Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Life Sci ; 351: 122781, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38848937

ABSTRACT

The hepatocellular carcinoma (HCC) features a remarkable epidemiological burden, ranking as the third most lethal cancer worldwide. As the HCC-related molecular and cellular complexity unfolds as the disease progresses, the use of a myriad of in vitro models available is mandatory in translational preclinical research setups. In this review paper, we will compile cutting-edge information on the in vitro bioassays for HCC research, (A) emphasizing their morphological and molecular parallels with human HCC; (B) delineating the advantages and limitations of their application; and (C) offering perspectives on their prospective applications. While bidimensional (2D) (co) culture setups provide a rapid low-cost strategy for metabolism and drug screening investigations, tridimensional (3D) (co) culture bioassays - including patient-derived protocols as organoids and precision cut slices - surpass some of the 2D strategies limitations, mimicking the complex microarchitecture and cellular and non-cellular microenvironment observed in human HCC. 3D models have become invaluable tools to unveil HCC pathophysiology and targeted therapy. In both setups, the recapitulation of HCC in different etiologies/backgrounds (i.e., viral, fibrosis, and fatty liver) may be considered as a fundamental guide for obtaining translational findings. Therefore, a "multimodel" approach - encompassing the advantages of different in vitro bioassays - is encouraged to circumvent "model-biased" outcomes in preclinical HCC research.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Liver Neoplasms/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/genetics , Animals , Carcinogenesis/pathology , Carcinogenesis/genetics , Organoids/pathology , Models, Biological
2.
Exp Cell Res ; 440(2): 114136, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38909881

ABSTRACT

Considering the importance of alternative methodologies to animal experimentation, we propose an organoid-based biological model for in vitro blood vessel generation, achieved through co-culturing endothelial and vascular smooth muscle cells (VSMCs). Initially, the organoids underwent comprehensive characterization, revealing VSMCs (α-SMA + cells) at the periphery and endothelial cells (CD31+ cells) at the core. Additionally, ephrin B2 and ephrin B4, genes implicated in arterial and venous formation respectively, were used to validate the obtained organoid. Moreover, the data indicates exclusive HIF-1α expression in VSMCs, identified through various methodologies. Subsequently, we tested the hypothesis that the generated blood vessels have the capacity to modulate the osteogenic phenotype, demonstrating the ability of HIF-1α to promote osteogenic signals, primarily by influencing Runx2 expression. Overall, this study underscores that the methodology employed to create blood vessel organoids establishes an experimental framework capable of producing a 3D culture model of both venous and arterial endothelial tissues. This model effectively guides morphogenesis from mesenchymal stem cells through paracrine signaling, ultimately leading to an osteogenic acquisition phenotype, with the dynamic involvement of HIF-1α.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Organoids , Osteogenesis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Osteogenesis/genetics , Organoids/metabolism , Organoids/cytology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Cells, Cultured , Blood Vessels/metabolism , Blood Vessels/cytology , Blood Vessels/growth & development , Coculture Techniques/methods , Cell Differentiation , Endothelial Cells/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology
3.
Cells ; 13(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38727281

ABSTRACT

This review delves into the groundbreaking impact of induced pluripotent stem cells (iPSCs) and three-dimensional organoid models in propelling forward neuropathology research. With a focus on neurodegenerative diseases, neuromotor disorders, and related conditions, iPSCs provide a platform for personalized disease modeling, holding significant potential for regenerative therapy and drug discovery. The adaptability of iPSCs, along with associated methodologies, enables the generation of various types of neural cell differentiations and their integration into three-dimensional organoid models, effectively replicating complex tissue structures in vitro. Key advancements in organoid and iPSC generation protocols, alongside the careful selection of donor cell types, are emphasized as critical steps in harnessing these technologies to mitigate tumorigenic risks and other hurdles. Encouragingly, iPSCs show promising outcomes in regenerative therapies, as evidenced by their successful application in animal models.


Subject(s)
Induced Pluripotent Stem Cells , Organoids , Organoids/pathology , Humans , Induced Pluripotent Stem Cells/cytology , Animals , Neuropathology/methods , Regenerative Medicine/methods , Neurodegenerative Diseases/therapy , Neurodegenerative Diseases/pathology , Cell Differentiation
4.
Int J Artif Organs ; 47(2): 107-114, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38182554

ABSTRACT

BACKGROUND: Congenital pseudoarthrosis of the tibia (CPT) is an uncommon disease associated with failure to achieve bone union and recurrent fractures. There is evidence showing that CPT is associated with decreased osteogenesis. Based on the capacity of mesenchymal stromal cells (MSCs) to induce osteogenesis, we develop an osteogenic organoid (OstO) constituted by these cells, and other components of the bone niche, for inducing bone formation in a child diagnosed with CPT. AIM: To evaluate the capacity of an OstO to induce bone formation in a patient with CPT. METHODS: The OstO was fabricated with allogeneic bone marrow MSCs from a healthy donor, collagen microbeads (CM) and PRP clot. The CM and PRP function as extracellular matrix and scaffolds for MSC. The OstO was placed at the site of non-union. Internal and external fixation was placed in the tibia. Radiological evaluation was performed after MSCs transplantation. RESULTS: After 4 months of MSCs transplantation, radiographic imaging showed evidence of osteogenesis at the site of CPT lesion. The tibia showed bone consolidation and complete healing of the non-union CPT lesion after 6 months. Functional improvement was observed after 1 year of MSC transplantation. CONCLUSIONS: The OstO is a bone-like niche which promote osteogenesis in patients with failure in bone formation, such as CPT. To our knowledge, these results provide the first evidence showing CPT healing induced by an OstO constituted by allogeneic MSCs. Future studies incorporating a larger number of patients may confirm these results.


Subject(s)
Osteogenesis , Pseudarthrosis/congenital , Tibia , Child , Humans , Tibia/diagnostic imaging , Tibia/surgery , Tibia/abnormalities , Bone Regeneration , Collagen , Organoids , Cell Differentiation
5.
Nat Rev Cancer ; 24(3): 166, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38228902
6.
Biol Res ; 56(1): 63, 2023 Dec 02.
Article in English | MEDLINE | ID: mdl-38041132

ABSTRACT

In December 2022 the US Food and Drug Administration (FDA) removed the requirement that drugs in development must undergo animal testing before clinical evaluation, a declaration that now demands the establishment and verification of ex vivo preclinical models that closely represent tumor complexity and that can predict therapeutic response. Fortunately, the emergence of patient-derived organoid (PDOs) culture has enabled the ex vivo mimicking of the pathophysiology of human tumors with the reassembly of tissue-specific features. These features include histopathological variability, molecular expression profiles, genetic and cellular heterogeneity of parental tissue, and furthermore growing evidence suggests the ability to predict patient therapeutic response. Concentrating on the highly lethal and heterogeneous gastrointestinal (GI) tumors, herein we present the state-of-the-art and the current methodology of PDOs. We highlight the potential additions, improvements and testing required to allow the ex vivo of study the tumor microenvironment, as well as offering commentary on the predictive value of clinical response to treatments such as chemotherapy and immunotherapy.


Subject(s)
Gastrointestinal Neoplasms , United States , Animals , Humans , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Organoids/metabolism , Organoids/pathology , Tumor Microenvironment
7.
Nat Commun ; 14(1): 6882, 2023 10 28.
Article in English | MEDLINE | ID: mdl-37898615

ABSTRACT

Bats are natural reservoirs for several zoonotic viruses, potentially due to an enhanced capacity to control viral infection. However, the mechanisms of antiviral responses in bats are poorly defined. Here we established a Jamaican fruit bat (JFB, Artibeus jamaicensis) intestinal organoid model of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. Upon infection with SARS-CoV-2, increased viral RNA and subgenomic RNA was detected, but no infectious virus was released, indicating that JFB organoids support only limited viral replication but not viral reproduction. SARS-CoV-2 replication was associated with significantly increased gene expression of type I interferons and inflammatory cytokines. Interestingly, SARS-CoV-2 also caused enhanced formation and growth of JFB organoids. Proteomics revealed an increase in inflammatory signaling, cell turnover, cell repair, and SARS-CoV-2 infection pathways. Collectively, our findings suggest that primary JFB intestinal epithelial cells mount successful antiviral interferon responses and that SARS-CoV-2 infection in JFB cells induces protective regenerative pathways.


Subject(s)
COVID-19 , Chiroptera , Interferon Type I , Viruses , Animals , SARS-CoV-2 , Jamaica , Antiviral Agents , Organoids
8.
Stem Cell Res Ther ; 14(1): 70, 2023 04 07.
Article in English | MEDLINE | ID: mdl-37024989

ABSTRACT

BACKGROUND: Adipose tissue-derived stromal vascular fraction (SVF) harbors multipotent cells with potential therapeutic relevance. We developed a method to form adipose spheroids (AS) from the SVF with complex organoid structure and enhanced leptin secretion upon insulin stimulation. METHODS: SVF was generated from the interscapular brown adipose tissue of newborn mice. Immunophenotype and stemness of cultured SVF were determined by flow cytometry and in vitro differentiation, respectively. Spheroids were generated in hanging drops and non-adherent plates and compared by morphometric methods. The adipogenic potential was compared between preadipocyte monolayers and spheroids. Extracellular leptin was quantified by immunoassay. Lipolysis was stimulated with isoprenaline and quantified by colorimetric methods. AS viability and ultrastructure were determined by confocal and transmission electron microscopy analyses. RESULTS: Cultured SVF contained Sca1 + CD29 + CD44 + CD11b- CD45- CD90- cells with adipogenic and chondrogenic but no osteogenic potential. Culture on non-adherent plates yielded the highest quantity and biggest size of spheroids. Differentiation of AS for 15 days in a culture medium supplemented with insulin and rosiglitazone resulted in greater Pparg, Plin1, and Lep expression compared to differentiated adipocytes monolayers. AS were viable and maintained leptin secretion even in the absence of adipogenic stimulation. Glycerol release after isoprenaline stimulation was higher in AS compared to adipocytes in monolayers. AS were composed of outer layers of unilocular mature adipocytes and an inner structure composed of preadipocytes, immature adipocytes and an abundant loose extracellular matrix. CONCLUSION: Newborn mice adipose SVF can be efficiently differentiated into leptin-secreting AS. Prolonged stimulation with insulin and rosiglitazone allows the formation of structurally complex adipose organoids able to respond to adrenergic lipolytic stimulation.


Subject(s)
Adipocytes , Adipose Tissue, Brown , Cell Differentiation , Leptin , Leptin/metabolism , Organoids , Insulin/pharmacology , Animals , Mice , Adipose Tissue, Brown/cytology , Rosiglitazone/pharmacology , Cells, Cultured , Animals, Newborn , Immunophenotyping , Osteogenesis , Chondrogenesis , Adipocytes/ultrastructure , Lipolysis , Primary Cell Culture
9.
Front Cell Infect Microbiol ; 13: 1082524, 2023.
Article in English | MEDLINE | ID: mdl-36968103

ABSTRACT

Chagas disease (CD) is a life-threatening illness caused by the parasite Trypanosoma cruzi (T. cruzi). With around seven million people infected worldwide and over 50,000 deaths per year, CD is a major public health issue in Latin America. The main route of transmission to humans is through a triatomine bug (vector-borne), but congenital and oral transmission have also been reported. The acute phase of CD presents mild symptoms but may develop into a long-lasting chronic illness, characterized by severely impaired cardiac, digestive, and neurological functions. The intestinal tissue appears to have a key role during oral transmission and chronic infection of CD. In this immune-privileged reservoir, dormant/quiescent parasites have been suggested to contribute to disease persistence, infection relapse, and treatment failure. However, the interaction between the intestinal epithelium and T. cruzi has not been examined in depth, in part, due to the lack of in vitro models that approximate to the biological and structural complexity of this tissue. Therefore, to understand the role played by the intestinal tissue during transmission and chronic infection, physiological models resembling the organ complexity are needed. Here we addressed this issue by establishing and characterizing adult stem cell-derived colonoid infection models that are clinically relevant for CD. 3D and 2D systems of murine intestinal organoids infected with T. cruzi Dm28c (a highly virulent strain associated with oral outbreaks) were analyzed at different time points by confocal microscopy. T. cruzi was able to invade and replicate in intestinal epithelial primary cells grown as intact organoids (3D) and monolayers (2D). The permissiveness to pathogen infection differed markedly between organoids and cell lines (primate and intestinal human cell lines). So far, this represents the first evidence of the potential that these cellular systems offer for the study of host-pathogen interactions and the discovery of effective anti-chagasic drugs.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Humans , Animals , Mice , Persistent Infection , Chagas Disease/parasitology , Intestinal Mucosa , Colon , Organoids
10.
Circ Res ; 132(4): 481-482, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36795848
11.
Biomolecules ; 13(2)2023 01 30.
Article in English | MEDLINE | ID: mdl-36830629

ABSTRACT

Autism spectrum disorders (ASD) are a group of complex neurodevelopmental disorders that affect communication and social interactions and present with restricted interests and repetitive behavior patterns. The susceptibility to ASD is strongly influenced by genetic/heritable factors; however, there is still a large gap in understanding the cellular and molecular mechanisms underlying the neurobiology of ASD. Significant progress has been made in identifying ASD risk genes and the possible convergent pathways regulated by these gene networks during development. The breakthrough of cellular reprogramming technology has allowed the generation of induced pluripotent stem cells (iPSCs) from individuals with syndromic and idiopathic ASD, providing patient-specific cell models for mechanistic studies. In the past decade, protocols for developing brain organoids from these cells have been established, leading to significant advances in the in vitro reproducibility of the early steps of human brain development. Here, we reviewed the most relevant literature regarding the application of brain organoids to the study of ASD, providing the current state of the art, and discussing the impact of such models on the field, limitations, and opportunities for future development.


Subject(s)
Autism Spectrum Disorder , Induced Pluripotent Stem Cells , Humans , Autism Spectrum Disorder/genetics , Reproducibility of Results , Brain , Organoids
12.
Stem Cell Rev Rep ; 19(4): 1116-1123, 2023 05.
Article in English | MEDLINE | ID: mdl-36652145

ABSTRACT

Down syndrome (DS, or trisomy 21, T21), is the most common genetic cause of intellectual disability. Alterations in the complex process of cerebral cortex development contribute to the neurological deficits in DS, although the underlying molecular and cellular mechanisms are not completely understood. Human cerebral organoids (COs) derived from three-dimensional (3D) cultures of induced pluripotent stem cells (iPSCs) provide a new avenue for gaining a better understanding of DS neuropathology. In this study, we aimed to generate iPSCs from individuals with DS (T21-iPSCs) and euploid controls using urine-derived cells, which can be easily and noninvasively obtained from most individuals, and examine their ability to differentiate into neurons and astrocytes grown in monolayer cultures, as well as into 3D COs. We employed nonintegrating episomal vectors to generate urine-derived iPSC lines, and a simple-to-use system to produce COs with forebrain identity. We observed that both T21 and control urine-derived iPSC lines successfully differentiate into neurons and astrocytes in monolayer, as well as into COs that recapitulate early features of human cortical development, including organization of neural progenitor zones, programmed differentiation of excitatory and inhibitory neurons, and upper-and deep-layer cortical neurons as well as astrocytes. Our findings demonstrate for the first time the suitability of using urine-derived iPSC lines to produce COs for modeling DS.


Subject(s)
Cerebrum , Down Syndrome , Induced Pluripotent Stem Cells , Neurogenesis , Organoids , Induced Pluripotent Stem Cells/cytology , Organoids/cytology , Organoids/growth & development , Cerebrum/cytology , Cerebrum/growth & development , Down Syndrome/genetics , Down Syndrome/pathology , Down Syndrome/urine , Cell Culture Techniques, Three Dimensional , Humans , Neurons/cytology , Astrocytes/cytology , Cell Lineage
13.
Int J Artif Organs ; 46(1): 29-39, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36349723

ABSTRACT

BACKGROUND: Under certain clinical and experimental conditions hematopoiesis occurs in other site than bone marrow (BM), such as the liver. Here, we develop a 3D organoid that mimics several components of the hematopoietic niche present during liver extramedullary hematopoiesis. AIM: To evaluate the capacity of a 3D hematopoietic organoid (3D-HO) to function as a hematopoietic like-niche allowing for blood cell production outside of the BM. METHODS: The 3D-HO is constituted by liver sinusoidal endothelial cells (LSEC) as the stromal component, BM isolated from 5-FU treated mice (FU-BMCs), collagen microspheres and plasma clot as scaffolds. The ability of the 3D-HO to support the survival and functionality of FU-BMCs was investigated by using confocal microscopy, histology analysis, flow cytometry, and clonogenic assays. RESULTS: After 15 and 30 days, post-ectopic implantation, histological studies of the 3D-HO showed the presence of cells with myeloid and lymphoid lineage morphology. Flow cytometry analysis of these cells showed the presence of cells expressing hematopoietic stem progenitor cells (HSPC) (Sca-1+/c-Kit+), myeloid (Gr-1+) and lymphoid (B220+ and CD19+) markers. Clonogenic assays showed that cells from the 3D-HO formed hematopoietic colonies. Expression of the Sry gene by cells from the 3D-HO, implanted for 30 days in female mice, indicated that male donor cells persist in this model of extramedullary hematopoiesis. CONCLUSIONS: The 3D-HO constitutes an extramedullary hematopoietic-like niche which supports the survival and functionality of FU-BMCs. It may constitute an efficient model for investigating, in vitro and in vivo, those factors that control hematopoiesis outside BM.


Subject(s)
Hematopoiesis, Extramedullary , Male , Female , Mice , Animals , Endothelial Cells , Hematopoiesis/genetics , Hematopoietic Stem Cells/physiology , Organoids , Bone Marrow Cells/metabolism
14.
Semin Cell Dev Biol ; 144: 77-86, 2023 07 30.
Article in English | MEDLINE | ID: mdl-36210260

ABSTRACT

Human-induced pluripotent stem cells (hiPSCs) have provided new methods to study neurodegenerative diseases. In addition to their wide application in neuronal disorders, hiPSCs technology can also encompass specific conditions, such as inherited retinal dystrophies. The possibility of evaluating alterations related to retinal disorders in 3D organoids increases the truthfulness of in vitro models. Moreover, both Alzheimer's (AD) and Parkinson's disease (PD) have been described as causing early retinal alterations, generating beta-amyloid protein accumulation, or affecting dopaminergic amacrine cells. This review addresses recent advances and future perspectives obtained from in vitro modeling of retinal diseases, focusing on retinitis pigmentosa (RP). Additionally, we depicted the possibility of evaluating changes related to AD and PD in retinal organoids obtained from potential patients long before the onset of the disease, constituting a valuable tool in early diagnosis. With this, we pointed out prospects in the study of retinal dystrophies and early diagnosis of AD and PD.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Parkinson Disease , Retinitis Pigmentosa , Humans , Induced Pluripotent Stem Cells/metabolism , Parkinson Disease/diagnosis , Parkinson Disease/metabolism , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Retinitis Pigmentosa/metabolism , Organoids , Early Diagnosis
15.
Methods Mol Biol ; 2561: 135-158, 2023.
Article in English | MEDLINE | ID: mdl-36399268

ABSTRACT

Alzheimer's disease (AD) is the primary cause of dementia, to date. The urgent need to understand the biological and biochemical processes related to this condition, as well as the demand for reliable in vitro models for drug screening, has led to the development of novel techniques, among which stem cell methods are of utmost relevance for AD research, particularly the development of human brain organoids. Brain organoids are three-dimensional cellular aggregates derived from induced pluripotent stem cells (iPSCs) that recreate different neural cell interactions and tissue characteristics in culture. Here, we describe the protocol for the generation of brain organoids derived from AD patients and for the analysis of AD-derived pathology. AD organoids can recapitulate beta-amyloid and tau pathological features, making them a promising model for studying the molecular mechanisms underlying disease and for in vitro drug testing.


Subject(s)
Alzheimer Disease , Induced Pluripotent Stem Cells , Humans , Organoids , Alzheimer Disease/pathology , Brain/pathology , Amyloid beta-Peptides/metabolism
16.
Semin Cell Dev Biol ; 144: 67-76, 2023 07 30.
Article in English | MEDLINE | ID: mdl-36115764

ABSTRACT

The use of antidepressants during pregnancy benefits the mother's well-being, but the effects of such substances on neurodevelopment remain poorly understood. Moreover, the consequences of early exposure to antidepressants may not be immediately apparent at birth. In utero exposure to selective serotonin reuptake inhibitors (SSRIs) has been related to developmental abnormalities, including a reduced white matter volume. Several reports have observed an increased incidence of autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD) after prenatal exposure to SSRIs such as sertraline, the most widely prescribed SSRI. The advent of human-induced pluripotent stem cell (hiPSC) methods and assays now offers appropriate tools to test the consequences of such compounds for neurodevelopment in vitro. In particular, hiPSCs can be used to generate cerebral organoids - self-organized structures that recapitulate the morphology and complex physiology of the developing human brain, overcoming the limitations found in 2D cell culture and experimental animal models for testing drug efficacy and side effects. For example, single-cell RNA sequencing (scRNA-seq) and electrophysiological measurements on organoids can be used to evaluate the impact of antidepressants on the transcriptome and neuronal activity signatures in developing neurons. While the analysis of large-scale transcriptomic data depends on dimensionality reduction methods, electrophysiological recordings rely on temporal data series to discriminate statistical characteristics of neuronal activity, allowing for the rigorous analysis of the effects of antidepressants and other molecules that affect the developing nervous system, especially when applied in combination with relevant human cellular models such as brain organoids.


Subject(s)
Autism Spectrum Disorder , Selective Serotonin Reuptake Inhibitors , Pregnancy , Female , Infant, Newborn , Animals , Humans , Selective Serotonin Reuptake Inhibitors/pharmacology , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/epidemiology , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain , Organoids
17.
Semin Cell Dev Biol ; 144: 87-96, 2023 07 30.
Article in English | MEDLINE | ID: mdl-36182613

ABSTRACT

Infectious diseases worldwide affect human health and have important societal impacts. A better understanding of infectious diseases is urgently needed. In vitro and in vivo infection models have brought notable contributions to the current knowledge of these diseases. Organoids are multicellular culture systems resembling tissue architecture and function, recapitulating many characteristics of human disease and elucidating mechanisms of host-infectious agent interactions in the respiratory and gastrointestinal systems, the central nervous system and the skin. Here, we discuss the applicability of the organoid technology for modeling pathogenesis, host response and features, which can be explored for the development of preventive and therapeutic treatments.


Subject(s)
Communicable Diseases , Organoids , Humans , Gastrointestinal Tract
18.
Article in Portuguese | LILACS | ID: biblio-1511478

ABSTRACT

Há muitos anos a cultura celular bidimensional (2D) é utilizada como modelo de estudo de doenças, possuindo grande importância na medicina regenerativa, apesar de ainda conter limitações significativas. A fim de contornar essas limitações, a cultura celular tridimensional (3D) propõe uma organização mais complexa e sustentável que pode ser produzida a partir de células-tronco adultas (ASCs), células-tronco embrionárias (ESCs) ou células-tronco pluripotentes induzidas (iPSCs). A cultura 3D possibilitou o cultivo de células em um ambiente mais próximo do fisiológico, levando à formação de distintos tecidos órgãos-específicos. Em outras palavras, a cultura de células 3D possibilita a criação de estruturas orgânicas muito semelhantes aos órgãos de um ser humano, tanto estruturalmente, quanto funcionalmente. Desse modo, tem-se o que é chamado de organoides. O uso dos organoides tem crescido exponencialmente em ambientes in vitro, permitindo a análise e observação dos diversos fenômenos fisiológicos existentes. Como exemplo, pode-se citar os organoides cerebrais ("mini-brains") reproduzidos in vitro buscando delinear as peculiaridades e complexidades do cérebro humano, com o objetivo de compreender algumas disfunções neurológicas que acometem esse sistema, como as duas principais doenças neurodegenerativas: Doenças de Alzheimer e Parkinson. Portanto, os organoides cerebrais podem permitir notável avanço da medicina regenerativa aplicada a doenças neurodegenerativas, já que esses "mini-brains" podem ser produzidos a partir de células do próprio paciente. Isso permitirá intervenções personalizadas, como testagens farmacológicas, a fim de definir qual seria o melhor tratamento medicamentoso. Consequentemente, essa tecnologia pode permitir terapias mais eficientes e individualizadas - o que é fundamental para a Medicina Personalizada (AU).


For many years, two-dimensional (2D) cell culture has been used as a model to study diseases, having great importance in regenerative medicine, despite still having significant limitations. In order to circumvent these limitations, three-dimensional (3D) cell culture proposes a more complex and sustainable organization that can be produced from adult stem cells (ASCs), embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs). The 3D culture enabled the cultivation of cells in an environment closer to the physiological one, leading to the formation of different organ-specific tissues. In other words, 3D cell culture makes it possible to create organic structures very similar to the organs of a human being, both structurally and functionally. In this way, we have what are called organoids. The use of organoids has grown exponentially in in vitro environments, allowing the analysis and observation of the various existing physiological phenomena. As an example, we can mention the brain organoids ("mini-brains") reproduced in vitro, seeking to delineate the peculiarities and complexities of the human brain, in order to understand some neurological dysfunctions that affect this system, such as the two main neurodegenerative diseases: Alzheimer's and Parkinson's Diseases. Therefore, brain organoids may allow a remarkable advance in regenerative medicine applied to neurodegenerative diseases, as these "mini-brains" can be produced from the patient's own cells. This will allow for personalized interventions, such as drug testing, in order to define what would be the best drug treatment. Consequently, this technology can enable more efficient and individualized therapies - which is fundamental for Personalized Medicine (AU).


Subject(s)
Humans , Parkinson Disease , Organoids , Concierge Medicine
19.
Sci Rep ; 12(1): 14425, 2022 08 24.
Article in English | MEDLINE | ID: mdl-36002565

ABSTRACT

Inflammation is an important process for epithelial barrier protection but when uncontrolled, it can also lead to tissue damage. The nuclear factor-kappa light chain enhancer of activated B cells (NF-κB) signaling pathway is particularly relevant in the intestine, as it seems to play a dual role. Whereas NF-κB protects intestinal epithelium against various noxious stimuli, the same pathway mediates intestinal inflammatory diseases by inducing pro-inflammatory gene expression. The availability of appropriate in vitro models of the intestinal epithelium is crucial for further understanding the contribution of NF-κB in physiological and pathological processes and advancing in the development of drugs and therapies against gut diseases. Here we established, characterized, and validated three-dimensional cultures of intestinal organoids obtained from biopsies of NF-κB-RE-Luc mice. The NF-κB-RE-Luc intestinal organoids derived from different intestine regions recreated the cellular composition of the tissue and showed a reporter responsiveness similar to the in vivo murine model. When stimulated with TNF-α, jejunum-derived NF-κB-RE-Luc-reporter organoids, provided a useful model to evaluate the anti-inflammatory effects of natural and synthetic compounds. These reporter organoids are valuable tools to explore the epithelial TNF-α-induced NF-κB contribution in the small intestine, being a reliable alternative method while helping to reduce the use of laboratory animals for experimentation.


Subject(s)
NF-kappa B , Tumor Necrosis Factor-alpha , Animals , Inflammation/metabolism , Jejunum/metabolism , Mice , NF-kappa B/metabolism , Organoids/metabolism , Tumor Necrosis Factor-alpha/metabolism
20.
Rev. peru. med. exp. salud publica ; 39(2): 227-235, abr.-jun. 2022. tab, graf
Article in Spanish | LILACS | ID: biblio-1395048

ABSTRACT

RESUMEN Los organoides son estructuras miniaturizadas, generadas principalmente a partir de células madre pluripotentes inducidas, que se cultivan en el laboratorio conservando sus características innatas o adquiridas. Tienen el potencial de reproducir procesos de desarrollo biológico, modelar procesos patológicos que permitirán el descubrimiento de nuevos fármacos y propicien la medicina regenerativa. Sin embargo, estas experiencias requieren perfeccionamiento constante porque pueden haberse realizado variaciones en la constitución de estos órganos. Por ello, el presente artículo tiene como objetivo revisar la información actualizada sobre organoides y sus procesos experimentales básicos y recientes, empezando por la gastrulación, para tratar de imitar, en lo posible, la formación de las tres capas: ectodermo, mesodermo y endodermo, incluyendo los factores que intervienen en la inducción, diferenciación y maduración en la generación de estos organoides. Asimismo, el diseño y preparación de medios de cultivo altamente especializados que permitan obtener el órgano seleccionado con la mayor precisión y seguridad. Se realizó una búsqueda de artículos originales y de revisión publicados en PubMed, Nature y Science. Los artículos se seleccionaron por sus resúmenes y por su texto completo. Las conclusiones de este articulo destacan las ventajas futuras en el uso y aplicaciones de los organoides.


ABSTRACT Organoids are tiny structures, mainly generated from induced pluripotent stem cells, which are cultured in the laboratory while retaining their innate or acquired characteristics. They have the potential to reproduce biological development processes, model pathological processes that will enable the discovery of new drugs and promote regenerative medicine. However, these processes require constant improvement because variations may have occurred in the constitution of the organs. Therefore, this article aims to review updated information on organoids and their basic and recent experimental processes, starting with gastrulation, in an attempt to mimic, as much as possible, the formation of the three layers: ectoderm, mesoderm and endoderm; as well as the information regarding the factors involved in the induction, differentiation and maturation during the generation of organoids. Likewise, the design and preparation of highly specialized culture media that allow obtaining the selected organ with the highest precision and safety. We searched for original and review articles published in PubMed, Nature and Science. Articles were selected for their abstracts and full text. The conclusions of this article highlight the future advantages in the use and applications of organoids.


Subject(s)
Organoids , Signal Transduction , Cell Differentiation , Gastrulation , Induced Pluripotent Stem Cells
SELECTION OF CITATIONS
SEARCH DETAIL