12 results on '"exofucosylation"'
Search Results
2. Optimizing cryopreservation conditions for use of fucosylated human mesenchymal stromal cells in anti-inflammatory/immunomodulatory therapeutics.
- Author
-
Gil-Chinchilla, Jesús I., Bueno, Carlos, Martínez, Carlos M., Ferrández-Múrtula, Ana, García-Hernández, Ana M., Blanquer, Miguel, Molina-Molina, Mar, Zapata, Agustín G., Sackstein, Robert, Moraleda, Jose M., and García-Bernal, David
- Subjects
STROMAL cells ,MANUFACTURING cells ,THERAPEUTICS ,BONE marrow ,ADIPOSE tissues ,CRYOPROTECTIVE agents - Abstract
Mesenchymal stem/stromal cells (MSCs) are being increasingly used in cellbased therapies due to their broad anti-inflammatory and immunomodulatory properties. Intravascularly-administered MSCs do not efficiently migrate to sites of inflammation/immunopathology, but this shortfall has been overcome by cell surface enzymatic fucosylation to engender expression of the potent E-selectin ligand HCELL. In applications of cell-based therapies, cryopreservation enables stability in both storage and transport of the produced cells from the manufacturing facility to the point of care. However, it has been reported that cryopreservation and thawing dampens their immunomodulatory/antiinflammatory activity even after a reactivation/reconditioning step. To address this issue, we employed a variety of methods to cryopreserve and thaw fucosylated human MSCs derived from either bone marrow or adipose tissue sources. We then evaluated their immunosuppressive properties, cell viability, morphology, proliferation kinetics, immunophenotype, senescence, and osteogenic and adipogenic differentiation. Our studies provide new insights into the immunobiology of cryopreserved and thawed MSCs and offer a readily applicable approach to optimize the use of fucosylated human allogeneic MSCs as immunomodulatory/anti-inflammatory therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Optimizing cryopreservation conditions for use of fucosylated human mesenchymal stromal cells in anti-inflammatory/immunomodulatory therapeutics
- Author
-
Jesús I. Gil-Chinchilla, Carlos Bueno, Carlos M. Martínez, Ana Ferrández-Múrtula, Ana M. García-Hernández, Miguel Blanquer, Mar Molina-Molina, Agustín G. Zapata, Robert Sackstein, Jose M. Moraleda, and David García-Bernal
- Subjects
mesenchymal stromal cells ,exofucosylation ,HCELL ,E-selectin ligand ,cryopreservation ,immunomodulation ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Mesenchymal stem/stromal cells (MSCs) are being increasingly used in cell-based therapies due to their broad anti-inflammatory and immunomodulatory properties. Intravascularly-administered MSCs do not efficiently migrate to sites of inflammation/immunopathology, but this shortfall has been overcome by cell surface enzymatic fucosylation to engender expression of the potent E-selectin ligand HCELL. In applications of cell-based therapies, cryopreservation enables stability in both storage and transport of the produced cells from the manufacturing facility to the point of care. However, it has been reported that cryopreservation and thawing dampens their immunomodulatory/anti-inflammatory activity even after a reactivation/reconditioning step. To address this issue, we employed a variety of methods to cryopreserve and thaw fucosylated human MSCs derived from either bone marrow or adipose tissue sources. We then evaluated their immunosuppressive properties, cell viability, morphology, proliferation kinetics, immunophenotype, senescence, and osteogenic and adipogenic differentiation. Our studies provide new insights into the immunobiology of cryopreserved and thawed MSCs and offer a readily applicable approach to optimize the use of fucosylated human allogeneic MSCs as immunomodulatory/anti-inflammatory therapeutics.
- Published
- 2024
- Full Text
- View/download PDF
4. Enforced sialyl‐Lewis‐X (sLeX) display in E‐selectin ligands by exofucosylation is dispensable for CD19‐CAR T‐cell activity and bone marrow homing
- Author
-
Diego Sánchez‐Martínez, Francisco Gutiérrez‐Agüera, Paola Romecin, Meritxell Vinyoles, Marta Palomo, Néstor Tirado, Samanta Romina Zanetti, Manel Juan, Michela Carlet, Irmela Jeremias, and Pablo Menéndez
- Subjects
BM homing ,CAR T‐cells ,E‐selectin ligands ,exofucosylation ,Medicine (General) ,R5-920 - Abstract
Abstract CD19‐directed chimeric antigen receptors (CAR) T cells induce impressive rates of complete response in advanced B‐cell malignancies, specially in B‐cell acute lymphoblastic leukemia (B‐ALL). However, CAR T‐cell‐treated patients eventually progress due to poor CAR T‐cell persistence and/or disease relapse. The bone marrow (BM) is the primary location for acute leukemia. The rapid/efficient colonization of the BM by systemically infused CD19‐CAR T cells might enhance CAR T‐cell activity and persistence, thus, offering clinical benefits. Circulating cells traffic to BM upon binding of tetrasaccharide sialyl‐Lewis X (sLeX)‐decorated E‐selectin ligands (sialofucosylated) to the E‐selectin receptor expressed in the vascular endothelium. sLeX‐installation in E‐selectin ligands is achieved through an ex vivo fucosylation reaction. Here, we sought to characterize the basal and cell‐autonomous display of sLeX in CAR T‐cells activated using different cytokines, and to assess whether exofucosylation of E‐selectin ligands improves CD19‐CAR T‐cell activity and BM homing. We report that cell‐autonomous sialofucosylation (sLeX display) steadily increases in culture‐ and in vivo‐expanded CAR T cells, and that, the cytokines used during T‐cell activation influence both the degree of such endogenous sialofucosylation and the CD19‐CAR T‐cell efficacy and persistence in vivo. However, glycoengineered enforced sialofucosylation of E‐selectin ligands was dispensable for CD19‐CAR T‐cell activity and BM homing in multiple xenograft models regardless the cytokines employed for T‐cell expansion, thus, representing a dispensable strategy for CD19‐CAR T‐cell therapy.
- Published
- 2021
- Full Text
- View/download PDF
5. Production via good manufacturing practice of exofucosylated human mesenchymal stromal cells for clinical applications.
- Author
-
LÓPEZ-LUCAS, MARÍA DOLORES, PACHÓN-PEÑA, GISELA, GARCÍA-HERNÁNDEZ, ANA MARÍA, PARRADO, ANTONIO, SÁNCHEZ-SALINAS, DARÍO, GARCÍA-BERNAL, DAVID, ALGUERÓ, MARIA DEL CARMEN, MARTINEZ, FRANCISCA INIESTA, BLANQUER, MIGUEL, CABAÑAS-PERIANES, VALENTÍN, MOLINA-MOLINA, MAR, ASÍN-AGUILAR, CIRA, MORALEDA, JOSÉ M, and SACKSTEIN, ROBERT
- Subjects
- *
CURRENT good manufacturing practices , *MESENCHYMAL stem cells , *CELLULAR therapy , *FUCOSYLTRANSFERASES , *PHOSPHORYLATION - Abstract
Abstract Background The regenerative and immunomodulatory properties of human mesenchymal stromal cells (hMSCs) have raised great hope for their use in cell therapy. However, when intravenously infused, hMSCs fail to reach sites of tissue injury. Fucose addition in α(1,3)-linkage to terminal sialyllactosamines on CD44 creates the molecule known as hematopoietic cell E-/L-selectin ligand (HCELL), programming hMSC binding to E-selectin that is expressed on microvascular endothelial cells of bone marrow (BM), skin and at all sites of inflammation. Here we describe how this modification on BM-derived hMSCs (BM-hMSCs) can be adapted to good manufacturing practice (GMP) standards. Methods BM-hMSCs were expanded using xenogenic-free media and exofucosylated using α(1,3)-fucosyltransferases VI (FTVI) or VII (FTVII). Enforced fucosylation converted CD44 into HCELL, and HCELL formation was assessed using Western blot, flow cytometry and cell-binding assays. Untreated (unfucosylated), buffer-treated and exofucosylated BM-hMSCs were each analyzed for cell viability, immunophenotype and differentiation potential, and E-selectin binding stability was assessed at room temperature, at 4°C, and after cryopreservation. Cell product safety was evaluated using microbiological testing, karyotype analysis, and c-Myc messenger RNA (mRNA) expression, and potential effects on genetic reprogramming and in cell signaling were analyzed using gene expression microarrays and receptor tyrosine kinase (RTK) phosphorylation arrays. Results Our protocol efficiently generates HCELL on clinical-scale batches of BM-hMSCs. Exofucosylation yields stable HCELL expression for 48 h at 4°C, with retained expression after cell cryopreservation. Cell viability and identity are unaffected by exofucosylation, without changes in gene expression or RTK phosphorylation. Discussion The described exofucosylation protocol using xenogenic-free reagents enforces HCELL expression on hMSCs endowing potent E-selectin binding without affecting cell viability or native phenotype. This described protocol is readily scalable for GMP-compliant clinical production. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
6. A Glycovariant of Human CD44 is Characteristically Expressed on Human Mesenchymal Stem Cells.
- Author
-
Pachón-Peña, Gisela, Donnelly, Conor, Ruiz-Cañada, Catalina, Katz, Adam, Fernández-Veledo, Sonia, Vendrell, Joan, and Sackstein, Robert
- Abstract
A bstract The clinical effectiveness of systemically administered human mesenchymal stem cells (hMSCs) depends on their capacity to engage vascular endothelium. hMSCs derived from bone marrow (BM-hMSCs) natively lack endothelial binding capacity, but express a CD44 glycovariant containing N-linked sialyllactosamines that can be α(1,3)-fucosylated using fucosyltransferase-VI (FTVI) to enforce sLe
X decorations, thereby creating hematopoietic cell E-/L-selectin ligand (HCELL). HCELL expression programs potent shear-resistant adhesion of circulating cells to endothelial beds expressing E-selectin. An alternative source of hMSCs is adipose tissue (A-hMSCs), and we assessed whether A-hMSCs bind E-selectin and/or possess sialyllactosamine-decorated CD44 accessible to α(1,3)-fucosylation. Similar to BM-hMSCs, we found that A-hMSCs natively lack E-selectin ligands, but FTVI-mediated cell surface α(1,3)-fucosylation induces sLeX expression and robust E-selectin binding secondary to conversion of CD44 into HCELL. Moreover, treatment with the α(1,3)-fucosyltransferase-FTVII also generated expression of HCELL on both BM-hMSCs and A-hMSCs, with sLeX decorations created on N-linked glycans of the 'standard' CD44 (CD44s) isoform. The finding that hMSCs from both source tissues each lack native E-selectin ligand expression prompted examination of the expression of glycosyltransferases that direct lactosaminyl glycan synthesis. These studies reveal that both types of hMSCs conspicuously lack transcripts encoding α(1,3)-fucosyltransferases, but equally express glycosyltransferases critical to creation of sialyllactosamines. Collectively, these data indicate that assembly of a sialyllactosaminyl-decorated CD44s glycovariant is a conserved feature of hMSCs derived from adipose tissue and marrow, thus identifying a CD44 glycosignature of these cells and supporting the applicability of cell surface α(1,3)-fucosylation in programming migration of systemically administered A-hMSCs to sites of tissue injury/inflammation. S tem C ells 2017;35:1080-1092 [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
7. Glycoengineering of E-Selectin Ligands by Intracellular versus Extracellular Fucosylation Differentially Affects Osteotropism of Human Mesenchymal Stem Cells.
- Author
-
Dykstra, Brad, Lee, Jungmin, Mortensen, Luke J., Yu, Haixiao, Wu, Zhengliang L., Lin, Charles P., Rossi, Derrick J., and Sackstein, Robert
- Subjects
MESENCHYMAL stem cell differentiation ,SELECTINS ,INTRACELLULAR membranes ,FUCOSYLATION ,LIGANDS (Biochemistry) ,MESSENGER RNA - Abstract
Human mesenchymal stem cells (MSCs) hold great promise in cellular therapeutics for skeletal diseases but lack expression of E-selectin ligands that direct homing of blood-borne cells to bone marrow. Previously, we described a method to engineer E-selectin ligands on the MSC surface by exofucosylating cells with fucosyltransferase VI (FTVI) and its donor sugar, GDP-Fucose, enforcing transient surface expression of the potent E-selectin ligand HCELL with resultant enhanced osteotropism of intravenously administered cells. Here, we sought to determine whether E-selectin ligands created via FTVI-exofucosylation are distinct in identity and function to those created by FTVI expressed intracellularly. To this end, we introduced synthetic modified mRNA encoding FTVI ( FUT6-modRNA) into human MSCs. FTVI-exofucosylation (i.e., extracellular fucosylation) and FUT6-modRNA transfection (i.e., intracellular fucosylation) produced similar peak increases in cell surface E-selectin ligand levels, and shear-based functional assays showed comparable increases in tethering/rolling on human endothelial cells expressing E-selectin. However, biochemical analyses revealed that intracellular fucosylation induced expression of both intracellular and cell surface E-selectin ligands and also induced a more sustained expression of E-selectin ligands compared to extracellular fucosylation. Notably, live imaging studies to assess homing of human MSC to mouse calvarium revealed more osteotropism following intravenous administration of intracellularly-fucosylated cells compared to extracellularly-fucosylated cells. This study represents the first direct analysis of E-selectin ligand expression programmed on human MSCs by FTVI-mediated intracellular versus extracellular fucosylation. The observed differential biologic effects of FTVI activity in these two contexts may yield new strategies for improving the efficacy of human MSCs in clinical applications. S tem C ells 2016;34:2501-2511 [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
8. Fulfilling Koch's postulates in glycoscience: HCELL, GPS and translational glycobiology.
- Author
-
Sackstein, Robert
- Subjects
- *
AXIOMS , *GLYCOMICS , *BONE marrow , *SELECTINS , *LIGANDS (Biochemistry) - Abstract
Glycoscience-based research that is performed expressly to address medical necessity and improve patient outcomes is called "translational glycobiology". In the 19th century, Robert Koch proposed a set of postulates to rigorously establish causality in microbial pathogenesis, and these postulates can be reshaped to guide knowledge into hownaturally-expressed glycoconjugates direct molecular processes critical to human well-being. Studies in the 1990s indicated that E-selectin, an endothelial lectin that binds sialofucosylated carbohydrate determinants, is constitutively expressed on marrow microvessels, and investigations in my laboratory indicated that human hematopoietic stem cells (HSCs) uniquely express high levels of a specialized glycoform of CD44 called "hematopoietic cell E-/L-selectin ligand" (HCELL) that functions as a highly potent E-selectin ligand. To assess the role of HCELL in directing HSC migration to marrow, a method called "glycosyltransferase-programmed stereosubstitution" (GPS) was developed to custom-modify CD44 glycans to enforce HCELL expression on viable cell surfaces. Human mesenchymal stem cells (MSCs) are devoid of E-selectin ligands, but GPS-based glycoengineering of CD44 on MSCs licenses homing of these cells to marrow in vivo, providing direct evidence that HCELL serves as a "bone marrow homing receptor". This review will discuss the molecular basis of cell migration in historical context, will describe the discovery of HCELL and its function as the bone marrow homing receptor, and will inform on how glycoengineering of CD44 serves as a model for adapting Koch's postulates to elucidate the key roles that glycoconjugates play in human biology and for realizing the immense impact of translational glycobiology in clinical medicine. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
9. Enforced sialyl-Lewis-X (sLeX) display in E-selectin ligands by exofucosylation is dispensable for CD19-CAR T-cell activity and bone marrow homing
- Author
-
Sánchez-Martínez, Diego, Gutiérrez-Agüera, Francisco, Romecin, Paola, Vinyoles, Meritxell, Palomo, Marta, Tirado, Néstor, Zanetti, SR, Juan, Manel, Carlet, Michela, Jeremias, Irmela, Menéndez, Pablo, Sánchez-Martínez, Diego, Gutiérrez-Agüera, Francisco, Romecin, Paola, Vinyoles, Meritxell, Palomo, Marta, Tirado, Néstor, Zanetti, SR, Juan, Manel, Carlet, Michela, Jeremias, Irmela, and Menéndez, Pablo
- Abstract
Altres ajuts: Fundació La Caixa, CD19-directed chimeric antigen receptors (CAR) T cells induce impressive rates of complete response in advanced B-cell malignancies, specially in B-cell acute lymphoblastic leukemia (B-ALL). However, CAR T-cell-treated patients eventually progress due to poor CAR T-cell persistence and/or disease relapse. The bone marrow (BM) is the primary location for acute leukemia. The rapid/efficient colonization of the BM by systemically infused CD19-CAR T cells might enhance CAR T-cell activity and persistence, thus, offering clinical benefits. Circulating cells traffic to BM upon binding of tetrasaccharide sialyl-Lewis X (sLeX)-decorated E-selectin ligands (sialofucosylated) to the E-selectin receptor expressed in the vascular endothelium. sLeX-installation in E-selectin ligands is achieved through an ex vivo fucosylation reaction. Here, we sought to characterize the basal and cell-autonomous display of sLeX in CAR T-cells activated using different cytokines, and to assess whether exofucosylation of E-selectin ligands improves CD19-CAR T-cell activity and BM homing. We report that cell-autonomous sialofucosylation (sLeX display) steadily increases in culture- and in vivo -expanded CAR T cells, and that, the cytokines used during T-cell activation influence both the degree of such endogenous sialofucosylation and the CD19-CAR T-cell efficacy and persistence in vivo. However, glycoengineered enforced sialofucosylation of E-selectin ligands was dispensable for CD19-CAR T-cell activity and BM homing in multiple xenograft models regardless the cytokines employed for T-cell expansion, thus, representing a dispensable strategy for CD19-CAR T-cell therapy. Glyco-engineered enforced sialofucosylation of E-selectin ligands was dispensable for CD19-CAR T-cell activity and BM homing in multiple xenograft models, thus representing an unnecessary strategy for CD19-CAR T-cell therapy
- Published
- 2021
10. Cell surface glycan engineering of neural stem cells augments neurotropism and improves recovery in a murine model of multiple sclerosis.
- Author
-
Merzaban, Jasmeen S., Imitola, Jaime, Starossom, Sarah C., Bing Zhu, Yue Wang, Jack Lee, Ali, Amal J., Olah, Marta, Abuelela, Ayman F., Khoury, Samia J., and Sackstein, Robert
- Subjects
- *
CELL membranes , *GLYCANS , *NEURAL stem cells , *MULTIPLE sclerosis , *CENTRAL nervous system diseases , *THERAPEUTICS , *GLYCOSYLTRANSFERASES , *GENE expression , *ANIMAL models in research - Abstract
Neural stem cell (NSC)-based therapies offer potential for neural repair in central nervous system (CNS) inflammatory and degenerative disorders. Typically, these conditions present with multifocal CNS lesions making it impractical to inject NSCs locally, thus mandating optimization of vascular delivery of the cells to involved sites. Here, we analyzed NSCs for expression of molecular effectors of cell migration and found that these cells are natively devoid of E-selectin ligands. Using glycosyltransferase- programmed stereosubstitution (GPS), we glycan engineered the cell surface of NSCs ("GPS-NSCs") with resultant enforced expression of the potent E-selectin ligand HCELL (hematopoietic cell E-/L-selectin ligand) and of an E-selectin-binding glycoform of neural cell adhesion molecule ("NCAM-E"). Following intravenous (i.v.) injection, short-term homing studies demonstrated that, compared with buffer-treated (control) NSCs, GPS-NSCs showed greater neurotropism. Administration of GPS-NSC significantly attenuated the clinical course of experimental autoimmune encephalomyelitis (EAE), with markedly decreased inflammation and improved oligodendroglial and axonal integrity, but without evidence of long-term stem cell engraftment. Notably, this effect of NSC is not a universal property of adult stem cells, as administration of GPS-engineered mouse hematopoietic stem/progenitor cells did not improve EAE clinical course. These findings highlight the utility of cell surface glycan engineering to boost stem cell delivery in neuroinflammatory conditions and indicate that, despite the use of a neural tissue-specific progenitor cell population, neural repair in EAE results from endogenous repair and not from direct, NSC-derived cell replacement. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
11. Enforced sialyl-Lewis-X (sLeX) display in E-selectin ligands by exofucosylation is dispensable for CD19-CAR T-cell activity and bone marrow homing
- Author
-
Pablo Menendez, Néstor Tirado, Michela Carlet, Diego Sánchez-Martínez, Francisco Gutierrez-Agüera, Manel Juan, Meritxell Vinyoles, Marta Palomo, Samanta Romina Zanetti, Irmela Jeremias, and Paola Alejandra Romecin
- Subjects
0301 basic medicine ,BM homing ,Medicine (miscellaneous) ,CAR T-cells ,Ligands ,Immunotherapy, Adoptive ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,E-selectin ligands ,Bone Marrow ,Mice, Inbred NOD ,E‐selectin ligands ,Receptor ,Research Articles ,lcsh:R5-920 ,Receptors, Chimeric Antigen ,biology ,Chemistry ,exofucosylation ,3. Good health ,Exofucosylation ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Models, Animal ,Molecular Medicine ,lcsh:Medicine (General) ,E-Selectin ,Research Article ,Antigens, CD19 ,CD19 ,03 medical and health sciences ,E-selectin ,medicine ,CAR T‐cells ,Animals ,Sialyl Lewis X Antigen ,Bm Homing ,Car T-cells ,E-selectin Ligands ,Chimeric antigen receptor ,030104 developmental biology ,Sialyl-Lewis X ,Cancer research ,biology.protein ,Bone marrow ,Endothelium, Vascular ,Ex vivo ,Homing (hematopoietic) - Abstract
CD19‐directed chimeric antigen receptors (CAR) T cells induce impressive rates of complete response in advanced B‐cell malignancies, specially in B‐cell acute lymphoblastic leukemia (B‐ALL). However, CAR T‐cell‐treated patients eventually progress due to poor CAR T‐cell persistence and/or disease relapse. The bone marrow (BM) is the primary location for acute leukemia. The rapid/efficient colonization of the BM by systemically infused CD19‐CAR T cells might enhance CAR T‐cell activity and persistence, thus, offering clinical benefits. Circulating cells traffic to BM upon binding of tetrasaccharide sialyl‐Lewis X (sLeX)‐decorated E‐selectin ligands (sialofucosylated) to the E‐selectin receptor expressed in the vascular endothelium. sLeX‐installation in E‐selectin ligands is achieved through an ex vivo fucosylation reaction. Here, we sought to characterize the basal and cell‐autonomous display of sLeX in CAR T‐cells activated using different cytokines, and to assess whether exofucosylation of E‐selectin ligands improves CD19‐CAR T‐cell activity and BM homing. We report that cell‐autonomous sialofucosylation (sLeX display) steadily increases in culture‐ and in vivo‐expanded CAR T cells, and that, the cytokines used during T‐cell activation influence both the degree of such endogenous sialofucosylation and the CD19‐CAR T‐cell efficacy and persistence in vivo. However, glycoengineered enforced sialofucosylation of E‐selectin ligands was dispensable for CD19‐CAR T‐cell activity and BM homing in multiple xenograft models regardless the cytokines employed for T‐cell expansion, thus, representing a dispensable strategy for CD19‐CAR T‐cell therapy., Glyco‐engineered enforced sialofucosylation of E‐selectin ligands was dispensable for CD19‐CAR T‐cell activity and BM homing in multiple xenograft models, thus representing an unnecessary strategy for CD19‐CAR T‐cell therapy
- Published
- 2021
12. Chaperone-Mediated Autophagy Ablation in Pericytes Reveals New Glioblastoma Prognostic Markers and Efficient Treatment Against Tumor Progression.
- Author
-
Molina ML, García-Bernal D, Salinas MD, Rubio G, Aparicio P, Moraleda JM, Martínez S, and Valdor R
- Abstract
Background: The lack of knowledge of the progression mechanisms of glioblastoma (GB), the most aggressive brain tumor, contributes to the absence of successful therapeutic strategies. Our team has recently demonstrated a crucial new role for chaperone-mediated autophagy (CMA) in pericytes (PC)-acquired immunosuppressive function, which prevents anti-tumor immune responses and facilitates GB progression. The possible impact that GB-induced CMA in PC has on other functions that might be useful for future GB prognosis/treatment, has not been explored yet. Thus, we proposed to analyze the contribution of CMA to other GB-induced changes in PC biology and determine if CMA ablation in PC is a key target mechanism for GB treatment. Methods: Studies of RNA-seq and secretome analysis were done in GB-conditioned PC with and without CMA (from knockout mice for LAMP-2A) and compared to control PC. Different therapeutic strategies in a GB mouse model were compared. Results: We found several gene expression pathways enriched in LAMP2A-KO PC and affected by GB-induced CMA in PC that correlate with our previous findings. Phagosome formation, cellular senescence, focal adhesion and the effector function to promote anti-tumor immune responses were the most affected pathways, revealing a transcriptomic profiling of specific target functions useful for future therapies. In addition, several molecules associated with tumor mechanisms and related to tumor immune responses such as gelsolin, periostin, osteopontin, lumican and vitamin D, were identified in the PC secretome dependent on GB-induced CMA. The CMA ablation in PC with GB cells showed an expected immunogenic phenotype able to phagocyte GB cells and a key strategy to develop future therapeutic strategies against GB tumor progression. A novel intravenous therapy using exofucosylated CMA-deficient PC was efficient to make PC reach the tumor niche and facilitate tumor elimination. Conclusion: Our results corroborate previous findings on the impaired immunogenic function of PC with GB-induced CMA, driving to other altered PC functions and the identifications of new target markers related to the tumor immune responses and useful for GB prognosis/therapy. Our work demonstrates CMA ablation in PC as a key target mechanism to develop a successful therapy against GB progression., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Molina, García-Bernal, Salinas, Rubio, Aparicio, Moraleda, Martínez and Valdor.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.