Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.311
Filtrar
1.
J Virol ; 97(11): e0149723, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37877719

RESUMO

IMPORTANCE: Duck Tembusu virus (DTMUV) is an emerging pathogenic flavivirus that replicates well in mosquito, bird, and mammalian cells. An in vivo study revealed that BALB/c mice and Kunming mice were susceptible to DTMUV after intracerebral inoculation. Moreover, there are no reports about DTMUV-related human disease, but antibodies against DTMUV and viral RNA were detected in the serum samples of duck industry workers. This information implies that DTMUV has expanded its host range and poses a threat to mammalian health. Thus, understanding the pathogenic mechanism of DTMUV is crucial for identifying potential antiviral targets. In this study, we discovered that NS3 can induce the mitochondria-mediated apoptotic pathway through the PERK/PKR pathway; it can also interact with voltage-dependent anion channel 2 to induce apoptosis. Our findings provide a theoretical basis for understanding the pathogenic mechanism of DTMUV infection and identifying potential antiviral targets and may also serve as a reference for exploring the pathogenesis of other flaviviruses.


Assuntos
Apoptose , Patos , Infecções por Flavivirus , Flavivirus , Especificidade de Hospedeiro , Animais , Humanos , Antivirais/farmacologia , Patos/virologia , eIF-2 Quinase/metabolismo , Flavivirus/enzimologia , Flavivirus/patogenicidade , Infecções por Flavivirus/diagnóstico , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/transmissão , Infecções por Flavivirus/virologia , Mitocôndrias/metabolismo , Terapia de Alvo Molecular/tendências , Zoonoses Virais/diagnóstico , Zoonoses Virais/imunologia , Zoonoses Virais/transmissão , Zoonoses Virais/virologia , Canal de Ânion 2 Dependente de Voltagem/metabolismo
2.
Int J Mol Sci ; 24(9)2023 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-37175935

RESUMO

Alzheimer's disease (AD) is a scourge for patients, caregivers and healthcare professionals due to the progressive character of the disease and the lack of effective treatments. AD is considered a proteinopathy, which means that aetiological and clinical features of AD have been linked to the deposition of amyloid ß (Aß) and hyperphosphorylated tau protein aggregates throughout the brain, with Aß and hyperphosphorylated tau representing classical AD hallmarks. However, some other putative mechanisms underlying the pathogenesis of the disease have been proposed, including inflammation in the brain, microglia activation, impaired hippocampus neurogenesis and alterations in the production and release of neurotrophic factors. Among all, microglia activation and chronic inflammation in the brain gained some attention, with researchers worldwide wondering whether it is possible to prevent and stop, respectively, the onset and progression of the disease by modulating microglia phenotypes. The following key points have been established so far: (i) Aß deposition in brain parenchyma represents repeated stimulus determining chronic activation of microglia; (ii) chronic activation and priming of microglia make these cells lose neuroprotective functions and favour damage and loss of neurons; (iii) quiescent status of microglia at baseline prevents chronic activation and priming, meaning that the more microglia are quiescent, the less they become neurotoxic. Many molecules are known to modulate the quiescent baseline state of microglia, attracting huge interest among scientists as to whether these molecules could be used as valuable targets in AD treatment. The downside of the coin came early with the observation that quiescent microglia do not display phagocytic ability, being unable to clear Aß deposits since phagocytosis is crucial for Aß clearance efficacy. A possible solution for this issue could be found in the modulation of microglia status at baseline, which could help maintain both neuroprotective features and phagocytic ability at the same time. Among the molecules known to influence the baseline status of microglia, C-X3-chemokine Ligand 1 (CX3CL1), also known as Fractalkine (FKN), is one of the most investigated. FKN and its microglial receptor CX3CR1 are crucial players in the interplay between neurons and microglia, modulating the operation of some neural circuits and the efficacy and persistence of immune response against injury. In addition, CX3CL1 regulates synaptic pruning and plasticity in the developmental age and in adulthood, when it strongly impacts the hippocampus neurogenesis of the adult. CX3CL1 has an effect on Aß clearance and tau phosphorylation, as well as in microglia activation and priming. For all the above, CX3CL1/CX3CR1 signalling has been widely studied in relation to AD pathogenesis, and its biochemical pathway could hide molecular targets for novel treatment strategies in AD. This review summarizes the possible role of CX3CL1 in AD pathogenesis and its use as a potential target for AD treatment.


Assuntos
Doença de Alzheimer , Quimiocina CXCL1 , Terapia de Alvo Molecular , Transdução de Sinais , Doença de Alzheimer/fisiopatologia , Doença de Alzheimer/terapia , Terapia de Alvo Molecular/tendências , Microglia/fisiologia , Quimiocina CXCL1/metabolismo , Receptor 1 de Quimiocina CX3C/metabolismo
3.
ESMO Open ; 8(2): 100882, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36806375

RESUMO

Estrogen receptor-positive (ER+)/HER2-negative (HER2-), the so-called luminal-type breast cancer, is the most frequent subset, accounting for around 70% of all breast cancer cases. Endocrine therapy (ET) combined with cyclin-dependent kinases (CDK) 4/6 inhibitors is the standard first option in the management of advanced luminal breast cancer independently of disease extension. Classically, patients undergo multiple lines of ET ± targeted treatments until endocrine resistance occurs and palliative chemotherapy is proposed. Understanding endocrine resistance mechanisms and development of novel ET options is one of the main challenges in current clinical research. Another area of utmost interest is the improvement of post-endocrine therapeutic approaches. Among others, the development of antibody-drug conjugates (ADCs) is very promising, and some of these drugs will probably soon become a part of the therapeutic arsenal against this incurable disease. This review paper provides an overview of currently available treatment options in ER+/HER2- metastatic breast cancer and extensively discusses new approaches in late clinical development.


Assuntos
Neoplasias da Mama , Terapia de Alvo Molecular , Feminino , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Imunoconjugados/uso terapêutico , Terapia de Alvo Molecular/tendências , Resistencia a Medicamentos Antineoplásicos , Antineoplásicos/uso terapêutico , Mutação , Moduladores Seletivos de Receptor Estrogênico/uso terapêutico
6.
Expert Rev Hematol ; 15(5): 431-441, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35523283

RESUMO

INTRODUCTION: Hemophilia A (HA) or B (HB) is an X-linked recessive disorder caused by a defect in the factor VIII (FVIII) or factor IX (FIX) gene which leads to the dysfunction of blood coagulation. Protein replacement therapy (PRT) uses recombinant proteins and plasma-derived products, which incurs high cost and inconvenience requiring routine intravenous infusions and life-time treatment. Understanding of detailed molecular mechanisms on FVIII gene function could provide innovative solutions to amend this disorder. In recent decades, gene therapeutics have advanced rapidly and a one-time cure solution has been proposed. AREAS COVERED: This review summarizes current understanding of molecular pathways involved in blood coagulation, with emphasis on FVIII's functional role. The existing knowledge and challenges on FVIII gene expression, from transcription, translation, post-translational modification including glycosylation to protein processing and secretion, and co-factor interactions are deciphered and potential molecular interventions discussed. EXPERT OPINION: This article reviews the potential treatment targets for HA and HB, including antibodies, small molecules and gene therapeutics, based on molecular mechanisms of FVIII biosynthesis, and further, assessing the pros and cons of these various treatment strategies. Understanding detailed FVIII protein synthesis and secretory pathways could provide exciting opportunities in identifying novel therapeutics to ameliorate hemophilia state.


Assuntos
Hemofilia A , Hemofilia B , Proteínas Recombinantes , Fator IX/genética , Fator IX/uso terapêutico , Fator VIII/genética , Fator VIII/uso terapêutico , Hemofilia A/genética , Hemofilia A/terapia , Hemofilia B/genética , Hemofilia B/terapia , Humanos , Terapia de Alvo Molecular/tendências , Proteínas Recombinantes/uso terapêutico
7.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35163273

RESUMO

TMPRSS2 is a type II transmembrane protease with broad expression in epithelial cells of the respiratory and gastrointestinal tract, the prostate, and other organs. Although the physiological role of TMPRSS2 remains largely elusive, several endogenous substrates have been identified. TMPRSS2 serves as a major cofactor in SARS-CoV-2 entry, and primes glycoproteins of other respiratory viruses as well. Consequently, inhibiting TMPRSS2 activity is a promising strategy to block viral infection. In this review, we provide an overview of the role of TMPRSS2 in the entry processes of different respiratory viruses. We then review the different classes of TMPRSS2 inhibitors and their clinical development, with a focus on COVID-19 treatment.


Assuntos
Tratamento Farmacológico da COVID-19 , Terapia de Alvo Molecular/tendências , Serina Endopeptidases/fisiologia , COVID-19/genética , Humanos , Terapia de Alvo Molecular/métodos , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/fisiologia , Serina Endopeptidases/genética , Inibidores de Serino Proteinase/farmacologia , Inibidores de Serino Proteinase/uso terapêutico , Internalização do Vírus/efeitos dos fármacos
8.
Neuropharmacology ; 202: 108870, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34742741

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder and is defined pathologically by the abnormal accumulation of the presynaptic protein alpha-synuclein (aSyn) in the form of Lewy bodies and Lewy neurites and loss of midbrain dopaminergic neurons in the substantia nigra pars compacta. Because of aSyn's involvement in both sporadic and familial forms of PD, it has become a key target for the development of novel therapeutics. Aberrant aSyn is associated with multiple mechanisms of neuronal dysfunction and degeneration including inflammation, impaired mitochondrial function, altered protein degradation systems, and oxidative stress. Inflammation, in particular, has emerged as a potential significant contributor early in the disease making it an attractive target for disease modification and neuroprotection. Thus, immunotherapies targeting aSyn are currently being investigated in pre-clinical and clinical trials. The focus of this review is to highlight the role of aSyn in neuroinflammation and discuss the current status of aSyn-directed immunotherapies in pre-clinical and clinical trials for PD.


Assuntos
Sistema Imunitário/imunologia , Imunoterapia Ativa/métodos , Imunoterapia/métodos , Terapia de Alvo Molecular/métodos , Doença de Parkinson/etiologia , Doença de Parkinson/terapia , alfa-Sinucleína/metabolismo , Animais , Ensaios Clínicos como Assunto , Neurônios Dopaminérgicos/patologia , Humanos , Imunoterapia/tendências , Imunoterapia Ativa/tendências , Corpos de Lewy/metabolismo , Camundongos Transgênicos , Terapia de Alvo Molecular/tendências , Doenças Neuroinflamatórias , Estresse Oxidativo , Doença de Parkinson/imunologia , Substância Negra/metabolismo , Substância Negra/patologia
9.
J Nippon Med Sch ; 89(2): 128-138, 2022 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-34840210

RESUMO

Among recent advances in cancer treatment, the emergence of novel drugs targeting specific molecules has considerably modulated therapeutic strategies. Despite the efficacy of these agents, renal complications that are distinct from those of conventional chemotherapeutic drugs have been reported. Targeted therapy drugs include monoclonal antibodies and small-molecule agents. Bevacizumab is a monoclonal antibody that targets vascular endothelial growth factor (VEGF) and blocks tumor angiogenesis. This anti-angiogenic effect causes endothelial injury, resulting in "thrombotic microangiopathy-like lesions" confined to the glomerulus. Segmental hyalinosis of the glomerular tuft is also observed. Small molecular agents, including tyrosine kinase inhibitors (TKIs) such as pazopanib, can cause endothelial injury and podocytopathy by blocking VEGF receptors and their downstream signaling. Minimal change nephrotic syndrome and focal segmental glomerulosclerosis are associated with TKI-induced renal complications. Immune checkpoint inhibitors (ICIs) such as PD-1, CTLA-4, and PD-L1 modulate immune checkpoints and are a novel form of immunotherapy against cancer. Owing to their unique function, ICIs cause inflammatory side effects referred to as immune-related adverse events (irAEs). irAEs in the kidney include acute tubulointerstitial nephritis and tubulitis, occasionally accompanied by granuloma formation. Vasculitis, thrombotic microangiopathy, and glomerulonephritis have also been reported. Renal toxicity associated with other molecular drugs, such as protease inhibitors and mammalian target of rapamycin inhibitors, has also been documented. In this article, we review the clinicohistopathological aspects of renal complications associated with molecular targeted therapies and focus on anti-VEGF agents and immune checkpoint inhibitors from a pathological perspective.


Assuntos
Anticorpos Monoclonais , Terapia de Alvo Molecular , Neoplasias , Microangiopatias Trombóticas , Anticorpos Monoclonais/efeitos adversos , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Rim/patologia , Terapia de Alvo Molecular/efeitos adversos , Terapia de Alvo Molecular/tendências , Neoplasias/tratamento farmacológico , Microangiopatias Trombóticas/induzido quimicamente , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
10.
Drug Discov Today ; 27(1): 17-30, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34537333

RESUMO

Durable cell and gene therapies potentially transform patient lives, but payers fear unsustainable costs arising from the more than 1000 therapies in the development pipeline. A novel multi-module Markov chain Monte Carlo-based model projects product-indication approvals, treated patients, and product revenues. We estimate a mean 63.5 (54-74 5th to 95th percentile range) cumulative US product-indication approvals through 2030, with a mean 93000 patients treated in 2030 generating a mean US$24.4 billion (US$17.0B-35.0B, US$73.0B extreme) list price product revenues not including ancillary medical costs or cost offsets. Thus, the likely dozens of durable cell and gene therapies developed through 2030 are unlikely to threaten US health system financial sustainability.


Assuntos
Produtos Biológicos , Custos de Medicamentos/tendências , Terapia Genética , Terapia de Alvo Molecular , Produtos Biológicos/economia , Produtos Biológicos/farmacologia , Aprovação de Drogas , Previsões , Terapia Genética/métodos , Terapia Genética/tendências , Humanos , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Estados Unidos
11.
Drug Discov Today ; 27(1): 82-101, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34252612

RESUMO

WNT/ß-catenin signaling orchestrates various physiological processes, including embryonic development, growth, tissue homeostasis, and regeneration. Abnormal WNT/ß-catenin signaling is associated with various cancers and its inhibition has shown effective antitumor responses. In this review, we discuss the pathway, potential targets for the development of WNT/ß-catenin inhibitors, available inhibitors, and their specific molecular interactions with the target proteins. We also discuss inhibitors that are in clinical trials and describe potential new avenues for therapeutically targeting the WNT/ß-catenin pathway. Furthermore, we introduce emerging strategies, including artificial intelligence (AI)-assisted tools and technology-based actionable approaches, to translate WNT/ß-catenin inhibitors to the clinic for cancer therapy.


Assuntos
Produtos Biológicos/farmacologia , Terapia de Alvo Molecular , Neoplasias , Via de Sinalização Wnt , Desenho de Fármacos , Humanos , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/fisiologia
12.
Br J Haematol ; 196(3): 488-506, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34472091

RESUMO

Treatment outcomes in multiple myeloma (MM) have improved dramatically over the past 10 years. However, patients with high-risk disease such as those with Stage III disease by the Revised International Staging System, the presence of adverse cytogenetics, or who are refractory to proteosome inhibitors, immunomodulatory drugs and monoclonal antibodies may have dismal outcomes. These patients represent an urgent ongoing need in MM. One of the hallmarks of MM is immune dysfunction and a tumour-permissive immune microenvironment. Ameliorating the immune-paresis could lead to improved outcomes. The role of immunotherapies has been growing at an exponential pace with numerous agents under development in clinical trials. In the present review, we provide an overview of immunotherapies in MM, focussing on bispecific antibodies (BsAbs). We review efficacy outcomes from the published clinical trials and consider the important safety aspects of these therapies, in particular the risk of cytokine-release syndrome and immune effector cell-associated neurotoxicity syndrome, and how these compare with patients receiving chimeric antigen receptor T cells. We discuss the MM epitopes being targeted by BsAbs, either in clinical or preclinical stages, and we consider where these therapies might best fit within the future ever-changing paradigm of MM treatment.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Imunoterapia , Mieloma Múltiplo/terapia , Animais , Anticorpos Biespecíficos/farmacologia , Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/farmacologia , Tomada de Decisão Clínica , Terapia Combinada/efeitos adversos , Terapia Combinada/métodos , Gerenciamento Clínico , Humanos , Imunoterapia/métodos , Imunoterapia/tendências , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/etiologia , Mieloma Múltiplo/mortalidade , Prognóstico , Resultado do Tratamento
13.
Drug Discov Today ; 27(1): 246-256, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34438075

RESUMO

Bromodomain-containing protein 4 (BRD4) is emerging as a therapeutic target that acts synergistically with other targets of small-molecule drugs in cancer. Therefore, the discovery of potential new dual-target inhibitors of BRD4 may be a promising strategy for cancer therapy. In this review, we highlight a series of strategies to design therapeutic dual-target inhibitors of BRD4 that focus on the synergistic functions of this protein. Drug combinations that exploit synthetic lethality, protein-protein interactions, functional complementarity, and blocking of resistance mechanisms could ultimately overcome the barriers inherent to the development of BRD4 inhibitors as future cancer drugs.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular , Desenho de Fármacos/métodos , Terapia de Alvo Molecular , Neoplasias , Fatores de Transcrição , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/metabolismo , Humanos , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Relação Estrutura-Atividade , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo
15.
Diabetes ; 70(12): 2721-2732, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34872936

RESUMO

Life-threatening hypoglycemia is a limiting factor in the management of type 1 diabetes. People with diabetes are prone to develop hypoglycemia because they lose physiological mechanisms that prevent plasma glucose levels from falling. Among these so-called counterregulatory responses, secretion of glucagon from pancreatic α-cells is preeminent. Glucagon, a hormone secreted in response to a lowering in glucose concentration, counteracts a further drop in glycemia by promoting gluconeogenesis and glycogenolysis in target tissues. In diabetes, however, α-cells do not respond appropriately to changes in glycemia and, thus, cannot mount a counterregulatory response. If the α-cell could be targeted therapeutically to restore its ability to prevent hypoglycemia, type 1 diabetes could be managed more efficiently and safely. Unfortunately, the mechanisms that allow the α-cell to respond to hypoglycemia have not been fully elucidated. We know even less about the pathophysiological mechanisms that cause α-cell dysfunction in diabetes. Based on published findings and unpublished observations, and taking into account its electrophysiological properties, we propose here a model of α-cell function that could explain its impairment in diabetes. Within this frame, we emphasize those elements that could be targeted pharmacologically with repurposed U.S. Food and Drug Administration-approved drugs to rescue α-cell function and restore glucose counterregulation in people with diabetes.


Assuntos
Diabetes Mellitus Tipo 1/terapia , Células Secretoras de Glucagon/efeitos dos fármacos , Hipoglicemia/prevenção & controle , Terapia de Alvo Molecular/métodos , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/complicações , Glucagon/metabolismo , Células Secretoras de Glucagon/metabolismo , Humanos , Hipoglicemia/sangue , Hipoglicemia/etiologia , Insulina/farmacologia , Insulina/uso terapêutico , Terapia de Alvo Molecular/tendências
16.
Front Endocrinol (Lausanne) ; 12: 777075, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34950105

RESUMO

Non-alcoholic fatty liver disease (NAFLD) is a continuous progression of pathophysiologic stages that is challenging to diagnose due to its inherent heterogeneity and poor standardization across a wide variety of diagnostic measures. NAFLD is heritable, and several loci have been robustly associated with various stages of disease. In the past few years, larger genetic association studies using new methodology have identified novel genes associated with NAFLD, some of which have shown therapeutic promise. This mini-review provides an overview of the heterogeneity in NAFLD phenotypes and diagnostic methods, discusses genetic associations in relation to the specific stages for which they were identified, and offers a perspective on the design of future genetic mapping studies to accelerate therapeutic target identification.


Assuntos
Genética Populacional , Terapia de Alvo Molecular/métodos , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/terapia , Biomarcadores/metabolismo , Estudos de Casos e Controles , Técnicas de Diagnóstico Endócrino/tendências , Progressão da Doença , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Genética Humana/métodos , Genética Humana/tendências , Humanos , Terapia de Alvo Molecular/tendências , Hepatopatia Gordurosa não Alcoólica/diagnóstico , Hepatopatia Gordurosa não Alcoólica/epidemiologia , Polimorfismo de Nucleotídeo Único
17.
Am J Psychiatry ; 178(11): 1014-1025, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34734743

RESUMO

Alzheimer's disease is a neuropsychiatric disorder with devastating clinical and socioeconomic consequences. Since the original description of the neuropathological correlates of the disorder, neuritic plaques and neurofibrillary tangles have been presumed to be critical to the underlying pathophysiology of the illness. The authors review the clinical and neuropathological origins of Alzheimer's disease and trace the evolution of modern biomarkers from their historical roots. They describe how technological innovations such as neuroimaging and biochemical assays have been used to measure and quantify key proteins and lipids in the brain, cerebrospinal fluid, and blood and advance their role as biomarkers of Alzheimer's disease. Together with genomics, these approaches have led to the development of a thematic and focused science in the area of degenerative disorders. The authors conclude by drawing distinctions between legitimate biomarkers of disease and molecular targets for therapeutic intervention and discuss future approaches to this complex neurobehavioral illness.


Assuntos
Doença de Alzheimer , Peptídeos beta-Amiloides/metabolismo , Encéfalo , Terapia de Alvo Molecular , Proteínas tau/metabolismo , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Biomarcadores/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências , Neuroimagem/métodos
18.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34768969

RESUMO

Inherited retinal degenerations (IRDs) are a diverse group of conditions that are often characterized by the loss of photoreceptors and blindness. Recent innovations in molecular biology and genomics have allowed us to identify the causative defects behind these dystrophies and to design therapeutics that target specific mechanisms of retinal disease. Recently, the FDA approved the first in vivo gene therapy for one of these hereditary blinding conditions. Current clinical trials are exploring new therapies that could provide treatment for a growing number of retinal dystrophies. While the field has had early success with gene augmentation strategies for treating retinal disease based on loss-of-function mutations, many novel approaches hold the promise of offering therapies that span the full spectrum of causative mutations and mechanisms. Here, we provide a comprehensive review of the approaches currently in development including a discussion of retinal neuroprotection, gene therapies (gene augmentation, gene editing, RNA modification, optogenetics), and regenerative stem or precursor cell-based therapies. Our review focuses on technologies that are being developed for clinical translation or are in active clinical trials and discusses the advantages and limitations for each approach.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/tendências , Terapia de Alvo Molecular/tendências , Degeneração Retiniana/genética , Degeneração Retiniana/terapia , Edição de Genes/tendências , Terapia Genética/tendências , Humanos , Neuroproteção , Optogenética/tendências , Medicina Regenerativa/tendências
19.
Int J Mol Sci ; 22(21)2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34768978

RESUMO

Histologically, bladder cancer is a heterogeneous group comprising urothelial carcinoma (UC), squamous cell carcinoma, adenocarcinomas (ACs), urachal carcinomas (UrCs), and small cell neuroendocrine carcinomas (SCCs). However, all bladder cancers have been treated so far uniformly, and targeted therapy options are still limited. Thus, we aimed to determine the protein expression/molecular status of commonly used cancer targets (programmed cell death 1 ligand 1 (PD-L1), mismatch repair (MMR), androgen and estrogen receptors (AR/ER), Nectin-4, tumor-associated calcium signal transducer 2 (Tacstd2, Trop-2), epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and fibroblast growth factor receptor 3 (FGFR3)) to give first insights into whether patients with SCC, AC/UrCs, and squamous-differentiated carcinomas (Sq-BLCA) of the bladder could be eligible for targeted therapies. In addition, for MMR-deficient tumors, microsatellite instability was analyzed. We completed our own data with molecular data from The Cancer Genome Atlas (TCGA). We present ratios for each drug and cumulative ratios for multiple therapeutic options for each nonurothelial subtype. For example, 58.9% of SCC patients, 33.5% of AC/UrCs patients, and 79.3% of Sq-BLCA patients would be eligible for at least one of the analyzed targets. In conclusion, our findings hold promise for targeted therapeutic approaches in selected patients in the future, as various drugs could be applied according to the biomarker status.


Assuntos
Terapia de Alvo Molecular/métodos , Neoplasias da Bexiga Urinária/tratamento farmacológico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Idoso , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células de Transição/tratamento farmacológico , Carcinoma de Células de Transição/genética , Carcinoma de Células de Transição/metabolismo , Reparo de Erro de Pareamento de DNA , Feminino , Humanos , Imuno-Histoquímica , Masculino , Instabilidade de Microssatélites , Terapia de Alvo Molecular/tendências , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia
20.
Genes (Basel) ; 12(11)2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34828306

RESUMO

Autism spectrum disorder (ASD) is a genetically heterogenous neurodevelopmental disorder. In the early years of next-generation sequencing, de novo germline variants were shown to contribute to ASD risk. These germline mutations are present in all of the cells of an affected individual and can be detected in any tissue, including clinically accessible DNA sources such as blood or saliva. In recent years, studies have also implicated de novo somatic variants in ASD risk. These somatic mutations arise postzygotically and are present in only a subset of the cells of an affected individual. Depending on the developmental time and progenitor cell in which a somatic mutation occurs, it may be detectable in some tissues and not in others. Somatic mutations detectable at relatively low sequencing coverage in clinically accessible tissues are suggested to contribute to 3-5% of simplex ASD diagnoses, and "brain limited" somatic mutations have been identified in postmortem ASD brain tissue. Somatic mutations likely represent the genetic diagnosis in a proportion of otherwise unexplained individuals with ASD, and brain limited somatic mutations can be used as markers to discover risk genes, cell types, brain regions, and cellular pathways important for ASD pathogenesis and to potentially target for therapeutics.


Assuntos
Transtorno do Espectro Autista/genética , Mosaicismo , Transtorno do Espectro Autista/diagnóstico , Transtorno do Espectro Autista/psicologia , Transtorno do Espectro Autista/terapia , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Terapia de Alvo Molecular/métodos , Terapia de Alvo Molecular/tendências
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...