hybrid cells
Recently Published Documents


TOTAL DOCUMENTS

839
(FIVE YEARS 56)

H-INDEX

54
(FIVE YEARS 4)

Cancers ◽  
2022 ◽  
Vol 14 (2) ◽  
pp. 287
Author(s):  
Carolina Rubio ◽  
José Avendaño-Ortiz ◽  
Raquel Ruiz-Palomares ◽  
Viktoriya Karaivanova ◽  
Omaira Alberquilla ◽  
...  

Bladder cancer (BC) is the second most frequent cancer of the genitourinary system. The most successful therapy since the 1970s has consisted of intravesical instillations of Bacillus Calmette–Guérin (BCG) in which the tumor microenvironment (TME), including macrophages, plays an important role. However, some patients cannot be treated with this therapy due to comorbidities and severe inflammatory side effects. The overexpression of histone deacetylases (HDACs) in BC has been correlated with macrophage polarization together with higher tumor grades and poor prognosis. Herein we demonstrated that phenylbutyrate acid (PBA), a HDAC inhibitor, acts as an antitumoral compound and immunomodulator. In BC cell lines, PBA induced significant cell cycle arrest in G1, reduced stemness markers and increased PD-L1 expression with a corresponding reduction in histone 3 and 4 acetylation patterns. Concerning its role as an immunomodulator, we found that PBA reduced macrophage IL-6 and IL-10 production as well as CD14 downregulation and the upregulation of both PD-L1 and IL-1β. Along this line, PBA showed a reduction in IL-4-induced M2 polarization in human macrophages. In co-cultures of BC cell lines with human macrophages, a double-positive myeloid–tumoral hybrid population (CD11b+EPCAM+) was detected after 48 h, which indicates BC cell–macrophage fusions known as tumor hybrid cells (THC). These THC were characterized by high PD-L1 and stemness markers (SOX2, NANOG, miR-302) as compared with non-fused (CD11b−EPCAM+) cancer cells. Eventually, PBA reduced stemness markers along with BMP4 and IL-10. Our data indicate that PBA could have beneficial properties for BC management, affecting not only tumor cells but also the TME.


2022 ◽  
Author(s):  
Ayalur Raghu Subbalakshmi ◽  
Bazella Ashraf ◽  
Mohit Kumar Jolly

Abstract The Epithelial-Mesenchymal Transition (EMT) is a biological phenomenon associated with explicit phenotypic and molecular changes in cellular traits. Unlike the earlier-held popular belief of it being a binary process, EMT is now thought of as a landscape including diverse hybrid E/M phenotypes manifested by varying degrees of the transition. These hybrid cells can co-express both epithelial and mesenchymal markers and/or functional traits, and can possess the property of collective cell migration, enhanced tumor-initiating ability, and immune/targeted therapy-evasive features, all of which are often associated with worse patient outcomes. These characteristics of the hybrid E/M cells have led to a surge in studies that map their biophysical and biochemical hallmarks that can be helpful in exploiting their therapeutic vulnerabilities. This review discusses recent advances made in investigating hybrid E/M phenotype(s) from diverse biophysical and biochemical aspects by integrating live cell-imaging, cellular morphology quantification and mathematical modelling, and highlights a set of questions that remain unanswered about the dynamics of hybrid E/M states.


2021 ◽  
Vol 10 (6) ◽  
pp. 2972-2979
Author(s):  
Satya Subrahmanya Ajay Dangeti ◽  
Chaitanya Kishore Patnaik Sekharamantry ◽  
Venkata Kowshik Bayanti ◽  
B. A. Raju Ch ◽  
KVS Ramachandra Murthy ◽  
...  

This paper introduces a cascaded converter structure using hybrid two-level cells to form a single-phase seven-level inverter. Compared to various conventional and existing multilevel (MLI) topologies, this topology requires a lower number of devices. It uses several DC sources integrated in two-stage cells to provide the required voltage. In this topology, the DC-link condensers are not necessary. The modes of operation and modulation of the structure proposed are described in depth. Finally, in the MATLAB/Simulink platform the new topology is evaluated and the results reported. 


2021 ◽  
Author(s):  
Maxim E. Menyailo ◽  
Viktoria R. Zainullina ◽  
Liubov A. Tashireva ◽  
Sofia Yu. Zolotareva ◽  
Tatiana S. Gerashchenko ◽  
...  

Circulating tumor cells (CTCs) and tumor hybrid cells, being the leading players in metastasis, have prognostic relevance and are potential antimetastatic targets. CTCs are identified as epithelial-positive and CD45 (leukocyte)-negative cells, whereas tumor hybrid cells usually have epithelial and leukocyte components. However, epithelial and hybrid cells are also observed in healthy subjects that complicate the detection of CTCs and tumor hybrid cells in cancer patients. This study evaluated the diversity of CD45-negative and CD45-positive circulating epithelial cells (CECs) in breast cancer patients (n=20) using single-cell RNA sequencing. We also tried to detect CTCs and tumor hybrid cells among CD45─ CECs and CD45+ CECs by analyzing DNA ploidy since aneuploidy is a hallmark of cancer cells. Aneuploid cells were predominantly detected in CD45─ CECs, whereas CD45+ CECs were mainly diploid. Most transcriptional cancer features, including many cancer-associated signaling pathways, were specific to aneuploid cells of one CD45─ CEC cluster. These cells were also enriched by platelet genes and signaling pathways that may indicate their increased potential to adhere with thrombocytes. In CD45+ CECs, only one cluster had many aneuploid cells that were surprisingly negative for transcriptional cancer features. Thus, CD45─ and CD45+ CECs are highly heterogeneous in breast cancer patients and consist of transcriptionally-distinct cell populations with varying degrees of DNA ploidy where aneuploid cells are likely CTCs and tumor hybrid cells.


Author(s):  
Maria S Tretyakova ◽  
Ayalur Raghu Subbalakshmi ◽  
Maxim E Menyailo ◽  
Mohit Kumar Jolly ◽  
Evgeny Denisov

Metastasis is the leading cause of cancer death and can be realized through the phenomenon of tumor cell fusion. The fusion of tumor cells with other tumor or normal cells leads to the appearance of tumor hybrid cells (THCs) exhibiting novel properties such as increased proliferation and migration, drug resistance, decreased apoptosis rate and avoiding immune surveillance. Experimental studies showed the association of THCs with a high frequency of cancer metastasis; however, the underlying mechanisms remain unclear. Many other questions also remain to be answered: the role of genetic alterations in tumor cell fusion, the molecular landscape of cells after fusion, the lifetime and fate of different THCs, and the specific markers of THCs, and their correlation with various cancers and clinicopathological parameters. In this review, we discuss the factors and potential mechanisms involved in the occurrence of THCs, the types of THCs, and their role in cancer drug resistance and metastasis, as well as potential therapeutic approaches for the prevention and targeting of tumor cell fusion. In conclusion, we emphasize the current knowledge gaps in the biology of THCs that should be addressed to develop highly effective therapeutics and strategies for metastasis suppression.


2021 ◽  
Author(s):  
Sue-Hwa Lin ◽  
Yu-Chen Lee ◽  
Song-Chang Lin ◽  
Guoyu Yu ◽  
Ming Zhu ◽  
...  

Metastatic prostate cancer (PCa) in bone induces bone-forming lesions that enhance PCa progression. How tumor-induced bone formation enhances PCa progression is not known. We have previously shown that PCa-induced bone originates from endothelial cells (EC) that have undergone endothelial-to-osteoblast (EC-to-OSB) transition by tumor-secreted BMP4. Here, we show that EC-to-OSB transition leads to changes in the tumor microenvironment that increases the metastatic potential of PCa cells. We found that conditioned medium (CM) from EC-OSB hybrid cells increases the migration, invasion and survival of PC3-mm2 and C4-2B4 PCa cells. Quantitative mass spectrometry (iTRAQ) identified Tenascin C (TNC) as one of the major proteins secreted from EC-OSB hybrid cells. TNC expression in tumor-induced osteoblasts was confirmed by immunohistochemistry of MDA-PCa118b xenograft and human bone metastasis specimens. Mechanistically, BMP4 increases TNC expression in EC-OSB cells through the Smad1-Notch/Hey1 pathway. How TNC promotes PCa metastasis was next interrogated by in vitro and in vivo studies. In vitro studies showed that a TNC neutralizing antibody inhibits EC-OSB-CM-mediated PCa cell migration and survival. TNC knockdown decreased, while addition of recombinant TNC or TNC overexpression increased migration and anchorage-independent growth of PC3 or C4-2b cells. When injected orthotopically, PC3-mm2-shTNC clones decreased metastasis to bone, while C4-2b-TNC overexpressing cells increased metastasis to lymph nodes. TNC enhances PCa cell migration through α5β1 integrin-mediated YAP/TAZ inhibition. These studies elucidate that tumor-induced stromal reprogramming generates TNC that enhances PCa metastasis and suggest that TNC may be a target for PCa therapy.


2021 ◽  
Author(s):  
Junmin Lee ◽  
Aly Ung ◽  
Hanjun Kim ◽  
KangJu Lee ◽  
Hyun-Jong Cho ◽  
...  

Abstract Increasing evidence from cancer cell fusion with different cell types in the tumor microenvironment has suggested a probable mechanism for how metastasis-initiating cells could be generated in tumors. Although human mesenchymal stem cells (hMSCs) have been known as promising candidates to create hybrid cells with cancer cells, the role of hMSCs in fusion with cancer cells is still controversial. Here, we fabricated a liver-on-a-chip platform to monitor the fusion of liver hepatocellular cells (HepG2) with hMSCs and study their invasive potential. We demonstrated that hMSCs might play dual roles in HepG2 spheroids. The analysis of tumor growth with different fractions of hMSCs in HepG2 spheroids revealed hMSCs’ role in preventing HepG2 growth and proliferation, while the hMSCs presented in the HepG2 spheroids led to the generation of HepG2-hMSC hybrid cells with much higher invasiveness compared to HepG2. These invasive HepG2-hMSC hybrid cells expressed high levels of markers associated with stemness, proliferation, epithelial to mesenchymal transition, and matrix deposition, which corresponded to the expression of these markers for hMSCs escaping from hMSC spheroids. In addition, these fused cells were responsible for collective invasion following HepG2 by depositing Collagen I and Fibronectin in their surrounding microenvironment. Furthermore, we showed that hepatic stellate cells (HSCs) could also be fused with HepG2, and the HepG2-HSC hybrid cells possessed similar features to those from HepG2-hMSC fusion. This fusion of HepG2 with liver-resident HSCs may propose a new potential mechanism of hepatic cancer metastasis.


Cancers ◽  
2021 ◽  
Vol 13 (18) ◽  
pp. 4636
Author(s):  
Ralf Hass ◽  
Juliane von der Ohe ◽  
Thomas Dittmar

Fusion of cancer cells either with other cancer cells (homotypic fusion) in local vicinity of the tumor tissue or with other cell types (e.g., macrophages, cancer-associated fibroblasts (CAFs), mesenchymal stromal-/stem-like cells (MSC)) (heterotypic fusion) represents a rare event. Accordingly, the clinical relevance of cancer-cell fusion events appears questionable. However, enhanced tumor growth and/or development of certain metastases can originate from cancer-cell fusion. Formation of hybrid cells after cancer-cell fusion requires a post-hybrid selection process (PHSP) to cope with genomic instability of the parental nuclei and reorganize survival and metabolic functionality. The present review dissects mechanisms that contribute to a PHSP and resulting functional alterations of the cancer hybrids. Based upon new properties of cancer hybrid cells, the arising clinical consequences of the subsequent tumor heterogeneity after cancer-cell fusion represent a major therapeutic challenge. However, cellular partners during cancer-cell fusion such as MSC within the tumor microenvironment or MSC-derived exosomes may provide a suitable vehicle to specifically address and deliver anti-tumor cargo to cancer cells.


Cancers ◽  
2021 ◽  
Vol 13 (17) ◽  
pp. 4496
Author(s):  
Ralf Hass ◽  
Juliane von der Ohe ◽  
Thomas Dittmar

The generation of cancer hybrid cells by intra-tumoral cell fusion opens new avenues for tumor plasticity to develop cancer stem cells with altered properties, to escape from immune surveillance, to change metastatic behavior, and to broaden drug responsiveness/resistance. Genomic instability and chromosomal rearrangements in bi- or multinucleated aneuploid cancer hybrid cells contribute to these new functions. However, the significance of cell fusion in tumorigenesis is controversial with respect to the low frequency of cancer cell fusion events and a clonal advantage of surviving cancer hybrid cells following a post-hybrid selection process. This review highlights alternative processes of cancer hybrid cell development such as entosis, emperipolesis, cannibalism, therapy-induced polyploidization/endoreduplication, horizontal or lateral gene transfer, and focusses on the predominant mechanisms of cell fusion. Based upon new properties of cancer hybrid cells the arising clinical consequences of the subsequent tumor heterogeneity after cancer cell fusion represent a major therapeutic challenge.


2021 ◽  
Vol 32 ◽  
pp. S367-S368
Author(s):  
E.V. Denisov ◽  
M.E. Menyailo ◽  
S.Y. Zolotareva ◽  
T.S. Gerashchenko ◽  
V.V. Alifanov ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document