TMEM173's function as an essential regulator of type I interferon (IFN) responses is fundamentally linked to its participation in immune regulation and the induction of cell death. JH-X-119-01 clinical trial Recent cancer immunotherapy research has established the activation of TMEM173 as a promising course of action. Despite this, the transcriptomic properties of TMEM173 within B-cell acute lymphoblastic leukemia (B-ALL) are not presently known.
Peripheral blood mononuclear cells (PBMCs) were analyzed for TMEM173 mRNA and protein expression using quantitative real-time PCR (qRT-PCR) and western blotting (WB). A Sanger sequencing analysis was conducted to determine the mutation status of TMEM173. Using single-cell RNA sequencing (scRNA-seq), the expression of TMEM173 was examined across a range of bone marrow (BM) cell types.
B-ALL patient PBMCs displayed a rise in the mRNA and protein expression of TMEM173. Incidentally, the TMEM173 gene sequences of two B-ALL patients had a frameshift mutation. ScRNA-seq data analysis demonstrated the unique expression profiles of TMEM173 mRNA in the bone marrow of high-risk B-ALL patients. Within the cell types of granulocytes, progenitor cells, mast cells, and plasmacytoid dendritic cells (pDCs), TMEM173 expression was found to be superior to that observed in B cells, T cells, natural killer (NK) cells, and dendritic cells (DCs). During the progression of B-ALL, a subset analysis indicated that proliferative precursor-B (pre-B) cells, expressing nuclear factor kappa-B (NF-κB), CD19, and Bruton's tyrosine kinase (BTK), showcased restricted expression of TMEM173 and pyroptosis effector gasdermin D (GSDMD). Moreover, TMEM173 was linked to the operational activation of NK cells and dendritic cells in B-ALL.
Our findings offer insights into the transcriptomic characterization of TMEM173 from the bone marrow (BM) of high-risk B-cell acute lymphoblastic leukemia (B-ALL) patients. Therapeutic strategies for B-ALL patients might emerge from the targeted activation of TMEM173 in specific cellular contexts.
In high-risk B-ALL patients, our study detailed the transcriptomic aspects of TMEM173 within the bone marrow (BM). A new era in B-ALL therapy could emerge with the targeted activation of TMEM173 in selected cellular targets.
The critical role of mitochondrial quality control (MQC) in the progression of tubulointerstitial injury within diabetic kidney disease (DKD) cannot be understated. The mitochondrial unfolded protein response (UPRmt), an essential mitochondrial quality control (MQC) process, is activated to preserve the integrity of mitochondrial protein homeostasis when faced with mitochondrial stress. The mammalian UPRmt (unfolded protein response in mitochondria) depends on activating transcription factor 5 (ATF5) to mediate the translocation from the mitochondria to the nucleus. Nevertheless, the part played by ATF5 and UPRmt in tubular impairment associated with DKD is unknown.
In DKD patients and db/db mice, ATF5 and UPRmt-related proteins, including heat shock protein 60 (HSP60) and Lon peptidase 1 (LONP1), were the subject of immunohistochemistry (IHC) and western blot investigation. Eight-week-old db/db mice received injections of ATF5-shRNA lentiviruses via the tail vein, whereas a control group was given a negative lentivirus. At the 12-week time point, mice were euthanized, and subsequent kidney section analyses involved dihydroethidium (DHE) for reactive oxygen species (ROS) assessment and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) for apoptosis evaluation. Under controlled in vitro conditions, the impact of ATF5 and HSP60 on tubular injury in HK-2 cells was assessed by transfecting the cells with ATF5-siRNA, ATF5 overexpression plasmids, or HSP60-siRNA under ambient hyperglycemic conditions. To evaluate mitochondrial oxidative stress, a MitoSOX staining technique was used, alongside the use of Annexin V-FITC kits to examine the early stage of apoptosis.
Elevated expression of ATF5, HSP60, and LONP1 proteins was evident in the renal tissues of both DKD patients and db/db mice, exhibiting a strong association with tubular damage severity. db/db mice, upon receiving lentiviral vectors expressing ATF5 shRNA, demonstrated a reduction in HSP60 and LONP1 activity, alongside enhancements in serum creatinine levels, along with less tubulointerstitial fibrosis and apoptosis. The expression of ATF5 in HK-2 cells elevated in a way directly related to exposure duration following high glucose exposure, accompanied by an increase in the production of HSP60, fibronectin, and cleaved caspase-3 in the in vitro setting. Upon ATF5-siRNA transfection, the expression of HSP60 and LONP1 was reduced in HK-2 cells chronically exposed to high exogenous glucose, thus mitigating oxidative stress and apoptosis. The detrimental effects of ATF5 overexpression were apparent in these impairments. The effect of ATF5 on HK-2 cells, exposed to sustained HG treatment, was negated by HSP60-siRNA transfection. An unexpected finding was that ATF5 blockage exacerbated mitochondrial reactive oxygen species (ROS) levels and apoptosis in HK-2 cells during the initial 6 hours of high-glucose intervention.
ATF5's initial protective action in very early diabetic kidney disease is counteracted by its influence on HSP60 and the UPRmt pathway, thereby inducing tubulointerstitial damage. This finding identifies a possible target to combat DKD progression.
ATF5's protective potential in the initial phase of DKD is potentially compromised by its action on HSP60 and the UPRmt pathway, which subsequently results in tubulointerstitial damage, suggesting this pathway as a potential target for managing DKD progression.
Photothermal therapy (PTT), activated by near-infrared-II (NIR-II, 1000-1700 nm) light, is being developed as a possible treatment for tumors, featuring deeper tissue penetration and higher allowable laser power density relative to the NIR-I (750-1000 nm) biological window. The promising applications of black phosphorus (BP) in photothermal therapy (PTT), due to its excellent biocompatibility and favorable biodegradability, are impacted by limitations in ambient stability and photothermal conversion efficiency (PCE). Its use in near-infrared-II (NIR-II) PTT is scarcely documented. Novel, fullerene-modified few-layer boron-phosphorus nanosheets (BPNSs), precisely 9 layers in thickness, are synthesized through a simple one-step esterification process. This new material, abbreviated as BP-ester-C60, demonstrates a dramatic improvement in ambient stability, owing to the strong bonding between the highly stable, hydrophobic C60 and the phosphorus atom's unshared electron pair. BP-ester-C60's application as a photosensitizer in NIR-II PTT yields a considerably higher PCE than that observed for the pristine BPNSs. In vitro and in vivo investigations of antitumor responses, utilizing 1064 nm NIR-II laser irradiation, indicated a dramatic enhancement in the photothermal therapeutic efficacy of BP-ester-C60 relative to the pristine BPNSs, while maintaining significant biosafety. The boost in NIR light absorption is a consequence of the intramolecular electron transfer from BPNSs to C60, which affects the band energy level.
Within the systemic disorder MELAS syndrome, mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes may manifest due to failure of mitochondrial metabolism, potentially causing multi-organ dysfunction. This disorder's most frequent causes are maternally inherited mutations within the MT-TL1 gene. The presence of stroke-like episodes, epilepsy, dementia, headache, and myopathy suggests potential clinical manifestations. Because of stroke-like events, acute visual loss, often accompanied by cortical blindness, can affect the occipital cortex or visual pathways. A characteristic symptom of mitochondrial diseases, including Leber hereditary optic neuropathy (LHON), is vision loss resulting from optic neuropathy.
A 55-year-old woman, whose sister has a history of MELAS with the m.3243A>G (p.0, MT-TL1) mutation, presented with an unremarkable medical history. This was followed by subacute, painful vision loss in one eye, and additional proximal muscular pain and a headache. A severe and continuous decline in vision, localized to one eye, manifested over the coming weeks. Following ocular examination, unilateral swelling of the optic nerve head was identified; fluorescein angiography further indicated a segmental perfusion delay in the optic disc and leakage from the papilla. A combination of neuroimaging, blood and CSF analysis, and temporal artery biopsy definitively excluded neuroinflammatory disorders and giant cell arteritis (GCA). Mitochondrial sequencing analysis unequivocally identified the m.3243A>G transition, while simultaneously excluding the three most common LHON mutations, as well as the m.3376G>A LHON/MELAS overlap syndrome mutation. JH-X-119-01 clinical trial Given the constellation of clinical symptoms and signs, including muscular involvement, observed in our patient, and the investigative findings, a diagnosis of optic neuropathy as a stroke-like event affecting the optic disc was established. L-arginine and ubidecarenone treatments were initiated with the objective of mitigating stroke-like episode symptoms and averting future occurrences. No further visual symptoms or deterioration were noted, maintaining the existing visual defect's stability.
Patients with mitochondrial disorders, even those with well-documented phenotypes and low mutational loads in peripheral tissue, should be assessed for any atypical clinical presentations. Knowledge of the precise heteroplasmy degree in distinct tissues, such as the retina and optic nerve, is not possible through observing the mitotic segregation of mitochondrial DNA (mtDNA). JH-X-119-01 clinical trial The therapeutic significance of an accurate diagnosis of atypically presenting mitochondrial disorders is undeniable.
Even in seemingly typical presentations of mitochondrial disorders, atypical clinical manifestations should be actively considered, particularly when the mutational burden in peripheral tissues is modest. The mitotic segregation of mitochondrial DNA (mtDNA) prevents a precise determination of heteroplasmy levels across various tissues, including the retina and optic nerve.