Figure 1 - uploaded by Lin Zhang
Content may be subject to copyright.
Morphological features of necroptosis and apoptosis in cancer cells HT29 colon cancer cells treated with an anticancer drug for 48 hours were analyzed by transmission electron microscopy. The cell undergoing necroptosis shows plasma membrane rupture and permeabilization, compared to the intact plasma membrane with blebbing in the apoptotic cell (red arrowheads). The necroptotic cell exhibits cytoplasm swelling and vacuolization, which are absent in the apoptotic cell (green arrowheads). The necroptotic cell has swelled mitochondria, in contrast to those in the apoptotic cells (yellow arrowheads). The necroptotic cell also lacks condensed and fragmented nuclei seen in the apoptotic cell (blue arrowheads).

Morphological features of necroptosis and apoptosis in cancer cells HT29 colon cancer cells treated with an anticancer drug for 48 hours were analyzed by transmission electron microscopy. The cell undergoing necroptosis shows plasma membrane rupture and permeabilization, compared to the intact plasma membrane with blebbing in the apoptotic cell (red arrowheads). The necroptotic cell exhibits cytoplasm swelling and vacuolization, which are absent in the apoptotic cell (green arrowheads). The necroptotic cell has swelled mitochondria, in contrast to those in the apoptotic cells (yellow arrowheads). The necroptotic cell also lacks condensed and fragmented nuclei seen in the apoptotic cell (blue arrowheads).

Contexts in source publication

Context 1
... has several distinctive characteristics compared to other types of cell death, in particular, apoptosis (Table 1). Necroptotic cells share several morphological features of necrosis, including early loss of plasma membrane integrity, translucent cytosol, and swelling mitochondria (Figure 1) [12]. In contrast, apoptotic cells lack these features, and are characterized by cell shrinkage, plasma membrane blebbing, and condensed and fragmented nuclei and organelles (Figure 1). ...
Context 2
... cells share several morphological features of necrosis, including early loss of plasma membrane integrity, translucent cytosol, and swelling mitochondria (Figure 1) [12]. In contrast, apoptotic cells lack these features, and are characterized by cell shrinkage, plasma membrane blebbing, and condensed and fragmented nuclei and organelles (Figure 1). At the biochemical level, necroptotic cells show marked depletion of cellular ATP and leakage of intracellular contents, in contrast to apoptosis, which is a more energy-consuming process requiring a relatively higher level of cellular ATP. ...
Context 3
... to lack of specific molecular markers of necroptosis, a combination of approaches is often required to distinguish necroptosis from other cell death modalities. For cultured cells, transmission electron microscopy (TEM) is commonly used to provide morphological evidence of necrosis ( Figure 1). Extracellular release of HMGB1, loss of cell viability, and depletion of ATP can be used as markers of necroptosis, but do not distinguish necroptosis from other types of necrotic death. ...

Citations

... The necrosome formed by phosphorylation and activation of RIP1 and RIP3 recruits the MLKL and initiates necroptosis [31][32][33][34][35][36]. Necroptosis releases damage-associated molecular patterns (DAMPs), including high-mobility group box 1 protein (HMGB1), crucial in initiating adaptive immunity, immune system activation, antigen presentation, and T-cell responses [37][38][39][40][41][42]. Therefore, necroptosis is distinguished from another form of cell death by releasing paracrine factors associated with the immune system [26,[37][38][39]. ...
Article
Full-text available
Abstracts Herein, we present human adipose-derived stem cells (ADSCs) inserted with the receptor-interacting protein kinase-3 (RIP3) gene (RP@ADSCs), which induces cell necroptosis, for tumor immunotherapy. Necroptosis has characteristics of both apoptosis, such as programmed cell death, and necrosis, such as swelling and plasma membrane rupture, during which damage-related molecular patterns are released, triggering an immune response. Therefore, necroptosis has the potential to be used as an effective anticancer immunotherapy. RP@ADSCs were programmed to necroptosis after a particular time after being injected in vivo, and various pro-inflammatory cytokines secreted during the stem cell death process stimulated the immune system, showing local and sustained anticancer effects. It was confirmed that RIP3 protein expression increased in ADSCs after RP transfection. RP@ADSCs continued to induce ADSCs death for 7 days, and various pro-inflammatory cytokines were secreted through ADSCs death. The efficacy of RP@ADSCs-mediated immunotherapy was evaluated in mouse models bearing GL-26 (glioblastoma) and K1735 (melanoma), and it was found that RP resulted in an increase in the population of long-term cytotoxic T cells and a decrease in the population of regulatory T cells. This shows that RP@ADSCs have potential and applicability as an excellent anticancer immunotherapy agent in clinical practice.
... Necroptosis is a novel programmed cell death pathway characterized by significant inflammatory outcomes and immune responses. It differs from apoptosis, and its common symptoms include cellular swelling, organelle dysfunction, extensive mitochondrial damage, and plasma membrane rupture 4,5 . Necroptosis is dependent on receptor-interacting protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain like pseudokinase (MLKL) from a morphological standpoint 6,7 . ...
Article
Full-text available
Ovarian cancer (OC) is one of the most prevalent and fatal malignant tumors of the female reproductive system. Our research aimed to develop a prognostic model to assist inclinical treatment decision-making.Utilizing data from The Cancer Genome Atlas (TCGA) and copy number variation (CNV) data from the University of California Santa Cruz (UCSC) database, we conducted analyses of differentially expressed genes (DEGs), gene function, and tumor microenvironment (TME) scores in various clusters of OC samples.Next, we classified participants into low-risk and high-risk groups based on the median risk score, thereby dividing both the training group and the entire group accordingly. Overall survival (OS) was significantly reduced in the high-risk group, and two independent prognostic factors were identified: age and risk score. Additionally, three genes—C-X-C Motif Chemokine Ligand 10 (CXCL10), RELB, and Caspase-3 (CASP3)—emerged as potential candidates for an independent prognostic signature with acceptable prognostic value. In Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, pathways related to immune responses and inflammatory cell chemotaxis were identified. Cellular experiments further validated the reliability and precision of our findings. In conclusion, necroptosis-related genes play critical roles in tumor immunity, and our model introduces a novel strategy for predicting the prognosis of OC patients.
... A high level of ATP is generated by cells in response to stress for the activation of proteins, such as caspases, and DNA repair, necessary conditions for apoptosis generation [37]. In contrast, necrosis is characterized by a significant decrease in ATP generation responding to electron transport chain uncoupling [53]. These features of necrotic cell death are similar to those shown by our results (Figure 3), where the T3SS2 of V. parahaemolyticus generates first an increase and then a decrease in ATP concentrations (Figure 3A), generating a decrease in cell viability ( Figure 3B) and damaging the integrity of the cell membrane from 3 hpi ( Figure 3C). ...
Article
Full-text available
Vibrio parahaemolyticus is an important human pathogen that is currently the leading cause of shellfish-borne gastroenteritis in the world. Particularly, the pandemic strain has the capacity to induce cytotoxicity and enterotoxicity through its Type 3 Secretion System (T3SS2) that leads to massive cell death. However, the specific mechanism by which the T3SS2 induces cell death remains unclear and its contribution to mitochondrial stress is not fully understood. In this work, we evaluated the contribution of the T3SS2 of V. parahaemolyticus in generating mitochondrial stress during infection in human intestinal HT-29 cells. To evaluate the contribution of the T3SS2 of V. parahaemolyticus in mitochondrial stress, infection assays were carried out to evaluate mitochondrial transition pore opening, mitochondrial fragmentation, ATP quantification, and cell viability during infection. Our results showed that the Δvscn1 (T3SS2+) mutant strain contributes to generating the sustained opening of the mitochondrial transition pore. Furthermore, it generates perturbations in the ATP production in infected cells, leading to a significant decrease in cell viability and loss of membrane integrity. Our results suggest that the T3SS2 from V. parahaemolyticus plays a role in generating mitochondrial stress that leads to cell death in human intestinal HT-29 cells. It is important to highlight that this study represents the first report indicating the possible role of the V. parahaemolyticus T3SS2 and its effector proteins involvement in generating mitochondrial stress, its impact on the mitochondrial pore, and its effect on ATP production in human cells.
... The cell deaths due to AMF treatment are classified based on the mapping from the flow cytometry analysis as necroptosis, early apoptosis, and late apoptosis. Necroptosis is a form of programmed cell death that occurs in response to various types of cellular stress, such as viral infection or oxidative stress or even mechanical stress [31]. Unlike apoptosis, which is a well-characterized form of programmed cell death that involves controlled dismantling of cells, necroptosis results in the rupture of cells and the release of cellular contents into the extracellular environment, triggering an inflammatory response. ...
... The cell deaths due to AMF treatment are classified based on the mapping from the flow cytometry analysis as necroptosis, early apoptosis, and late apoptosis. Necroptosis is a form of programmed cell death that occurs in response to various types of cellular stress, such as viral infection or oxidative stress or even mechanical stress [31]. Unlike apoptosis, which is a well-characterized form of programmed cell death that in-volves controlled dismantling of cells, necroptosis results in the rupture of cells and the release of cellular contents into the extracellular environment, triggering an inflammatory response. ...
Article
Full-text available
The PEG-coated ferrite nanoparticles Co0.2Mn0.6Zn0.2Fe2O4 (X1), Co0.4Mn0.4Zn0.2Fe2O4 (X2), and Co0.6Mn0.2Zn0.2Fe2O4 (X3) were synthesized by the coprecipitation method. The nanoparticles were characterized by XRD, Raman, VSM, XPS, and TEM. The magnetic hyperthermia efficiency (MH) was determined for PEG-coated nanoparticles using an alternating magnetic field (AMF). X2 nanoparticles displayed the highest saturation magnetization and specific absorption rate (SAR) value of 245.2 W/g for 2 mg/mL in a water medium. Based on these properties, X2 nanoparticles were further evaluated for antiproliferative activity against HCT116 cells at an AMF of 495.25 kHz frequency and 350 G strength, using MTT, colony formation, wound healing assays, and flow cytometry analysis for determining the cell viability, clonogenic property, cell migration ability, and cell death of HCT116 cells upon AMF treatment in HCT116 cells, respectively. We observed a significant inhibition of cell viability (2% for untreated control vs. 50% for AMF), colony-forming ability (530 cells/colony for untreated control vs. 220 cells/colony for AMF), abrogation of cell migration (100% wound closure for untreated control vs. 5% wound closure for AMF), and induction of apoptosis-mediated cell death (7.5% for untreated control vs. 24.7% for AMF) of HCT116 cells with respect to untreated control cells after AMF treatment. Collectively, these results demonstrated that the PEG-coated (CoMnZn-Fe2O4) mixed ferrite nanoparticles upon treatment with AMF induced a significant antiproliferative effect on HCT116 cells compared with the untreated cells, indicating the promising antiproliferative potential of the Co0.4Mn0.4Zn0.2Fe2O4 nanoparticles for targeting colorectal cancer cells. Additionally, these results provide appealing evidence that ferrite-based nanoparticles using MH could act as potential anticancer agents and need further evaluation in preclinical models in future studies against colorectal and other cancers.
... Necroptosis is a drug-and dose-dependent mechanism of tumor cell destruction achieved by continuous exposure to high chemotherapy levels. Necroptosis is characterized by loss of tumor cell membrane integrity, exposing tumorspecific antigens to immune surveillance which can stimulate a robust response of the adaptive immune system to tumor-specific antigens [16,[56][57][58]. Immunogenicity requires that tumor cells provide adequate levels of antigens and that the tumor cells effectively present antigens in a form that leads to immune activation instead of immune tolerance which can occur when antigen is presented intermittently, in low quantities, and with progressive modifications [59]. ...
Article
Full-text available
This review summarizes development of large surface area microparticle paclitaxel (LSAM-PTX) and docetaxel (LSAM-DTX) for local treatment of primary carcinomas with emphasis on immunomodulation. Intratumoral (IT) delivery of LSAM-PTX and LSAM-DTX provides continuous, therapeutic drug levels for several weeks. Preclinical studies and clinical trials reported a reduction in tumor volume (TV) and immunomodulation in primary tumor and peripheral blood with increases in innate and adaptive immune cells and decreases in suppressor cells. Increased levels of checkpoint expression of immune cells occurred in clinical trials of high-risk non-muscle-invasive bladder cancer (LSAM-DTX) and unresectable localized pancreatic cancer (LSAM-PTX). TV reduction and increases in immune effector cells occurred following IT LSAM-DTX and IT LSAM-PTX together with anti-mCTLA-4 and anti-mPD-1, respectively. Synergistic benefits from combinatorial therapy in a 4T1-Luc breast cancer model included reduction of metastasis with IT LSAM-DTX + anti-mCTLA-4. IT LSAM-PTX and LSAM-DTX are tumoricidal, immune enhancing, and may improve solid tumor response to immune checkpoint inhibitors without additional systemic toxicity.
... [25][26][27] Paclitaxel may be a particularly strong immunostimulant, as it is able to both activate CD8+ T cells and reduce immunosuppressive cells, such as regulatory T cells [21][22][23][28][29][30] and myeloid-derived suppressor cells (MDSC). 23,31,32 To maximize tumoricidal effects, it has been suggested that the immune system can be primed with chemotherapy ahead of immunotherapy to reinstate or enhance immunosurveillance. 24,25,[33][34][35][36] Infiltration of adaptive and innate immune cells as well as increases in checkpoint expression by immune cells were found following LSAM-PTX and LSAM-DTX treatment of patients with unresectable locally advanced pancreatic cancer and high-risk non-muscle invasive bladder cancer, respectively. ...
Article
Full-text available
The effects of intratumoral (IT) large surface area microparticle paclitaxel (LSAM-PTX) alone and in combination with systemic administration of the programmed cell death protein antibody (anti-mPD-1) were evaluated in a syngeneic murine model of melanoma. Groups of mice with subcutaneously implanted Clone M3 (Cloudman S91) tumors were treated with single and combination therapies. Tumor volume (TV) measurements, body weights, and clinical observations were followed in-life. At end of study, tumor-site tissues were collected, measured, and processed for flow cytometry along with blood and lymph nodes. The combination of LSAM-PTX + anti-mPD-1 resulted in an antitumoral response, which produced a significant decrease in TV compared to control animals. TV decreases also occurred in the LSAM-PTX and anti-mPD-1 groups. Flow cytometry analysis found increases in granulocytes and M2 macrophages and decreases in dendritic cells (DC) and monocytic myeloid-derived suppressor cells (M-MDSC) in tumor-site tissues. Increases in granulocytes and decreases in CD4+ T cells, macrophages, and M1 macrophages were found in the blood of animals administered the combination treatment. Increases in natural killer (NK) cells were found in lymph node tissue in the combination treatment group. These findings suggest that IT LSAM-PTX may provide benefit in the local treatment of melanomas and may synergize with systemic anti-PD-1 therapy, leading to additional tumoricidal outcomes without added systemic toxicity.
... Accumulating evidence suggests that necroptosis, similar to apoptosis, functions as a barrier against tumor development and plays a critical role in anticancer therapy (8). Genetic and epigenetic alterations of necroptosis regulators, such as RIP3, are frequently found in human tumors including colorectal cancers (8). ...
... Accumulating evidence suggests that necroptosis, similar to apoptosis, functions as a barrier against tumor development and plays a critical role in anticancer therapy (8). Genetic and epigenetic alterations of necroptosis regulators, such as RIP3, are frequently found in human tumors including colorectal cancers (8). A variety of anticancer agents can promote necroptosis in addition to apoptosis (8). ...
... Genetic and epigenetic alterations of necroptosis regulators, such as RIP3, are frequently found in human tumors including colorectal cancers (8). A variety of anticancer agents can promote necroptosis in addition to apoptosis (8). However, the functional role and regulatory mechanisms of necroptosis in tumor progression and anticancer therapy are poorly understood. ...
Article
Induction of programmed cell death (PCD) is a key cytotoxic effect of anticancer therapies. PCD is not confined to caspase-dependent apoptosis, but includes necroptosis, a regulated form of necrotic cell death controlled by Receptor-Interacting Protein (RIP) kinases 1 and 3, and Mixed Lineage Kinase Domain-Like (MLKL) pseudo-kinase. Necroptosis functions as a defense mechanism against oncogenic mutations and pathogens and can be induced by a variety of anticancer agents. However, the functional role and regulatory mechanisms of necroptosis in anticancer therapy are poorly understood. In this study, we found that RIP3-dependent but RIP1-independent necroptosis is engaged by 5-fluorouracil (5-FU) and other widely used antimetabolite drugs, and functions as a major mode of cell death in a subset of colorectal cancer (CRC) cells that express RIP3. We identified a novel 5-FU-induced necroptosis pathway involving p53-mediated induction of the BH3-only Bcl-2 family protein, p53 upregulated modulator of apoptosis (PUMA), which promotes cytosolic release of mitochondrial DNA and stimulates its sensor z-DNA-binding protein 1 (ZBP1) to activate RIP3. PUMA/RIP3-dependent necroptosis mediates the in vitro and in vivo antitumor effects of 5-FU and promotes a robust antitumor immune response. Our findings provide a rationale for stimulating necroptosis to enhance tumor cell killing and antitumor immune response leading to improved CRC treatments.
... Necroptosis, an alternative form of cell death, comes into play when the apoptotic pathway is compromised or suppressed (Chen et al. 2016). It can be instigated by chemical or physical injury, inflammation, or infection, resulting in cell membrane rupture, gradual cytoplasmic translucency, organelle swelling, and subsequent release of cellular contents (He et al. 2022). ...
Article
Full-text available
The potential benefits of adiponectin replacement therapy extend to numerous human diseases, with current research showing particular interest in its effectiveness against specific cancer forms, especially hormone-related. However, limitations in the pharmacological use of the intact protein have led to a focus on alternative options. AdipoRon is an extensively studied non-peptidic drug candidate for adiponectin replacement therapy. While researchers have explored the efficacy and therapeutic applications of AdipoRon in various disease conditions, their effects against cancer models advanced more, with no review regarding AdipoRon’s efficacy against hormone-related cancers being published. The present systematic review aims to fill this gap. Preclinical evidence was compiled from PubMed, EMBASE, COCHRANE, and Google Scholar following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, and the manuscript’s quality assessment was conducted using the Joanna Briggs Institute (JBI) Checklist Critical Appraisal Tool for Systematic Reviews’ Quality. The included nine studies incorporated various cell and animal models of the pancreas, gynaecological system, and osteosarcoma cancers. AdipoRon demonstrated effectiveness against pancreatic cancer by activating p44/42 MAPK, mitochondrial dysfunction, and AMPK-mediated inhibition of ACC1. In gynaecological cancers, it exhibited promising anticancer effects through the activation of AMPK, potential inhibition of mTOR, and modulation of the SET1B/BOD1/AdipoR1 signaling cascade. Against osteosarcoma, AdipoRon worked by perturbing ERK1/2 signaling and reducing p70S6K phosphorylation. AdipoRon shows promise in preclinical studies, but human trials are crucial for clinical safety and effectiveness. Caution is needed due to potential off-target effects, especially in cancer therapy with multi-target approaches. Structural biology and computational methods can help predict these effects.
... Morphological changes induced by 5-ALA-PDT were examined in 4T1 and DLD-1 cells by transmission electron microscopy (TEM) ( Figure 5). In both cell lines, we observed typical morphological features of pyroptosis and necroptosis such as plasma membrane permeabilization (red arrow), organelle swelling and vacuolization (green arrow), mitochondrial swelling (blue arrow) and deficiency of nuclear chromatin (yellow arrow) (50,51). In contrast, morphological features of apoptosis such as chromatin condensation and relocation were not found in the TEM analysis. ...
Article
Full-text available
Background Inflammatory cell death is a form of programmed cell death (PCD) that induces inflammatory mediators during the process. The production of inflammatory mediators during cell death is beneficial in standard cancer therapies as it can break the immune silence in cancers and induce anticancer immunity. Photodynamic therapy (PDT) is a cancer therapy with photosensitizer molecules and light sources to destroy cancer cells, which is currently used for treating different types of cancers in clinical settings. In this study, we investigated if PDT using 5-aminolevulinic (5-ALA-PDT) causes inflammatory cell death and, subsequently, increases the immunogenicity of cancer cells. Methods Mouse breast cancer (4T1) and human colon cancer (DLD-1) cells were treated with 5-ALA for 4 hours and then irradiated with a light source. PCD induction was measured by western blot analysis and FACS. Morphological changes were determined by transmission electron microscopy (TEM). BALB/c mice were injected with cell-free media, supernatant of freeze/thaw cells or supernatant of PDT cells intramuscular every week for 4 weeks and then challenged with 4T1 cells at the right hind flank of BALB/c. Tumor growth was monitored for 12 days. Results We found that 5-ALA-PDT induces inflammatory cell death, but not apoptosis, in 4T1 cells and DLD-1 cells in vitro. Moreover, when mice were pretreated with 5-ALA-PDT culture supernatant, the growth of 4T1 tumors was significantly suppressed compared to those pretreated with freeze and thaw (F/T) 4T1 culture supernatant. Conclusion These results indicate that 5-ALA-PDT induces inflammatory cell death which promotes anticancer immunity in vivo.
... Necroptotic cell death is characterized by the formation of a necrosome-containing RIPK and pore-forming mixed lineage kinase domain-like protein (MLKL). The molecular pathways downstream of the necrosome cause disintegration of mitochondria, lysosomes, and plasma membrane and the reduction of cytosolic adenosine triphosphate (ATP) and ROS burst [136][137][138][139][140][141]. The cytotoxic effect of ceramide nanoliposomes was mediated by the induction of MLKL-dependent necroptosis [142]. ...