Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Massimiliano Caiazzo is active.

Publication


Featured researches published by Massimiliano Caiazzo.


Nature | 2011

Direct generation of functional dopaminergic neurons from mouse and human fibroblasts

Massimiliano Caiazzo; Maria Teresa Dell’Anno; Elena Dvoretskova; Dejan Lazarevic; Stefano Taverna; Damiana Leo; Tatyana D. Sotnikova; Andrea Menegon; Paola Roncaglia; Giorgia Colciago; Giovanni Russo; Piero Carninci; Gianni Pezzoli; Raul R. Gainetdinov; Stefano Gustincich; Alexander Dityatev; Vania Broccoli

Transplantation of dopaminergic neurons can potentially improve the clinical outcome of Parkinson’s disease, a neurological disorder resulting from degeneration of mesencephalic dopaminergic neurons. In particular, transplantation of embryonic-stem-cell-derived dopaminergic neurons has been shown to be efficient in restoring motor symptoms in conditions of dopamine deficiency. However, the use of pluripotent-derived cells might lead to the development of tumours if not properly controlled. Here we identified a minimal set of three transcription factors—Mash1 (also known as Ascl1), Nurr1 (also known as Nr4a2) and Lmx1a—that are able to generate directly functional dopaminergic neurons from mouse and human fibroblasts without reverting to a progenitor cell stage. Induced dopaminergic (iDA) cells release dopamine and show spontaneous electrical activity organized in regular spikes consistent with the pacemaker activity featured by brain dopaminergic neurons. The three factors were able to elicit dopaminergic neuronal conversion in prenatal and adult fibroblasts from healthy donors and Parkinson’s disease patients. Direct generation of iDA cells from somatic cells might have significant implications for understanding critical processes for neuronal development, in vitro disease modelling and cell replacement therapies.


Nature Materials | 2016

Defined three-dimensional microenvironments boost induction of pluripotency

Massimiliano Caiazzo; Yuya Okawa; Adrian Ranga; Alessandra Piersigilli; Yoji Tabata; Matthias P. Lutolf

Since the discovery of induced pluripotent stem cells (iPSCs), numerous approaches have been explored to improve the original protocol, which is based on a two-dimensional (2D) cell-culture system. Surprisingly, nothing is known about the effect of a more biologically faithful 3D environment on somatic-cell reprogramming. Here, we report a systematic analysis of how reprogramming of somatic cells occurs within engineered 3D extracellular matrices. By modulating microenvironmental stiffness, degradability and biochemical composition, we have identified a previously unknown role for biophysical effectors in the promotion of iPSC generation. We find that the physical cell confinement imposed by the 3D microenvironment boosts reprogramming through an accelerated mesenchymal-to-epithelial transition and increased epigenetic remodelling. We conclude that 3D microenvironmental signals act synergistically with reprogramming transcription factors to increase somatic plasticity.


Stem cell reports | 2015

Direct Conversion of Fibroblasts into Functional Astrocytes by Defined Transcription Factors

Massimiliano Caiazzo; Serena G. Giannelli; Pierluigi Valente; Gabriele Lignani; Annamaria Carissimo; Alessandro Sessa; Gaia Colasante; Rosa Bartolomeo; Luca Massimino; Stefano Ferroni; Carmine Settembre; Fabio Benfenati; Vania Broccoli

Summary Direct cell reprogramming enables direct conversion of fibroblasts into functional neurons and oligodendrocytes using a minimal set of cell-lineage-specific transcription factors. This approach is rapid and simple, generating the cell types of interest in one step. However, it remains unknown whether this technology can be applied to convert fibroblasts into astrocytes, the third neural lineage. Astrocytes play crucial roles in neuronal homeostasis, and their dysfunctions contribute to the origin and progression of multiple human diseases. Herein, we carried out a screening using several transcription factors involved in defining the astroglial cell fate and identified NFIA, NFIB, and SOX9 to be sufficient to convert with high efficiency embryonic and postnatal mouse fibroblasts into astrocytes (iAstrocytes). We proved both by gene-expression profiling and functional tests that iAstrocytes are comparable to native brain astrocytes. This protocol can be then employed to generate functional iAstrocytes for a wide range of experimental applications.


Journal of Neurochemistry | 2007

Bdnf gene is a downstream target of Nurr1 transcription factor in rat midbrain neurons in vitro.

Floriana Volpicelli; Massimiliano Caiazzo; Dario Greco; Claudia Consales; Luigi Leone; Carla Perrone-Capano; Luca Colucci D’Amato; Umberto di Porzio

The transcription factor Nurr1 is essential for the generation of midbrain dopaminergic neurons (mDA). Only a few Nurr1‐regulated genes have so far been identified and it remains unclear how Nurr1 influences the development and function of dopaminergic neurons. To identify novel Nurr1 target genes we have used genome‐wide expression profiling in rat midbrain primary cultures, enriched in dopaminergic neurons, following up‐regulation of Nurr1 expression by depolarization. In this study we demonstrate that following depolarization the hyperexpression of Nurr1 and the brain derived neurotrophic factor (BDNF) are phospholipase C‐ and protein kinase C‐dependent. We show that Bdnf, which encodes a neurotrophin involved also in the phenotypic maturation of mDA neurons, is a novel Nurr1 target gene. By RNA interference experiments we show that a decreased Nurr1 expression is followed by tyrosine hydroxylase and BDNF mRNA and protein down‐regulation. Reporter gene assay experiments performed on midbrain primary cultures using four Bdnf promoter constructs show that Bdnf is a direct target gene of Nurr1. Taken together, our findings suggest that Nurr1 might also influence the development and the function of midbrain dopaminergic neurons via direct regulation of Bdnf expression.


Cell Stem Cell | 2015

Rapid Conversion of Fibroblasts into Functional Forebrain GABAergic Interneurons by Direct Genetic Reprogramming

Gaia Colasante; Gabriele Lignani; Alicia Rubio; Lucian Medrihan; Latefa Yekhlef; Alessandro Sessa; Luca Massimino; Serena G. Giannelli; Silvio Sacchetti; Massimiliano Caiazzo; Damiana Leo; Dimitra Alexopoulou; Maria Teresa Dell’Anno; Ernesto Ciabatti; Marta Orlando; Michèle Studer; Andreas Dahl; Raul R. Gainetdinov; Stefano Taverna; Fabio Benfenati; Vania Broccoli

Transplantation of GABAergic interneurons (INs) can provide long-term functional benefits in animal models of epilepsy and other neurological disorders. Whereas GABAergic INs can be differentiated from embryonic stem cells, alternative sources of GABAergic INs may be more tractable for disease modeling and transplantation. We identified five factors (Foxg1, Sox2, Ascl1, Dlx5, and Lhx6) that convert mouse fibroblasts into induced GABAergic INs (iGABA-INs) possessing molecular signatures of telencephalic INs. Factor overexpression activates transcriptional networks required for GABAergic fate specification. iGABA-INs display progressively maturing firing patterns comparable to cortical INs, form functional synapses, and release GABA. Importantly, iGABA-INs survive and mature upon being grafted into mouse hippocampus. Optogenetic stimulation demonstrated functional integration of grafted iGABA-INs into host circuitry, triggering inhibition of host granule neuron activity. These five factors also converted human cells into functional GABAergic INs. These properties suggest that iGABA-INs have potential for disease modeling and cell-based therapeutic approaches to neurological disorders.


Journal of Clinical Investigation | 2014

Remote control of induced dopaminergic neurons in parkinsonian rats.

Maria Teresa Dell’Anno; Massimiliano Caiazzo; Damiana Leo; Elena Dvoretskova; Lucian Medrihan; Gaia Colasante; Serena G. Giannelli; Ilda Theka; Giovanni Russo; Liudmila Mus; Gianni Pezzoli; Raul R. Gainetdinov; Fabio Benfenati; Stefano Taverna; Alexander Dityatev; Vania Broccoli

Direct lineage reprogramming through genetic-based strategies enables the conversion of differentiated somatic cells into functional neurons and distinct neuronal subtypes. Induced dopaminergic (iDA) neurons can be generated by direct conversion of skin fibroblasts; however, their in vivo phenotypic and functional properties remain incompletely understood, leaving their impact on Parkinsons disease (PD) cell therapy and modeling uncertain. Here, we determined that iDA neurons retain a transgene-independent stable phenotype in culture and in animal models. Furthermore, transplanted iDA neurons functionally integrated into host neuronal tissue, exhibiting electrically excitable membranes, synaptic currents, dopamine release, and substantial reduction of motor symptoms in a PD animal model. Neuronal cell replacement approaches will benefit from a system that allows the activity of transplanted neurons to be controlled remotely and enables modulation depending on the physiological needs of the recipient; therefore, we adapted a DREADD (designer receptor exclusively activated by designer drug) technology for remote and real-time control of grafted iDA neuronal activity in living animals. Remote DREADD-dependent iDA neuron activation markedly enhanced the beneficial effects in transplanted PD animals. These data suggest that iDA neurons have therapeutic potential as a cell replacement approach for PD and highlight the applicability of pharmacogenetics for enhancing cellular signaling in reprogrammed cell-based approaches.


Stem Cells Translational Medicine | 2013

Rapid Generation of Functional Dopaminergic Neurons From Human Induced Pluripotent Stem Cells Through a Single-Step Procedure Using Cell Lineage Transcription Factors

Ilda Theka; Massimiliano Caiazzo; Elena Dvoretskova; Damiana Leo; Federica Ungaro; Sebastiano Curreli; Francesca Managò; Maria Teresa Dell'Anno; Gianni Pezzoli; Raul R. Gainetdinov; Alexander Dityatev; Vania Broccoli

Current protocols for in vitro differentiation of human induced pluripotent stem cells (hiPSCs) to generate dopamine (DA) neurons are laborious and time‐expensive. In order to accelerate the overall process, we have established a fast protocol by expressing the developmental transcription factors ASCL1, NURR1, and LMX1A. With this method, we were able to generate mature and functional dopaminergic neurons in as few as 21 days, skipping all the intermediate steps for inducting and selecting embryoid bodies and rosette‐neural precursors. Strikingly, the resulting neuronal conversion process was very proficient, with an overall efficiency that was more than 93% of all the coinfected cells. hiPSC‐derived DA neurons expressed all the critical molecular markers of the DA molecular machinery and exhibited sophisticated functional features including spontaneous electrical activity and dopamine release. This one‐step protocol holds important implications for in vitro disease modeling and is particularly amenable for exploitation in high‐throughput screening protocols.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Neural tube morphogenesis in synthetic 3D microenvironments

Adrian Ranga; Mehmet Girgin; Andrea Meinhardt; Dominic Eberle; Massimiliano Caiazzo; Elly M. Tanaka; Matthias P. Lutolf

Significance In vitro organoids have become widely used model systems in basic research and for therapeutic applications due to their ability to recapitulate key elements of in vivo form and function. However, their full potential remains unfulfilled as a result of the poorly defined matrices in which they are grown. Here, we use modular synthetic 3D matrices to show that early neural morphogenesis can be precisely controlled by the extracellular microenvironment. Our approach is broadly applicable to gain a broader understanding of the multifactorial 3D cell–matrix interactions that coordinate multicellular growth and differentiation, and opens up avenues to discover and dissect the unique microenvironments that control morphogenesis in various organoid systems. Three-dimensional organoid constructs serve as increasingly widespread in vitro models for development and disease modeling. Current approaches to recreate morphogenetic processes in vitro rely on poorly controllable and ill-defined matrices, thereby largely overlooking the contribution of biochemical and biophysical extracellular matrix (ECM) factors in promoting multicellular growth and reorganization. Here, we show how defined synthetic matrices can be used to explore the role of the ECM in the development of complex 3D neuroepithelial cysts that recapitulate key steps in early neurogenesis. We demonstrate how key ECM parameters are involved in specifying cytoskeleton-mediated symmetry-breaking events that ultimately lead to neural tube-like patterning along the dorsal–ventral (DV) axis. Such synthetic materials serve as valuable tools for studying the discrete action of extrinsic factors in organogenesis, and allow for the discovery of relationships between cytoskeletal mechanobiology and morphogenesis.


Neural Plasticity | 2004

Enhancement of Dopaminergic Differentiation in Proliferating Midbrain Neuroblasts by Sonic Hedgehog and Ascorbic Acid

Floriana Volpicelli; Claudia Consales; Massimiliano Caiazzo; Luca Colucci-D'Amato; Carla Perrone-Capano; Umberto di Porzio

We analyzed the molecular mechanisms involved in the acquisition and maturation of dopaminergic (DA) neurons generated in vitro from rat ventral mesencephalon (MES) cells in the presence of mitogens or specific signaling molecules. The addition of basic fibroblast growth factor (bFGF) to MES cells in serum-free medium stimulates the proliferation of neuroblasts but delays DA differentiation. Recombinant Sonic hedgehog (SHH) protein increases up to three fold the number of tyrosine hydroxylase (TH)-positive cells and their differentiation, an effect abolished by anti-SHH antibodies. The expanded cultures are rich in nestin-positive neurons, glial cells are rare, all TH+ neurons are DA, and all DA and GABAergic markers analyzed are expressed. Adding ascorbic acid to bFGF/SHH-treated cultures resulted in a further five- to seven-fold enhancement of viable DA neurons. This experimental system also provides a powerful tool to generate DA neurons from single embryos. Our strategy provides an enriched source of MES DA neurons that are useful for analyzing molecular mechanisms controlling their function and for experimental regenerative approaches in DA dysfunction.


Experimental Cell Research | 2010

Transcription factor KLF7 regulates differentiation of neuroectodermal and mesodermal cell lineages.

Massimiliano Caiazzo; Luca Colucci-D'Amato; Maria Teresa Esposito; Silvia Parisi; Stefano Stifani; Francesco Ramirez; Umberto di Porzio

Previous gene targeting studies in mice have implicated the nuclear protein Krüppel-like factor 7 (KLF7) in nervous system development while cell culture assays have documented its involvement in cell cycle regulation. By employing short hairpin RNA (shRNA)-mediated gene silencing, here we demonstrate that murine Klf7 gene expression is required for in vitro differentiation of neuroectodermal and mesodermal cells. Specifically, we show a correlation of Klf7 silencing with down-regulation of the neuronal marker microtubule-associated protein 2 (Map2) and the nerve growth factor (NGF) tyrosine kinase receptor A (TrkA) using the PC12 neuronal cell line. Similarly, KLF7 inactivation in Klf7-null mice decreases the expression of the neurogenic marker brain lipid-binding protein/fatty acid-binding protein 7 (BLBP/FABP7) in neural stem cells (NSCs). We also report that Klf7 silencing is detrimental to neuronal and cardiomyocytic differentiation of embryonic stem cells (ESCs), in addition to altering the adipogenic and osteogenic potential of mouse embryonic fibroblasts (MEFs). Finally, our results suggest that genes that are key for self-renewal of undifferentiated ESCs repress Klf7 expression in ESCs. Together with previous findings, these results provide evidence that KLF7 has a broad spectrum of regulatory functions, which reflect the discrete cellular and molecular contexts in which this transcription factor operates.

Collaboration


Dive into the Massimiliano Caiazzo's collaboration.

Top Co-Authors

Avatar

Vania Broccoli

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Floriana Volpicelli

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Damiana Leo

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Matthias P. Lutolf

École Polytechnique Fédérale de Lausanne

View shared research outputs
Top Co-Authors

Avatar

Raul R. Gainetdinov

Saint Petersburg State University

View shared research outputs
Top Co-Authors

Avatar

Carla Perrone-Capano

University of Naples Federico II

View shared research outputs
Top Co-Authors

Avatar

Elena Dvoretskova

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Fabio Benfenati

Istituto Italiano di Tecnologia

View shared research outputs
Top Co-Authors

Avatar

Gaia Colasante

Vita-Salute San Raffaele University

View shared research outputs
Researchain Logo
Decentralizing Knowledge