptor interaction, and exosome communication. These behaviors benefit LSCs proliferation and survival and inhibit physiological hematopoiesis. Finally, we describe the recent advances in therapy targeting MSC hoping to provide new perspectives and therapeutic strategies for leukemia.

14.
Am J Transl Res ; 14(7): 4591-4605, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35958497

RESUMO

OBJECTIVE: To explore the synergistic effect and metabolic mechanism of chronic arsenic exposure and PTPN11 gain-of-function mutation on tumorigenesis. METHODS: Arsenic-transformed Ptpn11+/+ (WT-As) and Ptpn11D61G/+ -mutant (D61G-As) mouse embryonic fibroblasts (MEFs) were established by chronic treatment of low-dose arsenic. We used cell counting, plate colony and soft agar colony formation, and a nude mouse xenograft model to detect malignant transformation and tumorigenesis in vitro and in vivo. To detect mitochondrial oxidative phosphorylation (OXPHOS), we used Seahorse real-time cell metabolic analysis as well as adenosine triphosphate (ATP) and ROS production assays. Lastly, we examined mTOR signaling pathway changes by western blotting. RESULTS: Low-dose arsenic exposure promoted WT MEFs proliferation and exacerbated malignancy driven by Ptpn11D61G/+ mutation. Additionally, Ptpn11D61G/+ -mutant MEFs exhibited increased mitochondrial metabolism and low-dose arsenic amplified this malignant metabolic activity. Mechanistically, the mTOR signaling pathway was activated in Ptpn11D61G/+ -mutant MEFs and was further phosphorylated in arsenic-treated MEFs expressing Ptpn11D61G/+ . Critically, tumorigenesis induced by the synergistic effect of low-dose arsenic and Ptpn11D61G/+ mutation was prevented by mTOR pathway inhibition via rapamycin. CONCLUSION: This study found that metabolic reprogramming, particularly mitochondrial hyperactivation, is a core mechanism underlying tumorigenesis induced by the synergistic effect of Ptpn11D61G/+ mutation and arsenic exposure. Furthermore, these findings suggested mTOR is a therapeutic target for Ptpn11-associated cancers.

15.
Bone Res ; 10(1): 62, 2022 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-36289197

RESUMO

Heterotopic ossification (HO) is the abnormal formation of bone in extraskeletal sites. However, the mechanisms linking HO pathogenesis with bone mass dysfunction remain unclear. Here, we showed that mice harboring injury-induced and BMP4-dependent HO exhibit bone mass loss similar to that presented by patients with HO. Moreover, we found that injury-induced hyperinflammatory responses at the injury site triggered HO initiation but did not result in bone mass loss at 1 day post-injury (dpi). In contrast, a suppressive immune response promoted HO propagation and bone mass loss by 7 dpi. Correcting immune dysregulation by PD1/PDL1 blockade dramatically alleviated HO propagation and bone mass loss. We further demonstrated that fetuin-A (FetA), which has been frequently detected in HO lesions but rarely observed in HO-adjacent normal bone, acts as an immunomodulator to promote PD1 expression and M2 macrophage polarization, leading to immunosuppression. Intervention with recombinant FetA inhibited hyperinflammation and prevented HO and associated bone mass loss. Collectively, our findings provide new insights into the osteoimmunological interactions that occur during HO formation and suggest that FetA is an immunosuppressor and a potential therapeutic option for the treatment of HO.

16.
Arch Gerontol Geriatr ; 94: 104334, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33516077

RESUMO

Using the Pearlin stress process model, the present study aimed to test if there was an association between caregiving intensity and caregiver burden, to analyze what type of association existed, and to test if different indicators of social support moderated such association among caregivers of people with dementia. Data from the baseline assessment of the Resources for Enhancing Alzheimer's Caregiver Health (REACH II) (N = 637) were used. Caregiver burden (12-item Zarit caregiver burden scale), caregiving intensity (caregiving hours), and social support (Lubben social network, received support, satisfaction with support, and negative interactions) were the main measurements. Separate multivariate regression models were conducted with Stata 16. The results showed that the relationship between caregiving hours and caregiver burden was a nonlinear inversed U shape after controlling all of the socio-demographic variables. Further analyses showed that when caregiving hours reached 14 hours per day, the levels of burden were the highest. In addition, received support, satisfaction with support, and social network significantly buffered the relationship between caregiving hours and caregiver burden when they were examined separately. However, only social network played a significant buffering role when examining the four social support indicators simultaneously. These findings suggest the need for programs and practices that emphasize the importance of identifying, gaining, and strengthening positive aspect of social support, especially in how to broaden a caregiver's social network while caring for a family member with dementia.


Assuntos
Doença de Alzheimer , Cuidadores , Sobrecarga do Cuidador , Efeitos Psicossociais da Doença , Humanos , Apoio Social
17.
Bone Res ; 7: 33, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31700694

RESUMO

Heterotopic ossification (HO), true bone formation in soft tissue, is closely associated with abnormal injury/immune responses. We hypothesized that a key underlying mechanism of HO might be injury-induced dysregulation of immune checkpoint proteins (ICs). We found that the earliest stages of HO are characterized by enhanced infiltration of polarized macrophages into sites of minor injuries in an animal model of HO. The non-specific immune suppressants, Rapamycin and Ebselen, prevented HO providing evidence of the central role of the immune responses. We examined the expression pattern of ICs and found that they are dysregulated in HO lesions. More importantly, loss of function of inhibitory ICs (including PD1, PD-L1, and CD152) markedly inhibited HO, whereas loss of function of stimulatory ICs (including CD40L and OX-40L) facilitated HO. These findings suggest that IC inhibitors may provide a therapeutic approach to prevent or limit the extent of HO.

18.
Stem Cell Res Ther ; 10(1): 14, 2019 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-30635039

RESUMO

BACKGROUND: Heterotopic ossification (HO), either acquired (aHO) or hereditary, such as fibrodysplasia ossificans progressiva (FOP), is a serious condition without effective treatment. Understanding of the core process of injury-induced HO is still severely limited. METHODS: Double-pulse thymidine analog labeling was used to explore the distinctive domains evolved in injury-induced lesions in an animal model of HO (Nse-BMP4). Histological studies were performed to see whether a similar zonal pattern is also consistently found in biopsies from patients with aHO and FOP. In vivo clonal analysis with Rainbow mice, genetic loss-of-function studies with diphtheria toxin A (DTA)-mediated depletion and lineage tracing with Zsgreen reporter mice were used to obtain further evidence that Tie2-cre-, Gli1-creERT-, and Glast-creERT-labeled cells contribute to HO as niche-dwelling progenitor/stem cells. Immunohistochemistry was used to test whether vasculature, neurites, macrophages, and mast cells are closely associated with the proposed niche and thus are possible candidate niche supportive cells. Similar methods also were employed to further understand the signaling pathways that regulate the niche and the resultant HO. RESULTS: We found that distinctive domains evolved in injury-induced lesions, including, from outside-in, a mesenchymal stem cell (MSC) niche, a transient domain and an inner differentiated core in an animal model of HO (Nse-BMP4). A similar zonal structure was found in patients with aHO and FOP. In vivo clonal analysis with Rainbow mice and genetic loss-of-function studies with DTA provided evidence that Tie2-cre-, Gli1-creERT-, and Glast-creERT-labeled cells contribute to HO as niche-dwelling progenitor/stem cells; consistently, vasculature, neurites, macrophages, and mast cells are closely associated with the proposed niche and thus are possible candidate niche supportive cells. Further mechanistic study found that BMP and hedgehog (Hh) signaling co-regulate the niche and the resultant HO. CONCLUSIONS: Available data provide evidence of a potential core mechanism in which multiple disease-specific cellular and extracellular molecular elements form a unique local microenvironment, i.e., an injury-induced stem cell niche, which regulates the proliferation and osteogenic differentiation of mesenchymal stem cells (MSCs). The implication for HO is that therapeutic approaches must consider several different disease specific factors as parts of a functional unit, instead of treating one factor at a time.


Assuntos
Miosite Ossificante/genética , Ossificação Heterotópica/genética , Osteogênese/genética , Nicho de Células-Tronco/genética , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Toxina Diftérica/genética , Modelos Animais de Doenças , Transportador 1 de Aminoácido Excitatório/genética , Humanos , Mutação com Perda de Função/genética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Miosite Ossificante/patologia , Miosite Ossificante/terapia , Ossificação Heterotópica/patologia , Ossificação Heterotópica/terapia , Fragmentos de Peptídeos/genética , Receptor TIE-2/genética , Transdução de Sinais/genética , Proteína GLI1 em Dedos de Zinco/genética
19.
Stem Cells Int ; 2018: 8351374, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29983715

RESUMO

Stem cells, including embryonic stem cells (ESCs) and adult stem cells, play a central role in mammal organism development and homeostasis. They have two unique properties: the capacity for self-renewal and the ability to differentiate into many specialized cell types. Src homology region 2- (SH2-) containing protein tyrosine phosphatase 2 (SHP-2), a nonreceptor protein tyrosine phosphatase encoded by protein tyrosine phosphatase nonreceptor type 11 gene (PTPN11), regulates multicellular differentiation, proliferation, and survival through numerous conserved signal pathways. Gain-of-function (GOF) or loss-of-function (LOF) SHP2 in various cells, especially for stem cells, disrupt organism self-balance and lead to a plethora of diseases, such as cancer, maldevelopment, and excessive hyperblastosis. However, the exact mechanisms of SHP2 dysfunction in stem cells remain unclear. In this review, we intended to raise the attention and clarify the framework of SHP2-mediated signal pathways in various stem cells. Establishment of integrated signal architecture, from ESCs to adult stem cells, will help us to understand the changes of dynamic, multilayered pathways in response to SHP2 dysfunction. Overall, better understanding the functions of SHP2 in stem cells provides a new avenue to treat SHP2-associated diseases.

20.
Mol Med Rep ; 17(5): 6777-6783, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29512749

RESUMO

Allicin is an oxygenated carotenoid derivative that exhibits strong antioxidant activity, which effectively removes reactive oxygen species from the body and has important roles in disease prevention and treatment. Therefore, the present study aimed to investigate whether allicin attenuates lipopolysaccharide (LPS)­induced acute lung injury (ALI) in neonatal rats and the potential underlying mechanisms. An LPS­induced ALI neonatal rat model was utilized to assess the therapeutic value and mechanisms of allicin. Following allicin treatment, increases in lung wet/dry ratio and the lung protein concentration were significantly suppressed in LPS­induced ALI neonatal rats. Furthermore, ELISA results demonstrated that allicin significantly reduced the levels of malondialdehyde, tumor necrosis factor­α and interleukin­6, and increased superoxide dismutase activity, in the bronchoalveolar lavage fluid of LPS­treated rats. Additionally, allicin administration increased the protein expression of Bcl­2 and reduced the activity of caspase­3/-9, as determined by western blotting or ELISA, respectively, and increased phosphatidylinositol 3­kinase (PI3K) and phosphorylated­Akt protein levels, in LPS­treated ALI neonatal rats. The results of the present study indicate that allicin attenuate LPS­induced ALI in neonatal rats by ameliorating oxidative stress, inflammation and apoptosis via the PI3K/Akt pathway. Allicin may be used for development of a novel drug for treatment of ALI.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Lipopolissacarídeos/toxicidade , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ácidos Sulfínicos/farmacologia , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Dissulfetos , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA