Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
Más filtros

Intervalo de año de publicación
1.
Mol Ther ; 32(5): 1479-1496, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38429926

RESUMEN

Intense inflammatory response impairs bone marrow mesenchymal stem cell (BMSC)-mediated bone regeneration, with transforming growth factor (TGF)-ß1 being the most highly expressed cytokine. However, how to find effective and safe means to improve bone formation impaired by excessive TGF-ß1 remains unclear. In this study, we found that the expression of orphan nuclear receptor Nr4a1, an endogenous repressor of TGF-ß1, was suppressed directly by TGF-ß1-induced Smad3 and indirectly by Hdac4, respectively. Importantly, Nr4a1 overexpression promoted BMSC osteogenesis and reversed TGF-ß1-mediated osteogenic inhibition and pro-fibrotic effects. Transcriptomic and histologic analyses confirmed that upregulation of Nr4a1 increased the transcription of Wnt family member 4 (Wnt4) and activated Wnt pathway. Mechanistically, Nr4a1 bound to the promoter of Wnt4 and regulated its expression, thereby enhancing the osteogenic capacity of BMSCs. Moreover, treatment with Nr4a1 gene therapy or Nr4a1 agonist Csn-B could promote ectopic bone formation, defect repair, and fracture healing. Finally, we demonstrated the correlation of NR4A1 with osteogenesis and the activation of the WNT4/ß-catenin pathway in human BMSCs and fracture samples. Taken together, these findings uncover the critical role of Nr4a1 in bone formation and alleviation of inflammation-induced bone regeneration disorders, and suggest that Nr4a1 has the potential to be a therapeutic target for accelerating bone healing.


Asunto(s)
Regeneración Ósea , Inflamación , Células Madre Mesenquimatosas , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Osteogénesis , Proteína Wnt4 , Células Madre Mesenquimatosas/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Osteogénesis/genética , Regeneración Ósea/genética , Animales , Ratones , Proteína Wnt4/metabolismo , Proteína Wnt4/genética , Humanos , Inflamación/genética , Inflamación/metabolismo , Regulación de la Expresión Génica , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta1/genética , Vía de Señalización Wnt , Masculino , Transcripción Genética , Histona Desacetilasas/metabolismo , Histona Desacetilasas/genética , Modelos Animales de Enfermedad
2.
Immunology ; 172(4): 533-546, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38561001

RESUMEN

Radiation-induced fibrosis (RIF) is a severe chronic complication of radiotherapy (RT) manifested by excessive extracellular matrix (ECM) components deposition within the irradiated area. The lung, heart, skin, jaw, pelvic organs and so on may be affected by RIF, which hampers body functions and quality of life. There is accumulating evidence suggesting that the immune microenvironment may play a key regulatory role in RIF. This article discussed the synergetic or antagonistic effects of immune cells and mediators in regulating RIF's development. Several potential preventative and therapeutic strategies for RIF were proposed based on the immunological mechanisms to provide clinicians with improved cognition and clinical treatment guidance.


Asunto(s)
Microambiente Celular , Fibrosis , Traumatismos por Radiación , Radioterapia , Humanos , Animales , Traumatismos por Radiación/inmunología , Radioterapia/efectos adversos , Matriz Extracelular/metabolismo , Matriz Extracelular/inmunología , Matriz Extracelular/efectos de la radiación
3.
Curr Oncol Rep ; 26(5): 439-465, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38546941

RESUMEN

PURPOSE OF REVIEW: The purpose of this review is to discuss a wide variety of novel therapies recently studied or actively undergoing study in patients with glioblastoma. This review also discusses current and future strategies for improving clinical trial design in patients with glioblastoma to maximize efficacy in discovering effective treatments. RECENT FINDINGS: Over the years, there has been significant expansion in therapy modalities studied in patients with glioblastoma. These therapies include, but are not limited to, targeted molecular therapies, DNA repair pathway targeted therapies, immunotherapies, vaccine therapies, and surgically targeted radiotherapies. Glioblastoma is the most common malignant primary brain tumor in adults and unfortunately remains with poor overall survival following the current standard of care. Given the dismal prognosis, significant clinical and research efforts are ongoing with the goal of improving patient outcomes and enhancing quality and quantity of life utilizing a wide variety of novel therapies.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/uso terapéutico , Ensayos Clínicos como Asunto , Glioblastoma/inmunología , Glioblastoma/terapia , Inmunoterapia/métodos , Terapia Molecular Dirigida/métodos
4.
Mol Biol Rep ; 51(1): 219, 2024 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-38281269

RESUMEN

Despite the availability of technological advances in traditional anti-cancer therapies, there is a need for more precise and targeted cancer treatment strategies. The wide-ranging shortfalls of conventional anticancer therapies such as systematic toxicity, compromised life quality, and limited to severe side effects are major areas of concern of conventional cancer treatment approaches. Owing to the expansion of knowledge and technological advancements in the field of cancer biology, more innovative and safe anti-cancerous approaches such as immune therapy, gene therapy and targeted therapy are rapidly evolving with the aim to address the limitations of conventional therapies. The concept of immunotherapy began with the capability of coley toxins to stimulate toll-like receptors of immune cells to provoke an immune response against cancers. With an in-depth understating of the molecular mechanisms of carcinogenesis and their relationship to disease prognosis, molecular targeted therapy approaches, that inhibit or stimulate specific cancer-promoting or cancer-inhibitory molecules respectively, have offered promising outcomes. In this review, we evaluate the achievement and challenges of these technically advanced therapies with the aim of presenting the overall progress and perspective of each approach.


Asunto(s)
Terapia Molecular Dirigida , Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Inmunoterapia , Terapia Genética
5.
Mol Ther ; 31(12): 3389-3413, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37740493

RESUMEN

Long noncoding RNAs (lncRNAs) are a distinct subtype of RNA that lack protein-coding capacity but exert significant influence on various cellular processes. In non-small cell lung cancer (NSCLC), dysregulated lncRNAs act as either oncogenes or tumor suppressors, contributing to tumorigenesis and tumor progression. LncRNAs directly modulate gene expression, act as competitive endogenous RNAs by interacting with microRNAs or proteins, and associate with RNA binding proteins. Moreover, lncRNAs can reshape the tumor immune microenvironment and influence cellular metabolism, cancer cell stemness, and angiogenesis by engaging various signaling pathways. Notably, lncRNAs have shown great potential as diagnostic or prognostic biomarkers in liquid biopsies and therapeutic strategies for NSCLC. This comprehensive review elucidates the significant roles and diverse mechanisms of lncRNAs in NSCLC. Furthermore, we provide insights into the clinical relevance, current research progress, limitations, innovative research approaches, and future perspectives for targeting lncRNAs in NSCLC. By summarizing the existing knowledge and advancements, we aim to enhance the understanding of the pivotal roles played by lncRNAs in NSCLC and stimulate further research in this field. Ultimately, unraveling the complex network of lncRNA-mediated regulatory mechanisms in NSCLC could potentially lead to the development of novel diagnostic tools and therapeutic strategies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Humanos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , MicroARNs/genética , Oncogenes , Regulación Neoplásica de la Expresión Génica , Microambiente Tumoral
6.
Cancer ; 129(10): 1467-1472, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36825454

RESUMEN

Management of brain tumors has been challenging given the limited therapeutic options and disabling morbidities associated with central nervous system (CNS) dysfunction. This review focuses on recent developments in the field, with an emphasis on clinical management. The growing clinical trials landscape reflects advanced insights into cancer immunology and genomics and the need to address molecular and clinical heterogeneity. Recent phase 3 trials investigating anti-PD-1 immunotherapies, particularly nivolumab, have failed to demonstrate improved survival in glioblastoma, underscoring the need to better understand the complexity of CNS immunologic surveillance. Conversely, targeted therapies have accounted for several US Food and Drug Administration approvals extended to brain tumors, particularly therapies directed to BRAF V600E mutations and TRAK fusions, underscoring a need to routinely screen patients for these rare molecular abnormalities. In primary CNS lymphoma, attention has turned to long-term outcomes of consolidation therapies, and recent studies have highlighted the excellent disease control afforded by high-dose chemotherapy and stem cell transplantation. Meningiomas remain a focus of investigations, with preliminary promising results observed with octreotide combined with mTOR inhibition, and immunotherapy with single-agent pembrolizumab. Finally, proton radiotherapy has emerged as a novel alternative for leptomeningeal metastases from solid tumors, which can now be treated more safely with craniospinal irradiation and monitored by the enumeration of circulating tumor cells in the cerebrospinal fluid as a biomarker. Taken together, these incremental advances have improved outcomes in select brain tumor patient populations, whereas ongoing clinical trials hold the promise of meaningful advances and breakthroughs for larger proportions of patients with brain tumors.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Meníngeas , Meningioma , Humanos , Neoplasias Encefálicas/secundario
7.
Int J Mol Sci ; 24(21)2023 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-37958500

RESUMEN

Galectin-3 is the most studied member of the Galectin family, with a large range of mediation in biological activities such as cell growth, proliferation, apoptosis, differentiation, cell adhesion, and tissue repair, as well as in pathological processes such as inflammation, tissue fibrosis, and angiogenesis. As is known to all, inflammation, aberrant cell apoptosis, and neovascularization are the main pathophysiological processes in retinal degeneration and many ocular diseases. Therefore, the review aims to conclude the role of Gal3 in the retinal degeneration of various diseases as well as the occurrence and development of the diseases and discuss its molecular mechanisms according to research in systemic diseases. At the same time, we summarized the predictive role of Gal3 as a biomarker and the clinical application of its inhibitors to discuss the possibility of Gal3 as a novel target for the treatment of ocular diseases.


Asunto(s)
Galectina 3 , Degeneración Retiniana , Humanos , Galectina 3/genética , Degeneración Retiniana/tratamiento farmacológico , Galectinas , Biomarcadores , Inflamación
8.
Int J Mol Sci ; 24(2)2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36675267

RESUMEN

A prospective source of stem cells for bone tissue engineering is adipose-derived stem cells (ADSCs), and BMP-2 has been proven to be highly effective in promoting the osteogenic differentiation of stem cells. Rarely has research been conducted on the impact of lactoferrin (LF) on ADSCs' osteogenic differentiation. As such, in this study, we examined the effects of LF and BMP-2 to assess the ability of LF to stimulate ADSCs' osteogenic differentiation. The osteogenic medium was supplemented with the LF at the following concentrations to culture ADSCs: 0, 10, 20, 50, 100, and 500 µg/mL. The Cell Counting Kit-8 (CCK-8) assay was used to measure the proliferation of ADSCs. Calcium deposition, alkaline phosphatase (ALP) staining, real-time polymerase chain reaction (RT-PCR), and an ALP activity assay were used to establish osteogenic differentiation. RNA sequencing analysis was carried out to investigate the mechanism of LF boosting the osteogenic development of ADSCs. In the concentration range of 0-100 µg/mL, LF concentration-dependently increased the proliferative vitality and osteogenic differentiation of ADSCs. At a dose of 500 µg/mL, LF sped up and enhanced differentiation, but inhibited ADSCs from proliferating. LF (100 and 500 µg/mL) produced more substantial osteoinductive effects than BMP-2. The PI3 kinase/AKT (PI3K/AKT) and IGF-R1 signaling pathways were significantly activated in LF-treated ADSCs. The in vitro study results showed that LF could effectively promote osteogenic differentiation of ADSCs by activating the PI3K/AKT and IGF-R1 pathways. In our in vitro investigation, an LF concentration of 100 µg/mL was optimal for osteoinduction and proliferation. Our study suggests that LF is an attractive alternative to BMP-2 in bone tissue engineering. As a bioactive molecule capable of inducing adipose stem cells to form osteoblasts, LF is expected to be clinically used in combination with biomaterials as an innovative molecular and cellular therapy to promote bone repair.


Asunto(s)
Tejido Adiposo , Osteogénesis , Tejido Adiposo/metabolismo , Lactoferrina/farmacología , Lactoferrina/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Estudios Prospectivos , Células Cultivadas , Células Madre/metabolismo , Diferenciación Celular
9.
Cancer Cell Int ; 22(1): 358, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36376983

RESUMEN

BACKGROUND: Tumor suppressor CYLD dysfunction by loss of its expression, triggers malignant transformation, especially drug resistance and tumor invasion/metastasis. Although loss of CYLD expression is significantly associated with poor prognosis in a large variety of tumors, no clinically-effective treatment for CYLD-negative cancer patients is available. METHODS: We focused on oral squamous cell carcinoma (OSCC), and sought to develop novel therapeutic agents for CYLD-negative cancer patients with poor prognosis. CYLD-knockdown OSCC cells by using CYLD-specific siRNA, were used to elucidate and determine the efficacy of novel drug candidates by evaluating cell viability and epithelial-mesenchymal transition (EMT)-like change. Therapeutic effects of candidate drug on cell line-derived xenograft (CDX) model and usefulness of CYLD as a novel biomarker using patient-derived xenograft (PDX) model were further investigated. RESULTS: CYLD-knockdown OSCC cells were resistant for all currently-available cytotoxic chemotherapeutic agents for OSCC, such as, cisplatin, 5-FU, carboplatin, docetaxel, and paclitaxel. By using comprehensive proteome analysis approach, we identified epidermal growth factor receptor (EGFR), a receptor tyrosine kinase, played key roles in CYLD-knockdown OSCC cells. Indeed, cell survival rate in the cisplatin-resistant CYLD-knockdown OSCC cells was markedly inhibited by treatment with clinically available EGFR tyrosine kinase inhibitors (EGFR-TKIs), such as gefitinib. In addition, gefitinib was significantly effective for not only cell survival, but also EMT-like changes through inhibiting transforming growth factor-ß (TGF-ß) signaling in CYLD-knockdown OSCC cells. Thereby, overall survival of CYLD-knockdown CDX models was significantly prolonged by gefitinib treatment. Moreover, we found that CYLD expression was significantly associated with gefitinib response by using PDX models. CONCLUSIONS: Our results first revealed that EGFR-targeted molecular therapies, such as EGFR-TKIs, could have potential to be novel therapeutic agents for the CYLD-negative OSCC patients with poor prognosis.

10.
Curr Oncol Rep ; 24(12): 1677-1683, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36417134

RESUMEN

PURPOSE OF REVIEW: In advanced-stage and high-risk endometrial cancer, adjuvant treatment is the standard of care and typically includes chemotherapy with or without radiotherapy. Debate continues over the optimal use of these two treatment modalities together or separately. This review covers the historical literature leading to the current recommendation for adjuvant chemotherapy, in addition to looking forward to the relatively new field of targeted molecular treatment. RECENT FINDINGS: The review covers recent phase III trials comparing chemotherapy to radiotherapy in high-risk endometrial cancer. Additionally, the era of genomic medicine has a new foothold in endometrial cancer, and the review covers new discoveries on molecular classification and prognostic implications. Fortunately, the majority of endometrial cancer has a good prognosis. For advanced-stage and high-risk histologies, the prognosis can be guarded, with adjuvant treatment improving outcomes. Gynecologic oncologists continue to debate the optimal treatment modality/modalities, a debate which will likely become more robust as the field of molecular treatment in endometrial cancer evolves.


Asunto(s)
Neoplasias Endometriales , Oncología por Radiación , Femenino , Humanos , Terapia Combinada , Neoplasias Endometriales/tratamiento farmacológico , Quimioterapia Adyuvante , Medicina Genómica
11.
Int J Mol Sci ; 23(9)2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35563013

RESUMEN

Myotonic dystrophy type 1 (DM1) is the most common muscular dystrophy affecting many different body tissues, predominantly skeletal and cardiac muscles and the central nervous system. The expansion of CTG repeats in the DM1 protein-kinase (DMPK) gene is the genetic cause of the disease. The pathogenetic mechanisms are mainly mediated by the production of a toxic expanded CUG transcript from the DMPK gene. With the availability of new knowledge, disease models, and technical tools, much progress has been made in the discovery of altered pathways and in the potential of therapeutic intervention, making the path to the clinic a closer reality. In this review, we describe and discuss the molecular therapeutic strategies for DM1, which are designed to directly target the CTG genomic tract, the expanded CUG transcript or downstream signaling molecules.


Asunto(s)
Distrofia Miotónica , Edición Génica , Humanos , Distrofia Miotónica/tratamiento farmacológico , Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/metabolismo , Expansión de Repetición de Trinucleótido/genética
12.
Cancer Sci ; 112(10): 3935-3944, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34251718

RESUMEN

Homeostasis of the hematopoietic system is achieved in a hierarchy, with hematopoietic stem cells at the pinnacle. Because only hematopoietic stem cells (HSCs) can self-renew, the size of the hematopoietic system is strictly controlled. In hematopoietic reconstitution experiments, 1 HSC can reconstitute the entire hematopoietic system, whereas 50 multipotent progenitors cannot. This indicates that only HSCs self-renew, whereas non-HSC hematopoietic progenitors are programmed to differentiate or senesce. Oncogenic mutations of the mixed lineage leukemia gene (MLL) overcome this "programmed differentiation" by conferring the self-renewing ability to non-HSC hematopoietic progenitors. In leukemia, mutated MLL proteins constitutively activate a broad range of previously transcribed CpG-rich promoters by an MLL-mediated transcriptional activation system. This system promotes self-renewal by replicating an expression profile similar to that of the mother cell in its daughter cells. In this transcriptional activation system, MLL binds to unmethylated CpG-rich promoters and recruits RNA polymerase II. MLL recruits p300/CBP through its transcriptional activation domain, which acetylates histone H3 at lysines 9, 18, and 27. The AF4 family/ENL family/P-TEFb complex (AEP) binds to acetylated H3K9/18/27 to activate transcription. Gene rearrangements of MLL with AEP- or CBP/p300-complex components generate constitutively active transcriptional machinery of this transcriptional activation system, which causes aberrant self-renewal of leukemia stem cells. Inhibitors of the components of this system effectively decrease their leukemogenic potential.


Asunto(s)
Autorrenovación de las Células/fisiología , Células Madre Hematopoyéticas/fisiología , N-Metiltransferasa de Histona-Lisina/genética , Leucemia/etiología , Proteína de la Leucemia Mieloide-Linfoide/genética , Activación Transcripcional/fisiología , Acetilación , Diferenciación Celular , Autorrenovación de las Células/genética , Senescencia Celular , Islas de CpG/genética , Proteínas de Unión al ADN/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Reordenamiento Génico , Hematopoyesis/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Leucemia/prevención & control , Lisina/metabolismo , Células Madre Multipotentes/fisiología , Mutación , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , ARN Polimerasa II/metabolismo , Factores de Elongación Transcripcional/metabolismo
13.
BJU Int ; 127(5): 498-506, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33606327

RESUMEN

OBJECTIVES: To examine the current molecular therapeutics in the medical treatment of recurrent ischemic priapism (RIP). To propose a stepwise clinical management paradigm for the treatment of RIP. METHODS: We performed a literature search using the PubMed database for the terms 'recurrent ischemic priapism' and 'stuttering priapism' up until December 2020. We assessed pre-clinical and clinical studies regarding medical management of RIP and molecular pathophysiology. Case series and randomized trials were evaluated by study quality and patient outcomes to determine a potential clinical management scheme. RESULTS: Recent research has fostered an improved understanding of the underlying molecular pathophysiology of RIP that has paved the way forward for developing new therapeutic agents. Medications targeting neurovascular, hormonal and haematological mechanisms associated with RIP show great promise towards remedying this condition. A host of therapeutic agents operating across different mechanistic directions may be implemented according to a clinical management scheme to potentially optimize RIP outcomes. CONCLUSION: RIP remains a medically neglected condition with current management focused on treating the acute condition rather than modulating the course of disease. Continued research into the molecular mechanisms of RIP and standardized clinical pathways can improve the quality of care for patients suffering from this condition.


Asunto(s)
Algoritmos , Anemia de Células Falciformes/tratamiento farmacológico , Isquemia/tratamiento farmacológico , Priapismo/tratamiento farmacológico , Inhibidores de 5-alfa-Reductasa/uso terapéutico , Agonistas de Receptores Adrenérgicos alfa 1/uso terapéutico , Agonistas Adrenérgicos beta/uso terapéutico , Anemia de Células Falciformes/complicaciones , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antifúngicos/uso terapéutico , Antidrepanocíticos/uso terapéutico , Humanos , Hidroxiurea/uso terapéutico , Isquemia/complicaciones , Cetoconazol/uso terapéutico , Masculino , Selectina-P/antagonistas & inhibidores , Erección Peniana/fisiología , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Priapismo/etiología , Recurrencia
14.
Pharmacol Res ; 165: 105415, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33412279

RESUMEN

Genetically modified animal studies have revealed specific expression patterns and unequivocal roles of class I PI3K isoenzymes. PI3K(p110α), a catalytic subunit of class I PI3Ks is ubiquitously expressed and is well characterised in the cardiovascular system. Given that genetic inhibition of PI3K(p110α) causes lethal phenotype embryonically, the catalytic subunit is critically important in housekeeping and biological processes. A growing number of studies underpin crucial roles of PI3K(p110α) in cell survival, proliferation, hypertrophy and arrhythmogenesis. While the studies provide great insights, the precise mechanisms involved in PI3K(p110α) hypofunction and atrial fibrillation (AF) are not fully known. AF is a well recognised clinical problem with significant management limitations. In this translational review, we attempted a narration of PI3K(p110α) hypofunction in the molecular basis of AF pathophysiology. We sought to cautiously highlight the relevance of this molecule in the therapeutic approaches for AF management per se (i.e without conditions associate with cell proliferation, like cancer), and in mitigating effects of clinical risk factors in atrial substrate formation leading to AF progression. We also considered PI3K(p110α) in AF gene association, with the aim of identifying mechanistic links between the ever increasingly well-defined genetic loci (regions and genes) and AF. Such mechanisms will aid in identifying new drug targets for arrhythmogenic substrate and AF.


Asunto(s)
Antiarrítmicos/uso terapéutico , Fibrilación Atrial/fisiopatología , Fosfatidilinositol 3-Quinasa Clase Ia/fisiología , Animales , Antiarrítmicos/farmacología , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/etiología , Fosfatidilinositol 3-Quinasa Clase Ia/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Humanos
15.
Int J Med Sci ; 18(1): 207-215, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390789

RESUMEN

Background: Placental-like chondroitin sulfate A (pl-CSA) is exclusively expressed in cancerous and placental tissues and is highly correlated with the degree of malignancy. However, the mechanism through which pl-CSA regulates tumorigenesis and metastasis in choriocarcinoma remains unclear. Methods: Stable transfectants of the JEG3 choriocarcinoma cell line, including a negative control (NC) line and a cell line with knockout of the biosynthetic enzyme CS synthase-2 (ChSy-2) (ChSy-2-/-), were obtained using CRISPR/Cas9 systems and identified by immunofluorescence, flow cytometry, western blots and enzyme-linked immunosorbent assays (ELISAs). The proliferation, migration, invasion and colony formation of the cells were determined by a cell counting kit, scratch-wound assays, transwell assays and soft agar colony formation assays in vitro, respectively. The tumorigenesis and metastasis of choriocarcinoma were also investigated through two xenograft models in vivo. Results: The ChSy-2 protein in the ChSy-2-/-group was below the detection threshold, which was accompanied a significant reduction in the pl-CSA level. Reducing pl-CSA through ChSy-2 knockout significantly inhibited cell proliferation, migration, invasion and colony formation in vitro and tumorigenesis and metastasis of choriocarcinoma, with deceases in tumor volume and metastatic foci and a high percent survival compared to the NC in vivo. Conclusion: pl-CSA, as a necessary component of JEG-3 cells, was efficiently reduced through ChSy-2 knockout, which significantly inhibited the tumorigenesis and metastasis of choriocarcinoma. ChSy-2/pl-CSA could be alternative targets for tumor therapy.


Asunto(s)
Carcinogénesis/patología , Sulfatos de Condroitina/metabolismo , Coriocarcinoma/secundario , Glicosiltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias Uterinas/patología , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Técnicas de Silenciamiento del Gen , Glicosiltransferasas/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Embarazo , Organismos Libres de Patógenos Específicos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Radiologe ; 61(10): 955-967, 2021 Oct.
Artículo en Alemán | MEDLINE | ID: mdl-34550423

RESUMEN

Radiation therapy and more recently, drug-based molecular therapy in particular, are a central component of modern oncology. Both forms of therapy are suitable for effectively treating tumors with comparatively low systemic side effects. Nevertheless, even these treatment approaches have side effects, which are triggered by the toxicity of the radiation and by the immunomodulatory effects of the administered drugs. The pneumotoxic potential of these forms of therapy is reflected in the development of interstitial pneumonitis, which can transition into fibrotic changes in the lung structure. The clinical diagnosis of the disease is made more difficult as the symptoms are nonspecific. Computed tomography (CT) is an excellent means of diagnosing the corresponding consolidations and to monitor them over time. Therefore, in the interdisciplinary context the radiologist plays a central role in the diagnostics of this disease.


Asunto(s)
Enfermedades Pulmonares Intersticiales , Neoplasias , Humanos , Pulmón/diagnóstico por imagen , Enfermedades Pulmonares Intersticiales/inducido químicamente , Enfermedades Pulmonares Intersticiales/diagnóstico por imagen , Enfermedades Pulmonares Intersticiales/terapia , Tomografía Computarizada por Rayos X
17.
Int J Mol Sci ; 22(7)2021 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-33805154

RESUMEN

Intermediate junctional epidermolysis bullosa caused by mutations in the COL17A1 gene is characterized by the frequent development of blisters and erosions on the skin and mucous membranes. The rarity of the disease and the heterogeneity of the underlying mutations renders therapy developments challenging. However, the high number of short in-frame exons facilitates the use of antisense oligonucleotides (AON) to restore collagen 17 (C17) expression by inducing exon skipping. In a personalized approach, we designed and tested three AONs in combination with a cationic liposomal carrier for their ability to induce skipping of COL17A1 exon 7 in 2D culture and in 3D skin equivalents. We show that AON-induced exon skipping excludes the targeted exon from pre-mRNA processing, which restores the reading frame, leading to the expression of a slightly truncated protein. Furthermore, the expression and correct deposition of C17 at the dermal-epidermal junction indicates its functionality. Thus, we assume AON-mediated exon skipping to be a promising tool for the treatment of junctional epidermolysis bullosa, particularly applicable in a personalized manner for rare genotypes.


Asunto(s)
Autoantígenos/metabolismo , Epidermólisis Ampollosa de la Unión/genética , Colágenos no Fibrilares/metabolismo , Oligonucleótidos Antisentido/genética , Empalme del ARN , Empalme Alternativo , Biopsia , Línea Celular , Supervivencia Celular , Epidermólisis Ampollosa de la Unión/metabolismo , Epidermólisis Ampollosa de la Unión/terapia , Exones , Genotipo , Homocigoto , Humanos , Queratinocitos/citología , Liposomas/química , Mutación , Técnicas de Cultivo de Órganos , ARN Mensajero/metabolismo , Colágeno Tipo XVII
18.
Rinsho Ketsueki ; 62(8): 988-997, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34497239

RESUMEN

Leukemia is caused by uncontrolled proliferation of immature hematopoietic progenitors. MLL fusion proteins, generated by chromosomal translocations, activate a broad range of previously transcribed genes to achieve the same expression profile as that of the parent cell in the daughter cells, thereby promoting self-renewal. Normally, replication of the expression profile only occurs in the hematopoietic stem cells (HSCs). A transactivation system comprised of MLL and AF4/ENL/P-TEFb (AEP) complexes promotes it by reactivating CpG-rich promoters. In the normal hematopoietic development, this system is tightly regulated and progressively suppressed during the course of hematopoietic differentiation so that non-HSC hematopoietic cells would not self-renew. Genetic mutations such as fusions of MLL and AEP components generate a constitutively active form of the MLL transcriptional machinery, which aberrantly promotes self-renewal even in non-HSC hematopoietic cells. In this review, I depict a molecular mechanism of MLL fusion-mediated leukemogenesis from a standpoint that leukemogenesis is driven by aberrant self-renewal that is mediated by hyper-active transcriptional machinery, and introduce several molecularly targeted therapies in the making which specifically perturb this transactivation system.


Asunto(s)
Leucemia , Proteína de la Leucemia Mieloide-Linfoide , Carcinogénesis , Células Madre Hematopoyéticas , Humanos , Leucemia/genética , Leucemia/terapia , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Fusión Oncogénica/genética
19.
BMC Cancer ; 20(1): 129, 2020 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-32066410

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is a heterogeneous disease with a complex biology and a wide number of altered genes such as BRAF, KRAS and PIK3CA. Advances with new-targeted therapies have been achieved and available treating options have prolonged patient's survival. However, BRAF-mutated CRC patients remain unresponsive to available therapies with RAF inhibitors (RAFi) alone or combined with ErbB inhibitors (ErbBi). These unmet needs require further exploitation of oncogenic signaling in order to set up individualized treatments. METHODS: To this end, we tested the efficacy of single agent or combined treatments using the BRAFi, vemurafenib and two different ErbBi: panitumumab and afatinib in CRC cells characterized by different molecular phenotypes. RESULTS: Combination strategies with BRAFi and ErbBi achieved a better response in BRAFV600E mutated cells expressing high levels of ErbB2. CONCLUSIONS: Our findings support the importance of ErbB2 evaluation in BRAF-mutated CRC patients and its role as a positive predictor factor of response to BRAFi/ErbBi combination.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Mutación , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Afatinib/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Terapia Molecular Dirigida/métodos , Panitumumab/administración & dosificación , Receptor ErbB-2/metabolismo , Vemurafenib/administración & dosificación
20.
J Surg Oncol ; 122(3): 555-561, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32441371

RESUMEN

BACKGROUND AND OBJECTIVES: Metastasectomy for melanoma provides durable disease control in carefully selected patients. Similarly, BRAF-targeted and immune checkpoint inhibition has improved median overall survival (OS) in metastatic patients. We hypothesized that there is an increasing role for metastasectomy in melanoma patients responding to these therapies. METHODS: Retrospective analysis of a prospectively maintained database identified 128 patients with stage IV melanoma who received targeted molecular and/or checkpoint inhibitors at an academic institution from 2006 to 2017. Records were reviewed to characterize clinicopathologic characteristics, response to treatment, and intent of surgery for those who underwent metastasectomy. OS was analyzed by the Kaplan-Meier method. RESULTS: Median OS from stage IV diagnosis was 31.3 months. A total of 81 patients received checkpoint inhibitors, 11 received targeted inhibitors, and 36 received both. A total of 73 patients underwent metastasectomy. Indications for surgery included the intent to render disease-free (54%), palliation (34%), and diagnostic confirmation (11%). Responders to systemic therapy who underwent metastasectomy had improved OS compared to responders who did not (84.3 vs. 42.9 months, P = .018). CONCLUSIONS: Metastasectomy for melanoma is associated with improved OS in patients that respond to targeted molecular or immunotherapy. Resection should be strongly considered in this cohort as multimodality treatment results in excellent OS.


Asunto(s)
Melanoma/secundario , Melanoma/terapia , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Melanoma/mortalidad , Melanoma/cirugía , Metastasectomía , Terapia Molecular Dirigida/métodos , Estadificación de Neoplasias , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA