Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Endocr Relat Cancer ; 28(6): R173-R190, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33852432

ABSTRACT

Obesity is the strongest known risk factor to develop type 2 diabetes (T2D) and both share a state of chronic, diffuse and low-grade inflammation, impaired immune responses and alterations in the composition and function of the microbiome. Notably, these hallmarks are shared with colorectal cancer (CRC), which is epidemiologically associated to obesity and T2D. Gut barrier damages in T2D destabilize the microbiome that metabolizes the diet and modulates the host immune response triggering inflammatory and proliferative pathways. In this review, we discuss the pathways altered by defects in the immune response and microbiota that may link T2D to CRC development. Stressed adipocytes, metabolic incongruity in blood and gut barrier failure with dysbiosis cooperate to establish imbalances between immune innate and adaptive cells and cytokines such as interleukin 6 (IL6) or TNFA that define low-grade diffuse inflammation in T2D. Inflammation drives tissue repair through proliferation and migration (critical mechanisms for tumourigenesis) and under physiological conditions feeds anti-inflammatory cytokine production to resolve the process. The disproportion in pro- vs anti-inflammatory cells and cytokines imposed by T2D will impact the tumour micro- and macro-environment, favouring tumour proliferation, angiogenesis and decreased immune responses. Complex bidirectional relationships between the metabolic environment of T2D, gut microbiota, and immune dysfunctions may favour tumour cell demands and will define the outcome. Animal models developed to study the relationships between T2D and CRC in the context of microbiota and immune system are discussed.


Subject(s)
Colorectal Neoplasms , Diabetes Mellitus, Type 2 , Microbiota , Animals , Cytokines , Humans , Inflammation , Obesity
2.
Endocr Relat Cancer ; 28(6): R191-R206, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33910163

ABSTRACT

The existence of molecular links that facilitate colorectal cancer (CRC) development in the population with type 2 diabetes (T2D) is supported by substantial epidemiological evidence. This review summarizes how the systemic, metabolic and hormonal imbalances from T2D alter CRC cell metabolism, signalling and gene expression as well as their reciprocal meshing, with an overview of CRC molecular subtypes and animal models to study the diabetes-CRC cancer links. Metabolic and growth factor checkpoints ensure a physiological cell proliferation rate compatible with limited nutrient supply. Hyperinsulinaemia and hyperleptinaemia in prediabetes and excess circulating glucose and lipids in T2D overcome formidable barriers for tumour development. Increased nutrient availability favours metabolic reprogramming, alters signalling and generates mutations and epigenetic modifications through increased reactive oxygen species and oncometabolites. The reciprocal control between metabolism and hormone signalling is lost in diabetes. Excess adipose tissue at the origin of T2D unbalances adipokine (leptin/adiponectin) secretion ratios and function and disrupts the insulin/IGF axes. Leptin/adiponectin imbalances in T2D are believed to promote proliferation and invasion of CRC cancer cells and contribute to inflammation, an important component of CRC tumourigenesis. Disruption of the insulin/IGF axes in T2D targets systemic and CRC cell metabolic reprogramming, survival and proliferation. Future research to clarify the molecular diabetes-CRC links will help to prevent CRC and reduce its incidence in the diabetic population and must guide therapeutic decisions.


Subject(s)
Colorectal Neoplasms , Diabetes Mellitus, Type 2 , Adiponectin , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Diabetes Mellitus, Type 2/pathology , Humans , Insulin , Leptin
3.
PLoS One ; 14(4): e0215537, 2019.
Article in English | MEDLINE | ID: mdl-31017970

ABSTRACT

BACKGROUND: Methyl-aminolevulinate photodynamic therapy (MAL-PDT) is an excellent option for the treatment of basal cell carcinoma (BCC). However, up to 25% of cases are resistant to this treatment modality. OBJECTIVE: The aim of this study was to identify potential biomarkers of BCC response to MAL-PDT. MATERIAL AND METHODS: Clinical, histological, and immunohistochemical (p53, Ki-67, CD-31, COX2, ß-catenin, EGFR, and survivin) variables were analyzed in a retrospective study of consecutive BCC patients treated with MAL-PDT at the San Jorge Hospital, Huesca, Spain between January 2006 and December 2015. To deepen on these markers, the effects on p53 and cyclin D1 expression, in vitro response to MAL-PDT of 2 murine BCC cell lines (ASZ and BSZ), was also evaluated. RESULTS: The retrospective study examined the response to MAL-PDT of 390 BCCs from 182 patients. The overall clinical response rate was 82.8%, with a mean follow-up time of 35.96 months (SD = 23.46). Immunohistochemistry revealed positive p53 in 84.6% of responders but only 15.4% of nonresponsive tumors (p = 0.011). Tumors with increased peripheral palisading of basal cell islands to immunostaining ß-catenin responded poorly to PDT (p = 0.01). In line with our findings in patients, in vitro studies revealed a better response to PDT in the p53-positive ASZ cell line than the p53-negative BSZ cell line (p<0.01). Multivariate analysis revealed that the following variables were significantly associated with response to PDT: age, nBCC, presence of peritumoral inflammatory infiltrate, and p53 immunopositivity. Patients with positive p53 immunostaining were 68.54 times more likely to achieve cure than p53-negative patients (CI95% 2.94-159.8). CONCLUSION: Our finding suggest that certain clinicopathological and immunohistochemical variables, particularly p53 expression, may serve as indicators of BCC response to MAL-PDT, and thus facilitate the selection of patients who are most likely to benefit from this therapy.


Subject(s)
Aminolevulinic Acid/analogs & derivatives , Carcinoma, Basal Cell/drug therapy , Carcinoma, Basal Cell/metabolism , Photochemotherapy , Photosensitizing Agents/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Aged , Aged, 80 and over , Aminolevulinic Acid/therapeutic use , Animals , Biomarkers, Tumor/metabolism , Carcinoma, Basal Cell/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Retrospective Studies , Skin Neoplasms/pathology
4.
Int J Mol Sci ; 20(5)2019 Mar 11.
Article in English | MEDLINE | ID: mdl-30862116

ABSTRACT

Photodynamic therapy (PDT) constitutes a cancer treatment modality based on the administration of a photosensitizer, which accumulates in tumor cells. The subsequent irradiation of the tumoral area triggers the formation of reactive oxygen species responsible for cancer cell death. One of the compounds approved in clinical practice is methyl-aminolevulinate (MAL), a protoporphyrin IX (PpIX) precursor intermediate of heme synthesis. We have identified the mitotic catastrophe (MC) process after MAL-PDT in HeLa human carcinoma cells. The fluorescence microscopy revealed that PpIX was located mainly at plasma membrane and lysosomes of HeLa cells, although some fluorescence was also detected at endoplasmic reticulum and Golgi apparatus. Cell blockage at metaphase-anaphase transition was observed 24 h after PDT by phase contrast microscopy and flow cytometry. Mitotic apparatus components evaluation by immunofluorescence and Western blot indicated: multipolar spindles and disorganized chromosomes in the equatorial plate accompanied with dispersion of centromeres and alterations in aurora kinase proteins. The mitotic blockage induced by MAL-PDT resembled that induced by two compounds used in chemotherapy, taxol and nocodazole, both targeting microtubules. The alterations in tumoral cells provided evidence of MC induced by MAL-PDT, resolving mainly by apoptosis, directly or through the formation of multinucleate cells.


Subject(s)
Aminolevulinic Acid/analogs & derivatives , Mitosis/drug effects , Mitosis/radiation effects , Photochemotherapy , Photosensitizing Agents/pharmacology , Aminolevulinic Acid/pharmacology , Biomarkers , Cell Division/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , HeLa Cells , Humans , Microtubules/metabolism , Protein Transport , Protoporphyrins/metabolism , Spindle Apparatus
5.
Sci Rep ; 9(1): 4835, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30886381

ABSTRACT

Photodynamic Therapy (PDT) with methyl-aminolevulinate acid (MAL-PDT) is being used for the treatment of Basal cell carcinoma (BCC), but recurrences have been reported. In this work, we have evaluated resistance mechanisms to MAL-PDT developed by three BCC cell lines (ASZ, BSZ and CSZ), derived from mice on a ptch+/- background and with or without p53 expression, subjected to 10 cycles of PDT (10thG). The resistant populations showed mesenchymal-like structure and diminished proliferative capacity and size compared to the parental (P) cells. The resistance was dependent on the production of the endogenous photosensitiser protoporphyrin IX in the CSZ cell line and on its cellular localisation in ASZ and BSZ cells. Moreover, resistant cells expressing the p53 gene presented lower proliferation rate and increased expression levels of N-cadherin and Gsk3ß (a component of the Wnt/ß-catenin pathway) than P cells. In contrast, 10thG cells lacking the p53 gene showed lower levels of expression of Gsk3ß in the cytoplasm and of E-cadherin and ß-catenin in the membrane. In addition, resistant cells presented higher tumorigenic ability in immunosuppressed mice. Altogether, these results shed light on resistance mechanisms of BCC to PDT and may help to improve the use of this therapeutic approach.


Subject(s)
Aminolevulinic Acid/analogs & derivatives , Carcinoma, Basal Cell/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Skin Neoplasms/drug therapy , Aminolevulinic Acid/pharmacology , Aminolevulinic Acid/therapeutic use , Animals , Carcinogenesis/drug effects , Carcinogenesis/radiation effects , Carcinoma, Basal Cell/pathology , Cell Line, Tumor/transplantation , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Disease Models, Animal , Drug Resistance, Neoplasm , Humans , Mice , Mice, Transgenic , Patched-1 Receptor/genetics , Photosensitizing Agents/therapeutic use , Protoporphyrins/metabolism , Skin Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays , Wnt Signaling Pathway
6.
Int J Mol Sci ; 20(6)2019 Mar 18.
Article in English | MEDLINE | ID: mdl-30889822

ABSTRACT

Exposure to natural and artificial light and environmental pollutants are the main factors that challenge skin homeostasis, promoting aging or even different forms of skin cancer through a variety of mechanisms that include accumulation of reactive oxygen species (ROS), engagement of DNA damage responses, and extracellular matrix (ECM) remodeling upon release of metalloproteases (MMPs). Ultraviolet A radiation is the predominant component of sunlight causative of photoaging, while ultraviolet B light is considered a potentiator of photoaging. In addition, different chemicals contribute to skin aging upon penetration through skin barrier disruption or hair follicles, aryl hydrocarbon receptors (AhR) being a major effector mechanism through which toxicity is exerted. Deschampsia antarctica is a polyextremophile Gramineae capable of thriving under extreme environmental conditions. Its aqueous extract (EDA) exhibits anti- photoaging in human skin cells, such as inhibition of MMPs, directly associated with extrinsic aging. EDA prevents cellular damage, attenuating stress responses such as autophagy and reducing cellular death induced by UV. We demonstrate that EDA also protects from dioxin-induced nuclear translocation of AhR and increases the production of loricrin, a marker of homeostasis in differentiated keratinocytes. Thus, our observations suggest a potential use exploiting EDA's protective properties in skin health supplements.


Subject(s)
Dermis/pathology , Dermis/radiation effects , Plant Extracts/pharmacology , Poaceae/chemistry , Polychlorinated Dibenzodioxins/toxicity , Ultraviolet Rays , Caspase 3/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Cell Shape/drug effects , Cell Shape/radiation effects , DNA Damage , Fibroblasts/drug effects , Fibroblasts/pathology , Fibroblasts/radiation effects , Histones/metabolism , Humans , Keratinocytes/drug effects , Keratinocytes/pathology , Keratinocytes/radiation effects , Matrix Metalloproteinase 1/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Stress, Physiological/drug effects , Stress, Physiological/radiation effects
7.
Int J Mol Sci ; 16(10): 25912-33, 2015 Oct 28.
Article in English | MEDLINE | ID: mdl-26516853

ABSTRACT

Non-melanoma skin cancer (NMSC) is the most common form of cancer in the Caucasian population. Among NMSC types, basal cell carcinoma (BCC) has the highest incidence and squamous cell carcinoma (SCC) is less common although it can metastasize, accounting for the majority of NMSC-related deaths. Treatment options for NMSC include both surgical and non-surgical modalities. Even though surgical approaches are most commonly used to treat these lesions, Photodynamic Therapy (PDT) has the advantage of being a non-invasive option, and capable of field treatment, providing optimum cosmetic outcomes. Numerous clinical research studies have shown the efficacy of PDT for treating pre-malignant and malignant NMSC. However, resistant or recurrent tumors appear and sometimes become more aggressive. In this sense, the enhancement of PDT effectiveness by combining it with other therapeutic modalities has become an interesting field in NMSC research. Depending on the characteristics and the type of tumor, PDT can be applied in combination with immunomodulatory (Imiquimod) and chemotherapeutic (5-fluorouracil, methotrexate, diclofenac, or ingenol mebutate) agents, inhibitors of some molecules implicated in the carcinogenic process (COX2 or MAPK), surgical techniques, or even radiotherapy. These new strategies open the way to a wider improvement of the prevention and eradication of skin cancer.


Subject(s)
Carcinoma/drug therapy , Photochemotherapy/methods , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Carcinoma/metabolism , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Humans , Photochemotherapy/adverse effects , Photosensitizing Agents/therapeutic use , Skin Neoplasms/radiotherapy
SELECTION OF CITATIONS
SEARCH DETAIL
...