Category Archives: Embryonic Stem Cells

On this day: Mahmoud Abbas becomes leader of Fatah in 2004 – In-Cyprus

Following are some of the major events to have occurred on November 25:

1935 King George II returns to Greece as monarch.

1936 Germany and Japan sign anti-Comintern pact.

1952 Agatha Christies play The Mousetrap opened in London. Still playing to audiences today, it holds the record for the longest continuous run of any show in the world.

1963 U.S. President John F. Kennedy was buried with full military honours at Arlington National Cemetery, three days after his assassination.

1974 The Burmese diplomat U Thant died. He became U.N. secretary-general after the death of Dag Hammarskjold in 1961, and held the post until 1971.

1997 Malawis former leader, Kamuzu Banda, died aged 99. As Hastings Banda, he became president in 1966 and proclaimed himself ruler for life in 1971. He was defeated in 1994 in Malawis first democratic election.

1999 Six-year-old Cuban Elian Gonzalez survives smuggling boat shipwreck on its way to the United States, sparking a controversial custody case between the two countries.

2001 Advanced Cell Technology Inc. of Massachusetts became the first organisation to report the successful cloning of a human embryo. The company said it did not intend to create a human being but to use the stem cells to treat disease.

2004 The dominant Palestinian political faction, Fatah, approved Mahmoud Abbas as its candidate to succeed Yasser Arafat, who had died on Nov. 11.

2005 Richard Burns, the only Englishman to win the world rally championship, died of a brain tumour at the age of 34.

2015 Pope Francis arrives in Kenya on historic African visit.

(Reuters)

The rest is here:
On this day: Mahmoud Abbas becomes leader of Fatah in 2004 - In-Cyprus

Improving the therapeutic efficacy of oncolytic viruses for cancer … – Journal of Translational Medicine

Origin and distribution of macrophages

Macrophages are ubiquitous in any part of the body and perform three essential functions, namely phagocytosis, exogenous antigen presentation, and secretion of cytokines and growth factors for immunomodulation. They perform important duties in tissue development, homeostasis, clearance of dead cells and foreign pathogens, and modulation of inflammatory and tumoral immune responses [32,33,34]. Macrophages also have different names and functions in different tissues, such as circulating monocyte-derived macrophages, tissue-resident macrophages (TRMs), and tumor-associated macrophages, which have complex correlations in terms of classification and origin. TRMs perform appropriate functions in various tissues of the body, including microglia in the brain, Kupffer cells in the liver, and Langerhans cells in the skin [35, 36], and it is currently believed that most of the population of TRMs originates from embryonic precursors in the yolk sac and fetal liver and that they self-maintain independently of the myeloid cells in adulthood [37, 38]. TAMs, on the other hand, consist mainly of circulating monocyte-derived macrophages and RTMs recruited by tumors into TME and are one of the important targets for tumor immunotherapy [39].

Macrophages are significant plastic and their activation state is influenced by a multitude of factors, but they can usually be simplified into two classifications based on stimulatory factors and secretory products (Fig.2), namely classically activated M1 macrophages and alternatively activated M2 macrophages [40]. Although this M1/M2 dichotomization simplifies the differences in phenotypic and functional continuum changes in macrophages, this terminology is still more commonly used when discussing whether macrophages are more biased toward a pro-inflammatory or anti-inflammatory phenotype [41].

Macrophage activation and M1/M2 typing. Macrophages polarized into classically activated (M1) or alternatively activated (M2) macrophages under the influence of different cytokines or other factors secrete different cytokines to change the cellular microenvironment to a pro-inflammatory or anti-inflammatory state, exerting anti-tumor or pro-tumor effects at the tumor site

M1 macrophage polarization is usually driven by granulocytemacrophage colony-stimulating factor (GM-CSF), lipopolysaccharide (LPS), IFN-, TNF-, and PAMPs [42]. M1 phenotype macrophages have mainly pro-inflammatory properties, promoting the pro-inflammatory response of helper T cells 1 (Th1) by secreting cytokines, such as TNF-, IL-1, IL-12, and IL-18, and enhancing the recruitment of Th1 cells to sites of inflammation by secreting chemokines, such as chemokines CXC motif ligand 9 (CXCL9) and CXCL10 [43]. M1 macrophages can trigger an adaptive immune response through self-mediated cytotoxicity or cross-presentation of antigens (TAAs and TANs), triggering potent anti-tumor immunity. Therefore, M1 macrophages are considered a tumor-suppressive macrophage phenotype [44].

M2 macrophage polarization is usually driven by macrophage colony-stimulating factor (M-CSF), IL-4, IL-10, IL-13, and transforming growth factor- (TGF-) [45]. M2 macrophages have a critical position in appropriate immune function and homeostasis in vivo, with examples including stimulation of Th2 cell responses, mediation of parasite clearance, immunomodulation, wound healing and tissue repair [46]. However, the function of M2 macrophages can also be adversely affected by tumor exploitation by producing immunosuppressive and pro-angiogenic factors such as IL-10, arginase 1 (ARG1), TGF-, or vascular endothelial growth factors (VEGFs), which stimulate tumor cell proliferation, invasion, metastasis, and angiogenesis [41]. Therefore, M2 macrophages are considered a tumor-supporting macrophage phenotype[47].

TAMs are a collective term for macrophages that are prevalent in tumors and can account for up to 50% of some solid tumors [48]. TAMs also share the markers of M1/M2 macrophages [49], however, TAMs rarely exhibit a true M1 or M2 phenotype and are more aptly referred to as M1-like/M2-like TAMs [50]. Under the effects of tumor-secreted colony-stimulating factor 1 (CSF-1, or M-CSF), TAMs polarize to M2-like, allowing immunosuppressive M2-like TAMs to predominate in tumors [47, 51]. High infiltration of M2-like TAMs reduces therapeutic efficacy, shaping tumor-supportive TME, angiogenesis, fibrosis, immunosuppressive cell recruitment, lymphocyte rejection, drug resistance, invasion, and metastasis to enhance tumor progression [52,53,54], which are often associated with poor clinical outcomes [55,56,57].

TAMs are effective target cells in immunotherapy of tumors [12, 58]. This is because macrophages exert opposite anti-tumor or pro-tumor functions through a range of activation pathways and/or different macrophage populations [13, 59]. Different approaches can be taken to eliminate tumor-promoting macrophages and activate or transform them into tumor-suppressing macrophages. Common therapeutic strategies are inhibition of TAMs recruitment [60, 61], reprogramming of TAMs to an M1-like phenotype [62,63,64], and depletion of TAMs [65, 66].

Macrophage plasticity influences tumor progression and treatment outcome and has a similar effect in oncolytic virotherapy. When OVs are delivered to the body, the body triggers innate immunity in response to the foreign invasion of viral infection. Monocytes, macrophages and NK cells will recognize and remove some of the OVs and play a certain inhibitory role. However, in this process, macrophages will also act as carriers of OVs to tumor cells. At the same time macrophages enhance polarization toward a pro-inflammatory phenotype, and this local immune response is also critical for initiating initial anti-tumor immunity [67]. Therefore, we need to further comprehend the complex interactions among OVs, macrophages, and tumors (Fig.3), to elucidate the mechanisms of macrophages that limit or promote the tumoricidal effects of OVs, and to better utilize the advantages of macrophages to enhance the anti-tumor benefits in future oncolytic virus therapeutic strategies.

Interaction of OVs, macrophages, and tumor cells. After OVs are delivered, some OVs are attacked by activated monocytes/macrophages, causing the viral titer of OVs to decrease. Another portion of OVs can be transported to the tumor site for viral replication, lysing tumor cells and releasing viral progeny, damage-associated molecular patterns (DAMPs), pathogen-associated molecular patterns (PAMPs), and tumor-associated antigens (TAAs). Antigen-presenting cells (APCs) take up and present these antigens, and the resulting activated antigen-specific CD8+T cells as well as natural killer (NK) cells exert antitumor effects. Secreted IFN- and PAMPs repolarize pro-tumorigenic M2-like macrophages into anti-tumorigenic M1-like macrophages, and the anti-tumor/viral effects of the immune system can be further enhanced by secreting IFN- and TNF-

In general, macrophages show antiviral activity in the setting of oncolytic virotherapy, which is consistent with their defense against pathogens.

Among the routes of administration of OVs, intravenous has more potential than intra-tumoral injection in the treatment of systemic metastatic tumors. However, intravenously administered OVs are often hindered by circulating and tissue immune complexes, neutralizing antibodies, and innate immune cells before reaching the tumor site. Activated macrophages have multiple viral clearance mechanisms, including virus recognition through PRRs, cytokine responses such as IFN, phagocytosis, and activation of other immune cells to reduce viral titers delivered to the tumor site [68, 69].

In a glioma model, phagocytosis by macrophages limits the spread of OVs. Delivery of oncolytic herpes simplex virus (oHSV) after the depletion of macrophages can increase viral titers at tumor sites [70]. IFN and TNF- signaling is an important mechanism for the antiviral effects of macrophages [71, 72]. In ovarian and breast cancer models it was shown that macrophages can activate the tumor cell JAK/STAT pathway and upregulate the expression of interferon-stimulated genes (ISGs), with tumor cells thereby acquiring an antiviral status that makes them resistant to OVs [73]. In a study of glioblastoma (GBM) treated with oHSV, macrophages, and microglia were found to be the main producers of TNF-, which inhibits viral replication. Brief administration of TNF- blockers effectively enhances the killing of tumor cells while reducing inflammation-induced neurotoxicity, enhancing viral replication and survival in GBM intracranial tumors [69]. TAMs and microglia in malignant gliomas largely limit the activity of OVs [74].

Although inflammatory cytokines and phagocytosis produced by macrophages are powerful weapons to kill tumor cells, they also reduce the efficiency of transport of OVs to tumors, so direct delivery of OVs requires a larger viral load to counteract this clearance effect and increases the viral titer of transport to tumor sites.

However, on the other hand, the interaction between macrophages and OVs could enhance the antitumor effect.

First of all, macrophages can act as carriers of OVs for transport. Macrophages have shown antiviral effects to some extent, but interestingly, increasing studies have evidenced that viruses can utilize monocytes/macrophages as vectors for spreading and replication [75], and macrophages may be an integral part of the therapy of OVs, possibly due to the higher susceptibility of monocytes or nave macrophages to OVs [76]. Previous research has found that monocytes/macrophages in peripheral blood can act as viral vectors, transporting viable viral particles to tumor sites. Follow-up after intravenous administration of the eutherian virus recovered replicative and oncolytic eutherian virus in blood mononuclear cells even in the presence of neutralizing antibodies (nAbs) to the virus [77]. In another study with oncolytic adenovirus, it was shown that, possibly due to the very low expression of viral antigens, macrophages can act as silent vectors that hide and support viral replication, allowing adenovirus delivery to the tumor site and produce a long-lasting therapeutic effect [78]. More interestingly, recent preclinical studies have found that macrophages are not only capable of uptake and delivery of the tumor oncolytic virus HSV1716 but also support HSV1716 replication within macrophages, which could enhance the effect of viral therapy [79].

Second, OVs can enhance the phagocytic activity of macrophages on tumor cells. As mentioned earlier, TAMs are an important component of macrophages. Activation of TAMs to produce phagocytic activity is a novel mechanism of tumor killing [80], which can be activated by oncolytic virus treatment. CD47 is a membrane-bound protein that is highly expressed on tumor cells and binds to signal regulatory protein (SIRP) on macrophages, delivering a don't eat me signal that leads to immune evasion by the tumor [81]. After OVs infect cells, PAMPs are exposed to the host immune system, inducing endoplasmic reticulum stress and ICD, leading to the release of DAMPs [82,83,84], which include calreticulin (CRT). CRT, an endoplasmic reticulum-associated molecular chaperone, can also block the CD47 receptor on tumor cells, thereby reducing the don't eat me signals generated by macrophages and DCs in response to CD47 binding, and attenuating immune evasion by tumor cells [85]. In addition, after OVs interacted with the B cell receptor (BCR), activated B cells were able to release neutralizing antibodies that mediated NK cell antibody-dependent cytotoxicity (ADCC) and macrophage antibody-dependent cell phagocytosis (ADCP) of virus-infected tumor cells, activating phagocytosis of tumor cells by innate immune cells [86].

Most importantly, OVs can induce polarization of TAMs towards an anti-tumor phenotype. OVs induce activation of NK cells and macrophages through PRRs recognizing PAMPs and DAMPs, secretion of inflammatory cytokines such as IFN-, and induced macrophage polarization to M1-like, which results in diminished immunosuppression of TAMs [76, 87]. In an in vitro model of breast cancer, it was found that irrespective of the initial polarization state of macrophages, treatment with oncolytic measles virus (MeV) and mumps virus (MuV) resulted in a significant increase in the M1 macrophage marker, CD80, in human monocyte-derived macrophages (MDMs), while inducing anti-tumor cytokines IL-1, TNF-, CXCL9, CXCL10, and IL -6 concentrations were elevated [88]. Preclinical and clinical studies in gastric cancer or glioma have found that treatment with HSV-1 or oncolytic adenovirus rapidly recruited inflammatory cells to the injected lesions, significantly increased the intra-tumoral infiltration of M1-like macrophages and NK cells, with a reduction in the expression of M2-like macrophages, and a significant elevation of the pro-inflammatory cytokines IFN- and TNF- [89, 90]. Although oncolytic adenovirus shifts human macrophages from a more pro-tumor phenotype to a less favorable phenotype, this phenotypic shift is not complete and the M2 trait is not completely lost at the level of gene expression, immunophenotype, and cytokines, which is consistent with the concept that the M1/M2 typing of macrophages is not completely extreme, but rather sequential in phenotype and function [91].

Due to the multifaceted effects generated by macrophages in the treatment of OVs, eliminating the limiting effect of macrophages on OVs, exploiting the effectiveness of macrophages, and obtaining better therapeutic results require intensive research. The current directions are mainly the following: (1), arming OVs to enhance the beneficial effects (pro-inflammatory phenotypic polarization and phagocytosis) or attenuate the adverse effects (antiviral and pro-tumorigenic effects); (2), combining with other drugs to increase the antitumor efficacy; and (3), augmenting the targeting of OVs to tumor cells through effective carrier delivery.

OVs can be genetically engineered to arm viruses, and different immunomodulatory genes for arming OVs are being actively tested. Various OVs expressing pro-inflammatory cytokines, chemokines, and other immune checkpoint-associated molecules have been developed to enhance the anti-tumor effects of macrophages (Fig.4A).

Basic macrophage strategies in oncolytic virotherapy. Currently, there are two major directions of basic strategies for targeting the macrophage to optimize therapeutic response. On the one hand, armed OVs enhance the anti-tumor effect of macrophages. A Repolarization to an antitumor phenotype. Given the pro-tumorigenic role of M2-like tumor-associated macrophages (TAMs), the expression of pro-inflammatory cytokines or chemokines by genetically modified viruses was used to increase macrophage activity and promote the polarization of M2-like macrophages to M1-like macrophages. B Enhancement of phagocytosis by macrophages. The expression of anti-CD47 antibody or SIRP-Fc fusion protein after viral genetic modification can disrupt don't eat me signaling and enhance the killing of tumor cells by macrophages. On the other hand, weakening the clearance of OVs by macrophages contributes to higher viral titers at tumor sites. C Direct macrophage depletion. Since OVs are subject to phagocytosis by macrophages and/or clearance by antiviral cytokines after delivery, brief administration of macrophage depletion agents prior to OVs treatment can cause apoptosis of macrophages, increase the titer of OVs, and change the phenotype of TAMs. D Delivered through the carrier. In addition, the use of tumorophilic carrier cells or liposomes to deliver OVs, is also able to avoid the negative effects of neutralizing antibodies and/or innate immune cells and overcome the challenges of systemic administration of OVs

A high M2/M1 ratio in TAMs is strongly associated with tumor progression and poor prognosis. Although OVs can inherently promote polarization of M1-like TAMs and reduce the number of M2-like TAMs, armed OVs can further enhance this polarization.

Talimogene laherparepvec (T-VEC), a GM-CSF-expressing HSV-1, is the first OVs approved by the U.S. Food and Drug Administration (FDA) for the treatment of patients with advanced melanoma, with favorable safety and therapeutic outcomes [92]. This is due to the ability of GM-CSF-expressing OVs to attract monocytes and differentiate them into macrophages and DCs, repolarize TAMs from an M2-like phenotype to an M1-like phenotype, and increase the expression of the pro-inflammatory cytokines TNF-, IL-6, and IL-10 [93, 94].

IL-12 is one of the major regulators of anti-tumor immune responses, promoting the maturation of NK cells, DCs, and T cells, inducing M1-like polarization of macrophages, and increasing IFN- levels [95]. Many OVs are currently modified and produce IL-12 [96], and in a GBM model, the use of an oHSV expressing murine IL-12 (G47-mIL12) increased polarization of M1-like TAMs (iNOS+ and pSTAT1+), which may be due to IL-12-induced increases in IFN- in the TME [97].

Although IL-12 can effectively induce antitumor immunity, it has certain toxic side effects after systemic administration [95], and IL-21 may be a safer cytokine compared to IL-12. In a pancreatic cancer model study, it was demonstrated that treatment with VVL-21, an oncolytic vaccinia virus (VV) that expresses IL-21, increased the expression of M1-like macrophage marker major histocompatibility complex II (MHC II) and cytokine gene transcripts (IL-6/IL-12 and COX2), and decreased the expression of M2 macrophage marker (CD206) and cytokine gene transcripts (IL-10, TGF-, and CCL22) expression while also increasing M1 polarization in nave macrophages [98]. In addition, an IL-36-expressing VV (IL-36-OVs) was developed. It induces infiltration of lymphocytes and DCs, reduces MDSCs and M2-like TAMs, and has shown significant therapeutic effects in a variety of mouse tumor models [99].

OVs with chemokines are able to effectively recruit immune cells with antitumor effects to migrate to infected tumor sites. Chemokine CC motif ligand 5 (CCL5) promotes immune cell chemotaxis by interacting with chemokine CC motif receptor 1 (CCR1), CCR3, and CCR5 [100]. Infection of tumor cells with CCL5-expressing OVs significantly enhances the migration and activation of NK cells, macrophages, and T cells, and also activates the secretion of CXCL9 by macrophages and DCs aggregated in tumors by binding to tumor cells to activate Fc receptor-mediated ADCC in NK cells and ADCP in macrophages [101, 102], which in turn further promotes the infiltration of circulating T cells into tumor tissues [103].

Both CD40 and OX40 and their ligands CD40L and OX40L belong to the TNF receptor superfamily (TNFRSF). The interaction of CD40 and CD40L activates APCs [104], and the interaction of OX40 and OX40L activates T cells [105], which promotes antitumor effects through activated downstream signaling pathways. A CD40L-expressing oncolytic adenovirus (TMZ-CD40L) is effective in treating pancreatic cancer, a tumor with a high level of M2 macrophages, by increasing the infiltration of M1-like macrophages and T cells into the tumor, repolarizing M2-like macrophages, and controlling tumor progression [106]. Also in a pancreatic cancer model, the use of HSV-1 expressing murine OX40L ((OV-mOX40L) triggered an OX40-OX40L signaling pathway-mediated response that also reprogrammed macrophages and neutrophils to an anti-tumor state, enhanced the anti-tumor response of T cells, and significantly prolonged the survival time of mice [107].

At the same time, it is desired to modify OVs to further block the immunosuppressive effect and enhance phagocytosis of tumors by macrophages (Fig.4B). An engineered oHSV equipped with a full-length anti-CD47 antibody can be used to disrupt the don't eat me signaling generated by the CD47/SIRP pathway. This oHSV activated phagocytosis and cytotoxicity of tumor cells by macrophages and NK cells, prolonging the survival of glioblastoma and ovarian cancer model mice [108, 109]. Accordingly, investigators designed a VV capable of expressing a chimeric molecule (SIRP-Fc) consisting of the ectodomain of SIRP and the Fc structural domain of IgG4. SIRP-Fc was able to disrupt CD47/SIRP interactions by blocking CD47 in tumor cells, redirecting macrophages to the tumor site and killing the tumor cells. This VV exerted potent anti-tumor activity in a mouse model of osteosarcoma and can be broadly applied to tumors expressing CD47 [110].

Recently, in a study on cholesterol metabolism, progress has also been made in relation to macrophage phagocytic activity. This study found that TAMs in GBM accumulate cholesterol abnormally, leading to dysfunctional phagocytosis [111]. Apolipoprotein A1 (ApoA1) is a cholesterol reverse transporter protein that allows cholesterol efflux from TAMs, thereby restoring their phagocytosis and antigen-presenting role. Therefore, the investigators developed an ApoA1-expressing oncolytic adenovirus (AdVAPOA1) to intervene in cholesterol metabolism in GBM. AdVAPOA1 activated the TAM-T cell axis and downregulated immune checkpoints after intra-tumor administration, inducing systemic tumor-specific immune memory [111]. This study proposes an immunometabolic treatment approach to armed OVs.

Genetically modified OVs not only enhance anti-tumor immunity in macrophages, but also circumvent the detrimental effects of macrophages, including reducing M2-like TAMs and attenuating macrophage-restricted effects on OVs.

Currently, a panel of oncolytic adenoviruses (EnAd) expressing bivalent T-cell engagers (BiTEs) has been designed to target the immunosuppressive effects of M2-like TAMs. The BiTEs recognize CD3 on T cells and CD206 or folate receptor (FR) on M2-like macrophages. Use of such OVs in patients with malignant ascites activates T cells to selectively kill M2-like macrophages, thereby preserving M1-like macrophages and repolarizing the microenvironment toward a pro-inflammatory state [112].

Human species C adenovirus (HAdv-C5) is bound by immunoglobulin M (IgM) and coagulation factor X (FX) in the blood when delivered intravenously [113, 114], leading to the sequestration of OVs in liver-resident macrophages (Kupffer cells), limiting their tumor targeting and leading to hepatotoxicity [115]. Based on these, the investigators constructed the HAdv-C5 capsid-modified viral variant Ad5-3M. Ad5-3M is resistant to IgM- and complement-mediated inactivation, reduces internalization of the viral variant by Kupffer cells, and circumvents the adverse effects of innate immunity to OVs. In mice with disseminated lung tumors, Ad5-3M prolonged survival and improved safety and efficacy after intravenous administration of OVs [116]. Therefore, the use of genetic modification to change some protein sites in OVs to enhance their resistance is also a worthy direction.

In addition to modifying the OVs' own properties, finding the appropriate drugs for combination therapy opens up more possibilities. These strategies include combining immune checkpoint inhibitors to enhance antitumor effects, and combining macrophage depleting agents or immunosuppressive drugs to increase the titer of OVs.

Combination therapy with OVs and immune checkpoint inhibitors (ICIs) is a common combination strategy in clinical trials today (Table 1), due to the ability of OVs to increase the sensitivity of tumor cells to ICIs, which has demonstrated a strong therapeutic effect in a wide range of tumor treatments [117,118,119]. In a GBM model, the use of IL-21-expressing VV (VVDTK-STCDN1L-mIL21) in combination with systemic anti-programmed death receptor 1 (anti-PD1) therapy showed significant induction of M1-like macrophage polarization in the tumor during treatment, along with increased activation of M0 macrophages (MHC II+) in the spleen and DCs in the lymph nodes [120]. Similarly, in other GBM and triple-negative breast cancer models, combination treatment of engineered OVs with ICIs such as anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA-4) antibody, anti-PD-1 antibody and anti-programmed cell death ligand 1 (anti-PD-L1) significantly inhibited tumor growth. The results showed an increase in the proportion of M1-like TAMs, CD4+ and CD8+ cells, and a decrease in the number of immunosuppressive cells such as Tregs. The application of ICIs prevented immune escape from the tumor and overcame the immunosuppressive microenvironment, which is of great significance for the effective eradication of the tumor [97, 121].

OVs combined with macrophage-depleting agents have been reported to remodel TME. In macrophage-dependent tumors, investigators tested the effectiveness of clodronate liposomes and trabectedin in the oHSV treatment of Ewing's sarcoma [122]. Clodronate liposomes can transiently deplete macrophages throughout the body and have demonstrated their therapeutic potential in applications in a variety of tumors [70, 123]. Trabectedin is a chemotherapeutic agent that depletes monocytes/macrophages, including TAMs, by activating caspase-8-dependent apoptosis through the TRAIL receptor [65]. Both drugs were found to enhance antitumor efficacy after macrophage depletion (Fig.4C). Clodronate liposomes induced antitumor gene expression in TAMs, trabectedin lowered the number of intratumoral MDSCs and M2-like macrophages, and the combination of both drugs with OVs significantly changed the phenotype of TAMs and tended the immune microenvironment to an inflammatory state [122].

Inhibition of macrophage-associated pathways has also shown good efficacy in combination with other immunologic agents. The phosphatidylinositol-3-kinase (PI3K) pathway has an important part in tumor development. PI3K signaling is a key driver of macrophage M2 polarization [124, 125]. PI3K, one of the classes I PI3K isoforms, is hyper-enriched in leukocytes, of which macrophages are included [126]. Some investigators have demonstrated that treatment with PI3K inhibitors prior to intravenous delivery of VV significantly improves VV delivery to tumors and enhances tumor efficacy. This was achieved by interfering with the RhoA/ROCK, AKT, and Rac signaling pathways to inhibit viral attachment to macrophages, independent of viral internalization by macrophages [127]. They combined a PI3K inhibitor (CAL-101), engineered VV, and -PD1 for the treatment of pancreatic cancer in mice, and the results showed strong synergistic effects, demonstrated the effectiveness of systemic administration, and broke through a major limitation in the treatment of OVs [98]. In addition to this, the use of rapamycin in oncolytic virotherapy has added new possibilities. Rapamycin has immunosuppressive properties and it is able to reduce type I IFN production by inhibiting mammalian target of rapamycin complex 1 (mTORC1) [128], reduce infiltration of CD68+ microglia and CD163+ macrophages in gliomas, and increase viral replication and therapeutic efficacy within tumors [129].

Although suppression of the antiviral immune response of macrophages is beneficial in enhancing the therapeutic effect of OV, such immunosuppression may impair the functional balance of macrophages in vivo and diminish the effect of virus-mediated immune stimulation against cancer. Delivery of OVs using carrier cells with tumorophilic properties can effectively avoid the influence of the immune system and reduce the neutralization and clearance of OVs before they reach the tumor (Fig.4D). Therefore, this approach may be a more desirable strategy to improve the pharmacokinetics and biological distribution of OVs and has been extensively studied in carrier cells such as mesenchymal stem cells (MSCs), T cells, myeloid cells, and neural stem cells [130].

Moreover, the use of tumor cell tropism to enhance tumor targeting has also been studied accordingly. Membrane-encapsulated oncolytic adenovirus from cancer cells delivered intravenously was able to effectively avoid the antiviral effects of neutralizing antibodies and the innate immune system. This system increases viral replication and enhances the ability of macrophages and DCs to present tumor antigens, and has shown good efficacy in the treatment of different mouse tumor models [131]. When using VV in hosts with pre-existing antibodies to poxviruses, the transient use of a combination of multiple immunosuppressive drugs and cancer cells as carrier cells significantly improves therapeutic efficacy. Although this approach is achieved by increasing the polarization of immunosuppressive M2-like TAMs, such changes are necessary in the long run [132].

Encapsulation of OVs via liposomes (LPs) is also one of the attractive nano-delivery systems. Encapsulation of oncolytic adenovirus (Ad[I/PPT-E1A]) into liposomes coupled to chemokine CC motif ligand 2 (CCL2), which upon intravenous delivery binds to circulating monocytes expressing chemokine CC motif receptor 2 (CCR2), takes advantage of the aggregation of monocytes to hypoxic tumor vessels to deliver encapsulated OVs targeting tumor sites [133]. This system can avoid recognition and delivery to the tumor site by the immune system after intravenous delivery, reducing the number of TAMs located near the blood vessels [134].

Therefore, the use of carriers for adjuvant delivery of OVs is one of the promising strategies. This approach evades the capture of OVs by innate immune cells without affecting the body's immune function, while enhancing the targeting of tumors and reducing the viral delivery load.

In conclusion, macrophages are an important factor affecting the therapeutic effect of OVs, and in the face of this dual effect, how to seek benefits and avoid harm is something we need to consider.

Continue reading here:
Improving the therapeutic efficacy of oncolytic viruses for cancer ... - Journal of Translational Medicine

Patients accept therapy using embryonic stem cells for Parkinson’s … – BMC Medical Ethics

Discrete choice experiment

Preferences of patients with PD for potential cell-based therapies to treat PD were assessed by a Discrete Choice Experiment (DCE) in Swedish patients with PD. The DCE is a cross-sectional survey method to investigate individuals preferences and can be used to determine the relative importance of different characteristics of an intervention and predict uptake of different interventions [15]. Respondents of a DCE are faced with a set of hypothetical choice questions with two or more alternatives, characterized by different characteristics (i.e., attributes) with varying levels. The DCE method also allows for the calculation of attribute trade-offs [16].

We performed a scoping literature review to identify attributes of treatments for PD that potentially were of importance for patients with PD when choosing treatment. Qualitative and quantitative papers investigating preferences of patients with PD related to treatment for PD were included. All literature searches were performed in PubMed and the keywords used were Parkinson disease, patient preferences, preferences, treatment, medication, and attributes. We identified 193 papers, including 29 papers that were relevant for this project, of which 20 papers remained after excluding duplicates. After reading the full text papers, 209 potential attributes were identified. Out of the 209 attributes identified in the scoping literature review, 115 attributes were unique. These attributes were condensed down to 45 by merging similar concepts. The identified attributes were discussed in a group consisting of a representative patient of a Parkinson patient organization, neurologists, a research coordinator, a nurse working with patients with PD, and researchers knowledgeable in DCE methodology. Based on the discussions in this group, 11 attributes remained. We let 17 patients with PD rank the 11 attributes from most to least important, for their decision about PD treatment. Based on the mean ranks of the attributes and discussions with clinicians, eight attributes remained. These were re-categorized into the five attributes that were assigned relevant levels to be assessed by the DCE: (i) type of treatment, (ii) aim of treatment, (iii) available knowledge of the different types of treatments, (iv) effect on symptoms, and (v) risk for severe side effects (Table1).

We followed methodological guidelines to estimate the sample size needed to identify preferences of patients with PD and differences within those preferences [17]. We considered the number of attributes in the DCE (Table1) and the number of choice questions for each respondent (n=9). Based on the sample size requirements for a DCE and accounting for subgroup analysis, we aimed for a sample size of 500 respondents.

Patients with PD were recruited from neurology clinics at two university hospitals in Sweden. This study was approved by the Swedish Ethical Review Authority (Dnr 201906539). Information about the study was sent out by mail to all potential respondents fulfilling the inclusion criteria: patients diagnosed with PD, 18 years or older, able to read and understand Swedish. Patients with a known dementia diagnosis were excluded. Information about the study was sent out to 1266 patients. Patients who had not responded within two weeks were sent a reminder by mail. All respondents provided their informed consent before entering the survey. Two patients formally declined participation, and five patients were unable to participate due to technical or health-related restrictions. In total, 498 patients participated in the study (i.e., 39% response rate).

This survey was administered as a web-based survey that included three parts: (i) information about the attributes and levels, (ii) the DCE with hypothetical choice scenarios, and (iii) demographic and attitude questions (see supplementary file for survey). The survey was created for this study and administered using Sawtooth Software (Sawtooth Software Inc.). Each respondent was faced with nine hypothetical choice scenarios that each included three alternatives. The respondents were asked to select the alternative that they most preferred out of the three presented to them. The first two alternatives were experimentally designed to assess preferences for potential treatment alternatives for PD and the third was a fixed profile (i.e., nonexperimental) to represented standard care (drugs) for patients with PD (Fig.1). We used a Bayesian D-efficient design to construct the choice scenarios for the DCE using the NGene program (version1.2.1; ChoiceMetrics 2012). Prior information on the attribute importance was gathered from a pilot test (n=142) in patients with PD. The design used 500 Halton draws and 1000 repetitions. Using the pilot data, a multinomial logit (MNL) model was fitted, and the beta estimates was used as priors for the final experimental DCE design.

Some conditions were posted on the design: if the aim of treatment was to repair damage caused by disease, the treatment could not consist of electric stimulation or drug. If the aim of the treatment was to slow down disease progression, the treatment could not consist of electric stimulation. The final discrete choice survey consisted of 36 unique choice scenarios divided into four blocks; each respondent was randomized into one block and answered nine choice scenarios. The choice questions also included a hover function with further explanations of the attributes and the levels (see Table1 for full description of the attribute levels).

Example of a choice scenario

The demographical and attitude questions included background questions (e.g., age, gender, and education) and disease-related questions (e.g., disease duration, treatment, and side effects). Moreover, the respondents attitudes were gauged with a ranking exercise with eight statements that they were asked to place in the order they found most important. The attitude questions asked respondents about their moral stands on the status of an embryo, and a ranking exercise to prioritize eight statements.

The respondents were asked about their views on how to regard the products left over after IVF procedures, which may be used for hESC isolation, that is, the blastocyst. Whether this material was regarded as a lump of cells or something more was used to dichotomize the answers. Questions to assess respondents health literacy [18] and health numeracy [19] were also included to define the sample.

The statistical analyses, in particular the estimation of the latent class model were performed using R 4.0.2 (R Core Team, 2018), the mlogit (version 1.1-1; Yves Croissant, 2009) and the gmnl (version 1.13.3; Mauricio Sarrias, 2017) [20].

Demographics describing the populations age, gender, country of birth, occupational situation, education, health numeracy, health literacy, drug frequency, disease duration, number of experienced side effects, and experience of advanced treatment were presented in mean, median, and percentages. The overall level of health literacy and numeracy was calculated for each respondent. Individuals who responded strongly disagree or disagree to one of the items were categorized as having low health literacy. Individuals who responded with neither agree nor disagree with one of the items were categorized as having medium health literacy. Individuals responding agree or strongly agree to all the items were categorized as having high health literacy, and likewise for numeracy.

Respondents attitude toward the moral status of a couple of days old human embryo was assessed using this question: The human is perceived to have a special moral position, in the sense of having rights just by being human. What moral position does a human embryo that is only a few days old have? The respondents had four statements from which to select: (1) The embryo is just a lump of cells; it is meaningless to discuss its moral status, (2) The embryo has a moral status that is in between being just a lump of cells and being a human being, (3) The embryo in its moral status is closer to being a human than just a lump of cells, and (4) The embryo has the same moral status as a human being. The variable was dichotomized based on the frequency of the data. Respondents answering The embryo is just a lump of cells; it is meaningless to discuss its moral status formed one group, and the rest another group. One-way analysis of variance and nonparametric measures were used to test the differences between the personal characteristics and the different perceptions of whether an embryo is more than a lump or cells or not.

The most important attitudinal statement was given a 1, the second most important the number 2 and so forth. The ranking exercise was illustrated with a boxplot by the median value of each statement, stratified on the different perceptions of whether an embryo is more than a lump of cells.

The latent class analysis was based on the a priori hypothesis that the authors thought would be associated with the willingness to accept a new treatment. Five variables were tested for class membership: (1) a summary of experience of different treatment, (2) experience of the summary of different side effects, (3) the perception of the moral status of the embryo, (4) experience of advanced treatment, and (5) the importance of religion. A sum of how many treatments each respondent had was calculated, and also how many side effects they had experienced. Advanced treatment was based on treatment experience with one or more of apomorphine subcutaneous injection, apomorphine subcutaneous infusion, deep brain stimulation, levodopa-carbidopa intestinal infusion, and levodopa-entacapone-carbidopa intestinal infusion. The variable the perception of the moral status of the embryo did not influence class membership and was therefore not included in the final class assignment model.

The statistical analyses of the preference data were based on a latent class model. A preference weight (i.e., coefficient) and a corresponding SE were estimated for all but one level of each attribute (i.e., reference attribute level) [21]. Dummy coding of the variables was selected for this analysis (i.e., corresponding to zero as the reference value). Each p-value is a measure of the statistical significance of the difference between the estimated preference weights for each level of the attribute compared to the reference attribute level. All results were considered statistically significant at p<0.05. Confidence intervals (95%) were also provided for each preference weight. The Akaike information criterion (AIC) and the log-likelihood values were considered when selecting the appropriate model.

The latent class model was used to identify hidden (latent) classes of respondents preferences [22]. In latent class analysis, unobserved preference heterogeneity among respondents preferences is modeled as classes with similar preference patterns but with different variances across classes. Once preference patterns have been stratified into classes, the model determines the extent to which demographic characteristics impact the likelihood of belonging to a certain class. The systematic utility component (V) describes the latent construct that participant r belonging to class c reported for alternative A, B or C in choice task t. The final utility functions were as follows:

Vr,t,A&B|c=1 * consist_hESCr,t,A&B|c+2 * consist_iPSr,t,A&B|c+3 * consist_electricr,t,A&B|c+4 * aim_slowr,t,A&B|c+5 * aim_repairer,t,A&B|c+6 * know_500r,t,A&B|c+7 * know_5000r,t,A&B|c+8 * effect_50r,t,A&B|c+9 * effect_80r,t,A&B|c+10 * sideeffects_0.001r,t,A&B|c+11 * sideeffects_0.01r,t,A&B|c+.

Vr,t,C|c=1 * consist_drugr,t,C|c+2 ** aim_reliefr,t,C|c+3 * know_5000r,t,C|c+4 * effect_50r,t,C|c+5 * sideeffects_0.01r,t,C|c+.

A class assignment model was fitted after the specified utility function. The variables: experience in treatment, side effects, advanced treatment therapy and religious beliefs were tested for their potential impact on class membership in the model. The final class assignment function was:

Vn|c=0+1* treatment_sum|c+2 * experience_sideeffects|c+3 * advanced_treatment|c+4 * Religion_dum|c+.

The relative importance of the attributes included in the DCE was calculated by estimating the difference in preference weights of the latent class model between the most preferred level of an attribute and the least preferred level of the same attribute [21]. The highest difference value was normalized to 1, which represents the most important value. The difference values were divided by the highest value to reveal the relative distance between all other attributes.

We calculated the predicted acceptance uptake for a potential treatment scenario using hESCs to treat patients with PD. Predicted acceptability can be understood as the probability that a participant will accept a described scenario. The scenario represents a hypothetical treatment scenario of treatments with hESCs based on the attributes assessment in the DCE. Attribute estimates assessed by the latent class model were used to calculate the predicted acceptability of attribute levels (treatment with hESCs, risk of severe side effects is 1 out of 1000 and 50 patients received treatment) in relevant future scenarios; (A) effect on symptoms is 2 out of 10, (B) effect on symptoms is 5 out of 10, and (C) effect on symptoms is 8 out of 10.

The predicted acceptability is presented as the percentage of 100 who would accept the presented scenario. The utility for the specific scenario was calculated by using the following equation:

VScenario 1=A+B+C.

The predicted acceptability, the probability of accepting a specific scenario, was then calculated by using the following equation:

Predicted acceptance uptake=1/(1+expVScenario 1).

Read more:
Patients accept therapy using embryonic stem cells for Parkinson's ... - BMC Medical Ethics

DNA aptamer finds novel application in regulating cell differentiation – Science Daily

Generating specific cell lineages from induced pluripotent stem cells and embryonic stem cells is the holy grail of regenerative medicine. Guiding iPSCs toward a target cell line has garnered much attention, but the process remains challenging. Now, researchers from Japan have discovered that an anti-nucleolin DNA aptamer, iSN04, can determine a cell's lineage during differentiation. By demonstrating the generation of cardiomyocytes from murine pluripotent stem cells, their concept shows promise as a regenerative therapy.

Self-renewal and pluripotency-the capacity to form any cell lineage-are inherent characteristics of induced pluripotent stem cells (iPSCs). Furthermore, they are highly prized in regenerative therapies targeting cardiovascular, neurological, and metabolic diseases as they are immunologically suitable for transplantation back into a donor. Unfortunately, regenerative medicine is not yet feasible outside a laboratory setting as available protocols to generate target cells are complicated and expensive. This raises a pertinent question: Can regulating the fate of stem cells in clinical settings and at scale be made more economical?

A team of researchers from Shinshu University, the National Institute of Advanced Industrial Science and Technology, and the University of Shizuoka in Japan set out to address this question by leveraging nucleic acid aptamers. Aptamers are single-stranded pieces of DNA that bind to target proteins and are able to modulate signaling cascades during cell differentiation when a stem cell commits to a specific functional role or phenotype. They hold promise in regenerative medicine as they are easily modified, can be synthesized economically, and are suitable for long-term storage.

The team, led by Associate Professor Tomohide Takaya from the Department of Agricultural and Life Sciences at Shinshu University, recently discovered that an anti-nucleolin aptamer, myogenetic oligodeoxynucleotide iSN04, induced myocardial differentiation in embryonic stem cells (ESCs). The study was led by Mina Ishioka, a graduate student in Dr. Takaya's laboratory, and published in The International Journal of Molecular Sciences on 21 September 2023.

"We had previously found that iSN04 promoted myogenic precursor cells (myoblasts) to differentiate into skeletal muscle cells and had hypothesized that the aptamer also enhanced differentiation of pluripotent stem cells. We were intrigued by the prospect of using iSN04 to promote iPSC differentiation into cardiomyocytes as this could lead to regenerating heart tissue," says Dr. Takaya, elaborating on the team's motivation to pursue the research.

Using various assays like RNA sequencing, cell staining and imaging, and molecular interaction and pathway analysis, the researchers investigated iSN04's effect on murine ESCs and iPSCs. iSN04 treatment under differentiating conditions inhibited stem cell commitment to the cardiac lineage. However, when these pluripotent stem cells were treated after experiencing differentiating conditions for five days, specific marker genes were upregulated, and the cells committed to forming beating cardiomyocytes.

"Ours is the first report to confirm a DNA aptamer that allows cardiomyocytes to develop from iPSCs," explains Dr. Takaya when asked about the significance of the work. "We uncovered two mechanisms of nucleolin interference with iSN04 at play whereby early treatment inhibits cardiomyogenesis, while treatment at a later stage enhances the generation of cardiac progenitors. First, iSN04 governs the translocation of nucleolin protein between the cytoplasm, plasma membrane, and nucleus. Second, it results in the modulation of the Wnt signaling pathway that governs cell differentiation."

The immunostaining experiments revealed that nucleolin was retained in the nucleoli following iSN04 treatment. Nucleolar nucleolin has a role in chromatin remodeling and gene transcription, and interestingly enough, Wnt pathway genes were differentially expressed in the RNA-seq data following iSN04 suppression. The team postulates that the iSN04-anchored nucleolin alters gene expression and Wnt signaling. Ultimately, terminal cell differentiation commits to the cardiomyocyte lineage.

And how could these findings impact regenerative medicine and patients' lives in the long term? Dr. Takaya provides insights into the broader implications of their work. "We believe there is a strong case to be made for further studies evaluating DNA aptamers in regenerative medicine. Aptamers are cost-effective and open up the possibility of producing specific cells from the patient's stem cells. But it doesn't end there! Since the aptamers can regulate stem cell fate, they can serve as therapeutic agents for many conditions linked to stem cell dysfunction," he concludes.

See the original post here:
DNA aptamer finds novel application in regulating cell differentiation - Science Daily

Automation is the key to future advances, says the Head of the … – National Health Executive

Below, Lee highlights how his passion for stem cell research brought him to the UKSCB, its role in the development of advanced therapeutics and discusses how automation and machine learning in the manufacturing process could mean faster patient access to stem cell therapies in the future.

My career has been hugely varied, taking me around the world across the public, private and academic sectors, but one thing has remained constant: my fascination with cell and gene therapies, and through research, bringing these closer to patients.

Ive been lucky enough to study cell and gene therapies in many different ways over the last twenty years, from the potential of stem cells as a therapy for Type I Diabetes and the role of genes in the cancer cell cycle, to perfecting techniques that differentiate stem cells into precursors for adult blood cells for transfusion therapies. But theres always more to discover.

One application that excites me the most is the use of stem cells for immuno-oncology, where immune cells are used to treat cancer. Were now able to take immune cells from donors and use gene modification so they can fight cancer in patients where they would normally be rejected.

These stem cells can provide an unlimited supply of cancer-fighting T cells, available off-the-shelf to treat anyone quickly when referred to a clinic. This living drug has the potential to constantly adapt to cancer, combating resistance, and existing within patients to continue fighting the disease indefinitely.

In fact, this simplest form of cell can become almost any cell type for many applications in cell therapies, and the UKs national repository for all human embryonic stem cell lines derived within the UK, is here, at the MHRAs UK Stem Cell Bank.

Established 20 years ago to curate all human embryonic stem cells created in the UK and to regulate their use and provision for research and in the clinic, the UKSCB at the MHRAs South Mimms Laboratories is now recognised globally as a leading repository with over 180 different human embryonic stem cell lines.

During this time, weve received support from the Medical Research Council (MRC) and National Institute for Health Research (NIHR) to deliver our banking activities and, together with the lines available for research, weve banked over 30 lines now available for clinical application, making the UKSCB the largest source of clinical grade human embryonic stem cells in the world.

As part of the MHRA, the UKSCB plays an important role in providing regulatory guidance and workshops to the stem cell health sector across the UK, and weve established links with Harvard University to support international training programmes.

The UKSCB is a unique asset within the MHRA, sitting at the forefront of the advanced therapies landscape in the UK, and Im eager to build on its great history and legacy at a time where were only just starting to realise the full potential and application of advanced cell therapies.

As an aging population, we can expect to receive novel treatments for disease in the form of small molecules and prescription drugs but often these only serve to manage symptoms or limit disease progression. Using stem cells, we can now develop replacement joints, neurons for improved brain function and even entire hearts can be made in the lab without the need for human donors.

But what can we do to keep up with demand and ensure quality? The UKSCB has contributed to over 100 publications in international peer reviewed journals, establishing strong links with the World Health Organisation and international partners to improve standards and controls. Weve recently completed a successful international collaboration with Japanese partners, Sinfonia, to trial their automated cell processing robot and we aim to continue with these efforts towards the cost-effective scaling up of our manufacturing.

I believe that future improvements in customer service lies in automation. Reliance on scientists for the manufacture of stem cells is labour intensive, expensive and introduces human error. Automation will alleviate a lot of the manual aspects of cell culture, allowing us to scale-up manufacturing, drive down costs and ensure the highest quality and consistency, allowing stem cell-derived advanced therapies to be more accessible to patients and affordable for healthcare systems. By introducing automation into this process, we can free-up capacity for our leading experts to move away from labour-intensive manufacturing and instead work on improving and adapting novel advanced therapies.

When I look at my role as the third custodian of the UKSCB I hope to build on the significant past achievements of Glyn Stacey and Elsa Abranches that have established us as the leading stem cell institution in the UK today. I hope to move us forward with a sustainable business model that secures the long-term viability of the UKSCB. This means expanding our operations across various cell therapy platforms, supporting the development of specific cell types for use in human cell therapies, and developing new standards, enabling regulatory approval and eventual uptake by healthcare organisations.

I also want the UKSCB to continue making an impact around the world, supporting the development of stem cell therapies and reference materials for advanced therapies with the World Health Organisation. Over the last twenty years, weve delivered more than 370 cell line vials to 25 different countries for research, and in 2022, 54% of the stem cell lines requested have been of clinical grade, rising year on year. I want to see this trend continue as demand for these lines as starting materials for cell therapies grows further.

The future of the MHRAs UKSCB is diverse and exciting and, much like the cells we curate, there are endless possibilities for us to support research and clinical advances in the UK and around the world. I cannot wait to see where the next 20 years will take us.

Image credit: iStock

Visit link:
Automation is the key to future advances, says the Head of the ... - National Health Executive

A comprehensive review of human trophoblast fusion models: recent … – Nature.com

Huppertz B, Gauster M. Trophoblast fusion. Adv Exp Med Biol. 2011;713:8195.

Article CAS PubMed Google Scholar

Burton GJ, Jauniaux E. Sonographic, stereological and Doppler flow velocimetric assessments of placental maturity. Br J Obstet Gynaecol. 1995;102:81825.

Article CAS PubMed Google Scholar

Kosiska-Kaczyska K. Placental syndromesa new paradigm in perinatology. Int J Environ Res Public Health. 2022;19:7392.

Article PubMed PubMed Central Google Scholar

Carter AM, Enders AC. Comparative aspects of trophoblast development and placentation. Reprod Biol Endocrinol RBE. 2004;2:46.

Article Google Scholar

Jaremek A, Jeyarajah MJ, Jaju Bhattad G, Renaud SJ. Omics approaches to study formation and function of human placental syncytiotrophoblast. Front Cell Dev Biol. 2021;9:674162.

Article PubMed PubMed Central Google Scholar

Boss AL, Chamley LW, James JL. Placental formation in early pregnancy: how is the centre of the placenta made? Hum Reprod Update. 2018;24:75060.

Article CAS PubMed Google Scholar

Turco MY, Moffett A. Development of the human placenta. Dev Camb Engl. 2019;146:dev163428.

CAS Google Scholar

Farah O, Nguyen C, Tekkatte C, Parast MM. Trophoblast lineage-specific differentiation and associated alterations in preeclampsia and fetal growth restriction. Placenta. 2020;102:49.

Article PubMed PubMed Central Google Scholar

Karvas RM, McInturf S, Zhou J, Ezashi T, Schust DJ, Roberts RM, et al. Use of a human embryonic stem cell model to discover GABRP, WFDC2, VTCN1 and ACTC1 as markers of early first trimester human trophoblast. Mol Hum Reprod. 2020;26:42540.

Article CAS PubMed PubMed Central Google Scholar

Calvert SJ, Longtine MS, Cotter S, Jones CJP, Sibley CP, Aplin JD, et al. Studies of the dynamics of nuclear clustering in human syncytiotrophoblast. Reprod Camb Engl. 2016;151:65771.

Article CAS Google Scholar

Coleman SJ, Gerza L, Jones CJP, Sibley CP, Aplin JD, Heazell AEP. Syncytial nuclear aggregates in normal placenta show increased nuclear condensation, but apoptosis and cytoskeletal redistribution are uncommon. Placenta. 2013;34:44955.

Article CAS PubMed PubMed Central Google Scholar

Cantle SJ, Kaufmann P, Luckhardt M, Schweikhart G. Interpretation of syncytial sprouts and bridges in the human placenta. Placenta. 1987;8:22134.

Article CAS PubMed Google Scholar

Heazell AEP, Moll SJ, Jones CJP, Baker PN, Crocker IP. Formation of syncytial knots is increased by hyperoxia, hypoxia and reactive oxygen species. Placenta. 2007;28:S3340.

Article PubMed Google Scholar

Renaud SJ, Jeyarajah MJ. How trophoblasts fuse: an in-depth look into placental syncytiotrophoblast formation. Cell Mol Life Sci. 2022;79:433.

Article CAS PubMed Google Scholar

Aguilar PS, Baylies MK, Fleissner A, Helming L, Inoue N, Podbilewicz B, et al. Genetic basis of cell-cell fusion mechanisms. Trends Genet TIG. 2013;29:42737.

Article CAS PubMed Google Scholar

Lu X, Wang R, Zhu C, Wang H, Lin H-Y, Gu Y, et al. Fine-tuned and cell-cycle-restricted expression of fusogenic protein syncytin-2 maintains functional placental syncytia. Cell Rep. 2017;21:11509.

Article CAS PubMed Google Scholar

Saha B, Ganguly A, Home P, Bhattacharya B, Ray S, Ghosh A, et al. TEAD4 ensures postimplantation development by promoting trophoblast self-renewal: An implication in early human pregnancy loss. Proc Natl Acad Sci USA 2020;117:1786475.

Article CAS PubMed PubMed Central Google Scholar

Mizutani T, Orisaka M, Miyazaki Y, Morichika R, Uesaka M, Miyamoto K, et al. Inhibition of YAP/TAZ-TEAD activity induces cytotrophoblast differentiation into syncytiotrophoblast in human trophoblast. Mol Hum Reprod. 2022;28:gaac032.

Krendl C, Shaposhnikov D, Rishko V, Ori C, Ziegenhain C, Sass S, et al. GATA2/3-TFAP2A/C transcription factor network couples human pluripotent stem cell differentiation to trophectoderm with repression of pluripotency. Proc Natl Acad Sci USA 2017;114:E957988.

Article CAS PubMed PubMed Central Google Scholar

Wang L-J, Chen C-P, Lee Y-S, Ng P-S, Chang G-D, Pao Y-H, et al. Functional antagonism between Np63 and GCM1 regulates human trophoblast stemness and differentiation. Nat Commun. 2022;13:1626.

Article PubMed PubMed Central Google Scholar

Renaud SJ, Chakraborty D, Mason CW, Rumi MAK, Vivian JL, Soares MJ. OVO-like 1 regulates progenitor cell fate in human trophoblast development. Proc Natl Acad Sci USA 2015;112:E617584.

Article CAS PubMed PubMed Central Google Scholar

Pidoux G, Gerbaud P, Gnidehou S, Grynberg M, Geneau G, Guibourdenche J, et al. ZO-1 is involved in trophoblastic cell differentiation in human placenta. Am J Physiol Cell Physiol. 2010;298:C151726.

Article CAS PubMed Google Scholar

Aghababaei M, Hogg K, Perdu S, Robinson WP, Beristain AG. ADAM12-directed ectodomain shedding of E-cadherin potentiates trophoblast fusion. Cell Death Differ. 2015;22:197084.

Article CAS PubMed PubMed Central Google Scholar

Coutifaris C, Kao LC, Sehdev HM, Chin U, Babalola GO, Blaschuk OW, et al. E-cadherin expression during the differentiation of human trophoblasts. Dev Camb Engl. 1991;113:76777.

CAS Google Scholar

Borges M, Bose P, Frank H-G, Kaufmann P, Ptgens AJG. A two-colour fluorescence assay for the measurement of syncytial fusion between trophoblast-derived cell lines. Placenta. 2003;24:95964.

Article CAS PubMed Google Scholar

Dupressoir A, Vernochet C, Bawa O, Harper F, Pierron G, Opolon P, et al. Syncytin-A knockout mice demonstrate the critical role in placentation of a fusogenic, endogenous retrovirus-derived, envelope gene. Proc Natl Acad Sci USA 2009;106:1212732.

Article CAS PubMed PubMed Central Google Scholar

Papuchova H, Latos PA. Transcription factor networks in trophoblast development. Cell Mol Life Sci 2022;79.

Gupta SK, Malhotra SS, Malik A, Verma S, Chaudhary P. Cell signaling pathways involved during invasion and syncytialization of trophoblast cells. Am J Reprod Immunol. 2016;75:36171.

Article PubMed Google Scholar

Hertz R. Choriocarcinoma of women maintained in serial passage in hamster and rat. Proc Soc Exp Biol Med Soc Exp Biol Med. 1959;102:7781.

Article CAS Google Scholar

Pattillo RA, Gey GO, Delfs E, Mattingly RF. Human hormone production in vitro. Science. 1968;159:14679.

Article CAS PubMed Google Scholar

Pattillo RA, Gey GO. The establishment of a cell line of human hormone-synthesizing trophoblastic cells in vitro. Cancer Res.1968;28:7.

Friedman SJ, Skehan P. Morphological differentiation of human choriocarcinoma cells induced by methotrexate. Cancer Res. 1979;39:19607.

CAS PubMed Google Scholar

Wice B, Menton D, Geuze H, Schwartz AL. Modulators of cyclic AMP metabolism induce syncytiotrophoblast formation in vitro. Exp Cell Res. 1990;186:30616.

Article CAS PubMed Google Scholar

Baczyk D, Drewlo S, Proctor L, Dunk C, Lye S, Kingdom J. Glial cell missing-1 transcription factor is required for the differentiation of the human trophoblast. Cell Death Differ. 2009;16:71927.

Article CAS PubMed Google Scholar

Alfaifi AA, Heyder RS, Bielski ER, Almuqbil RM, Kavdia M, Gerk PM, et al. Megalin-targeting liposomes for placental drug delivery. J Control Release. 2020;324:36678.

Article CAS PubMed PubMed Central Google Scholar

Li H, Pinilla-Macua I, Ouyang Y, Sadovsky E, Kajiwara K, Sorkin A, et al. Internalization of trophoblastic small extracellular vesicles and detection of their miRNA cargo in P-bodies. J Extracell Vesicles. 2020;9:1812261.

Article CAS PubMed PubMed Central Google Scholar

Abdelkhaliq A, van der Zande M, Peters RJB, Bouwmeester H. Combination of the BeWo b30 placental transport model and the embryonic stem cell test to assess the potential developmental toxicity of silver nanoparticles. Part Fibre Toxicol. 2020;17:11.

Article CAS PubMed PubMed Central Google Scholar

Wu F, Tian F, Zeng W, Liu X, Fan J, Lin Y, et al. Role of peroxiredoxin2 downregulation in recurrent miscarriage through regulation of trophoblast proliferation and apoptosis. Cell Death Dis. 2017;8:e2908.

Article CAS PubMed PubMed Central Google Scholar

Kohler PO, Bridson WE. Isolation of hormone-producing clonal lines of human choriocarcinoma. J Clin Endocrinol Metab. 1971;32:6837.

Article CAS PubMed Google Scholar

Pattillo RA, Hussa RO, Garancis JC. Glycogen metabolism in human hormone-producing trophoblastic cells in continuous culture. I. Regulation of glycogen metabolism by glucose. Vitro. 1971;7:5967.

Article CAS Google Scholar

Shao X, Cao G, Chen D, Liu J, Yu B, Liu M, et al. Placental trophoblast syncytialization potentiates macropinocytosis via mTOR signaling to adapt to reduced amino acid supply. Proc Natl Acad Sci USA 2021;118:e2017092118.

Article CAS PubMed PubMed Central Google Scholar

Vargas A, Moreau J, Le Bellego F, Lafond J, Barbeau B. Induction of trophoblast cell fusion by a protein tyrosine phosphatase inhibitor. Placenta. 2008;29:1704.

Article CAS PubMed Google Scholar

Msheik H, El Hayek S, Bari MF, Azar J, Abou-Kheir W, Kobeissy F, et al. Transcriptomic profiling of trophoblast fusion using BeWo and JEG-3 cell lines. Mol Hum Reprod. 2019;25:81124.

Article CAS PubMed Google Scholar

Al-Nasiry S, Spitz B, Hanssens M, Luyten C, Pijnenborg R. Differential effects of inducers of syncytialization and apoptosis on BeWo and JEG-3 choriocarcinoma cells. Hum Reprod Oxf Engl. 2006;21:193201.

Article CAS Google Scholar

Jasmer B, Muschol-Steinmetz C, Kreis N-N, Friemel A, Kielland-Kaisen U, Brggmann D, et al. Involvement of the oncogene B-cell lymphoma 6 in the fusion and differentiation process of trophoblastic cells of the placenta. Oncotarget. 2017;8:10864354.

Article PubMed PubMed Central Google Scholar

Jaju Bhattad G, Jeyarajah MJ, McGill MG, Dumeaux V, Okae H, Arima T, et al. Histone deacetylase 1 and 2 drive differentiation and fusion of progenitor cells in human placental trophoblasts. Cell Death Dis. 2020;11:311.

Article CAS PubMed PubMed Central Google Scholar

Finney DJ. Statistics in the university and in the community. Biom Z. 1965;7:7889.

Article CAS PubMed Google Scholar

Gilles JA, Carnes DL, Windeler AS. Development of an in vitro culture system for the study of osteoclast activity and function. J Endod. 1994;20:32731.

Article CAS PubMed Google Scholar

Cheng Y-H, Handwerger S. A placenta-specific enhancer of the human syncytin gene. Biol Reprod. 2005;73:5009.

Article CAS PubMed Google Scholar

Lv B, An Q, Zeng Q, Zhang X, Lu P, Wang Y, et al. Single-cell RNA sequencing reveals regulatory mechanism for trophoblast cell-fate divergence in human peri-implantation conceptuses. PLoS Biol. 2019;17:e3000187.

Article CAS PubMed PubMed Central Google Scholar

Knerr I, Schubert SW, Wich C, Amann K, Aigner T, Vogler T, et al. Stimulation of GCMa and syncytin via cAMP mediated PKA signaling in human trophoblastic cells under normoxic and hypoxic conditions. FEBS Lett. 2005;579:39918.

Article CAS PubMed Google Scholar

Chang C-W, Chang G-D, Chen H. A novel cyclic AMP/Epac1/CaMKI signaling cascade promotes GCM1 desumoylation and placental cell fusion. Mol Cell Biol. 2011;31:382031.

View original post here:
A comprehensive review of human trophoblast fusion models: recent ... - Nature.com

World Arthritis Day sheds light on innovative treatments – Omnia Health Insights

In the UAE, arthritis affects one in five people and is the top cause of disability globally. Arthritis includes over 100 autoimmune and rheumatic musculoskeletal conditions, and while it is commonly associated with ageing, younger patients are also affected.

World Arthritis Day, held on October 12 annually, emphasises the importance of recognising the symptoms of this painful disease, which can help lead to early intervention. Renowned organisations such as the Middle East Arthritis Foundation (MEAF) host focused events to further raise awareness and assist those who suffer from this debilitating disease.

While traditional treatments such as medications, physiotherapy, and surgery can alleviate symptoms, as World Arthritis Day is observed, we examine some alternatives and advancements that may offer hope for an enhanced quality of life.

Related:Understanding Systemic Juvenile Idiopathic Arthritis and gut health

The use of remote monitoring offers a promising way to decrease hospital visits for arthritic patients through a combination of self-management and telemedicine. With the potential to replace labour-intensive outpatient clinic visits, this could positively impact healthcare utilisation while keeping disease activity low.

Regenerative medicine in arthritis could utilise cell therapy, bioengineering and gene therapy to stimulate the body's natural healing response. A key development is Platelet-Rich Plasma (PRP), derived from a patient's blood, PRP can be used to treat pain, damaged tendons, hair loss and ageing skin. While it can relieve symptoms and boost healing, its effectiveness may vary based on preparation methods and patient factors.

Similar to fat-based PRP, Autologous Micro-fragmented Adipose Tissue (AMAT) involves liposuction to extract fat, which is then injected into areas needing treatment. Studies show improvements in osteoarthritis pain and function, but consistency in AMAT quality remains a challenge.

Stem cell therapy holds great potential for tissue repair and regeneration. The various stem cell types include: embryonic, adult, induced pluripotent, and very small embryonic-like stem cells. Clinical trials are ongoing and the FDA cautions against unproven therapies from for-profit clinics. It emphasises the need for standardised guidelines and further research to discover the full potential of stem cells in arthritis care.

Related:Gen AI may power the next generation of immunotherapies

Surgical options such as Osteochondral Autograft Transplantation Surgery (OATS) and Matrix-Induced Autologous Chondrocyte Implantation (MACI) use a patient's own or donor tissue to repair localised cartilage damage, preventing arthritis progression. Researchers are exploring gene editing tools like CRISPR-Cas9 to create custom-designed cells and gene therapies that target inflammatory proteins in osteoarthritis.

While these treatments in regenerative medicine demonstrate substantial potential, continuous research, protocol refinement, and establishing standardised methodologies are imperative to fully discover their benefits in the field of arthritis care.

Get the latest insights from the healthcare industry straight to your inbox.Subscribefor a bi-weekly roundup of the latest healthcare tech, trends, innovation and patient experience, accompanied by exclusive interviews from the industrys leaders.

___

Back to Technology

Read the rest here:
World Arthritis Day sheds light on innovative treatments - Omnia Health Insights

Cell atlases of the human brain – Science Daily

In two parallel projects, researchers at Karolinska Institutet have been involved in creating the most comprehensive atlases of human brain cells to date. The two studies, which are published in Science, provide clues on different brain diseases and give hope for medical advancements in the future, such as new cancer drugs.

Knowing what cells constitute the healthy brain, where different cell types are located and how the brain develops from the embryo stage is fundamental to the ability to compare and better understand how diseases arise. There are at present advanced atlases of the mouse brain, but not for the human brain. Until now.

A brain-cell census

"We've created the most detailed cell atlases of the adult human brain and of brain development during the first months of pregnancy," says Sten Linnarsson, professor of molecular system biology at the Department of Medical Biochemistry and Biophysics at Karolinska Institutet in Sweden. "You could say that we've taken a kind of brain-cell census."

The first project was led by Kimberly Siletti from Linnarsson's group. It was conducted in close collaboration with Ed Lein at the Allen Institute for Brain Science in Seattle, USA, as part of the international Human Cell Atlas initiative, and based on three donated human brains from adults. The researchers analysed more than three million individual cell nuclei using the technique of RNA sequencing, which reveals each cell's genetic identity. All in all, the researchers studied cells from just over a hundred brain regions and found over 3,000 cell types, some 80 per cent of which were neurons, the remainder being different kinds of glial cells.

"A lot of research has focused on the cerebral cortex, but the greatest diversity of neurons we found in the brainstem," says Professor Linnarsson. "We think that some of these cells control innate behaviours, such as pain reflexes, fear, aggression and sexuality."

Groundwork for medical advances

The researchers could also see that the cells' identity reflects the place in the brain where they first developed in the fetus, which links to the second project. Here, Emelie Braun and Miri Danan-Gotthold from Sten Linnarsson's group collaborated with the Swedish consortium for the Human Developmental Cell Atlas to analyse over a million individual cell nuclei from 27 embryos at different stages of development (between 5 and 14 weeks of fertilisation). The study enabled the researchers to show how the entire brain develops and is organised over time.

Even though the results are examples of molecular biological basic research, the new knowledge generated can also lay the groundwork for medical advances. Professor Linnarsson's research group has used similar methods to examine different kinds of brain tumours, one of which was a glioblastoma -- a cancer with a poor prognosis.

"The tumour cells resemble immature stem cells and it looks like they're trying to form a brain, but in a totally disorganised way," he explains. "What we observed was that these cancer cells activated hundreds of genes that are specific to them, and it might be interesting to dig into whether there is any potential for finding new therapeutic targets."

Freely available brain atlases

The brain atlases will be freely available to researchers around the world so that they can compare the brain diseases they are researching with what a normally developed brain looks like.

The studies are part of a larger package of articles published simultaneously in the scientific journal Science. The study on the adult brain was supported by a grant from the National Institutes of Health, while the embryo study was financed by the Knut and Alice Wallenberg and Erling-Persson foundations.

Sten Linnarsson is a scientific advisor at Moleculent, Combigene and Oslo University Center of Excellence in Immunotherapy. He and co-authors Alejandro Mossi Albiach and Lars E. Borm also hold shares in EEL Transcriptomics AB, which owns the intellectual property rights to the EEL method ("Spatial RNA localization"). Co-authors aneta Andrusivov and Joakim Lundeberg are scientific consultants for 10x Genomics, which holds the intellectual property rights to the Spatial Transcriptomics (Visium) technique.

Follow this link:
Cell atlases of the human brain - Science Daily

Adipose stromal cells bioproducts as cell-free therapies … – Journal of Translational Medicine

Cell culture

ASC from lipoaspirates from three donors were processed by the group of Prof. Karen Bieback (University of Heidelberg, Heidelberg, Germany) after informed consent. The Mannheim Ethics Commission II approved the study (vote 2011-215N-MA). The ASC were cultured using MEM- media, Gibco, ThermoFisher Scientific, 2,561,029) and 10% Fetal Bovine Serum (FBS, 10270-106, Gibco, MA, USA) at 37 with 5% CO2 and controlled humidity. These three ASC batches (referred to a N=3 biological replicates in the figure legends) were shipped as cryo-aliquots to the other two centers to be cultured under identical harmonized culture conditions from passage 46 as detailed previously [29]. Bioproducts were derived from the conditioned medium of either 3D cultured ASC, processed by size exclusion chromatography to yield (1) EV-SEC or the (2) protein-rich fraction, or 2D-cultured cells, processed by ultracentrifugation to yield (3) EV-UC or after concentration to yield (4) the conditioned medium (CM) or (5) the respective wash-off (CM-WO) (Fig.1).

At 80% confluence, ASC were passaged and seeded in a hollow-fibber bioreactor at a concentration of 14106 cells/cartridge (20kDa MWCO, 450cm2, C2025D, FiberCell System-KD Bio, France). Prior to injecting the cells, a pre-culture step was carried out to initiate and activate the bioreactor, first Dulbeccos phosphate-buffered saline (PBS) for 24h, followed by fibronectin coating over-night. After the pre-culture process, ASC were seeded in serum-free MEM- in the extra-capillary space, at 37 with 5% CO2 and controlled humidity for 7days without harvesting the supernatant, with continuous monitoring of glucose levels. Serum-containing medium was used as circulating medium, given that EVs and high molecular weight proteins cannot cross the 20 kD MWCO filter fiber and thus do not contaminate the cell-derived EVs harvested from the extra-capillary-space (according to the Hollow Fiber Bioreactor Protocol for Mesenchymal Stem Cells by fibercellsystems.com) ASC were cultured for 4weeks in the bioreactor and during this period, the supernatant was collected daily. Following centrifugation to remove cell debris (5min at 420g), the supernatant was stored at 80 until EV isolation by size exclusion chromatography (see below) was performed. Cells were harvested and counted to calculate the bioproduct per producer cell concentration.

The secretome obtained in vitro, also named conditioned media (CM), was generated from ASC at passage 4 to 6. Upon reaching 80% confluence, cells were washed with PBS and incubated for 24h in serum-free MEM- medium. The supernatant was collected and centrifuged for 5min at 400g to remove cell debris before being placed in centrifugal concentrator units of 3KDa molecular weight cut-off (UFC9003, Merck Millipore, USA). The CM was centrifuged for 90min at 3,000g, 4 using an Eppendorf 5810 R Centrifuge to achieve tenfold concentration. The flow-through resulted from the concentration step (thereafter named wash-off, CM-WO) was kept and used as a control. Concentrated conditioned media samples were stored at 80 until further use. Cells were harvested and counted to calculate the bioproduct per producer cell concentration.

When the cells reached 80% confluence, they were starved for 1624h in serum-free medium. The supernatant was collected and centrifuged for 20min at 3000g to remove cell debris and apoptotic cells. The supernatant was then ultracentrifuged for 2h at 100,000g, 4 using Beckman Coulter Optima L-100K Ultracentrifuge (Beckman Coulter, CA, USA) with the rotor type 70Ti. The EV pellet was resuspended in PBS supplemented with 1% DMSO. The suspension of EVs (EV-UC) was then stored at80 until further use. EVs were collected from ASC at 4-6th passage. Cells were harvested and counted to calculate the cell equivalents used for cell treatments.

After thawing at 4, samples were centrifuged for 10min at 300g and 20min at 4000g. After, the supernatant was filtered through a 0.2m syringe filter and concentrated with a 100kD MWCO concentration filter to a final volume of 10mL. The qEV10-IZON column 35mm was initially washed with sterile PBS, and then 10mL of the sample was added to concentrate it to the final volume of 1.5mL (Vivaspin 20, 100,000 MWCO PE, Sartorius). Each EV sample (EV-SEC) and the resultant supernatant containing the protein fraction (Protein-Rich Fraction) were collected, concentrated (Vivaspin 20, 100,000 MWCO PE, Sartorius) and stored at 80 until further use.

ASC derived bioproducts were used at a ratio of 2:1 and 20:1 relative to recipient cells. To do so, we counted the number of ASC after harvesting and used it to relate the number of particles/volumes generated of EVs and CM respectively for each bioproduct.

After the isolation, the concentration of all the samples was measured (a) by Nanosight NS300 or (b) ZetaView.

After the isolation, the concentration of all the samples was measured (a) by Nanosight NS300 (Malvern Instruments Ltd., Malvern, UK) equipped with a 488nm laser module that utilizes Brownian motion and refraction index. The particle size scatters 10nm to 1000nm, although the optimized size range is 70300nm. It uses the scattered light to detect a particle and tracks its motion as a function of time. The particles scattered light was recorded with a light-sensitive camera under a 90 angle to the irradiation plane. This angle allows the Brownian motion of the EVs. Samples were diluted 1:100 in physiologic solution. For each sample, 3 videos of 60s at camera level 15 and threshold 5 were captured using a syringe pump 30. All the samples were characterized with NTA 3.2.16 Analytical software. The NTA settings were kept constant between samples.

After the isolation, the concentration of all the samples was measured b) by ZetaView (Particle Metrix GmbH, Germany). 1L of concentrated EVs was diluted in sterile-filtered PBS in a dilution 1:1,000 and visualized using the ZetaView (sensitivity 80%, shutter 100, 11 positions, 2 cycles; Particle Metrix, Germany).

Super-resolution microscopy pictures of EVs were obtained using a temperature-controlled Nanoimager S Mark II microscope from ONI (Oxford Nanoimaging, Oxford, UK) equipped with a 100 , 1.4NA oil immersion objective, an XYZ closed-loop piezo 736 stage, and 405nm/150mW, 473nm/1W, 560nm/1W, 640nm/1W lasers and triple emission channels split at 640/and 555nm. For sample preparation, we followed the manufacturers protocol using EV profiler Kit ONI (Alfatest, Rome, Italy). Before each imaging session, bead slide calibration was performed for aligning the channels, to achieve a channel mapping precision smaller than 12nm. Images were taken in dSTORM mode using 50% laser power for the 647nm channel, 30% laser power for the 488nm laser channel, and 30% for the 555 channel. Three-channels (2000 frames per channel) (647, 555 and 488) were acquired sequentially at 30Hz (Hertz) in total reflection fluorescence (TIRF) mode. Single-molecule data was filtered using NimOSsoftware (v.1.18.3, ONI) based on the point spread function shape, photon count and localization precision to minimize background noise and remove low-precision and non-specific colocalization. Data has been processed with the Collaborative Discovery (CODI) online analysis platform https://www.alto.codi.bio/ from ONI and the drift correction pipeline version 0.2.3 was used. Clustering analysis was performed on localizations and BD clustering-constrained parameters were defined (photon count 300-max, sigma 0200nm, p-value 01, localization precision 020nm). Colocalization was defined by a minimum number of localizations for each fluorophore/protein within a distance of 100nm or a distance used from the centroid position of a cluster.

MACSPlex Exosome Kit (Miltenyi Biotec, Bergisch Gladbach, Germany) containing fluorescent labeled (FITC-PE) capture beads coupled to 37 exosomal surface epitopes and 2 isotope controls was used, following the manufacturers instructions (in detail: CD3, CD4, CD19, CD8, HLA-DR, CD56, CD105, CD2, CD1c, CD25, CD49e, ROR1, CD209, CD9, SSEA-4, HLA-ABC, CD63, CD40, CD62P, CD11c, CD81, MCSP, CD146, CD41b, CD42a, CD24, CD86, CD44, CD326, CD133-1, CD29, CD69, CD142, CD45, CD31, REA control, CD20, CD14, mIgG1 control). Briefly, 15L of beads were added to 120L of buffer or sample, including a total of 1109 EVs, and the complex was then incubated on a rotor overnight at 4. After the incubation and washing steps, a cocktail of APC fluorescent antibodies against tetraspanins (CD9, CD63 and CD81) was added (allowing the detection of beads bound EVs) and set on the rotor for 1h at room temperature. After washing, samples were detected using BD FACSCelestaTM Flow Cytometer (BD Bioscience, NJ, USA). Median background values of buffer control were subtracted, and samples were normalized to the median fluorescence intensity of tetraspanins.

Proteins extracted from Hela cells were used as cellular control, the pellet was resuspended in RIPA buffer (50mM TrisHCl, pH7.4, 150mM NaCl, 1% Triton X-100, 1% Na-deoxycholate, 0.1% SDS, 0.1mM CaCl2, and 0.01mM MgCl2 supplemented with protease inhibitor cocktail (Thermo Fisher Scientific), incubate 30min in ice vortexing every 10min and centrifuge 20min at 20,000g. An equal volume of bioproducts (38L) was loaded and separated on 415% Mini-PROTEAN TGX Precast Gels (Bio-Rad, USA). Bioproducts and cell lysates were treated with protein loading dye (Laemmli sample buffer; Bio-Rad) with freshly added -mercaptoethanol 10%; v/v; Sigma, Germany) and boiled for 5min at 95 before SDS-PAGE. Proteins were subsequently blotted to a nitrocellulose blotting membrane (0.2m; 1,060,000; GE Healthcare, USA). Membranes were blocked in 5% BSA (Carl Roth, Germany) in 0.1% Tween in TBS (TBS-T). After blocking, blots were probed with the following primary antibodies diluted in 5% BSA/TBS-T: Calnexin (1:500 dilution, E-10, Santa Cruz Biotechnology). After overnight incubation at 4, membranes were washed 3 times with TBS-T and subsequently incubated with the secondary antibody dilution: Polyclonal Goat anti-mouse HRP (1:5000 dilution; P0447) for 1h at room temperature followed by washing. Blots were then developed using Western Bright ECL (541,004; Biozym Scientific, Germany) and protein bands were detected using the FusionCapt Advanced Solo 4 (Vilber, Germany).

The capacity of ASC or their bioproducts to inhibit induced proliferation of peripheral blood mononuclear cells (PBMCs) was analyzed as described before [27]. PBMCs were isolated from leukapheresis samples from healthy donors, provided by the German Red Cross Blood Donor Service in Mannheim (Mannheim Ethics Commission; vote number 2018-594N-MA). To assess their proliferation, PBMCs were labelled with proliferation dye Cytotell Green (ATT Bioquest, 22,253) (1:500 dilution) and seeded at a 1:10 ASC/bioproduction:PBMCs ratio in RPMI, supplemented with 10% FBS, 2% l-glutamine (PAN Biotech, P04-80100), 1% Penicillin/Streptomycin (PAN Biotech, P06-07100), and 200U/mL IL-2 (Promokine, C61240). PBMC proliferation was stimulated with phytohemagglutinin-L (PHA, 4.8g/mL (Biochrom, Merck Millipore, M5030)). PBMCs cultured alone without ASC in the absence and presence of PHA served as negative and positive controls, respectively. After 5days, PBMC proliferation was measured based on the dilution of Cytotell Green dye using a FACS Canto II (BD Biosciences) and the data were analyzed with FlowJo Software.

THP-1 monocyte-like cell line (ATCC, Manassas, VA, USA) were cultured in RPMI-1640 growth medium with l-Glutamine (Sigma-Aldrich Ireland Ltd. Wicklow, Ireland) supplemented with 10% FBS (Sigma-Aldrich), 1% penicillin G (100U/mL) and streptomycin (100g/mL) solution (Sigma-Aldrich). In vitro assessment of phagocytic activity was done as described before [30]. THP-1 cells were seeded at a density of 5104 cells/well in dark 96 well-plates (Perkin Elmer Ireland Ltd. Dublin, Ireland) and exposed to 1g/mL of para-methoxyamphetamine (PMA, Sigma-Aldrich, St. Louis, MO, USA) for 48h to induce a macrophage-like phenotype. Cultures were washed with DPBS and fed with growth media for 24h. Afterwards, cells were activated with 100ng/mL of lipopolysaccharide (LPS, Sigma-Aldrich) for 24h. To measure the phagocytic capacity, Zymosan A FITC BioParticles (Thermo Fisher Ltd.) were used. Particles were opsonized with human serum (2mg/mL per 2107 particles, Sigma-Aldrich) for 1h and added to the cells in experimental media containing ASC bioproducts and growth media for 4h. Then, cells were washed twice with DPBS, fixed with 4% PFA for 15min and stained with Hoechst 33,342 (Invitrogen, Thermo Fisher Ltd). Images were taken on the Cytation 1 Imaging Reader at 20X (BioTek, with Gen5 Version 3.04 software, Swindon, UK). Six replicates were undertaken per condition and particle analysis was done by counting particle opsonization in a minimum of 200 cells per well.

HUVEC were seeded in 48-well plates at 84,000 cells/cm2 and cultured overnight. Subsequently, a p200 tip was used to create a scratch in each monolayer. Cultures were washed with DPBS before adding EVs/CM as described before. Complete EndoGRO-LS medium was used as a positive control, while EndoGRO-LS without FBS and VEGF served as a negative control. Scratches were imaged immediately after the addition of CM (0h) and after 8- and 24-h incubation using the automated Cytation 1 Imaging Reader at 4X. Six replicates were undertaken, and the total area of each scratch was measured using Image J. The percentage of closure was calculated relative to time 0h.

20,000 ASC were seeded in a 96-well Essen ImageLock plate and cultured overnight. Then, a 96-pin WoundMaker was used to create precise and reproducible wounds in all the wells. After the wound, the cells were washed 2 times with DPBS and ASC bioproducts added in different concentrations. Plates were then cultured in an IncuCyte ZOOM incubator and every 3h were taken a picture with the software. The results were analyzed after 24h. Relative Wound Density algorithm was used to report data.

Human umbilical vascular endothelial cells (HUVEC) either from Lonza or prepared as described before [31] and cultured until the 6th passage in EndoGRO-LS Complete Culture Media Kit (SCME001, Sigma-Aldrich, St. Louis, MO, USA). In vitro formation of capillary-like structures was performed on growth factorreduced Matrigel (356,231, Corning, NY, USA, center 1 and 3) or geltrex (Geltrex LDEV-free reduced growth factor matrix; Thermo Fisher Scientific, United States, center 2) HUVEC cells were treated with EVs or CM as described before, seeded at a density of 10103cells/well on a 48-well plate. Positive control was full EndoGro-LS medium, negative control medium without VEGF and FBS (as used for all the conditions). Cells were periodically observed with a Nikon TE2000E inverted microscope (Nikon, Tokyo, Japan), and experimental results were recorded after 16h; 3 images were taken per well. Image analysis was performed with the ImageJ software v.1.53c, using the Angiogenesis Analyzer (center 1,3). The data from three independent experiments were expressed as the meanSD of tube length in arbitrary units per field. Center 2 used live cell imaging (Incucyte Zoom) to assess network formation as described before [32].

Presence of vascular endothelial growth factor (VEGF) on ASC bioproducts was determined by solid phase sandwich ELISA using the human VEGF DuoSet ELISA (R&D Systems, USA) according to manufacturers instructions. The samples were read immediately at 450nm with a wavelength correction at 570nm using a VICTOR X4 multilabel plate reader (Perkin Elmer, Waltham, Massachusetts, USA). Levels of cytokines were quantified against an eight-point standard curve using twofold serial dilutions in reagent diluent.

The Pierce BCA Protein Assay Kit (ThermoFisher Scientific, UK) was used to determine protein concentration. In order to quantify the total amount of protein, samples were first lysed with RIPA buffer 4:1 (ThermoFisher Scientific, UK) for 30min on ice. The assay was carried out as per manufacturers instructions. The absorbance values were read in a VICTOR X4 plate reader (Perkin Elmer) at a 550nm wavelength, and the protein concentrations of the samples were quantified against the standard curve.

Statistical analysis was performed using GraphPad prism v9.4.2 (GraphPad software, USA). Data are expressed as meanstandard deviation (SD). N indicates biological replicates; n indicates technical replicates. Statistical differences among groups were calculated using ordinary two-way analysis of variance (ANOVA) and Tukeys post-hoc test when group distributions were normal (ShapiroWilks test) and variances of populations were equal (Bartletts test). When either or both assumptions were violated, non-parametric analysis was conducted; KruskalWallis test used to perform multiple comparison analysis and Dunns multiple comparison test for pairwise comparison. Results were considered statistically significant when p>0.05.

Originally posted here:
Adipose stromal cells bioproducts as cell-free therapies ... - Journal of Translational Medicine

Bayer Opens First Cell Therapy Manufacturing Facility to Advance … – BioSpace

BERLIN, Germany & BERKELEY, Calif.--(BUSINESS WIRE)-- Bayer AG announced today the opening of its first Cell Therapy Launch Facility in Berkeley, California to create the capacity to bring cell therapies to patients on a global scale. The $250 million (USD), 100,000-square-foot facility will supply the material required for late-stage clinical trials and potential commercial launch of BlueRock Therapeutics bemdaneprocel (BRT-DA01), an investigational cell therapy currently in evaluation for treating Parkinsons disease. In addition, it includes space for a second module of production suites to support additional cell therapies as Bayers portfolio advances. BlueRock Therapeutics LP is a clinical stage, cell therapy company and wholly owned, independently operated subsidiary of Bayer AG.

This press release features multimedia. View the full release here: https://www.businesswire.com/news/home/20231009274687/en/

(Photo: Business Wire)

Cell therapy represent a groundbreaking class of medicines and is an area where Bayer is making a significant investment to research potentially transformative treatment approaches for people with unmet medical needs, said Sebastian Guth, President of Bayer U.S.A. and Pharmaceuticals North America, and member of the Pharmaceutical Executive Committee. Our new cell therapy facility represents true innovation in product development and manufacturing in addition to contributing to Bayers sustainability goal as our first fully electric pharmaceutical manufacturing plant.

The new Cell Therapy Launch Facility is part of a transformation at the companys dedicated biotechnology site in Berkeley, where Bayer has invested nearly USD 500 million in infrastructure over the past five years.

Our teams are driving innovation in late-stage development and manufacturing with a goal of bringing transformational cell and gene therapies to patients on a global scale, and this facility will enable us to make it real, said Jens Vogel, Sr. Vice President and Global Head of Biotech for Bayers Pharmaceutical Division. Bayer is collaborating with biotech innovators, academia, and equipment and automation suppliers to establish platforms that would help bring more therapies to patients faster.

Bayers global biotech organization recognizes the importance of helping innovators transfer their product candidates from the laboratory bench to the clinical study and commercial launch settings. The Biotech team provides its biologic development and manufacturing capabilities for Bayers larger biotherapeutics portfolio, including commercial products and late-stage protein and cell therapies in development. As part of Bayers larger mission of Health for All, the company is now also helping early-stage U.S. and European companies to enable patient trials and commercial launches through its BioPartnering Solutions offerings.

Having access to this Cell Therapy Launch Facility is central to our goal to deliver impactful cell therapies from our pipeline to patients in need, said Seth Ettenberg, President & CEO of BlueRock Therapeutics. Our team is excited to be working shoulder to shoulder with Bayers biotech scientists and manufacturing experts as we look to scale up manufacturing for our first investigational therapy, bemdaneprocel for Parkinsons disease, as it advances through clinical trials.

The new Cell Therapy Launch Facility, launched in conjunction with manufacturing day in the United States, is among several recent investments to advance Bayers biologic pipeline of protein therapeutics, cell and gene therapies including a new Cell Culture Technology Center and Cell Therapy Labs. The new Cell Therapy Launch Facility features flexible, modular space for cell culture, viral transduction and automated filling of cell therapies leveraging Biotech@Bayer expertise in iPSC and CAR-T characterization, process development, analytics and clinical to commercial production.

Beyond Berkeley, the companys global biotech network includes biologic development, manufacturing science, industrialization and advanced manufacturing engineering teams in Wuppertal and Leverkusen, Germany; and Basel, Switzerland; with a full complement of labs and clinical production suites.

About bemdaneprocel and Parkinsons Disease

Bemdaneprocel (BRT-DA01) is an investigational cell therapy designed to replace the dopamine producing neurons that are lost in Parkinsons disease. These dopaminergic neuron precursors are derived from pluripotent stem cells (PSC) that are human embryonic stem cells. In a surgical procedure, these neuron precursors are implanted into the brain of a person with Parkinsons disease. When transplanted, they have the potential to reform neural networks that have been severely affected by Parkinsons and restore motor and non-motor function to patients. Planning is underway for BlueRock Therapeutics Phase II study that is expected to begin enrolling participants in H1 (first half) 2024.

Parkinsons disease is a progressive neurodegenerative disorder caused by the death of nerve cells in the brain, leading to decreased dopamine levels. At diagnosis, it is estimated that patients have already lost 50-80% of their dopaminergic neurons. The loss of these neurons leads to a progressive loss of motor function and symptoms such as tremors, muscle rigidity, and slowness of movement. Even with medication, the symptoms of Parkinsons disease can fluctuate during the course of the day. According to the Parkinsons Foundation, more than 10 million people worldwide suffer from Parkinsons disease, with approximately one million living in the United States. There is no cure, and the effectiveness of current treatments decreases over time.

About BioPartnering Solutions for Biotech Innovators in U.S. and Europe

Through Bayers BioPartnering Solutions, innovators can leverage industry-leading biotech process development and biomanufacturing capabilities to make their therapeutic candidates a reality. Bayers highly skilled Biotech teams and infrastructure which includes preclinical, clinical and commercial launch scale manufacturing are available to advance promising cell therapy, monoclonal antibody and protein therapeutic candidates from the discovery bench to patients. Through BioPartnering Solutions, Bayer provides early-stage companies with a single source of IND- and BLA-enabling development; bioprocess and biochemical engineering; clinical and commercial manufacturing. A range of complementing support service functions such as supply chain management, procurement, quality, and CMC strategy support for regulatory filings are also available. For more information visit: https://www.bayer.com/en/us/BioPartneringSolutions.

About Bayers Biotech Campus in Berkeley, CA

Innovation happens in communities which foster it. Bayer has a 46-acre site located in Berkeley, CA where a team of about 1,000 employees drives the development and manufacturing of traditional protein therapeutics as well as novel cell and gene therapies. It has also served as the global commercial supply center for manufacturing and supplying Bayers biotherapeutics for people living with hemophilia A around the world for 30 years.

During todays event, Bayer celebrated its continuing relationship with the City of Berkeley through a 30-year extension of its Development Agreement which will support up to one million square feet of additional infrastructure. Through the agreement, Bayer will invest more than $30 million in the local community in career technical education; initiatives focused on health equity, economic resiliency and climate action; and more. For more information visit: https://www.bayer.com/en/us/berkeley-site-updates.

About Bayer

Bayer is a global enterprise with core competencies in the life science fields of health care and nutrition. Its products and services are designed to help people and the planet thrive by supporting efforts to master the major challenges presented by a growing and aging global population. Bayer is committed to driving sustainable development and generating a positive impact with its businesses. At the same time, the Group aims to increase its earning power and create value through innovation and growth. The Bayer brand stands for trust, reliability and quality throughout the world. In fiscal 2022, the Group employed around 101,000 people and had sales of 50.7 billion euros. R&D expenses before special items amounted to 6.2 billion euros. For more information, go to http://www.bayer.com.

Find more information at https://pharma.bayer.com Follow us on Facebook: http://www.facebook.com/bayer Follow us on Twitter: @BayerPharma

Forward-Looking Statements

This release may contain forward-looking statements based on current assumptions and forecasts made by Bayer management. Various known and unknown risks, uncertainties and other factors could lead to material differences between the actual future results, financial situation, development or performance of the company and the estimates given here. These factors include those discussed in Bayers public reports which are available on the Bayer website at http://www.bayer.com. The company assumes no liability whatsoever to update these forward-looking statements or to conform them to future events or developments.

View source version on businesswire.com: https://www.businesswire.com/news/home/20231009274687/en/

Bayer U.S.-based Media Contacts: Cathy Keck, +1-206-249-5191 Email: cathy.keck@bayer.com

See more here:
Bayer Opens First Cell Therapy Manufacturing Facility to Advance ... - BioSpace