Cognitive impairment in various mental illnesses, particularly neuropsychiatric disorders, has adverse functional and clinical consequences. While genetic mutations and epigenetic dysregulations of several genes during embryonic and adult periods are linked to cognitive impairment in mental disorders, the composition and diversity of resident bacteria in the gastrointestinal tract—shaped by environmental factors—also influence the brain epigenome, affecting behavior and cognitive functions. Accordingly, many recent studies have provided evidence that human gut microbiota may offer a potential avenue for improving cognitive deficits. In this review, we provide an overview of the relationship between cognitive impairment, alterations in the gut microbiome, and epigenetic alterations during embryonic and adult periods. We examine how various factors beyond genetics—such as lifestyle, age, and maternal diet—impact the composition, diversity, and epigenetic functionality of the gut microbiome, consequently influencing cognitive performance. Additionally, we explore the potential of maternal gut microbiome signatures and epigenetic biomarkers for predicting cognitive impairment risk in older adults. This article also explores the potential roles of nutritional deficiencies in programming cognitive disorders during the perinatal period in offspring, as well as the promise of gut microbiome-targeted therapeutics with epigenetic effects to prevent or alleviate cognitive dysfunctions in infants, middle-aged adults, and older adults. Unsolved challenges of gut microbiome-targeted therapeutics in mitigating cognitive dysfunctions for translation into clinical practice are discussed, lastly.
Mutant forms of the gene encoding the tumor suppressor p53 are found in numerous human malignancies, but the physiologic function of p53 and the effects of mutations on this function are unknown. The p53 protein binds DNA in a sequence-specific manner and thus may regulate gene transcription. Cotransfection experiments showed that wild-type p53 activated the expression of genes adjacent to a p53 DNA binding site. The level of activation correlated with DNA binding in vitro. Oncogenic forms of p53 lost this activity. Moreover, all mutants inhibited the activity of coexpressed wild-type p53, providing a basis for the selection of such mutants during tumorigenesis.
<p>S1: SMAD4+/+ and SMAD4-/- colon cancer cells were serum-starved and seeded on transwells and allowed to cross the membrane for 24 hours (transwell migration; RFU, relative fluorescence unit; mean {plus minus} SD, n = 3 biological replicates; **P < 0.01). S2: RICTOR depletion in SMAD4-negative colon cancer cells using shRNA#2 was measured by RT-qPCR (RT-qPCR; mean {plus minus} SD, n = 3 technical replicates; ***P < 0.001). B, Western blotting shows the relative levels of RICTOR, p-AKTS473, and cleaved-caspase 3 upon knockdown of RICTOR and in response to irinotecan. C, The indicated cell lines were treated with irinotecan (10μM) for 72 hours. S3: Cells depleted of RICTOR were serum starved and seeded on transwells and allowed to cross the membrane for 24 hours (transwell migration; RFU, relative fluorescence unit; mean {plus minus} SD, n = 6 biological replicates; ***P < 0.001). S4: A, Western blot shows the relative levels of RICTOR and p-AKTS47 in SW403 cells upon transfection with the indicated siRNAs. B, siRNA-transfected SW403 cells were treated with irinotecan (10μM) for 120 hours. Viability of cells was normalized to DMSO-treated controls (cell viability assay; mean {plus minus} SD, n = 3 biological replicates; **P < 0.01). S5: High RICTOR/AKT1 expression correlates with worse survival in colon cancer. S6: Targeting AKT with MK2206 sensitizes SMAD4-negative SW403 colon cancer cells to irinotecan. S7: Targeting AKT with either AKTi-1/2 or MK2206 sensitizes SMAD4-negative colon cancer cells to irinotecan. S8: No toxicity observed in mice exposed to MK2206 and irinotecan. S9: Targeting AKT with MK2206 sensitizes SMAD4-negative ASPC1 and CFPAC1 pancreatic cancer cells to irinotecan. S10: In silico Kaplan-Meier analyses showing the correlation between SMAD4, RICTOR, or AKT1 expression and overall survival (OS) in pancreatic cancer patients.</p>
It has been universally believed that spindle assembly checkpoint (SAC) proteins which include the kinetochore proteins are involved in monitoring the faithful segregation of sister chromatids during cell division and hence defects in these proteins result in anueploidy. Furthermore, there are multiple sources of experimental data to suggest that a defect in p53 could also promote genomic instability leading to anueploidy. Despite these observations, a molecular basis for the prevention of aneuploidy to maintain genomic integrity upon activation of SAC has largely remained elusive. In this report, we demonstrate a novel mechanism for the maintenance of a balance between cell survival and apoptosis upon activation of SAC. We found that depletion of the outer kinetochore protein hBub1 upon activation of SAC primarily triggers early cell death mediated by p53. This phenomenon is further supported by the up-regulation of p53 down-stream pro-apoptotic genes, BAX and PUMA as well as a corresponding increase in the cleavage products of PARP and caspase 3, markers of apoptosis upon depletion of hBub1 in SAC activated cells. On the other hand, as expected, concomitant loss of both hBub1 and p53 resulted in disabling of the p53 mediated cell death pathway leading to the accumulation of cells with aneuploidy/polyploidy.Commentary also to:hBub1 negatively regulates p53 mediated early cell death upon mitotic checkpoint activationFangming Gao, Jose F Ponte, Panagiotis Papageorgis, Mary Levy, Sait Ozturk, Arthur W. Lambert, Arunthathi Thiagalingam, Hamid Mostafavi Abdolmaleky, Beth A Sullivan and Sam Thiagalingam