Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.314
1.
Neurosurg Focus ; 56(5): E15, 2024 May.
Article En | MEDLINE | ID: mdl-38691867

The role of systemic therapy in primary or advanced and metastatic chordoma has been traditionally limited because of the inherent resistance to cytotoxic therapies and lack of specific or effective therapeutic targets. Despite resection and adjuvant radiation therapy, local recurrence rates in clival chordoma remain high and the risk of systemic metastases is not trivial, leading to significant morbidity and mortality. Recently, molecular targeted therapies (MTTs) and immune checkpoint inhibitors (ICIs) have emerged as promising therapeutic avenues in chordoma. In recent years, preclinical studies have identified potential targets based on intrinsic genetic dependencies, epigenetic modulators, or newly identified tumor-associated cell populations driving treatment resistance and recurrence. Nonetheless, the role of systemic therapies in the neoadjuvant or adjuvant setting for primary, locally progressive, and distant metastatic chordomas is still being investigated. Herein, an overview of current and emerging systemic treatment strategies in advanced clival chordoma is provided. Furthermore, several molecular biomarkers have been recently uncovered as potential predictors of the response to specific molecular therapeutics. The authors describe the recently discovered role of 1p36 and 9p21 deletions as biomarkers capable of guiding drug selection. Then they discuss completed and ongoing clinical trials of MTTs, including several tyrosine kinase inhibitors used as monotherapy or in combination, such as imatinib, sorafenib, dasatinib, and lapatinib, among others, as well as mammalian target of rapamycin inhibitors such as everolimus and rapamycin. They present their experience and other recent studies demonstrating vast benefits in advanced chordoma from ICIs. Additionally, they provide a brief overview of novel systemic strategies such as adoptive cell transfer (CAR-T and NK cells), oncolytic viruses, epigenetic targeting (KDM6, HDAC, and EZH2 inhibitors), and several promising preclinical studies with high translational potential. Finally, the authors present their institutional multidisciplinary protocol for the incorporation of systemic therapy for both newly diagnosed and recurrent chordomas based on molecular studies including upfront enrollment in MTT trials in patients with epidermal growth factor receptor upregulation or INI-1 deficiency or enrollment in ICI clinical trials for patients with high tumor mutational burden or high PD-L1 expression on tumor cells or in the tumor microenvironment.


Chordoma , Skull Base Neoplasms , Humans , Chordoma/therapy , Chordoma/drug therapy , Skull Base Neoplasms/therapy , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Immune Checkpoint Inhibitors/therapeutic use
2.
J Virol ; 97(11): e0149723, 2023 Nov 30.
Article En | MEDLINE | ID: mdl-37877719

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.


Apoptosis , Ducks , Flavivirus Infections , Flavivirus , Host Specificity , Animals , Humans , Antiviral Agents/pharmacology , Ducks/virology , eIF-2 Kinase/metabolism , Flavivirus/enzymology , Flavivirus/pathogenicity , Flavivirus Infections/diagnosis , Flavivirus Infections/immunology , Flavivirus Infections/transmission , Flavivirus Infections/virology , Mitochondria/metabolism , Molecular Targeted Therapy/trends , Viral Zoonoses/diagnosis , Viral Zoonoses/immunology , Viral Zoonoses/transmission , Viral Zoonoses/virology , Voltage-Dependent Anion Channel 2/metabolism
3.
ESMO Open ; 8(3): 101573, 2023 Jun.
Article En | MEDLINE | ID: mdl-37263082

Increasing evidence suggests that patients with diabetes, particularly type 2 diabetes (T2D), are characterized by an increased risk of developing different types of cancer, so cancer could be proposed as a new T2D-related complication. On the other hand, cancer may also increase the risk of developing new-onset diabetes, mainly caused by anticancer therapies. Hyperinsulinemia, hyperglycemia, and chronic inflammation typical of T2D could represent possible mechanisms involved in cancer development in diabetic patients. MicroRNAs (miRNAs) are a subset of non-coding RNAs, ⁓22 nucleotides in length, which control the post-transcriptional regulation of gene expression through both translational repression and messenger RNA degradation. Of note, miRNAs have multiple target genes and alteration of their expression has been reported in multiple diseases, including T2D and cancer. Accordingly, specific miRNA-regulated pathways are involved in the pathogenesis of both conditions. In this review, a panel of experts from the Italian Association of Medical Oncology (AIOM), Italian Association of Medical Diabetologists (AMD), Italian Society of Diabetology (SID), Italian Society of Endocrinology (SIE), and Italian Society of Pharmacology (SIF) provide a critical view of the evidence about the involvement of miRNAs in the pathophysiology of both T2D and cancer, trying to identify the shared miRNA signature and pathways able to explain the strong correlation between the two conditions, as well as to envision new common pharmacological approaches.


Diabetes Mellitus, Type 2 , MicroRNAs , Neoplasms , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/therapy , Neoplasms/complications , Neoplasms/genetics , Neoplasms/therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Insulin-Secreting Cells/pathology , Insulin Resistance/genetics , Molecular Targeted Therapy/trends
4.
Int J Mol Sci ; 24(9)2023 May 04.
Article En | MEDLINE | ID: mdl-37175935

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.


Alzheimer Disease , Chemokine CXCL1 , Molecular Targeted Therapy , Signal Transduction , Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Molecular Targeted Therapy/trends , Microglia/physiology , Chemokine CXCL1/metabolism , CX3C Chemokine Receptor 1/metabolism
5.
ESMO Open ; 8(2): 100882, 2023 04.
Article En | MEDLINE | ID: mdl-36806375

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.


Breast Neoplasms , Molecular Targeted Therapy , Female , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Immunoconjugates/therapeutic use , Molecular Targeted Therapy/trends , Drug Resistance, Neoplasm , Antineoplastic Agents/therapeutic use , Mutation , Selective Estrogen Receptor Modulators/therapeutic use
6.
Blood Rev ; 60: 101056, 2023 07.
Article En | MEDLINE | ID: mdl-36805300

Approval of new agents to treat higher risk (HR) myelodysplastic syndrome (MDS) has stalled since the approval of DNA methyltransferase inhibitors (DNMTi). In addition, the options for patients with lower risk (LR) MDS who have high transfusion needs and do not harbor ring sideroblasts or 5q- syndrome are limited. Here, we review the current treatment landscape in MDS and identify areas of unmet need, such as treatment after failure of erythropoiesis-stimulating agents or DNMTis, TP53-mutated disease, and MDS with potentially targetable mutations. We discuss how our understanding of MDS pathogenesis can inform therapy development, including treating HR-MDS similarly to AML and pursuing therapies to address splicing factor mutations and dysregulated inflammation. We then bring a critical lens to current methodology of MDS studies and propose solutions to improve the efficiency and yield of these clinical trials, including using the most meaningful response metrics and expanding enrollment.


Drug Development , Myelodysplastic Syndromes , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/physiopathology , Drug Development/standards , Drug Development/trends , Mutation , Molecular Targeted Therapy/trends , Clinical Trials as Topic , Humans
9.
Expert Rev Hematol ; 15(5): 431-441, 2022 05.
Article En | MEDLINE | ID: mdl-35523283

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.


Hemophilia A , Hemophilia B , Recombinant Proteins , Factor IX/genetics , Factor IX/therapeutic use , Factor VIII/genetics , Factor VIII/therapeutic use , Hemophilia A/genetics , Hemophilia A/therapy , Hemophilia B/genetics , Hemophilia B/therapy , Humans , Molecular Targeted Therapy/trends , Recombinant Proteins/therapeutic use
10.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article En | MEDLINE | ID: mdl-35163273

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.


COVID-19 Drug Treatment , Molecular Targeted Therapy/trends , Serine Endopeptidases/physiology , COVID-19/genetics , Humans , Molecular Targeted Therapy/methods , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Serine Endopeptidases/genetics , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/therapeutic use , Virus Internalization/drug effects
11.
Drug Discov Today ; 27(1): 17-30, 2022 01.
Article En | MEDLINE | ID: mdl-34537333

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.


Biological Products , Drug Costs/trends , Genetic Therapy , Molecular Targeted Therapy , Biological Products/economics , Biological Products/pharmacology , Drug Approval , Forecasting , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , United States
13.
Drug Discov Today ; 27(1): 246-256, 2022 01.
Article En | MEDLINE | ID: mdl-34438075

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.


Antineoplastic Agents/pharmacology , Cell Cycle Proteins , Drug Design/methods , Molecular Targeted Therapy , Neoplasms , Transcription Factors , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Neoplasms/drug therapy , Neoplasms/metabolism , Structure-Activity Relationship , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
14.
Br J Haematol ; 196(3): 488-506, 2022 02.
Article En | MEDLINE | ID: mdl-34472091

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.


Antineoplastic Agents, Immunological/therapeutic use , Immunotherapy , Multiple Myeloma/therapy , Animals , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/pharmacology , Clinical Decision-Making , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Disease Management , Humans , Immunotherapy/methods , Immunotherapy/trends , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Multiple Myeloma/diagnosis , Multiple Myeloma/etiology , Multiple Myeloma/mortality , Prognosis , Treatment Outcome
15.
Drug Discov Today ; 27(1): 82-101, 2022 01.
Article En | MEDLINE | ID: mdl-34252612

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.


Biological Products/pharmacology , Molecular Targeted Therapy , Neoplasms , Wnt Signaling Pathway , Drug Design , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Neoplasms/drug therapy , Neoplasms/metabolism , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology
16.
Neuropharmacology ; 202: 108870, 2022 01 01.
Article En | MEDLINE | ID: mdl-34742741

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.


Immune System/immunology , Immunotherapy, Active/methods , Immunotherapy/methods , Molecular Targeted Therapy/methods , Parkinson Disease/etiology , Parkinson Disease/therapy , alpha-Synuclein/metabolism , Animals , Clinical Trials as Topic , Dopaminergic Neurons/pathology , Humans , Immunotherapy/trends , Immunotherapy, Active/trends , Lewy Bodies/metabolism , Mice, Transgenic , Molecular Targeted Therapy/trends , Neuroinflammatory Diseases , Oxidative Stress , Parkinson Disease/immunology , Substantia Nigra/metabolism , Substantia Nigra/pathology
17.
J Nippon Med Sch ; 89(2): 128-138, 2022 May 12.
Article En | MEDLINE | ID: mdl-34840210

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.


Antibodies, Monoclonal , Molecular Targeted Therapy , Neoplasms , Thrombotic Microangiopathies , Antibodies, Monoclonal/adverse effects , Humans , Immune Checkpoint Inhibitors/adverse effects , Kidney/pathology , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/trends , Neoplasms/drug therapy , Thrombotic Microangiopathies/chemically induced , Vascular Endothelial Growth Factor A/antagonists & inhibitors
18.
Front Endocrinol (Lausanne) ; 12: 777075, 2021.
Article En | MEDLINE | ID: mdl-34950105

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.


Genetics, Population , Molecular Targeted Therapy/methods , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/therapy , Biomarkers/metabolism , Case-Control Studies , Diagnostic Techniques, Endocrine/trends , Disease Progression , Genetic Predisposition to Disease , Genome-Wide Association Study , Human Genetics/methods , Human Genetics/trends , Humans , Molecular Targeted Therapy/trends , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/epidemiology , Polymorphism, Single Nucleotide
19.
Diabetes ; 70(12): 2721-2732, 2021 12.
Article En | MEDLINE | ID: mdl-34872936

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.


Diabetes Mellitus, Type 1/therapy , Glucagon-Secreting Cells/drug effects , Hypoglycemia/prevention & control , Molecular Targeted Therapy/methods , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/complications , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Humans , Hypoglycemia/blood , Hypoglycemia/etiology , Insulin/pharmacology , Insulin/therapeutic use , Molecular Targeted Therapy/trends
20.
Genes (Basel) ; 12(11)2021 10 26.
Article En | MEDLINE | ID: mdl-34828306

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.


Autism Spectrum Disorder/genetics , Mosaicism , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/psychology , Autism Spectrum Disorder/therapy , Genetic Predisposition to Disease , Germ-Line Mutation , High-Throughput Nucleotide Sequencing , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends
...