Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 242
Filter
1.
Cells ; 13(18)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39329722

ABSTRACT

Giardia doudenalis (lamblia, intestinalis) is a protozoan parasite that inhabits the lumen of the upper small intestine of vertebrates, causing chronic abdominal pains and severe diarrhea, symptoms of giardiasis, a persistent and recurrent infection. This characteristic is mainly due to the presence of membrane variant-specific surface proteins (VSPs) that give this parasite the ability to successively infect the host through antigenic variation. Using high-resolution scanning microscopy (HR-SM), we observed the presence, formation, and extension of tunneling-nanotube-like surface structures in Giardia, especially following parasite challenges with VSP antibodies. They were seen all over the parasite surface, both in vitro and in vivo, showing that G. duodenalis nanotube formation occurs in complex environments such as the gut. In addition, we also observed that some of these nanotubes displayed a periodic strangulation that produces 100 nm vesicles that seemed to be released in a process similar to that previously observed in Trypanosoma brucei. The presence of nanotube-like structures in G. duodenalis highlights yet another strategy of cellular communication utilized by these parasites, whether between themselves or with the host cell.


Subject(s)
Giardia lamblia , Nanotubes , Nanotubes/chemistry , Animals , Protozoan Proteins/metabolism , Protozoan Proteins/chemistry , Giardiasis/parasitology , Giardiasis/immunology , Mice , Humans
2.
Traffic ; 25(9): e12951, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39238078

ABSTRACT

Mitochondria, the dynamic organelles responsible for energy production and cellular metabolism, have the metabolic function of extracting energy from nutrients and synthesizing crucial metabolites. Nevertheless, recent research unveils that intercellular mitochondrial transfer by tunneling nanotubes, tumor microtubes, gap junction intercellular communication, extracellular vesicles, endocytosis and cell fusion may regulate mitochondrial function within recipient cells, potentially contributing to disease treatment, such as nonalcoholic steatohepatitis, glioblastoma, ischemic stroke, bladder cancer and neurodegenerative diseases. This review introduces the principal approaches to intercellular mitochondrial transfer and examines its role in various diseases. Furthermore, we provide a comprehensive overview of the inhibitors and activators of intercellular mitochondrial transfer, offering a unique perspective to illustrate the relationship between intercellular mitochondrial transfer and diseases.


Subject(s)
Mitochondria , Humans , Mitochondria/metabolism , Animals , Cell Communication , Extracellular Vesicles/metabolism , Biological Transport , Endocytosis/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/therapy
3.
Elife ; 132024 Sep 09.
Article in English | MEDLINE | ID: mdl-39250349

ABSTRACT

Tunneling nanotubes (TNTs) are open actin- and membrane-based channels, connecting remote cells and allowing direct transfer of cellular material (e.g. vesicles, mRNAs, protein aggregates) from the cytoplasm to the cytoplasm. Although they are important especially, in pathological conditions (e.g. cancers, neurodegenerative diseases), their precise composition and their regulation were still poorly described. Here, using a biochemical approach allowing to separate TNTs from cell bodies and from extracellular vesicles and particles (EVPs), we obtained the full composition of TNTs compared to EVPs. We then focused on two major components of our proteomic data, the CD9 and CD81 tetraspanins, and further investigated their specific roles in TNT formation and function. We show that these two tetraspanins have distinct non-redundant functions: CD9 participates in stabilizing TNTs, whereas CD81 expression is required to allow the functional transfer of vesicles in the newly formed TNTs, possibly by regulating docking to or fusion with the opposing cell.


Subject(s)
Proteomics , Tetraspanin 28 , Tetraspanin 29 , Tetraspanin 28/metabolism , Tetraspanin 28/genetics , Tetraspanin 29/metabolism , Tetraspanin 29/genetics , Proteomics/methods , Humans , Animals , Extracellular Vesicles/metabolism , Nanotubes/chemistry , Mice , Cell Communication
4.
Results Probl Cell Differ ; 73: 155-171, 2024.
Article in English | MEDLINE | ID: mdl-39242379

ABSTRACT

Tunneling nanotubes (TNTs) are open-ended, membrane-encased extensions that connect neighboring cells. They have diameters up to 1 µm but are able to expand to convey large cargos. Lengths vary depending on the distance of the cells but have been reported to be capable of extending beyond 300 µm. They have actin cytoskeletons that are essential for their formation, and may or may not have microtubule networks. It is thought that thin TNTs lack microtubules, while thicker TNTs have microtubular highways that use motor proteins to convey materials, including proteins, mitochondria, and nanoparticles between cells. Specifically, the presence of dynein and myosin support trafficking of cargo in both directions. The purpose of these connections is to enable cells to work as a unit or to extend cell life by diluting cytotoxic agents or acquiring biological material needed to survive.


Subject(s)
Cell Communication , Microtubules , Nanotubes , Microtubules/metabolism , Humans , Animals , Cell Communication/physiology , Cell Membrane Structures
5.
Results Probl Cell Differ ; 73: 353-373, 2024.
Article in English | MEDLINE | ID: mdl-39242386

ABSTRACT

Tunneling nanotubes (TNTs) are thin, membranous protrusions that connect cells and allow for the transfer of various molecules, including proteins, organelles, and genetic material. TNTs have been implicated in a wide range of biological processes, including intercellular communication, drug resistance, and viral transmission. In cancer, they have been investigated more deeply over the past decade for their potentially pivotal role in tumor progression and metastasis. TNTs, as cell contact-dependent protrusions that form at short and long distances, enable the exchange of signaling molecules and cargo between cancer cells, facilitating communication and coordination of their actions. This coordination induces a synchronization that is believed to mediate the TNT-directed evolution of drug resistance by allowing cancer cells to coordinate, including through direct expulsion of chemotherapeutic drugs to neighboring cells. Despite advances in the overall field of TNT biology since the first published report of their existence in 2004 (Rustom A, Saffrich R, Markovic I, Walther P, Gerdes HH, Science. 303:1007-10, 2004), the mechanisms of formation and components vital for the function of TNTs are complex and not yet fully understood. However, several factors have been implicated in their regulation, including actin polymerization, microtubule dynamics, and signaling pathways. The discovery of TNT-specific components that are necessary and sufficient for their formation, maintenance, and action opens a new potential avenue for drug discovery in cancer. Thus, targeting TNTs may offer a promising therapeutic strategy for cancer treatment. By disrupting TNT formation or function, it may be possible to inhibit tumor growth and metastasis and overcome drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Neoplasms , Humans , Animals , Neoplasms/drug therapy , Neoplasms/pathology , Cell Communication , Nanotubes/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Membrane Structures
6.
Results Probl Cell Differ ; 73: 435-474, 2024.
Article in English | MEDLINE | ID: mdl-39242389

ABSTRACT

Viruses are vehicles to exchange genetic information and proteins between cells and organisms by infecting their target cells either cell-free, or depending on cell-cell contacts. Several viruses like certain retroviruses or herpesviruses transmit by both mechanisms. However, viruses have also evolved the properties to exchange proteins between cells independent of viral particle formation. This exchange of viral proteins can be directed to target cells prior to infection to interfere with restriction factors and intrinsic immunity, thus, making the target cell prone to infection. However, also bystander cells, e.g. immune cell populations, can be targeted by viral proteins to dampen antiviral responses. Mechanistically, viruses exploit several routes of cell-cell communication to exchange viral proteins like the formation of extracellular vesicles or the formation of long-distance connections like tunneling nanotubes. Although it is known that viral nucleic acids can be transferred between cells as well, this chapter concentrates on viral proteins of human pathogenic viruses covering all Baltimore classes and summarizes our current knowledge on intercellular transport of viral proteins between cells.


Subject(s)
Viral Proteins , Humans , Viral Proteins/metabolism , Animals , Biological Transport , Viruses/immunology , Viruses/metabolism , Cell Communication
7.
Results Probl Cell Differ ; 73: 419-434, 2024.
Article in English | MEDLINE | ID: mdl-39242388

ABSTRACT

Tunneling nanotubes (TNTs) are cellular connections, which represent a novel route for cell-to-cell communication. Strong evidence points to a role for TNTs in the intercellular transfer of signals, molecules, organelles, and pathogens, involving them in many cellular functions. In myeloid cells (e.g., monocytes/macrophages, dendritic cells, and osteoclasts), intercellular communication via TNT contributes to their differentiation and immune functions, by favoring material and pathogen transfer, as well as cell fusion. This chapter addresses the complexity of the definition and characterization of TNTs in myeloid cells, the different processes involved in their formation, their existence in vivo, and finally their function(s) in health and infectious diseases, with the example of HIV-1 infection.


Subject(s)
Cell Communication , Myeloid Cells , Humans , Cell Communication/physiology , Animals , HIV Infections/immunology , HIV-1/physiology , Cell Membrane Structures , Nanotubes
9.
Mater Today Bio ; 28: 101175, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39171100

ABSTRACT

The repair of bone defects in the elderly individuals is significantly delayed due to cellular senescence and dysfunction, which presents a challenge in clinical settings. Furthermore, there are limited effective methods available to promote bone repair in older individuals. Herein, melatonin-loaded mesoporous bioactive glasses microspheres (MTBG) were successfully prepared based on their mesoporous properties. The repair of bone defects in aged rats was significantly accelerated by enhancing mitochondrial function through the sustained release of melatonin and bioactive ions. MTBG effectively rejuvenated senescent bone marrow mesenchymal stem cells (BMSCs) by scavenging excessive reactive oxygen species (ROS), stabilizing the mitochondrial membrane potential (ΔΨm), and increasing ATP synthesis. Analysis of the underlying mechanism revealed that the formation of tunneling nanotubes (TNTs) facilitated the intercellular transfer of mitochondria, thereby resulting in the recovery of mitochondrial function. This study provides critical insights into the design of new biomaterials for the elderly individuals and the biological mechanism involved in aged bone regeneration.

10.
Acta Biomater ; 185: 312-322, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38969079

ABSTRACT

Mutation in oncogene KRas plays a crucial role in the occurrence and progression of numerous malignant tumors. Malignancy involves changes in cell mechanics for extensive cellular deformation during metastatic dissemination. We hypothesize that oncogene KRas mutations are intrinsic to alterations in cellular mechanics that promote malignant tumor generation and progression. Here, we demonstrate the use of optical tweezers coupled with a confocal fluorescence imaging system and gene interference technique to reveal that the mutant KRas protein can be transported between homogeneous and heterogeneous tumor cells by tunneling nanotubes (TNTs), resulting in a significant reduction of membrane tension and acceleration of membrane phospholipid flow in the recipient cells. Simultaneously, the changes in membrane mechanical properties of the tumor cells also enhance the metastatic and invasive ability of the tumors, which further contribute to the deterioration of the tumors. This finding helps to clarify the association between oncogene mutations and changes in the mechanical properties of tumor cells, which provides a theoretical basis for the development of cancer treatment strategies. STATEMENT OF SIGNIFICANCE: Here, we present a laser confocal fluorescence system integrated with optical tweezers to observe the transfer of mutant KRasG12D protein from mutant cells to wild-type cells through TNTs. Malignancy involves changes in cell mechanics for extensive cellular deformation during metastatic dissemination. Our results demonstrate a significant decrease in membrane tension and an increase in membrane phospholipid flow in recipient cells. These alterations in mechanical properties augment the migration and invasive capabilities of tumor cells, contributing to tumor malignancy. Our findings propose that cellular mechanical properties could serve as new markers for tumor development, and targeting membrane tension may hold potential as a therapeutic strategy.


Subject(s)
Cell Membrane , Nanotubes , Proto-Oncogene Proteins p21(ras) , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Humans , Nanotubes/chemistry , Cell Membrane/metabolism , Animals , Cell Line, Tumor , Mutation/genetics , Optical Tweezers , Mice , Protein Transport , Neoplasms/pathology , Neoplasms/metabolism
11.
Neuron ; 112(18): 3106-3125.e8, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39059388

ABSTRACT

Microglia are crucial for maintaining brain health and neuron function. Here, we report that microglia establish connections with neurons using tunneling nanotubes (TNTs) in both physiological and pathological conditions. These TNTs facilitate the rapid exchange of organelles, vesicles, and proteins. In neurodegenerative diseases like Parkinson's and Alzheimer's disease, toxic aggregates of alpha-synuclein (α-syn) and tau accumulate within neurons. Our research demonstrates that microglia use TNTs to extract neurons from these aggregates, restoring neuronal health. Additionally, microglia share their healthy mitochondria with burdened neurons, reducing oxidative stress and normalizing gene expression. Disrupting mitochondrial function with antimycin A before TNT formation eliminates this neuroprotection. Moreover, co-culturing neurons with microglia and promoting TNT formation rescues suppressed neuronal activity caused by α-syn or tau aggregates. Notably, TNT-mediated aggregate transfer is compromised in microglia carrying Lrrk22(Gly2019Ser) or Trem2(T66M) and (R47H) mutations, suggesting a role in the pathology of these gene variants in neurodegenerative diseases.


Subject(s)
Microglia , Neurons , alpha-Synuclein , tau Proteins , Microglia/metabolism , Microglia/drug effects , Animals , Neurons/metabolism , Neurons/drug effects , tau Proteins/metabolism , tau Proteins/genetics , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Coculture Techniques , Mice , Mitochondria/metabolism , Mitochondria/drug effects , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Cell Death/drug effects , Cell Death/physiology , Nanotubes , Cells, Cultured , Cell Communication/physiology , Cell Communication/drug effects , Oxidative Stress/drug effects , Oxidative Stress/physiology , Cell Membrane Structures
12.
Stem Cell Res Ther ; 15(1): 189, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956646

ABSTRACT

BACKGROUND: Recent studies have proved the role of autophagy in mesenchymal stem cell (MSCs) function and regenerative properties. How and by which mechanism autophagy modulation can affect the juxtacrine interaction of MSCs should be addressed. Here, the role of autophagy was investigated in the formation of tunneling nanotubes (TNTs) and homotypic mitochondrial donation. METHODS: MSCs were incubated with 15 µM Metformin (Met) and/or 3 µM 3-methyladenine (3-MA) for 48 h. The formation of TNTs was assessed using bright-field and SEM images. The mitochondria density and ΔΨ values were monitored using flow cytometry analysis. Using RT-PCR and protein array, the close interaction and shared mediators between autophagy, apoptosis, and Wnt signaling pathways were also monitored. The total fatty acid profile was assessed using gas chromatography. RESULT: Data indicated the increase of TNT length and number, along with other cell projections after the induction of autophagy while these features were blunted in 3-MA-treated MSCs (p < 0.05). Western blotting revealed the significant reduction of Rab8 and p-FAK in 3-MA-treated MSCs (p < 0.05), indicating the inhibition of TNT assembly and vesicle transport. Likewise, the stimulation of autophagy increased autophagic flux and mitochondrial membrane integrity compared to 3-MA-treated MSCs. Despite these findings, protein levels of mitochondrial membrane Miro1 and 2 were unchanged after autophagy inhibition/stimulation (p > 0.05). We found that the inhibition/stimulation of autophagy can affect the protein, and transcription levels of several mediators related to Wnt and apoptosis signaling pathways involved in different cell bioactivities. Data confirmed the profound increase of mono and polyunsaturated/saturated fatty acid ratio in MSCs exposed to autophagy stimulator. CONCLUSIONS: In summary, autophagy modulation could affect TNT formation which is required for homotypic mitochondrial donation. Thus, the modulation of autophagy creates a promising perspective to increase the efficiency of cell-based therapies.


Subject(s)
Autophagy , Mesenchymal Stem Cells , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mitochondria/metabolism , Adenine/pharmacology , Adenine/analogs & derivatives , Humans , Nanotubes/chemistry , Apoptosis/drug effects , Animals , Metformin/pharmacology , Cells, Cultured , Wnt Signaling Pathway/drug effects , Cell Membrane Structures
13.
Theranostics ; 14(10): 3843-3858, 2024.
Article in English | MEDLINE | ID: mdl-38994028

ABSTRACT

Rationale: Extracellular vesicles (EVs) are thought to mediate intercellular communication during development and disease. Yet, biological insight to intercellular EV transfer remains elusive, also in the heart, and is technically challenging to demonstrate. Here, we aimed to investigate biological transfer of cardiomyocyte-derived EVs in the neonatal heart. Methods: We exploited CD9 as a marker of EVs, and generated two lines of cardiomyocyte specific EV reporter mice: Tnnt2-Cre; double-floxed inverted CD9/EGFP and αMHC-MerCreMer; double-floxed inverted CD9/EGFP. The two mouse lines were utilized to determine whether developing cardiomyocytes transfer EVs to other cardiac cells (non-myocytes and cardiomyocytes) in vitro and in vivo and investigate the intercellular transport pathway of cardiomyocyte-derived EVs. Results: Genetic tagging of cardiomyocytes was confirmed in both reporter mouse lines and proof of concept in the postnatal heart showed that, a fraction of EGFP+/MYH1- non-myocytes exist firmly demonstrating in vivo cardiomyocyte-derived EV transfer. However, two sets of direct and indirect EGFP +/- cardiac cell co-cultures showed that cardiomyocyte-derived EGFP+ EV transfer requires cell-cell contact and that uptake of EGFP+ EVs from the medium is limited. The same was observed when co-cultiring with mouse macrophages. Further mechanistic insight showed that cardiomyocyte EV transfer occurs through type I tunneling nanotubes. Conclusion: While the current notion assumes that EVs are transferred through secretion to the surroundings, our data show that cardiomyocyte-derived EV transfer in the developing heart occurs through nanotubes between neighboring cells. Whether these data are fundamental and relate to adult hearts and other organs remains to be determined, but they imply that the normal developmental process of EV transfer goes through cell-cell contact rather than through the extracellular compartment.


Subject(s)
Cell Communication , Coculture Techniques , Extracellular Vesicles , Myocytes, Cardiac , Animals , Extracellular Vesicles/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Mice , Cell Communication/physiology , Nanotubes , Heart/physiology , Tetraspanin 29/metabolism , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/genetics , Animals, Newborn , Mice, Transgenic
14.
Biochem Soc Trans ; 52(4): 1757-1764, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39034648

ABSTRACT

Intercellular communication within the tumor microenvironment (TME) is essential for establishing, mediating, and synchronizing cancer cell invasion and metastasis. Cancer cells, individually and collectively, react at the cellular and molecular levels to insults from standard-of-care treatments used to treat patients with cancer. One form of cell communication that serves as a prime example of cellular phenotypic stress response is a type of cellular protrusion called tunneling nanotubes (TNTs). TNTs are ultrafine, actin-enriched contact-dependent forms of membrane protrusions that facilitate long distance cell communication through transfer of various cargo, including genetic materials, mitochondria, proteins, ions, and various other molecules. In the past 5-10 years, there has been a growing body of evidence that implicates TNTs as a novel mechanism of cell-cell communication in cancer that facilitates and propagates factors that drive or enhance chemotherapeutic resistance in a variety of cancer cell types. Notably, recent literature has highlighted the potential of TNTs to serve as cellular conduits and mediators of drug and nanoparticle delivery. Given that TNTs have also been shown to form in vivo in a variety of tumor types, disrupting TNT communication within the TME provides a novel strategy for enhancing the cytotoxic effect of existing chemotherapies while suppressing this form of cellular stress response. In this review, we examine current understanding of interplay between cancer cells occurring via TNTs, and even further, the implications of TNT-mediated tumor-stromal cross-talk and the potential to enhance chemoresistance. We then examine tumor microtubes, an analogous cell protrusion heavily implicated in mediating treatment resistance in glioblastoma multiforme, and end with a brief discussion of the effects of radiation and other emerging treatment modalities on TNT formation.


Subject(s)
Cell Communication , Drug Resistance, Neoplasm , Nanotubes , Neoplasms , Tumor Microenvironment , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Cell Membrane Structures
15.
Methods Mol Biol ; 2825: 333-343, 2024.
Article in English | MEDLINE | ID: mdl-38913319

ABSTRACT

Cancer cytogenetic analyses often involve cell culture. However, many cytogeneticists overlook interesting phenotypes associated with cultured cells. Given that cytogeneticists need to focus more on phenotypes to comprehend the genotypes, the biological significance of seemingly trivial cellular variations deserves attention. One example is the formation of cellular tunneling tubes (TTs) in cultured cancer cells, which likely play a role in cell-to-cell communication and material transport. In this chapter, we describe protocols for studying these TTs as well as cellular spheres. In addition to diverse chromosomal variants, these different types of variations should be considered for understanding cancer heterogeneity and dynamics, as they illustrate the importance of various forms of fuzzy inheritance.


Subject(s)
Cell Communication , Spheroids, Cellular , Humans , Spheroids, Cellular/cytology , Cell Culture Techniques/methods , Cell Line, Tumor
16.
J Exp Clin Cancer Res ; 43(1): 166, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877575

ABSTRACT

BACKGROUND: Breast cancer (BC) is a complex disease, showing heterogeneity in the genetic background, molecular subtype, and treatment algorithm. Historically, treatment strategies have been directed towards cancer cells, but these are not the unique components of the tumor bulk, where a key role is played by the tumor microenvironment (TME), whose better understanding could be crucial to obtain better outcomes. METHODS: We evaluated mitochondrial transfer (MT) by co-culturing Adipose stem cells with different Breast cancer cells (BCCs), through MitoTracker assay, Mitoception, confocal and immunofluorescence analyses. MT inhibitors were used to confirm the MT by Tunneling Nano Tubes (TNTs). MT effect on multi-drug resistance (MDR) was assessed using Doxorubicin assay and ABC transporter evaluation. In addition, ATP production was measured by Oxygen Consumption rates (OCR) and Immunoblot analysis. RESULTS: We found that MT occurs via Tunneling Nano Tubes (TNTs) and can be blocked by actin polymerization inhibitors. Furthermore, in hybrid co-cultures between ASCs and patient-derived organoids we found a massive MT. Breast Cancer cells (BCCs) with ASCs derived mitochondria (ADM) showed a reduced HIF-1α expression in hypoxic conditions, with an increased ATP production driving ABC transporters-mediated multi-drug resistance (MDR), linked to oxidative phosphorylation metabolism rewiring. CONCLUSIONS: We provide a proof-of-concept of the occurrence of Mitochondrial Transfer (MT) from Adipose Stem Cells (ASCs) to BC models. Blocking MT from ASCs to BCCs could be a new effective therapeutic strategy for BC treatment.


Subject(s)
Breast Neoplasms , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Mitochondria , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Female , Mitochondria/metabolism , Stem Cells/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Cell Line, Tumor , Tumor Microenvironment
17.
Nutrients ; 16(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38931195

ABSTRACT

Understanding the role of biased taste T1R2/T1R3 G protein-coupled receptors (GPCR) agonists on glycosylated receptor signaling may provide insights into the opposing effects mediated by artificial and natural sweeteners, particularly in cancer and metastasis. Sweetener-taste GPCRs can be activated by several active states involving either biased agonism, functional selectivity, or ligand-directed signaling. However, there are increasing arrays of sweetener ligands with different degrees of allosteric biased modulation that can vary dramatically in binding- and signaling-specific manners. Here, emerging evidence proposes the involvement of taste GPCRs in a biased GPCR signaling crosstalk involving matrix metalloproteinase-9 (MMP-9) and neuraminidase-1 (Neu-1) activating glycosylated receptors by modifying sialic acids. The findings revealed that most natural and artificial sweeteners significantly activate Neu-1 sialidase in a dose-dependent fashion in RAW-Blue and PANC-1 cells. To confirm this biased GPCR signaling crosstalk, BIM-23127 (neuromedin B receptor inhibitor, MMP-9i (specific MMP-9 inhibitor), and oseltamivir phosphate (specific Neu-1 inhibitor) significantly block sweetener agonist-induced Neu-1 sialidase activity. To assess the effect of artificial and natural sweeteners on the key survival pathways critical for pancreatic cancer progression, we analyzed the expression of epithelial-mesenchymal markers, CD24, ADLH-1, E-cadherin, and N-cadherin in PANC-1 cells, and assess the cellular migration invasiveness in a scratch wound closure assay, and the tunneling nanotubes (TNTs) in staging the migratory intercellular communication. The artificial and natural sweeteners induced metastatic phenotype of PANC-1 pancreatic cancer cells to promote migratory intercellular communication and invasion. The sweeteners also induced the downstream NFκB activation using the secretory alkaline phosphatase (SEAP) assay. These findings elucidate a novel taste T1R2/T1R3 GPCR functional selectivity of a signaling platform in which sweeteners activate downstream signaling, contributing to tumorigenesis and metastasis via a proposed NFκB-induced epigenetic reprogramming modeling.


Subject(s)
Epithelial-Mesenchymal Transition , Matrix Metalloproteinase 9 , Neoplasm Metastasis , Receptors, G-Protein-Coupled , Sweetening Agents , Humans , Epithelial-Mesenchymal Transition/drug effects , Receptors, G-Protein-Coupled/metabolism , Sweetening Agents/pharmacology , Cell Line, Tumor , Matrix Metalloproteinase 9/metabolism , Glycosylation/drug effects , Signal Transduction/drug effects , Phenotype , Animals , Taste/drug effects , Cell Movement/drug effects , Neuraminidase
18.
Tissue Cell ; 89: 102431, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38870572

ABSTRACT

Tunneling nanotubes (TNTs) represent an innovative way for cells to communicate with one another, as they act as long conduits between cells. However, their roles in human dermal microvascular pericytes (HDMPCs) interaction remain elusive in vitro. In this work, we identified and characterized the TNT-like structures that connected two or more pericytes in two-dimensional cultures and formed a functional network in the human dermis. Immunofluorescence assay indicated that the F-actin was an essential element to form inter-pericyte TNT-like structures, as it decreased in actin polymer inhibitor-cytochalasin B treated groups, and microtubules were present in almost half of the TNT-like structures. Most importantly, we only found the presence of mitochondrial in TNT-like structures containing α-tubulin, and the application of microtubule assembly inhibitor-Nocodazole significantly reduced the percentage of TNT-like structures that contain α-tubulin, resulting in a sudden decrease in the positive rate of cytochrome c oxidase subunit 4 isoform 1 (COX IV, a marker of mitochondria) in TNT-like structures. In summary, we described a novel intercellular communication-TNT-like structures-between HDMPCs in vitro, and this work allows us to properly understand the cellular mechanisms of spreading materials between HDMPCs, shedding light on the role of HDMPCs.


Subject(s)
Pericytes , Humans , Pericytes/cytology , Pericytes/metabolism , Tubulin/metabolism , Microtubules/metabolism , Dermis/cytology , Dermis/metabolism , Cell Communication , Mitochondria/metabolism , Actins/metabolism , Nanotubes/chemistry , Microvessels/cytology , Microvessels/metabolism , Cells, Cultured , Cell Membrane Structures
19.
Cell Signal ; 121: 111274, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38936787

ABSTRACT

Tumor-associated macrophages (TAMs) secrete cytokines, chemokines, and growth factors in the tumor microenvironment (TME) to support cancer progression. Higher TAM infiltration in the breast TME is associated with a poor prognosis. Previous studies have demonstrated the role of macrophages in stimulating long-range intercellular bridges referred to as tunneling nanotubes (TNTs) in cancer cells. Intercellular communication between cancer cells via TNTs promotes cancer growth, invasion, metastasis, and therapy resistance. Given the important role of TNTs and macrophages in cancer, the role of macrophage-induced TNTs in chemotherapy drug doxorubicin resistance is not known. Furthermore, the mechanism of macrophage-mediated TNT formation is elusive. In this study, it is shown that the macrophage-conditioned medium (MΦCM) partially mimicked inflammatory TME, induced an EMT phenotype, and increased migration in MCF-7 breast cancer cells. Additionally, secreted proteins in MΦCM induced TNT formation in MCF-7 cells, which led to increased resistance to doxorubicin. Transcriptomic analysis of MΦCM-treated MCF-7 cells showed enrichment of the NF-κB and focal adhesion pathways, as well as upregulation of genes involved in EMT, extracellular remodeling, and actin cytoskeleton reorganization. Interestingly, inhibitors of PKC, Src, NF-κB, and p38 decreased macrophage-induced TNT formation in MCF-7 cells. These results reveal the novel role of PKC and Src in inducing TNT formation in cancer cells and suggest that inhibition of PKC and Src activity may likely contribute to reduced macrophage-breast cancer cell interaction and the potential therapeutic strategy of cancer.


Subject(s)
Breast Neoplasms , NF-kappa B , Protein Kinase C , p38 Mitogen-Activated Protein Kinases , Humans , NF-kappa B/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Culture Media, Conditioned/pharmacology , MCF-7 Cells , p38 Mitogen-Activated Protein Kinases/metabolism , Female , Protein Kinase C/metabolism , Doxorubicin/pharmacology , Macrophages/metabolism , Macrophages/drug effects , src-Family Kinases/metabolism , Tumor Microenvironment , Nanotubes/chemistry , Signal Transduction/drug effects , Cell Movement/drug effects , MAP Kinase Signaling System/drug effects
20.
J Exp Clin Cancer Res ; 43(1): 147, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38769583

ABSTRACT

A century ago, the Warburg effect was first proposed, revealing that cancer cells predominantly rely on glycolysis during the process of tumorigenesis, even in the presence of abundant oxygen, shifting the main pathway of energy metabolism from the tricarboxylic acid cycle to aerobic glycolysis. Recent studies have unveiled the dynamic transfer of mitochondria within the tumor microenvironment, not only between tumor cells but also between tumor cells and stromal cells, immune cells, and others. In this review, we explore the pathways and mechanisms of mitochondrial transfer within the tumor microenvironment, as well as how these transfer activities promote tumor aggressiveness, chemotherapy resistance, and immune evasion. Further, we discuss the research progress and potential clinical significance targeting these phenomena. We also highlight the therapeutic potential of targeting intercellular mitochondrial transfer as a future anti-cancer strategy and enhancing cell-mediated immunotherapy.


Subject(s)
Mitochondria , Neoplasms , Humans , Neoplasms/metabolism , Neoplasms/therapy , Neoplasms/pathology , Neoplasms/drug therapy , Mitochondria/metabolism , Tumor Microenvironment , Animals , Nanotubes
SELECTION OF CITATIONS
SEARCH DETAIL