Analysis of the influence of DYRK1A inhibitors on proliferation of INS-1E and MIN6 cells. In Ki67 staining, 5 μM of each inhibitor and harmine as a positive control were used. Data are expressed as mean ± SEM; * p < 0.05, ** p < 0.01, *** p < 0.001 compared to the control.

Analysis of the influence of DYRK1A inhibitors on proliferation of INS-1E and MIN6 cells. In Ki67 staining, 5 μM of each inhibitor and harmine as a positive control were used. Data are expressed as mean ± SEM; * p < 0.05, ** p < 0.01, *** p < 0.001 compared to the control.

Source publication
Article
Full-text available
The rising prevalence of diabetes is threatening global health. It is known not only for the occurrence of severe complications but also for the SARS-Cov-2 pandemic, which shows that it exacerbates susceptibility to infections. Current therapies focus on artificially maintaining insulin homeostasis, and a durable cure has not yet been achieved. We...

Contexts in source publication

Context 1
... study the impact of DYRK1A on β-cell replication, we incubated INS-1E and MIN6 cells with DYRK1A inhibitors ( Figure 5). The rate of proliferation of INS-1E and MIN6 cells was assessed by the Ki67 staining analysis. ...
Context 2
... we tested DYRK1A inhibitors in an in vitro GSIS assay (Figure 7). Harmine was used as a control as it causes enhanced insulin secretion and increases C-peptide levels ( Figure S5). Treatment of INS-1E and MIN6 cells with each compound increased insulin release at low-and high-glucose challenges. ...

Citations

... While DYRK1A affects the cell cycle state of neurons and plays an essential role in neurogenesis 8 , it was also found to regulate cyclin-dependent kinase-1 (CDK1) activity in glioblastoma cells and DYRK1A inhibition exhibit anti-tumor effects in glioblastoma 9,10 . The therapeutic use of DYRK1A-mediated cell cycle regulation was recently extended to other diseases, such as diabetes and myocardial infarction, as DYRK1A inhibition was found to sustain pancreatic beta cells and cardiomyocytes growth [11][12][13][14][15][16] . Despite having important cell cycle regulatory functions, a role for DYRK1A in cancer, other than glioblastoma remains to be fully investigated. ...
Article
Full-text available
Metastatic cancer remains incurable as patients eventually loose sensitivity to targeted therapies and chemotherapies, further leading to poor clinical outcome. Thus, there is a clear medical gap and urgent need to develop efficient and improved targeted therapies for cancer patients. In this study, we investigated the role of DYRK1A kinase in regulating cancer progression and evaluated the therapeutic potential of DYRK1A inhibition in invasive solid tumors, including colon and triple-negative breast cancers. We uncovered new roles played by the DYRK1A kinase. We found that blocking DYRK1A gene expression or pharmacological inhibition of its kinase activity via harmine efficiently blocked primary tumor formation and the metastatic tumor spread in preclinical models of breast and colon cancers. Further assessing the underlying molecular mechanisms, we found that DYRK1A inhibition resulted in increased expression of the G1/S cell cycle regulators while decreasing expression of the G2/M regulators. Combined, these effects release cancer cells from quiescence, leading to their accumulation in G1/S and further delaying/preventing their progression toward G2/M, ultimately leading to growth arrest and tumor growth inhibition. Furthermore, we show that accumulation of cancer cells in G1/S upon DYRK1A inhibition led to significant potentiation of G1/S targeting chemotherapy drug responses in vitro and in vivo. This study underscores the potential for developing novel DYRK1A-targeting therapies in colon and breast cancers and, at the same time, further defines DYRK1A pharmacological inhibition as a viable and powerful combinatorial treatment approach for improving G1/S targeting chemotherapy drugs treatments in solid tumors.
... While DYRK1A affects the cell cycle state of neurons and plays an essential role in neurogenesis 8 , it was also found to regulate cyclin-dependent kinase-1 (CDK1) activity in glioblastoma cells and DYRK1A inhibition exhibit anti-tumor effects in glioblastoma 9,10 . The therapeutic use of DYRK1A-mediated cell cycle regulation was recently extended to other diseases, such as diabetes and myocardial infarction, as DYRK1A inhibition was found to sustain pancreatic beta cells and cardiomyocytes growth [11][12][13][14][15][16] . Despite having important cell cycle regulatory functions, a role for DYRK1A in cancer, other than glioblastoma remains to be fully investigated. ...
Article
Full-text available
Metastatic cancer remains incurable as patients eventually loose sensitivity to targeted therapies and chemotherapies, further leading to poor clinical outcome. Thus, there is a clear medical gap and urgent need to develop efficient and improved targeted therapies for cancer patients. In this study, we investigated the role of DYRK1A kinase in regulating cancer progression and evaluated the therapeutic potential of DYRK1A inhibition in invasive solid tumors, including colon and triple-negative breast cancers. We uncovered new roles played by the DYRK1A kinase. We found that blocking DYRK1A gene expression or pharmacological inhibition of its kinase activity via harmine efficiently blocked primary tumor formation and the metastatic tumor spread in preclinical models of breast and colon cancers. Further assessing the underlying molecular mechanisms, we found that DYRK1A inhibition resulted in increased expression of the G1/S cell cycle regulators while decreasing expression of the G2/M regulators. Combined, these effects release cancer cells from quiescence, leading to their accumulation in G1/S and further delaying/preventing their progression toward G2/M, ultimately leading to growth arrest and tumor growth inhibition. Furthermore, we show that accumulation of cancer cells in G1/S upon DYRK1A inhibition led to significant potentiation of G1/S targeting chemotherapy drug responses in vitro and in vivo. This study underscores the potential for developing novel DYRK1A-targeting therapies in colon and breast cancers and, at the same time, further defines DYRK1A pharmacological inhibition as a viable and powerful combinatorial treatment approach for improving G1/S targeting chemotherapy drugs treatments in solid tumors. Cell cycle is one of the main pathways to be dysregulated in tumorigenesis and cancer progression. As such targeting cell cycle remains a favored approach for developing anti-cancer therapeutics 1. Protein phosphorylation has been implicated in carcinogenesis by regulating many cellular processes such as proliferation, apoptosis, differentiation, and metabolism 2. Because protein kinases are easily druggable, extensive efforts have been spent to explore their potential as targeted therapy in various types of cancer and in fact, many current anti-cancer drugs and treatments rely upon protein kinase inhibition 3. The dual-specificity tyrosine phosphorylation-regulated kinases (DYRKs) can auto-phosphorylate their activation loop on tyrosine residue while phosphorylating their specific substrates on threonine and serine residues 4. Of all the 7 DYRK family members, DYRK1A has been the most extensively studied 5. The DYRK1A gene maps to human chromosome 21 within the Down syndrome critical region (DSCR) and its overexpression has been implicated in neuronal development deficits and brain abnormalities in Down syndrome 6. DYRK1A regulates cell cycle and differentiation of neuronal cells by inducing G0/G1 arrest through phosphorylation and subsequent degradation of cyclin D as well as through stabilization of the
... In vitro and in vivo, Harmine and INDY stimulated human beta cells to enter the cell cycle, with beta cell labeling indices that are similar to those seen in people during the first year of life (54). Harmine, Torin and other DYRK1A inhibitors found to promote the activity and proliferation of β cell during study of their effects on cell culture (55). Harmine increased β cell mass and regeneration in a mice model and improved glycemic control and β cell proliferation in vivo in two additional typical human islet transplant models, one euglycemic and one diabetic (54). ...
Article
Background: Diabetes mellitus is a common metabolic disorder characterized by chronic high blood sugar levels due to impaired insulin secretion or action. Existing diabetic medications have limitations, including high costs and the risk of hypoglycemia. Aim: To overcome these challenges, researchers are exploring advanced treatments, and one potential path is studying plants and natural sources. Many plants include green tea (Camellia sinensis), rich in catechin derivatives, particularly epigallocatechin-3-gallate (EGCG), have shown promising effect because this agent may enhance beta cell proliferation, so it can produce dramatic response in management of diabetes mellitus and it is expected to reduce complication of this disease. Thorough data searching from September 2021 to June 2023 was used to conduct this study. The key terms diabetes mellitus, herbal treatment of diabetes, DYRK1A inhibitor, Epigallocatechin-3-gallate, and beta cell proliferation were concomitantly searched in Google Scholar, Web of Science, and PubMed in order to find relevant material. The publications that are presented here were published between 2014 and 2023. Conclusion: Collectively EGCG properties as a DYRK1A inhibitor may enhance β cell proliferation that is promising effects in diabetes mellitus treatment
... DYRK1A regulates multiple key biochemical pathways involved in neurodegenerative diseases and tumorigenesis, and has been identified as a therapeutic target for Alzheimer's disease, leukemia, pancreatic ductal adenocarcinoma, and many other malignancies 9,28,29 . Powerful regulation of pancreatic β cells and cardiomyocyte proliferation further makes it a promising target for promoting cardiac and pancreatic β cell regeneration in heart injury and diabetes 16,30 . We report for the first time that DYRK1A regulates pulmonary vascular remodeling and may represent a novel therapeutic target for PAH. ...
Article
Full-text available
Pulmonary arterial hypertension (PAH) is characterized by progressive vascular remodeling caused by the excessive proliferation and survival of pulmonary artery smooth muscle cells (PASMCs). Dual-specificity tyrosine regulated kinase 1A (DYRK1A) is a pleiotropic kinase involved in the regulation of multiple biological functions, including cell proliferation and survival. However, the role and underlying mechanisms of DYRK1A in PAH pathogenesis remain unclear. We found that DYRK1A was upregulated in PASMCs in response to hypoxia, both in vivo and in vitro. Inhibition of DYRK1A by harmine significantly attenuated hypoxia-induced pulmonary hypertension and pulmonary artery remodeling. Mechanistically, we found that DYRK1A promoted pulmonary arterial remodeling by enhancing the proliferation and survival of PASMCs through activating the STAT3/Pim-1/NFAT pathway, because STAT3 gain-of-function via adeno-associated virus serotype 2 (AAV2) carrying the constitutively active form of STAT3 (STAT3C) nearly abolished the protective effect of harmine on PAH. Collectively, our results reveal a significant role for DYRK1A in pulmonary arterial remodeling and suggest it as a drug target with translational potential for the treatment of PAH.
... The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) (Invitrogen) assay was used to quantify cell survival and inhibitor-mediated cytotoxicity as described previously 37 . After cell attachment, compounds in the growth medium at concentrations from 0 to 100 μM were added to the cell cultures. ...
... Glucose-Stimulated Insulin Secretion assay was carried out as described previously 37 . Briefly, cells were cultured in a low glucose medium (5.5 mM) for 24 h. ...
... The presence of insulin and Ki-67 was assessed in MIN6 pancreatic cell line as described previously 37 . Before imaging, cells were seeded onto slides at a density of 1 × 10 5 cells and maintained at 37 °C in 95% atmospheric air and 5% CO 2 in a humidified atmosphere for 24 h. ...
Article
Full-text available
The selective inhibition of kinases from the diabetic kinome is known to promote the regeneration of beta cells and provide an opportunity for the curative treatment of diabetes. The effect can be achieved by carefully tailoring the selectivity of inhibitor toward a particular kinase, especially DYRK1A, previously associated with Down syndrome and Alzheimer's disease. Recently DYRK1A inhibition has been shown to promote both insulin secretion and beta cells proliferation. Here, we show that commonly available flavones are effective inhibitors of DYRK1A. The observed biochemical activity of flavone compounds is confirmed by crystal structures solved at 2.06 Å and 2.32 Å resolution, deciphering the way inhibitors bind in the ATP-binding pocket of the kinase, which is driven by the arrangement of hydroxyl moieties. We also demonstrate antidiabetic properties of these biomolecules and prove that they could be further improved by therapy combined with TGF-β inhibitors. Our data will allow future structure-based optimization of the presented scaffolds toward potent, bioavailable and selective anti-diabetic drugs.
... This improvement of glucose homeostasis by the synergistic inhibition of DYRK1A and GSK3β hints at an opportunity for the curative therapy of diabetes. 23 −25 DYRK1A and GSK3β are serine−threonine kinases of the CMGC family and share many structural features. 26 This opens the possibility for developing dual inhibitors, which is especially attractive since the latter may benefit from the synergistic effects mentioned above. ...
Article
A clinical casein kinase 2 inhibitor, CX-4945 (silmitasertib), shows significant affinity toward the DYRK1A and GSK3β kinases, involved in down syndrome phenotypes, Alzheimer's disease, circadian clock regulation, and diabetes. This off-target activity offers an opportunity for studying the effect of the DYRK1A/GSK3β kinase system in disease biology and possible line extension. Motivated by the dual inhibition of these kinases, we solved and analyzed the crystal structures of DYRK1A and GSK3β with CX-4945. We built a quantum-chemistry-based model to rationalize the compound affinity for CK2α, DYRK1A, and GSK3β kinases. Our calculations identified a key element for CK2α's subnanomolar affinity to CX-4945. The methodology is expandable to other kinase selectivity modeling. We show that the inhibitor limits DYRK1A- and GSK3β-mediated cyclin D1 phosphorylation and reduces kinase-mediated NFAT signaling in the cell. Given the CX-4945's clinical and pharmacological profile, this inhibitory activity makes it an interesting candidate with potential for application in additional disease areas.
... However, numerous biological targets have been studied in this context, including DYRK1A. Studies show that DYRK1A small molecule inhibitors induce human βcell proliferation both in vitro and in vivo [54,78,[114][115][116][117][118]. Several studies have demonstrated that DYRK1A overexpression attenuated β-cell proliferation through NFAT dysregulation, a transcription factor that transactivates cell cycle-activating genes and represses cell cycle inhibitor genes including other CMGC, cyclins and p57 (Table 1) [30,68,114,115]. ...
Article
Full-text available
The increasing population will challenge healthcare, particularly because the worldwide population has never been older. Therapeutic solutions to age-related disease will be increasingly critical. Kinases are key regulators of human health and represent promising therapeutic targets for novel drug candidates. The dual-specificity tyrosine-regulated kinase (DYRKs) family is of particular interest and, among them, DYRK1A has been implicated ubiquitously in varied human diseases. Herein, we focus on the characteristics of DYRK1A, its regulation and functional role in different human diseases, which leads us to an overview of future research on this protein of promising therapeutic potential.
... Harmine is a plant-derived β-carboline alkaloid with a wide spectrum of pharmacological actions including antioxidant (Jalili et al., 2020), immunomodulatory (Wang et al., 2021), antidiabetic (Barzowska et al., 2012), and anticancer effects (Chin et al., 2021). Harmine activates AMPK pathway and increases the expression of p62 in gastric cancer cells (Li et al., 2017). ...
Article
Oxidative stress induced neurotoxicity is increasingly perceived as an important neuropathologic mechanism underlying the motor and behavioral phenotypes associated with Huntington's disease (HD). Repeated exposure to 3-nitropropionic acid (3-NP) induces neurotoxic changes which closely simulate the neuropathological and behavioral characteristics of HD. This study aimed at evaluating the prophylactic effects of the dual-specificity tyrosine phosphorylation regulated kinase 1A (DYRK1A) inhibitor “harmine” against 3-NP-indued neurotoxicity and HD-like symptoms. The potential prophylactic effect of harmine (10 mg/kg/day; intraperitoneal) was investigated on 3-NP-induced motor and cognitive HD-like deficits, nuclear factor erythroid 2 like 2 (NRF2), AMP kinase (AMPK) and p21 protein levels and the gene expression of haem oxygenase-1 (Ho-1), NAD(P)H: quinone oxidoreductase-1 (Nqo-1) and p62 in addition to redox imbalance and histological neurotoxic changes in the striatum, prefrontal cortex, and hippocampus of male Wistar rats. Harmine successfully increased the protein levels of NRF2, AMPK and p21 and the gene expression of Ho-1, Nqo-1 and p62, restored redox homeostasis, and reduced CASPASE-3 level. This was reflected in attenuation of 3-NP-induced neurodegenerative changes and improvement of rats' motor and cognitive performance. This study draws attention to the protective role of harmine against 3-NP-induced motor and cognitive dysfunction that could be mediated via enhancing NRF2-mediated signaling with subsequent amelioration of oxidative stress injury via NRF2 activators, p21 and AMPK, in the striatum, prefrontal cortex, and hippocampus which could offer a promising therapeutic tool to slow the progression of HD.
... It was discovered that the drug candidate galunisertib rescued β-cell death associated with GLIS3 mutations, which is expected to be applied to the precision treatment of diabetes related to GLIS3 mutations [79]. In another study, research based on human islet organoid models confirmed that a small molecule inhibitor of the dual-specificity tyrosine phosphorylation regulated kinase 1 A (DYRK1A) kinase effectively promotes β-cell proliferation within organoids, enhancing their long-term insulin secretion and balanced glucagon levels maintenance, which offers promising prospects for islet repair treatment in diabetic patients [80]. ...
Article
Full-text available
Diabetes mellitus, a significant global public health challenge, severely impacts human health worldwide. The organoid, an innovative in vitro three-dimensional (3D) culture model, closely mimics tissues or organs in vivo. Insulin-secreting islet organoid, derived from stem cells induced in vitro with 3D structures, has emerged as a potential alternative for islet transplantation and as a possible disease model that mirrors the human body’s in vivo environment, eliminating species difference. This technology has gained considerable attention for its potential in diabetes treatment. Despite advances, the process of stem cell differentiation into islet organoid and its cultivation demonstrates deficiencies, prompting ongoing efforts to develop more efficient differentiation protocols and 3D biomimetic materials. At present, the constructed islet organoid exhibit limitations in their composition, structure, and functionality when compared to natural islets. Consequently, further research is imperative to achieve a multi-tissue system composition and improved insulin secretion functionality in islet organoid, while addressing transplantation-related safety concerns, such as tumorigenicity, immune rejection, infection, and thrombosis. This review delves into the methodologies and strategies for constructing the islet organoid, its application in diabetes treatment, and the pivotal scientific challenges within organoid research, offering fresh perspectives for a deeper understanding of diabetes pathogenesis and the development of therapeutic interventions.